Cancer Letters

Cancer Letters

Volume 424, 28 June 2018, Pages 46-69
Cancer Letters

Original Articles
Phytochemicals as potent modulators of autophagy for cancer therapy

https://doi.org/10.1016/j.canlet.2018.02.030Get rights and content

Highlights

  • Targeting autophagy is a promising therapeutic strategy for cancer therapy.

  • Phytochemicals represent a rich source as autophagy modulators.

  • Autophagic effects of phytochemicals depend on dose, cell type and context.

  • Modulation of autophagy by phytochemicals targets cancer at different stages.

  • Nanotechnology solves problem of bioavailability of phytochemicals.

Abstract

The dysregulation of autophagy is involved in the pathogenesis of a broad range of diseases, and accordingly universal research efforts have focused on exploring novel compounds with autophagy-modulating properties. While a number of synthetic autophagy modulators have been identified as promising cancer therapy candidates, autophagy-modulating phytochemicals have also attracted attention as potential treatments with minimal side effects. In this review, we firstly highlight the importance of autophagy and its relevance in the pathogenesis and treatment of cancer. Subsequently, we present the data on common phytochemicals and their mechanism of action as autophagy modulators. Finally, we discuss the challenges associated with harnessing the autophagic potential of phytochemicals for cancer therapy.

Introduction

Phytochemicals (phyton means “plant” in Greek) are naturally occurring plant-based compounds. They are mostly composed of non-nutrient chemicals that are found in grains, vegetables, and fruits [1,2]. While over ten thousand phytochemicals have been identified, many others remain unknown and need to be identified [[1], [2], [3]]. Important groups of phytochemicals include phenolic compounds, alkaloids, terpenes, organosulfides, and glucosinolates [1,4,5]. The health benefits of phytochemicals are mostly associated with reducing the risk of developing diverse human diseases [3,5,6]. Such plant-based compounds are easily accessible, often with less toxic effects than synthetic molecules, and possess a wide range of biological and pharmacological effects, including anti-microbial, anti-tumoral, anti-mutagenic, and anti-oxidant or pro-oxidant activities [1,3,[5], [6], [7], [8], [9]]. Positive outcomes in clinical trials have led to the introduction of several phytochemicals into clinical practice in recent decades [6,10]. Particularly in cancer and other chronic diseases that are associated with excessive production of reactive oxygen species (ROS), both the anti-oxidant and pro-oxidant properties of phytochemicals can be utilized to prevent or eradicate cancer, respectively [3,9,[11], [12], [13]]. For example, enriched flavonoids from vegetables and fruits show potent ROS modulating and anti-cancer effects, alone or in combination with standard chemotherapy [14]. Mechanistically, the anti-cancer effects of phytochemicals are mostly mediated through induction of cell-cycle arrest or apoptotic cell death [[7], [8], [9],15]. In addition, recent studies have uncovered an important role for phytochemicals in governing the autophagy network [7,[15], [16], [17]]. In both normal and malignant cells, autophagy is induced by certain cellular stresses in order to preserve cell survival [18,19]. However, if the stress is not resolved, autophagy leads ultimately to programmed cell death [[18], [19], [20], [21], [22]]. In the context of cancer, autophagy acts as a tumor suppressor at early steps of tumorigenesis. Alternatively, autophagy can promote the growth and survival of established tumors during migration and epithelial-to-mesenchymal transition (EMT) as well as in response to chemotherapy [18,23,24]. In addition, autophagy is involved in cancer stem cell (CSC) survival, escape from immune surveillance and resistance to anoikis [25]. Therefore, autophagy with a double face role inhibits early stages of tumorigenesis while it becomes a driver of tumor invasion and metastasis at later stages [[26], [27], [28]]. Accordingly, manipulation of key factors in the autophagy pathway may be exploited as a novel therapeutic strategy for cancer therapies [20]. However, before proposing phytochemical as anti-cancer and autophagy-modulating agents, a better understanding of their complex mechanism of action needs to be addressed more deeply [5,16,17]. In this review, we represent common phytochemicals as a group of promising autophagy modulators and discuss their therapeutic importance in treating various cancers.

Section snippets

Autophagy machinery and its regulation

The term “autophagy” was coined by Christian de Duve in 1963. It was derived from Greek meaning “eating of self” [29]. Macroautophagy (hereafter called autophagy) is responsible for the turnover of cellular components that are sequestered into the double-membrane-bound vesicle, called autophagosome that originates from a precursor structure called phagophore [19,30]. The primarily goal of autophagy is to sustain cell survival under stressful conditions (e.g., starvation and nutrient

Phytochemicals as a valuable source of autophagy modulation agents

Epidemiological studies demonstrated that there is a strong association between diet and human cancer mortality so that daily consumption of phytochemicals declines the incidence of different types of cancer [2,52]. Therefore, a lot of research efforts focus on nutrients and non-nutritive phytochemicals to evaluate their chemopreventive and chemotherapeutic potential in in vitro and in vivo models of cancer [2,6,9,10]. In clinical oncology, natural compounds induce cell death pathways, mostly

Challenges in using phytochemicals as drugs

Despite the therapeutic benefits of phytochemicals, two main obstacles including, poor bioavailability [316] and lack of correlation between in vitro and in vivo concentrations, hinder their clinical use [317]. The term “bioavailability” means the concentration of the absorbed compound, or its metabolite, in a specific organ that is related to the primary target for the therapeutic action and is determined by the chemical structure of the compounds [318]. Pharmacokinetic studies showed that low

Conclusion and prospective

Despite the growing knowledge of cancer pathogenesis and its treatment, developing targeted chemotherapeutics remains a major challenge in modern medicine. Current cancer therapies are unable to target cancer stem cells, and in most cases, a relapse of the disease might be observed. Additionally, multi-drug resistance is another substantial problem in the clinical application of current chemotherapeutic drugs. In this line, autophagy modulators may serve as promising anti-cancer agents to

Conflicts of interest

None.

Acknowledgments

The authors acknowledge National Institute for Medical Research Development (NIMAD) project no. 940943 and National Research Institute for Science Policy (NRISP) no. 1456 for financial support of this work. AM appreciates National Institute for Genetic Engineering and Biotechnology grants. MR appreciates Cancer Biology Research Center grants. Atanas G. Atanasov acknowledges the support by the Polish KNOW (Leading National Research Centre) Scientific Consortium “Healthy Animal—Safe Food,”

References (356)

  • S. Arico

    The tumor suppressor PTEN positively regulates macroautophagy by inhibiting the phosphatidylinositol 3-kinase/protein kinase B pathway

    J. Biol. Chem.

    (2001)
  • M. Li et al.

    Crosstalk between autophagy and apoptosis: potential and emerging therapeutic targets for cardiac diseases

    Int. J. Mol. Sci.

    (2016)
  • L.-S. Wang et al.

    Anthocyanins and their role in cancer prevention

    Canc. Lett.

    (2008)
  • E.J. Choi et al.

    Apigenin causes G 2/M arrest associated with the modulation of p21 Cip1 and Cdc2 and activates p53-dependent apoptosis pathway in human breast cancer SK-BR-3 cells

    J. Nutr. Biochem.

    (2009)
  • B.D. Sahu

    Baicalein alleviates doxorubicin-induced cardiotoxicity via suppression of myocardial oxidative stress and apoptosis in mice

    Life Sci.

    (2016)
  • M. Tillhon et al.

    Berberine: new perspectives for old remedies

    Biochem. Pharmacol.

    (2012)
  • J.-w. Zhang

    Autophagy inhibition switches low-dose camptothecin-induced premature senescence to apoptosis in human colorectal cancer cells

    Biochem. Pharmacol.

    (2014)
  • M. Russo et al.

    Phytochemicals in cancer prevention and therapy: truth or dare?

    Toxins

    (2010)
  • G.-L. Russo et al.

    The pleiotropic flavonoid quercetin: from its metabolism to the inhibition of protein kinases in chronic lymphocytic leukemia

    Food Funct.

    (2014)
  • B.Y.K. Law

    New potential pharmacological functions of Chinese herbal medicines via regulation of autophagy

    Molecules

    (2016)
  • M.-K. Kim

    Autophagy as a target for anticancer therapy and its modulation by phytochemicals

    J. Food Drug Anal.

    (2012)
  • H. Gali-Muhtasib et al.

    Cell death mechanisms of plant-derived anticancer drugs: beyond apoptosis

    Apoptosis

    (2015)
  • M.M. Grabacka et al.

    Phytochemical modulators of mitochondria: the search for chemopreventive agents and supportive therapeutics

    Pharmaceuticals

    (2014)
  • J. Wang et al.

    Cancer cell killing via ROS: to increase or decrease, that is the question

    Canc. Biol. Ther.

    (2008)
  • D. Trachootham et al.

    Targeting cancer cells by ROS-mediated mechanisms: a radical therapeutic approach?

    Nat. Rev. Drug Discov.

    (2009)
  • M.A. Moosavi

    Photodynamic N-TiO2 nanoparticle treatment induces controlled Ros-mediated autophagy and terminal differentiation of leukemia cells

    Sci. Rep.

    (2016)
  • K. Jiang

    Silibinin, a natural flavonoid, induces autophagy via ROS-dependent mitochondrial dysfunction and loss of ATP involving BNIP3 in human MCF7 breast cancer cells

    Oncol. Rep.

    (2015)
  • S.-F. Zhang et al.

    Autophagy-associated targeting pathways of natural products during cancer treatment

    Asian Pac. J. Cancer Prev. APJCP: Asian Pac. J. Cancer Prev. APJCP

    (2014)
  • V. Naponelli et al.

    Roles of autophagy induced by natural compounds in prostate cancer

    BioMed Res. Int.

    (2015)
  • N. Hasima et al.

    Regulation of autophagy by polyphenolic compounds as a potential therapeutic strategy for cancer

    Cell Death Dis.

    (2014)
  • D.J. Klionsky

    Autophagy: from phenomenology to molecular understanding in less than a decade

    Nat. Rev. Mol. Cell Biol.

    (2007)
  • D.C. Rubinsztein et al.

    Potential therapeutic applications of autophagy

    Nat. Rev. Drug Discov.

    (2007)
  • B.Y.K. Law

    Natural small-molecule enhancers of autophagy induce autophagic cell death in apoptosis-defective cells

    Sci. Rep.

    (2014)
  • K. Sun

    Paradoxical roles of autophagy in different stages of tumorigenesis: protector for normal or cancer cells

    Cell Biosci.

    (2013)
  • E.E. Mowers et al.

    Autophagy in cancer metastasis

    Oncogene

    (2017)
  • B. Janji et al.
    (2013)
  • R. Mathew et al.

    Role of autophagy in cancer

    Nat. Rev. Canc.

    (2007)
  • R.L. Deter et al.

    Influence of glucagon, an inducer of cellular autophagy, on some physical properties of rat liver lysosomes

    J. Cell Biol.

    (1967)
  • D.J. Klionsky

    The molecular machinery of autophagy: unanswered questions

    J. Cell Sci.

    (2005)
  • P. Mokarram

    New frontiers in the treatment of colorectal cancer: autophagy and the unfolded protein response as promising targets

    Autophagy

    (2017)
  • P.-M. Coly et al.

    The autophagy machinery: a new player in chemotactic cell migration

    Front. Neurosci.

    (2017)
  • H. Heath-Engel et al.

    The endoplasmic reticulum in apoptosis and autophagy: role of the BCL-2 protein family

    Oncogene

    (2008)
  • D. Gozuacik et al.

    Autophagy as a cell death and tumor suppressor mechanism

    Oncogene

    (2004)
  • Y. Ávalos

    Tumor suppression and promotion by autophagy

    BioMed Res. Int.

    (2014)
  • R. Errafiy

    PTEN Increases Autophagy and Inhibits the Ubiquitin-Proteasome Pathway in Glioma Cells Independently of its Lipid Phosphatase Activity

    (2013)
  • E.J. White

    Autophagy regulation in cancer development and therapy

    Am. J. Canc. Res.

    (2011)
  • F. Kruiswijk et al.

    p53 in survival, death and metabolic health: a lifeguard with a licence to kill

    Nat. Rev. Mol. Cell Biol.

    (2015)
  • E. Morselli

    Mutant p53 protein localized in the cytoplasm inhibits autophagy

    Cell Cycle

    (2008)
  • B. Levine et al.

    p53: the Janus of autophagy?

    Nat. Cell Biol.

    (2008)
  • M. Abedin et al.

    Autophagy delays apoptotic death in breast cancer cells following DNA damage

    Cell Death Differ.

    (2007)
  • Cited by (0)

    1

    These two authors contributed equally.

    View full text