Cancer Letters

Cancer Letters

Volume 349, Issue 1, 10 July 2014, Pages 8-14
Cancer Letters

Mini-review
Multi-targeted therapy of cancer by niclosamide: A new application for an old drug

https://doi.org/10.1016/j.canlet.2014.04.003Get rights and content

Abstract

The rapid development of new anticancer drugs that are safe and effective is a common goal shared by basic scientists, clinicians and patients. The current review discusses one such agent, namely niclosamide, which has been used in the clinic for the treatment of intestinal parasite infections. Recent studies repeatedly identified niclosamide as a potential anticancer agent by various high-throughput screening campaigns. Niclosamide not only inhibits the Wnt/β-catenin, mTORC1, STAT3, NF-κB and Notch signaling pathways, but also targets mitochondria in cancer cells to induce cell cycle arrest, growth inhibition and apoptosis. A number of studies have established the anticancer activities of niclosamide in both in vitro and in vivo models. Moreover, the inhibitory effects of niclosamide on cancer stem cells provide further evidence for its consideration as a promising drug for cancer therapy. This article reviews various aspects of niclosamide as they relate to its efficacy against cancer and associated molecular mechanisms.

Introduction

Niclosamide (trade name Niclocide), a teniacide in the anthelmintic family which is especially effective against cestodes, has been approved for use in humans for nearly 50 years (Fig. 1) [1], [2]. Niclosamide inhibits oxidative phosphorylation and stimulates adenosine triphosphatase activity in the mitochondria of cestodes (eg. tapeworm), killing the scolex and proximal segments of the tapeworm both in vitro and in vivo [2]. Niclosamide is well tolerated in humans. The treatment of Taenia saginata (beef tapeworm), Diphyllobothrium latum (fish tapeworm) and Dipylidium caninum (dog tapeworm) in adult is 2 g as a single oral dose. For the treatment of Hymenolepis nana (dwarf tapeworm), the same oral dose is used for 7 days [2].

Drug development, from the initial lead discovery to the final medication, is an expensive, lengthy and incremental process [3]. Finding new uses for old or failed drugs is much faster and more economical than inventing a new drug from scratch, as existing drugs have known pharmacokinetics and safety profiles and have often been approved for human use, therefore any newly identified use(s) can be rapidly evaluated in clinical trials [4]. In the last 5 years niclosamide has been identified as a potential anticancer agent by various high-throughput screening campaigns. This article reviews the current studies regarding various aspects of niclosamide as they relate to its potential new use in cancer therapy.

Section snippets

Niclosamide – a multiple pathway inhibitor for anti-cancer efficacy

Recently, several studies reported the inhibitory effects of niclosamide on multiple intracellular signaling pathways. The signaling molecules in these pathways are either over-expressed, constitutively active or mutated in many cancer cells, and thus render niclosamide as a potential anticancer agent. The effects of niclosamide on these pathways are described below.

Niclosamide targeting of mitochondria

Mitochondria are vital for cellular bioenergetics and play a central role in determining the point-of-no-return of the apoptotic process. It has been proposed that targeting mitochondria is an efficient strategy for cancer chemotherapy [45]. Khanim et al. screened a panel of 100 off-patent licensed oral drugs for anti-myeloma activity, and identified niclosamide as a killer of multiple myeloma cell lines and primary multiple myeloma cells [30]. Interestingly, niclosamide anti-multiple myeloma

Anti-cancer activity of niclosamide: in vitro studies

Anti-cancer activity of niclosamide has been demonstrated in human breast cancer [10], [11], [18], [48], [49], prostate cancer [10], [29], colon cancer [9], [15], ovarian cancer [47], multiple myeloma [30], acute myelogenous leukemia [38], glioblastoma [12], head and neck cancer [21] and lung cancer cells [31]. As summarized in the earlier sections, niclosamide is able to block the multiple signaling pathways that govern cancer initiation and progression, thus it is not surprising that

Anti-cancer activity of niclosamide: in vivo studies

The efficacy of niclosamide has been shown against tumor growth and metastases in several xenograft models. Jin et al. demonstrated for the first time that niclosamide has in vivo anti-cancer activities [38]. As niclosamide has limited solubility in water, Jin et al. synthesized a niclosamide analog – phosphate of niclosamide (p-niclosamide), and found p-niclosamide showed significant inhibition of xenograft tumor growth of acute myeloid leukemia HL-60 cells by suppressing the NF-κB pathway [38]

Niclosamide as a drug for targeting CSCs

Current cancer treatments such as chemotherapy, targeted therapy and radiotherapy are successful at destroying bulk cancer cells, but fail to eliminate cancer stem cells (CSCs). CSCs are characterized by tumorigenic properties and the ability to self-renew, form differentiated progeny, and develop resistance to therapy. The inability to eradicate CSCs is thought to be the reason for cancer relapse and chemo-resistance, the major obstacles in current cancer therapy [57], [58], [59]. The

Challenges to using niclosamide as an anti-cancer agent in humans

The oral dose of niclosamide for adult in cestocidal treatment is 2 g as a single dose, leading to maximal serum concentrations of 0.25–6.0 μg/ml (corresponding to 0.76–18.35 μM) [1], which is well within the anti-cancer active concentration range. The reason for this wide range of serum concentrations is considered to be due to intra-individual difference in absorption rate [1]. Niclosamide has poor water solubility, and the oral bioavailability of niclosamide was only 10% in male Sprague-Dawley

Future directions

While considerable high-throughput screening campaigns have identified niclosamide is a potent inhibitor of a number of biological signaling pathways that mediate niclosamide’s anti-cancer effects in vitro and in vivo, direct targets of niclosamide still remain unclear. Niclosamide was tested in vitro against a panel of 95 protein kinases, and it did not significantly inhibit any of these kinases at concentrations (1 or 10 μM) efficiently inhibiting the Wnt/β-catenin, mTORC1, STAT3, NF-κB and

Conflict of Interest

The authors declare that they have no conflict of interest.

Acknowledgments

We thank Meredith S. Plaxco and Tommie A. Gamble for running the 60 human tumor cell line anticancer drug screen. This work was completed with the support of Grants from the National Institute of Health (R01CA124531 and R21CA182056 to Y. Li).

References (77)

  • H. Sakurai et al.

    Functional interactions of transforming growth factor beta-activated kinase 1 with IkappaB kinases to stimulate NF-kappaB activation

    J. Biol. Chem.

    (1999)
  • S.Y. Kim et al.

    Role of the IL-6-JAK1-STAT3-Oct-4 pathway in the conversion of non-stem cancer cells into cancer stem-like cells

    Cell. Signal.

    (2013)
  • S. Gongoll et al.

    Prognostic significance of calcium-binding protein S100A4 in colorectal cancer

    Gastroenterology

    (2002)
  • M.R. Alison et al.

    Cancer stem cells: in the line of fire

    Cancer Treat. Rev.

    (2012)
  • C. Ginestier et al.

    ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome

    Cell Stem Cell

    (2007)
  • P.B. Gupta et al.

    Identification of selective inhibitors of cancer stem cells by high-throughput screening

    Cell

    (2009)
  • R.A. Mook et al.

    Small molecule modulators of Wnt/beta-catenin signaling

    Bioorg. Med. Chem. Lett.

    (2013)
  • S. Kraljevic et al.

    Accelerating drug discovery

    EMBO Rep.

    (2004)
  • C.R. Chong et al.

    New uses for old drugs

    Nature

    (2007)
  • J.N. Anastas et al.

    WNT signalling pathways as therapeutic targets in cancer

    Nat. Rev. Cancer

    (2013)
  • P. Polakis

    Drugging Wnt signalling in cancer

    EMBO J.

    (2012)
  • M. Chen et al.

    The anti-helminthic niclosamide inhibits Wnt/Frizzled1 signaling

    Biochemistry

    (2009)
  • T. Osada et al.

    Antihelminth compound niclosamide downregulates Wnt signaling and elicits antitumor responses in tumors with activating APC mutations

    Cancer Res.

    (2011)
  • W. Lu et al.

    Niclosamide suppresses cancer cell growth by inducing Wnt co-receptor LRP6 degradation and inhibiting the Wnt/beta-catenin pathway

    PLoS ONE

    (2011)
  • A. Londoño-Joshi et al.

    Effect of niclosamide on basal-like breast cancers

    Mol. Cancer Ther.

    (2014)
  • A. Wieland et al.

    Anticancer effects of niclosamide in human glioblastoma

    Clin. Cancer Res.

    (2013)
  • U. Sack et al.

    Novel effect of antihelminthic niclosamide on S100A4-mediated metastatic progression in colon cancer

    J. Natl. Cancer Inst.

    (2011)
  • J. Dancey

    MTOR signaling and drug development in cancer

    Nat. Rev. Clin. Oncol.

    (2010)
  • A.D. Balgi et al.

    Screen for chemical modulators of autophagy reveals novel therapeutic inhibitors of mTORC1 signaling

    PLoS ONE

    (2009)
  • J.L. Jewell et al.

    Amino acid signalling upstream of mTOR

    Nat. Rev. Mol. Cell Biol.

    (2013)
  • H. Yu et al.

    STATs in cancer inflammation and immunity: a leading role for STAT3

    Nat. Rev. Cancer

    (2009)
  • J. Sasse et al.

    Mutational analysis of acute-phase response factor/Stat3 activation and dimerization

    Mol. Cell. Biol.

    (1997)
  • Z. Zhong et al.

    Stat3: a STAT family member activated by tyrosine phosphorylation in response to epidermal growth factor and interleukin-6

    Science

    (1994)
  • J.E. Darnell et al.

    Jak-STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins

    Science

    (1994)
  • V. Boudny et al.

    JAK/STAT signaling pathways and cancer. Janus kinases/signal transducers and activators of transcription

    Neoplasma

    (2002)
  • L.S. Steelman et al.

    Contributions of the Raf/MEK/ERK, PI3K/PTEN/Akt/mTOR and Jak/STAT pathways to leukemia

    Leukemia

    (2008)
  • R. Ren et al.

    Identification of niclosamide as a new small-molecule inhibitor of the STAT3 signaling pathway

    ACS Med. Chem. Lett.

    (2010)
  • F.L. Khanim et al.

    Redeployment-based drug screening identifies the anti-helminthic niclosamide as anti-myeloma therapy that also reduces free light chain production

    Blood Cancer J.

    (2011)
  • Cited by (0)

    View full text