Cancer Letters

Cancer Letters

Volume 412, 1 January 2018, Pages 118-130
Cancer Letters

Original Article
Flubendazole overcomes trastuzumab resistance by targeting cancer stem-like properties and HER2 signaling in HER2-positive breast cancer

https://doi.org/10.1016/j.canlet.2017.10.020Get rights and content

Highlights

  • Flubendazole induces apoptosis in HER2-positive breast cancer cells.

  • Trastuzumab-resistant cells are sensitive to flubendazole.

  • Flubendazole effectively targets cancer stem-like properties.

  • Flubendazole downregulates truncated p95HER2, phospho-HER2, and phospho-HER3.

  • Flubendazole suppresses tumor growth in trastuzumab-resistant xenografts.

Abstract

Although trastuzumab provides significant clinical benefit for HER2-positive breast cancers, responses are limited by the emergence of resistance. Trastuzumab resistance is a multi-factorial phenomenon thought to arise from the presence of cancer stem cells and interactions between truncated p95HER2 and HER family members. Flubendazole (FLU) is a potent anthelmintic agent with an exceptional safety profile. Evidence also suggests that it can act as an anticancer agent in several cancer cell types. We sought to investigate the effect of FLU on apoptosis, HER2/Akt signaling, breast cancer stem cell (BCSC)-like properties and trastuzumab resistance in HER2-positive breast cancer cells. FLU treatment induced apoptosis, associated with a significant downregulation of truncated p95HER2, phospho-HER2, phospho-HER3 and phospho-Akt levels, as well as suppression of HER2/HER3 hetero-dimerization in both trastuzumab-sensitive and –resistant lines. FLU effectively targeted BCSC-like properties including aldehyde dehydrogenase 1 (ALDH1) expression and the CD44high/CD24low phenotype, concomitant with a suppression of mammosphere-forming ability. FLU administration also caused significant tumor suppression in trastuzumab-resistant xenografts, coinciding with the downregulation of BCSC-related markers and intracellular HER2. These findings highlight the mechanisms of action of FLU in overcoming trastuzumab resistance in breast cancer.

Introduction

The human epidermal growth factor receptor 2 (HER2) is amplified in 20–30% of all breast cancers, and its overexpression is linked to aggressive tumor behavior and a poorer prognosis [1], [2]. Although HER2 has no known direct activating ligand, it can be activated through homo- and hetero-dimerization with other HER family receptors including EGFR (HER1), HER3 and HER4 [3], [4]. The HER2/HER3 dimer is the most potent oncogenic pair, and dimerization leads to activation of their kinase domains and subsequent activation of downstream signaling pathways, including phosphatidylinositol-3 kinase (PI3K)/Akt and mitogen-activated protein kinase (MAPK) [3], [4], [5], [6]. As a consequence, these signaling cascades promote the expression of genes that orchestrate cell survival, proliferation and differentiation, which fosters tumor progression [5], [7].

Trastuzumab is a humanized monoclonal antibody that disrupts HER2 activation by binding to its extracellular domain IV, leading to antibody-dependent cell-mediated cytotoxicity (ADCC) [8], [9], [10]. It has a significant clinical impact on disease-free and overall survival in patients with HER2-amplified metastatic and early breast cancer [11], [12], [13]. However, despite these outcomes in some cases, nearly 70% of patients experience primary or acquired resistance [12], and this issue remains a significant unmet medical need. Several potential mechanisms of resistance to trastuzumab include dimerization between members of the HER family, hyperactivation of PI3K/Akt signaling, mutation or loss of PTEN, and the upregulation of other receptor tyrosine kinases such as insulin-like growth factor 1 receptor (IGF-1R) and c-MET [14]. Of particular note, oncogenic p95HER2 is a major factor in trastuzumab resistance and is frequently found in 30% of HER2-positive breast cancers [15]. p95HER2 is a C-terminal fragment of HER2 that lacks the extracellular trastuzumab-binding domain but retains constitutive kinase activity, activating downstream signaling [16].

Emerging evidence suggests that there is a significant correlation between HER2 overexpression and breast cancer stem cell (BCSC)-like populations [17], [18]. BCSCs constitute a small subset of tumor cells with tumor-initiating potential, characterized by the CD44high/CD24low phenotype and high aldehyde dehydrogenase 1 (ALDH1) activity [17], [19]. These cells give rise to tumorigenesis, tumor growth and propagation, and are also associated with tumor recurrence and resistance to treatment [20], [21]. Preclinical studies have demonstrated that BCSCs are sensitive to trastuzumab treatment in HER2-positive breast cancer cells, but trastuzumab fails to eliminate trastuzumab-resistant BCSCs, driving recurrence and metastatic spread [18], [22]. Therefore, new treatment options are needed for the targeting of BCSC-like properties for a sustainable therapeutic approach to HER2-positive breast cancers.

Flubendazole (FLU) belongs to a large group of benzimidazole derivatives and is currently available in a number of countries worldwide for human use to treat gastrointestinal nematodes [23], [24], [25], [26]. Recent preclinical studies have shown that FLU exerts anti-proliferative activity in various types of cancers including leukemia, myeloma, neuroblastoma, colorectal and triple-negative breast cancer in vitro and in vivo [27], [28], [29], [30]. FLU is an inhibitor of tubulin polymerization that disrupts microtubule structure [27], however, its effects on HER2-positive breast cancers have not been elucidated. In the present study, we sought to characterize the mechanism of action responsible for FLU's novel effects in targeting BCSC and the HER2/Akt signaling pathway in trastuzumab-resistant HER2-positive breast cancer in vitro and in vivo.

Section snippets

Reagents and antibodies

Flubendazole, dimethyl sulfoxide (DMSO) and Triton X-100 were purchased from Sigma-Aldrich (St. Louis, MO). Primary antibodies used targeted the following proteins: β-tubulin, phospho-histone H3 (Ser10), ALDH1A1 and CD31 (Abcam, Cambridge, MA); Akt, phospho-Akt, PARP, cleaved-PARP, cleaved-caspase-3, cleaved-caspase-7, cleaved-caspase-8, HER2, phospho-HER2 (Tyr1221/1222), phospho-HER3 (Tyr1289), HER3 (Cell Signaling Technology, Beverly, CA); CB11 (Thermo Fisher Scientific Fremont, CA);

FLU reduces cell viability and induces G2/M phase arrest in HER2-positive breast cancer cells in vitro

To evaluate the cytotoxic effects of FLU in HER2-positive breast cancer cells, BT474 and SKBR3 cells were treated with various concentrations of FLU for up to 72 h. The cells treated with FLU (0.1–0.5 μM, 48 h) exhibited apoptotic morphological changes (Fig. 1A). Cell viability was significantly reduced in response to FLU (0.1–8 μM) in a dose- and time-dependent manner (p < 0.05, Fig. 1B). An analysis of the cell cycle distribution of BT474 and SKBR3 cells showed that FLU significantly

Discussion

Our findings suggest that FLU can overcome trastuzumab resistance in HER2-positive breast cancer cells via the suppression of HER2/Akt signaling and BCSC-like properties. FLU has been widely used for the treatment of intestinal parasites in humans and animals, and has a low acute toxicity [25], [49]. In rodents, the oral lethal dose (LD50) is more than 5000 mg/kg, and the intraperitoneal LD50 values are more than 400 mg/kg [50]. Several clinical studies have shown that patients can receive up

Author contributions

Conception and design: JY. Kim, Y-J. Kim, E. Oh and JH. Seo.

Performed the experiments: D. Sung, Y-J. Kim, E. Oh, Y. Cho and T-M, Cho and JY. Kim.

Analyzed the data: D. Sung, Y-J. Kim, E. Oh, L. Farrand and JY. Kim.

Contributed reagents, materials and analysis tools: D Sung, Y-J. Kim, E. Oh, Y. Cho, T-M. Cho, JH. Seo and JY. Kim.

Wrote the paper: Y-J. Kim, E. Oh, L. Farrand and JY. Kim.

Final approval manuscript: Y-J. Kim, D Sung, E. Oh, Y. Cho, T-M. Cho, L. Farrand, JH. Seo and JY. Kim.

Acknowledgements

This research was supported by a grant from the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (grant number: HI12C1852), the National Research Foundation of Korea (NRF) grant, funded by the Ministry of Science, ICT & Future Planning (MSIP, grant number: 2015R1C1A2A01053747) and the Ministry of Education (grant number: 2017R1A6A3A11029467), and was supported by a Korea University

References (68)

  • J.Y. Kim et al.

    Disulfiram targets cancer stem-like properties and the HER2/Akt signaling pathway in HER2-positive breast cancer

    Cancer Lett.

    (2016)
  • B. van der Vegt et al.

    Validation of the 4B5 rabbit monoclonal antibody in determining Her2/neu status in breast cancer

    Mod. Pathol. Off. J. U. S. Can. Acad. Pathol. Inc.

    (2009)
  • Z. Palyi-Krekk et al.

    Hyaluronan-induced masking of ErbB2 and CD44-enhanced trastuzumab internalisation in trastuzumab resistant breast cancer

    Eur. J. Cancer

    (2007)
  • S. Ghatak et al.

    Hyaluronan oligosaccharides inhibit anchorage-independent growth of tumor cells by suppressing the phosphoinositide 3-kinase/Akt cell survival pathway

    J. Biol. Chem.

    (2002)
  • Y. Wu et al.

    Impact of lapatinib plus trastuzumab versus single-agent lapatinib on quality of life of patients with trastuzumab-refractory HER2+ metastatic breast cancer

    Ann. Oncol. Off. J. Eur. Soc. Med. Oncol.

    (2011)
  • D.J. Slamon et al.

    Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene

    Science

    (1987)
  • S. Menard et al.

    HER2 as a prognostic factor in breast cancer

    Oncology

    (2001)
  • Y. Yarden et al.

    Untangling the ErbB signalling network

    Nat. Rev. Mol. Cell Biol.

    (2001)
  • M.A. Olayioye et al.

    The ErbB signaling network: receptor heterodimerization in development and cancer

    EMBO J.

    (2000)
  • T. Holbro et al.

    The ErbB2/ErbB3 heterodimer functions as an oncogenic unit: ErbB2 requires ErbB3 to drive breast tumor cell proliferation

    Proc. Natl. Acad. Sci. U. S. A.

    (2003)
  • S.T. Lee-Hoeflich et al.

    A central role for HER3 in HER2-amplified breast cancer: implications for targeted therapy

    Cancer Res.

    (2008)
  • M.M. Moasser

    The oncogene HER2: its signaling and transforming functions and its role in human cancer pathogenesis

    Oncogene

    (2007)
  • M.A. Molina et al.

    Trastuzumab (herceptin), a humanized anti-Her2 receptor monoclonal antibody, inhibits basal and activated Her2 ectodomain cleavage in breast cancer cells

    Cancer Res.

    (2001)
  • R. Gennari et al.

    Pilot study of the mechanism of action of preoperative trastuzumab in patients with primary operable breast tumors overexpressing HER2

    Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res.

    (2004)
  • M.J. Piccart-Gebhart et al.

    Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer

    N. Engl. J. Med.

    (2005)
  • D.J. Slamon et al.

    Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2

    N. Engl. J. Med.

    (2001)
  • I.J. Dahabreh et al.

    Trastuzumab in the adjuvant treatment of early-stage breast cancer: a systematic review and meta-analysis of randomized controlled trials

    Oncologist

    (2008)
  • P.R. Pohlmann et al.

    Resistance to trastuzumab in breast cancer

    Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res.

    (2009)
  • R. Saez et al.

    p95HER-2 predicts worse outcome in patients with HER-2-positive breast cancer

    Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res.

    (2006)
  • J. Arribas et al.

    p95HER2 and breast cancer

    Cancer Res.

    (2011)
  • H. Korkaya et al.

    HER2 regulates the mammary stem/progenitor cell population driving tumorigenesis and invasion

    Oncogene

    (2008)
  • M. Al-Hajj et al.

    Prospective identification of tumorigenic breast cancer cells

    Proc. Natl. Acad. Sci. U. S. A.

    (2003)
  • X. Li et al.

    Intrinsic resistance of tumorigenic breast cancer cells to chemotherapy

    J. Natl. Cancer Inst.

    (2008)
  • T.M. Phillips et al.

    The response of CD24(-/low)/CD44+ breast cancer-initiating cells to radiation

    J. Natl. Cancer Inst.

    (2006)
  • Cited by (40)

    • Emerging insights on functions of the anthelmintic flubendazole as a repurposed anticancer agent

      2021, Cancer Letters
      Citation Excerpt :

      However, trastuzumab's efficacy is limited by resistance involving breast cancer stem cells [25]. Kim et al. [6] reported that flubendazole can potentially overcome resistance to trastuzumab [26] by targeting HER2 signaling and cancer stem-like properties. Flubendazole caused G2/M phase arrest and apoptosis (activated caspase-3, caspase-7, caspase-8) in HER2+ breast cancer cells (BT474 and SKBR3).

    • Flubendazole, FDA-approved anthelmintic, elicits valid antitumor effects by targeting P53 and promoting ferroptosis in castration-resistant prostate cancer

      2021, Pharmacological Research
      Citation Excerpt :

      Flubendazole, a member of benzimidazole families, has been widely used to treat infections of the worm and intestinal parasites in clinical practice [5–9], which means that it has already undergone anthelmintic efficacy and safety trials in humans. In addition to its anthelmintic effects, several studies have reported that flubendazole might be beneficial in treating different types of tumors, including colon cancer, breast cancer, leukemia, and glioma, due to its ability to inhibit tubulin polymerization, resulting in mitotic catastrophe [10–17]. However, the specific roles of flubendazole and its precise mechanism of treating CRPC have not been elucidated.

    • Eucannabinolide, a novel sesquiterpene lactone, suppresses the growth, metastasis and BCSCS-like traits of TNBC via inactivation of STAT3

      2021, Neoplasia (United States)
      Citation Excerpt :

      Surprisingly, 80% of TNBC cells expressed activated STAT3 [10,11]. Notably, a significant relationship between constitutive activation of STAT3 and breast cancer stem cell (BCSC)-like properties was observed [11–14]. BCSCs are proved to promote tumor initiation, growth and propagation [15].

    View all citing articles on Scopus
    1

    Y-J. Kim, D. Sung and E. Oh contributed equally to this work.

    View full text