Original article
Temsirolimus improves cytotoxic efficacy of cisplatin and gemcitabine against urinary bladder cancer cell lines

https://doi.org/10.1016/j.urolonc.2013.04.012Get rights and content

Abstract

Objectives

To analyze the cytotoxic action of temsirolimus using 3 established human bladder cancer cell lines and to assess whether temsirolimus potentiates the anticancer activity of gemcitabine and cisplatin.

Methods

Temsirolimus (500, 1,000, 2,000, and 4,000 nM), in isolation, and combined with gemcitabine (100 nM) and cisplatin (2.5 µg/ml), was given to 5637, T24, and HT1376 bladder cancer cell lines. Cell proliferation, autophagy, early apoptosis, and cell cycle distribution were analyzed after a 72-hour period. The expression of mammalian target of rapamycin baseline, Akt, and their phosphorylated forms, before and after treatment with temsirolimus, was evaluated by immunoblotting.

Results

Temsirolimus slightly decreased the bladder cancer cell proliferation in all 3 cell lines. No significant differences in the expression of mammalian target of rapamycin, Akt, and their phosphorylated forms because of temsirolimus exposure were found in the 3 cell lines. As part of a combined regime along with gemcitabine, and especially with cisplatin, there was a more pronounced antiproliferative effect. This pattern of response was similar to the other parameters analyzed (increased autophagy and apoptosis). Also, in the combined regime, an enhanced cell cycle arrest in the G0/G1 phase was observed. The non–muscle invasive 5637 bladder cancer cell line was most sensitive to both combinations.

Conclusions

Temsirolimus makes a moderate contribution in terms of cell proliferation, apoptosis, and autophagy. However, it does potentiate the activity of gemcitabine and particularly cisplatin. Therefore, cisplatin- or gemcitabine-based chemotherapy regimen used in combination with temsirolimus to treat bladder cancer represents a novel and valuable treatment option, which should be tested for future studies in urinary bladder xenograft models.

Introduction

Temsirolimus belongs to the second generation of rapamycin derivatives (rapalogs), which has a better bioavailability than rapamycin, and this facilitates its clinical use [1], [2]. It is a selective inhibitor of the mammalian target of rapamycin (mTOR) [3]. This biological pathway regulates some vital cellular processes, such as cell survival, growth, protein synthesis, cellular metabolism, and angiogenesis, all of which are crucial to urothelial bladder cancer progression [4], [5], [6], [7].

In 2007, the US Food and Drug Administration approved temsirolimus as the first-in-class mTOR inhibitors in oncology and as a first-line treatment for renal cell cancer (RCC). In 2012, Sun et al. [8] presented a study in which they tested temsirolimus in phase II studies in East Asian patients with advanced RCC, having obtained promising results. This drug has also been used to treat cancer in multiple clinical studies, such as breast cancer [9], glioma [10], endometrial [11], and mantle cell lymphoma [12], as well as in preclinical studies, such as ovarian, lung, colon, breast, and prostate cell lines [13]. To enhance its action, combinations of temsirolimus with other antineoplastic drugs, such as sorafenib, bevacizumab, and tivozanib, have been explored in clinical trials in patients with metastatic RCC [14]. Temsirolimus and gemcitabine have already been tested in xenograft models of human pancreatic cancer in a promising therapeutic strategy [15]. However, a randomized phase III placebo-controlled trial of letrozole plus oral temsirolimus as a first-line endocrine therapy in postmenopausal women with locally advanced or metastatic breast cancer concluded that adding temsirolimus to letrozole did not improve progression-free survival, as first-line therapy in patients with nonsteroidal aromatase inhibitor-naive advanced breast cancer [16].

In bladder carcinoma, Gerullis et al. [17] applied a combined treatment of temsirolimus and vinflunine with a long-term response in a patient with bladder cancer. Rapamycin alone seems to be an effective drug in urothelial cell lines [18], [19] and its action when used in combination with cisplatin exhibited potential results [20]. Previous studies with other rapalog drugs, such as everolimus, on bladder cancer cell lines showed heterogeneous effects [21], [22]. In addition, rapalogs’ activity has been discussed and it has been suggested that its effectiveness may depend on the drug concentration (nanomolar vs. micromolar) [13] and on the genetic tumor profile, such as a loss of phosphatase and tensin homolog (PTEN) or of von Hippel-Lindau tumor suppressor genes, as well as the overexpression of Akt signaling [23], [24], [25].

In this study, we analyzed the effect of temsirolimus, in isolation and in combination, with cisplatin and gemcitabine (the cytotoxic backbone drugs of several chemotherapeutic treatments against bladder cancer) on 5637, T24, and HT1376 human bladder cancer cell lines, evaluating their action during cell proliferation, apoptosis, autophagy, and cell cycle distribution.

Section snippets

Cell culture

This study was conducted on a non–muscle invasive bladder cancer cell line (5637) and 2 human muscle invasive bladder cancer cell lines: T24 (DSMZ, Düsseldorf, Germany) and HT1376 (the 5637 and HT1376 cell lines were kindly provided by Dr. Paula Videira of the Universidade Nova de Lisboa, Lisboa, Portugal). All cell lines were cultured in RPMI 1640 culture medium (PAA, Pasching, Austria), supplemented with 10% heat-inactivated fetal bovine serum (Biological Industries, Kibbutz Beit Haemek,

Cytotoxic analysis

Using the MTT assay, temsirolimus treatment showed cell growth inhibition effect, in a dose-dependent manner (Fig. 1A). Its activity was similar in the 3 cell lines, achieving an average of IC30 at 4,000 nM. With the exception of 2 concentrations (500 nM and 1,000 nM) used in the 5637 cell line, the antiproliferative activity of temsirolimus was statistically significant (P<0.05) in all the cell lines, at all concentrations, when compared to the control group.

Autophagy assay

In the control cells, a basal level of

Discussion

Gemcitabine and cisplatin combination is the standard chemotherapy regimen used in the treatment of muscle invasive bladder cancer. This combination had efficacy similar to methotrexate, vinblastine, doxorubicin, and cisplatin in terms of objective response rate, progression-free survival, and overall survival and demonstrated a more favorable toxicity profile [31]. Even so, urinary bladder cancer does recur with most patients who unluckily do succumb to the disease [31], [32]. Therefore, it is

Acknowledgments

We would like to thank the Portuguese Association of Urology and FCT Pest-OE/AGR/UI0772/2011 unity.

References (55)

  • D. Hanahan et al.

    Hallmarks of cancer: the next generation

    Cell

    (2011)
  • R.M. Bukowski

    Temsirolimus: a safety and efficacy review

    Expert Opin Drug Saf

    (2012)
  • V. Wacheck

    mTOR pathway inhibitors in cancer therapy: moving past rapamycin

    Pharmacogenomics

    (2010)
  • J.S. Carew et al.

    Mechanisms of mTOR inhibitor resistance in cancer therapy

    Target Oncol

    (2011)
  • T. O'Reilly et al.

    Evaluation of the mTOR inhibitor, everolimus, in combination with cytotoxic antitumor agents using human tumor models in vitro and in vivo

    Anticancer Drugs

    (2011)
  • H. Pópulo et al.

    The mTOR signalling pathway in human cancer

    Int J Mol Sci

    (2012)
  • S.M. Baxi et al.

    Targeting the mTOR pathway in tumor malignancy

    Curr Cancer Drug Targets

    (2013)
  • Y. Sun et al.

    Phase II study of the safety and efficacy of Temsirolimus in East Asian patients with advanced renal cell carcinoma

    Jpn J Clin Oncol

    (2012)
  • G.F. Fleming et al.

    Phase II trial of temsirolimus in patients with metastatic breast cancer

    Breast Cancer Res Treat

    (2012)
  • A. Oza et al.

    Phase II study of temsirolimus in women with recurrent or metastatic endometrial cancer: a trial of the NCIC Clinical Trials Group

    J Clin Oncol

    (2011)
  • B. Shor et al.

    A new pharmacologic action of CCI-779 involves FKBP12-independent inhibition of mTOR kinase activity and profound repression of global protein synthesis

    Cancer Res

    (2008)
  • S. Pal et al.

    Future directions of mammalian target of rapamycin (mTOR) inhibitor therapy in renal cell carcinoma

    Target Oncol

    (2011)
  • D. Ito et al.

    In vivo antitumor effect of the mTOR inhibitor CCI-779 and gemcitabine in xenograft models of human pancreatic cancer

    Int J Cancer

    (2006)
  • A. Wolff et al.

    Randomized Phase III placebo-controlled trial of Letrozole plus oral temsirolimus as first-line endocrine therapy in postmenopausal women with locally advanced or metastatic breast cancer

    J Clin Oncol

    (2013)
  • H. Gerullis et al.

    Long-term response in advanced bladder cancer involving the use of temsirolimus and vinflunine after platin resistance

    Anticancer Drugs

    (2011)
  • R. Pinto-Leite et al.

    Effect of sirolimus on urinary bladder cancer T24 cell line

    J Exp Clin Cancer Res

    (2009)
  • I. Makhlin et al.

    The mTOR pathway affects proliferation and chemosensitivity of urothelial carcinoma cells and is upregulated in a subset of human bladder cancers

    BJU Int

    (2011)
  • Cited by (22)

    • Promising hepatoprotective effects of lycopene in different liver diseases

      2022, Life Sciences
      Citation Excerpt :

      Moreover, LYC downregulated MAPK in the liver tissues of rats exposed to toxic doses of tramadol [169]. The anti-neoplastic drug cisplatin is a common and potent chemotherapeutic used to treat testicular, ovarian, cervical, and head and neck cancers and bladder malignancies [170–174]. It is one of the most popular anticancer drugs.

    • Long noncoding RNA FOXD2-AS1 accelerates the gemcitabine-resistance of bladder cancer by sponging miR-143

      2018, Biomedicine and Pharmacotherapy
      Citation Excerpt :

      The chemotherapy is one of the most common clinical therapeutic methods for advanced malignancies. Combination drug therapy for bladder cancer, for example cisplatin and gemcitabine, is the effective treatment for bladder cancer [16,17]. In this study, we found that lncRNA FOXD2-AS1 was over-expressed in gemcitabine-resistant bladder cancer cells accompanied by dose-dependent pattern.

    • Pirarubicin induces an autophagic cytoprotective response through suppression of the mammalian target of rapamycin signaling pathway in human bladder cancer cells

      2015, Biochemical and Biophysical Research Communications
      Citation Excerpt :

      Temsirolimus, pazopanib and sunitinib are reported to trigger autophagic death of bladder cancer cells. By contrast, the AKT inhibitor AZ7328 and mTORc1/TORc2 inhibitor OSI-027 are found to support cell survival by increasing the autophagy level [18–21]. These reports have proved that the apparent conundrum of the balance between autophagy and apoptosis exists in bladder cancer.

    • Treatment of muscle invasive urinary bladders tumors: A potential role of the mTOR inhibitors

      2014, Biomedicine and Aging Pathology
      Citation Excerpt :

      Park et al. (2011) revealed that mTOR pathway activation, as assessed by 4E-BP1 and/or S6K1 phosphorylation, is involved in urinary bladder cancer tumorigenesis and is a predictor of disease progression and poor cancer-specific survival [30]. We have previously evaluated everolimus and temsirolimus actions on mTOR and Akt, as well as on their phosphorylated forms, in the three cell lines, without encouraging results [18,24]. Therefore, we decided to analyze, the p70S6 K and 4E-BP1 and its phosphorylated forms.

    View all citing articles on Scopus
    View full text