Next Article in Journal
An In Vitro Potency Assay for Monitoring the Immunomodulatory Potential of Stromal Cell-Derived Extracellular Vesicles
Next Article in Special Issue
Impact of Autophagy in Oncolytic Adenoviral Therapy for Cancer
Previous Article in Journal
The Nucleolus: In Genome Maintenance and Repair
Previous Article in Special Issue
Autophagic Mechanism in Anti-Cancer Immunity: Its Pros and Cons for Cancer Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Natural Compounds from Herbs that can Potentially Execute as Autophagy Inducers for Cancer Therapy

1
Department of Life Science and Institute of Biotechnology, National Dong Hwa University, 97401 Hualien, Taiwan
2
Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, 807 Kaohsiung city, Taiwan
3
Neural Regeneration Laboratory, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, 11221 Taipei, Taiwan
4
Center for Neural Regeneration, Neurological Institute, Taipei Veterans General Hospital, 11221 Taipei, Taiwan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2017, 18(7), 1412; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18071412
Submission received: 26 May 2017 / Revised: 19 June 2017 / Accepted: 27 June 2017 / Published: 1 July 2017
(This article belongs to the Special Issue Autophagy at the Intersection of the Immune System and Cancer)

Abstract

:
Accumulated evidence indicates that autophagy is a response of cancer cells to various anti-cancer therapies. Autophagy is designated as programmed cell death type II, and is characterized by the formation of autophagic vacuoles in the cytoplasm. Numerous herbs, including Chinese herbs, have been applied to cancer treatments as complementary and alternative medicines, supplements, or nutraceuticals to dampen the side or adverse effects of chemotherapy drugs. Moreover, the tumor suppressive actions of herbs and natural products induced autophagy that may lead to cell senescence, increase apoptosis-independent cell death or complement apoptotic processes. Hereby, the underlying mechanisms of natural autophagy inducers are cautiously reviewed in this article. Additionally, three natural compounds—curcumin, 16-hydroxycleroda-3,13-dien-15,16-olide, and prodigiosin—are presented as candidates for autophagy inducers that can trigger cell death in a supplement or alternative medicine for cancer therapy. Despite recent advancements in therapeutic drugs or agents of natural products in several cancers, it warrants further investigation in preclinical and clinical studies.

Graphical Abstract

1. Introduction

Cancer is a group of diseases involving out-of-control of cell growth due to the accumulation of defects, or mutations, in their DNA and with an impendence to invade or spread to other parts of the body [1]. In 2015, about 90.5 million people were diagnosed with cancer [2]. About 14.1 million new cases occur each year (not including skin cancer other than melanoma) [3]. Consequently, it causes about 8.8 million (15.7%) human deaths [4]. Anti-cancer drugs including 5-fluorouracil (5-FU), cisplatin, etoposide, paclitaxel, and doxorubicin are commonly used to treat various cancers, such as cisplatin and doxorubicin in ovarian cancer, 5-FU in colon and gastric cancer, paclitaxel and doxorubicin in breast cancer, and etoposide in small-cell lung cancer. However, these chemotherapeutic agents have evident side effects such as nausea, vomiting, loss of appetite, decreased immunity, oral ulcers, and other adverse effects [5]. In general, the anti-cancer drugs, such as cisplatin and doxorubicin favor abnormal triggering via programmed cell death (PCD) such as apoptosis, necrosis, necroptosis, and autophagy in normal cells as well as abolishing inflammation of damaged cells. Remarkably, apoptosis and autophagy are traditionally considered the most prominent cell death or cell death-related mechanisms for anti-cancer drug discovery [6,7].
In contrast to apoptosis, autophagy is a homeostatic process, and is one of the earliest responses to pharmacologically active compounds lead to ultra-structural perturbations; changes in membrane composition, cytoskeleton integrity, alterations of the endoplasmic reticulum, mitochondria and nucleus all result in the formation of morphological alterations. This biological process of autophagy is designated as type II PCD, and has several features, such as removing damaged or excessive proteins, utilization by various types of cells to maintain cellular homeostasis, high conservation across different types of mammalian cells, and is a potentiated cancer type preceding cell death. Moreover, autophagy can relieve tumor cells from nutrient and oxidative stress during a rapid expansion of a cancer. Excessive and sustained autophagy may lead to cell death and tumor shrinkage. Interestingly, autophagy also occurs in cancer cells and exerts anti-survival or pro-survival effects depending on stimuli, nutrients, and context.

2. The Significance of Autophagy in Cancer

Autophagy is an important process in many functions such as maintaining protein quality, presenting antigens, responding to endoplasmic reticulum (ER) stress, and supplying energy [8]. These functions represent a stress in cells so that autophagy usually links to degenerative diseases like dementia and Parkinson’s disease [9]. There are two opposite mechanisms for autophagy in cancer development. Firstly, autophagy can clean up damaged organelles or protein accumulations and further activate programmed cell death when the cell is severely damaged. This anti-cancer mechanism provides another checkpoint to avoid tumorigenesis. On the other hand, autophagy also affords a proportional way of overcoming chemotherapeutic drugs and a low energy state. Autophagy could be activated by ER stress and some reactive oxygen species (ROS), inducing chemotherapeutic drugs like cisplatin and doxorubicin, where it can assist cancer stem cells in cleaning up ROS-damaged proteins [10]. Autophagy, a catabolic degradation process via lysosomes, plays an important role in tumorigenesis and cancer therapy.

2.1. Natural Autophagy Inhibitor from Herbs

Currently, many herbs, including Chinese herbs, have been applied as cancer treatments to complement and act as alternative medicines, supplements, or nutraceuticals to dampen the aforementioned problems. It has been shown in the literature that many anti-cancer natural compounds and extracts could initiate autophagy in tumor cells. As summarized in this paper, tumor suppressive actions of natural product-induced autophagy may lead to cell senescence, provoke apoptosis-independent cell death, and complement apoptotic cell death by robust or target-specific mechanisms. Notably, natural compounds are fundamental for pharmacological treatments, and more than 50% of all anti-cancer drugs are of natural origins, or at least derived from scaffolds present in nature. Emerging research shows that molecules of natural origins are useful for preventive and therapeutic purposes by targeting essential hallmarks and enabling described characteristics. Moreover, natural compounds can change the differentiation status of selected cell types. In the past decade, some autophagy-related inducers and inhibitors have been extensively investigated. Of note, some autophagy inhibitors including 3-methyladenine (3-MA), pepstatin A, bafilomycin A1, and betulinic acid have been well studied, and have been applied as suppressors to examine the modality of autophagy formation in autophagy research [11]. Curiously, other natural autophagy inhibitors have also been elucidated as: (1) Matrine, a natural compound extract used in traditional Chinese medicine, can modulate the maturation process of lysosomal proteases in gastric cancer cell line (SGC-7901) cells [12]; (2) Elaiophylin promotes autophagosome accumulation but blocks autophagic flux by attenuating lysosomal cathepsin activity, resulting in the accumulation of SQSTM1/p62 in various human ovarian cancer cell lines [13]; (3) Oblongifolin C (OC), a natural small molecule compound extracted from Garcinia yunnanensis Hu, is a potent autophagic flux inhibitor. Exposure to OC results in an increased number of autophagosomes and impaired degradation of SQSTM1/p62 [14]; (4) p53 siRNA and epigallocatechin gallate (EGCG) dual therapy leads to the activation of pro-apoptotic genes, the inhibition of pro-survival autophagy and cell network formation [15]; (5) Frondoside A, a triterpenoid saponin with a sugar-steroid structure, is derived from the orange-footed sea cucumber, Cucumaria frondosa and inhibits pro-survival autophagy, a known mechanism of drug resistance in the human urothelial carcinoma cell lines and showed the synergistic activity with cisplatin and gemcitabine [16]; and (6) Rhizochalinin (Rhiz) from the marine sponge Rhizochalina incrustata, a novel sphingolipid-like marine compound is characterized by a unique combination of anti-cancer properties via one scenario of pro-survival autophagy inhibition in the human prostate cancer cells [17]. Thereby, natural products have also been demonstrated as autophagy inducers.

2.2. Natural Autophagy Inducers from Herbs

Conversely, the autophagy cascade may play a regulatory or major role resulting in cell death, particularly when natural products are employed. Apparently, more recent studies have elicited the benefits and molecular mechanisms triggered by natural active components for anti-cancer activity, particularly in the induction of autophagy. Sirolimus, also known as rapamycin, is isolated from the bacterium Streptomyces hygroscopicus, and may be the most famous natural autophagy inducer in autophagy research [18]. The extract of Emblica officinalis (Amla) inhibits ovarian cancer (OC) cell growth in vitro and in vivo, possibly via inhibition of angiogenesis and activation of autophagy in OC [19]. The natural compound lipoic acid (LA) inhibits O (6)-methylguanine-DNA methyltransferase (MGMT) and induces autophagy and subsequently LA enhances the cytotoxic effects of temozolomide in HCT-116 cells [20]. The combination of gossypol and BRD4770 increased LC3-II levels and the autophagosome number in PANC-1 cells. The compound combination appears to act in a BNIP3 (B-cell lymphoma 2 19-kDa interacting protein)-dependent manner, suggesting that these compounds act together to induce autophagy-related cell death in pancreatic cancer cells [21]. The synthesized natural alkaloid berberine derivatives are able to induce autophagy for human colon carcinoma HCT-116 and SW613-B3 cell lines [22]. C-1 acetoxymethyl—a derivative of 7-deoxypancratistatin—and JC-TH-acetate-4 (JCTH-4) are novel compounds capable of selectively inducing apoptosis and autophagy in human colorectal cancer (CRC) cells alone and in combination with tamoxifen (TAM) [23]. Honokiol (HNK), a biphenolic natural compound, significantly inhibits melanoma cell proliferation, viability, clonogenicity, and induces autophagy on melanoma cells [24]. Isobavachalcone (IBC), a natural chalcone compound, induces apoptosis- and autophagy-related cell death in myeloma cells [25]. In numerous breast cancer studies, MCF-7 is frequently used as a cell model for testing the efficacy of anti-cancer agents. Several natural compounds have been shown to be autophagy inducers, such as rottlerin [26], chrysin-organotin based on chrysin [27], betanin/isobetanin from beetroots [28], cucurbitacin B from cucurbitaceous plants [29], and 2-ethyl-3-O-sulphamoyl-estra-1,3,5(10)16-tetraene, a 17-β-estradiol analogue [30]. Additionally, the functions of rottlerin in caspase-3-deficient MCF-7 and caspases-3-transfected MCF-7 are significantly different. In caspase-3-deficient MCF-7, rottlerin acts as an autophagy inducer, while rottlerin acts as an apoptotic activator in caspases-3-transfected MCF-7 [26]. Furthermore, chrysin-organotin [27] and cucurbitacin B [29]. Recently, pancratistatin (PST) analogue, a C-1 acetoxymethyl derivative of 7-deoxypancratistatin (JCTH-4) induced apoptosis and autophagy, and accelerates cell death with combinatorial treatment using time-lapse microscopy in human breast adenocarcinoma and neuroblastoma cells with tamoxifen [31]. Table 1 summarizes our collection of compound names, herb sources, and cancer types to address natural autophagic inducers for anti-cancer activity.

3. Underlying Mechanisms of Natural Compound-Induced Autophagy

Accordingly, the mechanisms of natural product actions including plant-derived anti-cancer drug-induced apoptotic cell death may be intrinsic or extrinsic; p53-dependent or -independent pathways; and caspase-dependent or -independent manner in cancer research. Alternative modes of non-apoptotic cell death by plant-derived anti-cancer drugs are emerging, and mainly comprise autophagy, necrosis-like programmed cell death, mitotic catastrophe, and senescence leading to cell death. Considering that non-apoptotic cell death mechanisms of plant-derived anticancer drugs are less reviewed than the apoptotic ones, this article attempts to focus on such alternative cell death pathways for some representative anti-cancer plant-based compounds in clinical development. The understanding of autophagy-induced mechanisms of natural products could afford deep insight into the possibility of exploiting novel molecular pathways and targets of plant-derived compounds for future cancer therapeutics [45]. Various targets or molecules, including sequestosome-1 (SQSTM1), also known as p62, mammalian target of rapamycin (mTOR), uncoordinated-51-like kinase 1 (ULK1), and autophagy-related protein 12 (Atg12), are commonly applied in observations of autophagy, and the Akt/mTOR/p70 ribosomal protein S6 kinase (p70S6K) and the extracellular signal-regulated kinases 1/2 (ERK1/2) pathways are two major pathways that regulate autophagy induced by nutrient starvation [11,46]. In autophagy, rapamycin specifically targets mTOR and further inhibits the activity of mTOR. The inhibition of mTOR subsequently activates Beclin-1 and down-regulates PI3K-ClassIII to finally induce autophagy [47]. In addition to mTOR inhibition, sirolimus (rapamycin) also regulates microRNA expression, which is involved in tumor growth and drug resistance, including miR-7a, miR-17-92, miR-99a, and miR-100 [48]. Some reports have focused on the inhibitory effect of specific cancer cell types and an understanding of mechanical actions in addition to the abovementioned signal pathways, while several other potential signal pathways have been explored (Table 2). On the other hand, we also highlighted three exemplified natural compounds such as curcumin, 16-hydroxycleroda-3,13-dien-15,16-olide, and prodigiosin as interesting autophagy inducers to discuss more facets in the following section.

3.1. Curcumin

Curcumin is a yellow pigment isolated from turmeric (Curcuma longa L.). Recent study has shown that curcumin provides numerous biological activities including anti-oxidation, antiprotozoal, antimicrobial, immunomodulation, anti-angiogenesis, and antitumor, among others [89]. On anti-tumor activity, curcumin regulates metastasis-relating protein MMP-9 for reducing tumor metastasis [90]. Moreover, curcumin induces G2/M phase arresting in glioma cell via upregulation of p21 and ING4, and further induces apoptosis through up-regulating BAX and down-regulating the Bcl-2 and NF-κB signaling pathway in glioma cells [91]. In autophagy, curcumin induces autophagy via the ERK1/2 signaling pathway (Table 2). In glioblastoma, curcumin induces autophagy in vitro and in vivo, and is less toxic to normal cells, especially in glioma-initiating cells (GICs) [92]. A curcumin derivative—2E,6E-2-(1H-indol-3-yl)methylene)-6-(4-hydroxy-3-methoxybenzylidene)-cyclohexanone (IHCH)—inhibited A549 cell growth and induced the formation of characteristic autophago-lysosomes in a dose- and time-dependent manner [93].

3.2. 16-Hydroxycleroda-3,13-dien-15,16-olide (HCD)

Polyalthia longifolia var. pendula Linn is popularly known as ulta Ashok in India and is widely grown in gardens in tropical and subtropical Asia, such as the southern part of Taiwan, Pakistan, and Sri Lanka, as an evergreen ornamental tree. Many parts of P. longifolia var. pendula Linn tree are important in traditional Indian medicine [94], and encompass various biological functions, such as anti-inflammatory activity in neutrophils, cytotoxicity towards breast cancer cells, and hepatoma cancer cells [95]. The bark has shown to have medicinal value in the treatment of skin diseases, fever, hypertension, diabetes, and helminthiasis [96]. Recently, the chemical components of P. longifolia var. pendula such as diterpenes (clerodane and triterpenes) and aporphine alkaloids have been isolated. Diterpenoids in the hexane extract of P. longifolia seeds has exhibited significant anti-bacterial and anti-fungal activity [97]. Clerodane diterpenes can induce apoptosis in human leukemia HL-60 cells [98]. Moreover, 16-Hydroxycleroda-3,13-dien-15,16-olide (HCD) and its analogs extracted from the bark of P. longifolia have strong anti-inflammatory activity [99]. The enhanced expression of cyto-protective HO-1 factor and anti-inflammatory enzyme in microglia has been reported [100]. Hereby, the induction of apoptosis in leukemia K562 cells via a reduction in histone-modifying enzymes, PRC2-mediated gene silencing, the reactivation of downstream tumor suppressor gene expressions [101], via the PI3K/Akt pathway, and Aurora B resulting in gene silencing and cell cycle disturbance [102]. Our previous studies have demonstrated that HCD could cause apoptosis of two brain cancer cell lines, N18 and C6, via inhibition of focal adhesion kinase (FAK)-related signaling pathway and accordingly induced the autophagic cell death through ROS generation and p38/ERK1/2 signaling pathway activation [103,104]. In oral squamous cell carcinoma, HCD induced autophagy by activating AMPKα and inhibiting Akt, PI3K-ClassIII, and Beclin-1 activity [105].

3.3. Prodigiosin (PG)

Prodigiosin (PG, PubChem CID: 5377753) is an alkaloid and natural red pigment, which is a secondary metabolite of Serratia marcescens and also from actinomycete bacteria [106]. It is characterized by a common pyrrolyl pyrromethene skeleton [107,108]. The biological role of these pigments in the producer organisms remains unclear. Bacterial PGs and their synthetic derivatives have antimicrobial (bactericidal and bacteriostatic) [109,110,111,112], antimalarial [109,110,113], and antitumor [109,110,114,115,116] activities. In addition, they have been shown to be effective apoptotic agents against various cancer cell lines [117], with multiple cellular targets including multi-drug resistant cells with little or no toxicity towards normal cell lines and induce apoptosis in T and B lymphocytes [118,119]. Recently, PG can induce apoptosis in various cancer cells with low toxicity on normal cells and PG-induced apoptosis may ascribe to Bcl-2 and survivin inhibition in colorectal cancer HT-29 cells [120]. Moreover, PG and its structural analogue (compound R) have induced the expression of p53 target genes accompanied by cell-cycle arrest and apoptosis in p53-deficient cancer cells [121]. A previous study has indicated that PG could be effective as a potential inhibitor compound against COX-2 protein, and can be applied as an anti-inflammatory drug [122]. In melanoma cells, PG activates the mitochondrial apoptotic pathway by disrupting an anti-apoptotic member of the BCL-2 family-MCL-1/BAK complexes by binding to the BH3 domain [123]. Additionally, PG exerts nearly identical cytotoxic effects on the resistant cells in comparison to their parental lines to reveal that this pro-apoptotic agent acts independently on the overexpression of multi-drug resistance transporters—MDR1, BCRP, or MRP [124]. Mechanistically, PG engages the IRE1-JNK and PERK-eIF2α branches of the unfolded protein response (UPR) signaling to up-regulate CHOP that, in turn, mediates BCL2 suppression to induce cell death in multiple human breast carcinoma cell lines [125].

4. Perspectives: Natural Autophagy Inducers Potentiate a New Era of Chemotherapeutic Drug Discovery

This review is valuable in terms of clarifying important directions for research on the major role of autophagy inducers resulting in cell death and the underlying mechanisms as seen with numerous natural products. Notably, natural products are an important resource in the discovery of lead compound for anti-cancer drug development, and study on the role of autophagy in the tumor suppressive effects of natural products continues to produce insights into and emerging from difficulties. It is noteworthy that technical variations in detecting autophagy might affect data quality, and study should focus on elaborating the role of natural inducers in deciding cell fate. In vivo study monitoring of natural autophagy inducers in cancer treatment is expected to be a critical effort for the future. Furthermore, the clinically relevant action of autophagy-inducing natural products should be emphasized in translational study.

Author Contributions

Yaw-Syan Fu, May-Jywan Tsai, Henrich Cheng, and Ching-Feng Weng contributed to the literature collection, review and present idea; Shian-Ren Lin and Ching-Feng Weng wrote the paper; all authors have read and approved this manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

3-MA3-Methyladenine
5-FU5-Fluorouracil
HCD16-Hydroxycleroda-3,13-dien-15,16-olide
AMPKAMP-activated protein kinase
CaMKKCalcium/calmodulin-dependent protein kinase kinase
JNKc-Jun N-terminal kinases
eIF2αEukaryotic translation initiation factor 2 subunit 1
EGCGEpigallocatechin gallate
ERKExtracellular signal–regulated kinases
GICsGlioma-initiating cells
HSF1Heat shock factor 1
Hsp70Heat shock protein 70
HK2Hexokinase 2
HNKHonokiol
CRCHuman colorectal cancer
IBCIsobavachalcone
mTORMammalian target of rapamycin
LC3Microtubule-associated protein 1 light chain 3 α
MEKMitogen-activated protein kinase kinase
MGMTO (6)-methylguanine-DNA methyltransferase
TR3Nerve growth factor IB
NF-κBNuclear factor κ-light-chain-enhancer of activated B cells
OCOblongifolin C
p70S6Kp70 ribosomal protein S6 kinase β-1
PSTPancratistatin
PI3KPhosphoinositide 3-kinase
PGProdigiosin
PCDProgramed cell death
AktProtein kinase B
PERKProtein kinase RNA-like endoplasmic reticulum kinase
RhizRhizochalinin
ROSReactive oxygen species
SQSTM1Sequestosome-1
TAMTamoxifen
ULKUncoordinated-51-like kinase
WntWingless/Integrated

References

  1. Bast, R.C. Cardinal manifestations of cancer. In Holland-Frei Cancer Medicine, 9th ed.; Holland, J.F., Hong, W.K., Kufe, D.W., Bast, R.C., Jr., Hait, W.N., Pollock, R.E., Weichselbaum, R.R., Eds.; John Wiley & Sons, Inc.: Hoboken, NJ, USA, 2016; pp. 3–7. [Google Scholar]
  2. Disease, G.B.D.; Injury, I.; Prevalence, C. Global, regional, and national incidence, prevalence, and years lived with disability for 310 diseases and injuries, 1990–2015: A systematic analysis for the global burden of disease study 2015. Lancet 2016, 388, 1545–1602. [Google Scholar]
  3. Stewart, B.W.; Wild, C. World Cancer Report 2014; International Agency for Research on Cancer, WHO Press: Lyon, France; Geneva, Switzerland, 2014. [Google Scholar]
  4. Feigin, V.; GBD 2015 Mortality; Causes of Death Collaborators. Global, regional, and national life expectancy, all-cause mortality, and cause-specific mortality for 249 causes of death, 1980–2015: A systematic analysis for the global burden of disease study 2015. Lancet 2016, 388, 1459–1544. [Google Scholar]
  5. Von Hoff, D.D.; Hanauske, A.R. Preclinical and early clinical development of chemotherapeutic drugs, mechanism-based agents and biologics. In Holland-Frei Cancer Medicine, 8th ed.; Hong, K.W., Kufe, D.W., Bast, R.C., Jr., Hait, W., Pollock, R.E., Weichselbaum, R.R., Holland, J.F., Frei, E., III, Eds.; People’s Medical Publishing House-USA: Shelton, CT, USA, 2010; pp. 568–587. [Google Scholar]
  6. Barry, M.A.; Behnke, C.A.; Eastman, A. Activation of programmed cell death (apoptosis) by cisplatin, other anticancer drugs, toxins and hyperthermia. Biochem. Pharmacol. 1990, 40, 2353–2362. [Google Scholar] [CrossRef]
  7. Tacar, O.; Sriamornsak, P.; Dass, C.R. Doxorubicin: An update on anticancer molecular action, toxicity and novel drug delivery systems. J. Pharm. Pharmacol. 2013, 65, 157–170. [Google Scholar] [CrossRef] [PubMed]
  8. Mizushima, N. Autophagy: Process and function. Genes Dev. 2007, 21, 2861–2873. [Google Scholar] [CrossRef] [PubMed]
  9. Mizushima, N.; Levine, B.; Cuervo, A.M.; Klionsky, D.J. Autophagy fights disease through cellular self-digestion. Nature 2008, 451, 1069–1075. [Google Scholar] [CrossRef] [PubMed]
  10. Choi, K.S. Autophagy and cancer. Exp. Mol. Med. 2012, 44, 109–120. [Google Scholar] [CrossRef] [PubMed]
  11. Klionsky, D.J.; Abdelmohsen, K.; Abe, A.; Abedin, M.J.; Abeliovich, H.; Acevedo Arozena, A.; Adachi, H.; Adams, C.M.; Adams, P.D.; Adeli, K.; et al. Guidelines for the use and interpretation of assays for monitoring autophagy, 3rd ed. Autophagy 2016, 12, 1–222. [Google Scholar] [CrossRef] [PubMed]
  12. Wang, Z.; Zhang, J.; Wang, Y.; Xing, R.; Yi, C.; Zhu, H.; Chen, X.; Guo, J.; Guo, W.; Li, W.; et al. Matrine, a novel autophagy inhibitor, blocks trafficking and the proteolytic activation of lysosomal proteases. Carcinogenesis 2013, 34, 128–138. [Google Scholar] [CrossRef] [PubMed]
  13. Zhao, X.; Fang, Y.; Yang, Y.; Qin, Y.; Wu, P.; Wang, T.; Lai, H.; Meng, L.; Wang, D.; Zheng, Z.; et al. Elaiophylin, a novel autophagy inhibitor, exerts antitumor activity as a single agent in ovarian cancer cells. Autophagy 2015, 11, 1849–1863. [Google Scholar] [CrossRef] [PubMed]
  14. Lao, Y.; Wan, G.; Liu, Z.; Wang, X.; Ruan, P.; Xu, W.; Xu, D.; Xie, W.; Zhang, Y.; Xu, H.; et al. The natural compound oblongifolin C inhibits autophagic flux and enhances antitumor efficacy of nutrient deprivation. Autophagy 2014, 10, 736–749. [Google Scholar] [CrossRef] [PubMed]
  15. Braicu, C.; Pileczki, V.; Pop, L.; Petric, R.C.; Chira, S.; Pointiere, E.; Achimas-Cadariu, P.; Berindan-Neagoe, I. Dual targeted therapy with p53 siRNA and epigallocatechingallate in a triple negative breast cancer cell model. PLoS ONE 2015, 10, e0120936. [Google Scholar] [CrossRef] [PubMed]
  16. Dyshlovoy, S.A.; Madanchi, R.; Hauschild, J.; Otte, K.; Alsdorf, W.H.; Schumacher, U.; Kalinin, V.I.; Silchenko, A.S.; Avilov, S.A.; Honecker, F.; et al. The marine triterpene glycoside frondoside A induces p53-independent apoptosis and inhibits autophagy in urothelial carcinoma cells. BMC Cancer 2017, 17, 93–103. [Google Scholar] [CrossRef] [PubMed]
  17. Dyshlovoy, S.A.; Otte, K.; Alsdorf, W.H.; Hauschild, J.; Lange, T.; Venz, S.; Bauer, C.K.; Bahring, R.; Amann, K.; Mandanchi, R.; et al. Marine compound rhizochalinin shows high in vitro and in vivo efficacy in castration resistant prostate cancer. Oncotarget 2016, 7, 69703–69717. [Google Scholar] [CrossRef] [PubMed]
  18. Vezina, C.; Kudelski, A.; Sehgal, S.N. Rapamycin (AY-22, 989), a new antifungal antibiotic. I. Taxonomy of the producing streptomycete and isolation of the active principle. J. Antibiot (Tokyo) 1975, 28, 721–726. [Google Scholar] [CrossRef] [PubMed]
  19. De, A.; De, A.; Papasian, C.; Hentges, S.; Banerjee, S.; Haque, I.; Banerjee, S.K. Emblica officinalis extract induces autophagy and inhibits human ovarian cancer cell proliferation, angiogenesis, growth of mouse xenograft tumors. PLoS ONE 2013, 8, e72748. [Google Scholar] [CrossRef] [PubMed]
  20. Goder, A.; Nagel, G.; Kraus, A.; Dorsam, B.; Seiwert, N.; Kaina, B.; Fahrer, J. Lipoic acid inhibits the DNA repair protein O6-methylguanine-DNA methyltransferase (MGMT) and triggers its depletion in colorectal cancer cells with concomitant autophagy induction. Carcinogenesis 2015, 36, 817–831. [Google Scholar] [CrossRef] [PubMed]
  21. Yuan, Y.; Tang, A.J.; Castoreno, A.B.; Kuo, S.Y.; Wang, Q.; Kuballa, P.; Xavier, R.; Shamji, A.F.; Schreiber, S.L.; Wagner, B.K. Gossypol and an HMT G9a inhibitor act in synergy to induce cell death in pancreatic cancer cells. Cell Death Dis. 2013, 4, e690. [Google Scholar] [CrossRef] [PubMed]
  22. Guaman Ortiz, L.M.; Croce, A.L.; Aredia, F.; Sapienza, S.; Fiorillo, G.; Syeda, T.M.; Buzzetti, F.; Lombardi, P.; Scovassi, A.I. Effect of new berberine derivatives on colon cancer cells. Acta Biochim. Biophys. Sin. (Shanghai) 2015, 47, 824–833. [Google Scholar] [CrossRef] [PubMed]
  23. Ma, D.; Tremblay, P.; Mahngar, K.; Akbari-Asl, P.; Collins, J.; Hudlicky, T.; McNulty, J.; Pandey, S. A novel synthetic C-1 analogue of 7-deoxypancratistatin induces apoptosis in p53 positive and negative human colorectal cancer cells by targeting the mitochondria: Enhancement of activity by tamoxifen. Investig. New Drugs 2012, 30, 1012–1027. [Google Scholar] [CrossRef] [PubMed]
  24. Kaushik, G.; Venugopal, A.; Ramamoorthy, P.; Standing, D.; Subramaniam, D.; Umar, S.; Jensen, R.A.; Anant, S.; Mammen, J.M. Honokiol inhibits melanoma stem cells by targeting notch signaling. Mol. Carcinog. 2015, 54, 1710–1721. [Google Scholar] [CrossRef] [PubMed]
  25. Zhao, S.; Ma, C.M.; Liu, C.X.; Wei, W.; Sun, Y.; Yan, H.; Wu, Y.L. Autophagy inhibition enhances isobavachalcone-induced cell death in multiple myeloma cells. Int. J. Mol. Med. 2012, 30, 939–944. [Google Scholar] [PubMed]
  26. Torricelli, C.; Salvadori, S.; Valacchi, G.; Soucek, K.; Slabáková, E.; Muscettola, M.; Volpi, N.; Maioli, E. Alternative pathways of cancer cell death by rottlerin: Apoptosis versus autophagy. Evid. Based Complement Alternat. Med. 2012, 2012, 11. [Google Scholar] [CrossRef] [PubMed]
  27. Xuan, H.Z.; Zhang, J.H.; Wang, Y.H.; Fu, C.L.; Zhang, W. Anti-tumor activity evaluation of novel chrysin-organotin compound in MCF-7 cells. Bioorg. Med. Chem. Lett. 2016, 26, 570–574. [Google Scholar] [CrossRef] [PubMed]
  28. Nowacki, L.; Vigneron, P.; Rotellini, L.; Cazzola, H.; Merlier, F.; Prost, E.; Ralanairina, R.; Gadonna, J.P.; Rossi, C.; Vayssade, M. Betanin-enriched red beetroot (Beta vulgaris L.) extract induces apoptosis and autophagic cell death in MCF-7 cells. Phytother. Res. 2015, 29, 1964–1973. [Google Scholar] [CrossRef] [PubMed]
  29. Ren, G.; Sha, T.; Guo, J.; Li, W.; Lu, J.; Chen, X. Cucurbitacin B induces DNA damage and autophagy mediated by reactive oxygen species (ROS) in MCF-7 breast cancer cells. J. Nat. Med. 2015, 69, 522–530. [Google Scholar] [CrossRef] [PubMed]
  30. Nkandeu, D.S.; Mqoco, T.V.; Visagie, M.H.; Stander, B.A.; Wolmarans, E.; Cronje, M.J.; Joubert, A.M. In vitro changes in mitochondrial potential, aggresome formation and caspase activity by a novel 17-β-estradiol analogue in breast adenocarcinoma cells. Cell Biochem. Funct. 2013, 31, 566–574. [Google Scholar] [PubMed]
  31. Ma, D.; Collins, J.; Hudlicky, T.; Pandey, S. Enhancement of apoptotic and autophagic induction by a novel synthetic C-1 analogue of 7-deoxypancratistatin in human breast adenocarcinoma and neuroblastoma cells with tamoxifen. J. Vis. Exp. 2012, 63. [Google Scholar] [CrossRef] [PubMed]
  32. Wang, R.; Xiao, X.; Wang, P.Y.; Wang, L.; Guan, Q.; Du, C.; Wang, X.J. Stimulation of autophagic activity in human glioma cells by anti-proliferative ardipusilloside I isolated from Ardisia pusilla. Life Sci. 2014, 110, 15–22. [Google Scholar] [CrossRef] [PubMed]
  33. Keshmiri-Neghab, H.; Goliaei, B.; Nikoofar, A. Gossypol enhances radiation induced autophagy in glioblastoma multiforme. Gen. Physiol. Biophys. 2014, 33, 433–442. [Google Scholar] [CrossRef] [PubMed]
  34. Dyshlovoy, S.A.; Hauschild, J.; Amann, K.; Tabakmakher, K.M.; Venz, S.; Walther, R.; Guzii, A.G.; Makarieva, T.N.; Shubina, L.K.; Fedorov, S.N.; et al. Marine alkaloid monanchocidin a overcomes drug resistance by induction of autophagy and lysosomal membrane permeabilization. Oncotarget 2015, 6, 17328–17341. [Google Scholar] [CrossRef] [PubMed]
  35. Chan, M.L.; Liang, J.W.; Hsu, L.C.; Chang, W.L.; Lee, S.S.; Guh, J.H. Zerumbone, a ginger sesquiterpene, induces apoptosis and autophagy in human hormone-refractory prostate cancers through tubulin binding and crosstalk between endoplasmic reticulum stress and mitochondrial insult. Naunyn Schmiedebergs Arch. Pharmacol. 2015, 388, 1223–1236. [Google Scholar] [CrossRef] [PubMed]
  36. Kumar, S.; Guru, S.K.; Pathania, A.S.; Kumar, A.; Bhushan, S.; Malik, F. Autophagy triggered by magnolol derivative negatively regulates angiogenesis. Cell Death Dis. 2013, 4, e889. [Google Scholar] [CrossRef] [PubMed]
  37. Kasukabe, T.; Honma, Y.; Okabe-Kado, J.; Higuchi, Y.; Kato, N.; Kumakura, S. Combined treatment with cotylenin A and phenethyl isothiocyanate induces strong antitumor activity mainly through the induction of ferroptotic cell death in human pancreatic cancer cells. Oncol. Rep. 2016, 36, 968–976. [Google Scholar] [PubMed]
  38. Zhang, A.; He, W.; Shi, H.; Huang, X.; Ji, G. Natural compound oblongifolin C inhibits autophagic flux, and induces apoptosis and mitochondrial dysfunction in human cholangiocarcinoma QBC939 cells. Mol. Med. Rep. 2016, 14, 3179–3183. [Google Scholar] [CrossRef] [PubMed]
  39. Kim, W.K.; Pyee, Y.; Chung, H.J.; Park, H.J.; Hong, J.Y.; Son, K.H.; Lee, S.K. Antitumor activity of spicatoside A by modulation of autophagy and apoptosis in human colorectal cancer cells. J. Nat. Prod. 2016, 79, 1097–1104. [Google Scholar] [CrossRef] [PubMed]
  40. Kumar, A.; Singh, B.; Sharma, P.R.; Bharate, S.B.; Saxena, A.K.; Mondhe, D.M. A novel microtubule depolymerizing colchicine analogue triggers apoptosis and autophagy in HCT-116 colon cancer cells. Cell Biochem. Funct. 2016, 34, 69–81. [Google Scholar] [CrossRef] [PubMed]
  41. Ko, H.; Kim, Y.J.; Amor, E.C.; Lee, J.W.; Kim, H.C.; Kim, H.J.; Yang, H.O. Induction of autophagy by dimethyl cardamonin is associated with proliferative arrest in human colorectal carcinoma HCT116 and LOVO cells. J. Cell. Biochem. 2011, 112, 2471–2479. [Google Scholar] [CrossRef] [PubMed]
  42. Guo, S.; Luo, W.; Liu, L.; Pang, X.; Zhu, H.; Liu, A.; Lu, J.; Ma, D.L.; Leung, C.H.; Wang, Y.; et al. Isocryptotanshinone, a STAT3 inhibitor, induces apoptosis and pro-death autophagy in A549 lung cancer cells. J. Drug Target 2016, 24, 934–942. [Google Scholar] [CrossRef] [PubMed]
  43. Wang, W.B.; Feng, L.X.; Yue, Q.X.; Wu, W.Y.; Guan, S.H.; Jiang, B.H.; Yang, M.; Liu, X.; Guo, D.A. Paraptosis accompanied by autophagy and apoptosis was induced by celastrol, a natural compound with influence on proteasome, ER stress and Hsp90. J. Cell. Physiol. 2012, 227, 2196–2206. [Google Scholar] [CrossRef] [PubMed]
  44. De Fatima, A.; Zambuzzi, W.F.; Modolo, L.V.; Tarsitano, C.A.; Gadelha, F.R.; Hyslop, S.; de Carvalho, J.E.; Salgado, I.; Ferreira, C.V.; Pilli, R.A. Cytotoxicity of goniothalamin enantiomers in renal cancer cells: Involvement of nitric oxide, apoptosis and autophagy. Chem. Biol. Interact. 2008, 176, 143–150. [Google Scholar] [CrossRef] [PubMed]
  45. Chen, N.; Karantza, V. Autophagy as a therapeutic target in cancer. Cancer Biol. Ther. 2011, 11, 157–168. [Google Scholar] [CrossRef] [PubMed]
  46. Glick, D.; Barth, S.; Macleod, K.F. Autophagy: Cellular and molecular mechanisms. J. Pathol. 2010, 221, 3–12. [Google Scholar] [CrossRef] [PubMed]
  47. Choi, A.M.; Ryter, S.W.; Levine, B. Autophagy in human health and disease. N. Engl. J. Med. 2013, 368, 651–662. [Google Scholar] [CrossRef] [PubMed]
  48. Granata, S.; Dalla Gassa, A.; Carraro, A.; Brunelli, M.; Stallone, G.; Lupo, A.; Zaza, G. Sirolimus and everolimus pathway: Reviewing candidate genes influencing their intracellular effects. Int. J. Mol. Sci. 2016, 17, 735. [Google Scholar] [CrossRef] [PubMed]
  49. Ueda, J.Y.; Athikomkulchai, S.; Miyatake, R.; Saiki, I.; Esumi, H.; Awale, S. (+)-grandifloracin, an antiausterity agent, induces autophagic PANC-1 pancreatic cancer cell death. Drug Des. Devel. Ther. 2014, 8, 39–47. [Google Scholar] [PubMed]
  50. Rasul, A.; Yu, B.; Khan, M.; Zhang, K.; Iqbal, F.; Ma, T.; Yang, H. Magnolol, a natural compound, induces apoptosis of SGC-7901 human gastric adenocarcinoma cells via the mitochondrial and PI3K/Akt signaling pathways. Int. J. Oncol. 2012, 40, 1153–1161. [Google Scholar] [PubMed]
  51. Ruela-de-Sousa, R.R.; Fuhler, G.M.; Blom, N.; Ferreira, C.V.; Aoyama, H.; Peppelenbosch, M.P. Cytotoxicity of apigenin on leukemia cell lines: Implications for prevention and therapy. Cell Death Dis. 2010, 1, e19. [Google Scholar] [CrossRef] [PubMed]
  52. Deng, Q.; Yu, X.; Xiao, L.; Hu, Z.; Luo, X.; Tao, Y.; Yang, L.; Liu, X.; Chen, H.; Ding, Z.; et al. Neoalbaconol induces energy depletion and multiple cell death in cancer cells by targeting PDK1-PI3-K/Akt signaling pathway. Cell Death Dis. 2013, 4, e804. [Google Scholar] [CrossRef] [PubMed]
  53. Jing, Z.; Fei, W.; Zhou, J.; Zhang, L.; Chen, L.; Zhang, X.; Liang, X.; Xie, J.; Fang, Y.; Sui, X.; et al. Salvianolic acid B, a novel autophagy inducer, exerts antitumor activity as a single agent in colorectal cancer cells. Oncotarget 2016, 7, 61509–61519. [Google Scholar] [CrossRef] [PubMed]
  54. Wu, M.; Lao, Y.; Xu, N.; Wang, X.; Tan, H.; Fu, W.; Lin, Z.; Xu, H. Guttiferone K induces autophagy and sensitizes cancer cells to nutrient stress-induced cell death. Phytomedicine 2015, 22, 902–910. [Google Scholar] [CrossRef] [PubMed]
  55. Wang, M.; Yu, T.; Zhu, C.; Sun, H.; Qiu, Y.; Zhu, X.; Li, J. Resveratrol triggers protective autophagy through the ceramide/Akt/mTOR pathway in melanoma B16 cells. Nutr. Cancer 2014, 66, 435–440. [Google Scholar] [CrossRef] [PubMed]
  56. Kaushik, G.; Ramalingam, S.; Subramaniam, D.; Rangarajan, P.; Protti, P.; Rammamoorthy, P.; Anant, S.; Mammen, J.M. Honokiol induces cytotoxic and cytostatic effects in malignant melanoma cancer cells. Am. J. Surg. 2012, 204, 868–873. [Google Scholar] [CrossRef] [PubMed]
  57. Aoki, H.; Takada, Y.; Kondo, S.; Sawaya, R.; Aggarwal, B.B.; Kondo, Y. Evidence that curcumin suppresses the growth of malignant gliomas in vitro and in vivo through induction of autophagy: Role of Akt and extracellular signal-regulated kinase signaling pathways. Mol. Pharmacol. 2007, 72, 29–39. [Google Scholar] [CrossRef] [PubMed]
  58. Wang, W.J.; Wang, Y.; Hou, P.P.; Li, F.W.; Zhou, B.; Chen, H.Z.; Bian, X.L.; Cai, Q.X.; Xing, Y.Z.; He, J.P.; et al. Induction of autophagic death in cancer cells by agonizing TR3 and attenuating Akt2 activity. Chem. Biol. 2015, 22, 1040–1051. [Google Scholar] [CrossRef] [PubMed]
  59. Sun, Q.; Yao, G.D.; Song, X.Y.; Qi, X.L.; Xi, Y.F.; Li, L.Z.; Huang, X.X.; Song, S.J. Autophagy antagonizes apoptosis induced by flavan enantiomers from Daphne giraldii in hepatic carcinoma cells in vitro. Eur. J. Med. Chem. 2017, 133, 1–10. [Google Scholar] [CrossRef] [PubMed]
  60. Yao, Z.; Xie, F.; Li, M.; Liang, Z.; Xu, W.; Yang, J.; Liu, C.; Li, H.; Zhou, H.; Qu, L.H. Oridonin induces autophagy via inhibition of glucose metabolism in p53-mutated colorectal cancer cells. Cell Death Dis. 2017, 8, e2633. [Google Scholar] [CrossRef] [PubMed]
  61. Park, S.; Fudhaili, A.; Oh, S.S.; Lee, K.W.; Madhi, H.; Kim, D.H.; Yoo, J.; Ryu, H.W.; Park, K.H.; Kim, K.D. Cytotoxic effects of kazinol a derived from Broussonetia papyrifera on human bladder cancer cells, T24 and T24R2. Phytomedicine 2016, 23, 1462–1468. [Google Scholar] [CrossRef] [PubMed]
  62. Park, I.J.; Yang, W.K.; Nam, S.H.; Hong, J.; Yang, K.R.; Kim, J.; Kim, S.S.; Choe, W.; Kang, I.; Ha, J. Cryptotanshinone induces G1 cell cycle arrest and autophagic cell death by activating the AMP-activated protein kinase signal pathway in HepG2 hepatoma. Apoptosis 2014, 19, 615–628. [Google Scholar] [CrossRef] [PubMed]
  63. Yun, S.M.; Jung, J.H.; Jeong, S.J.; Sohn, E.J.; Kim, B.; Kim, S.H. Tanshinone IIA induces autophagic cell death via activation of AMPK and ERK and inhibition of mTOR and p70 S6K in KBM-5 leukemia cells. Phytother. Res. 2014, 28, 458–464. [Google Scholar] [CrossRef] [PubMed]
  64. Law, B.Y.; Wang, M.; Ma, D.L.; Al-Mousa, F.; Michelangeli, F.; Cheng, S.H.; Ng, M.H.; To, K.F.; Mok, A.Y.; Ko, R.Y.; et al. Alisol B, a novel inhibitor of the sarcoplasmic/endoplasmic reticulum Ca2+ ATPase pump, induces autophagy, endoplasmic reticulum stress, and apoptosis. Mol. Cancer Ther. 2010, 9, 718–730. [Google Scholar] [CrossRef] [PubMed]
  65. Amini, A.; Masoumi-Moghaddam, S.; Ehteda, A.; Morris, D.L. Bromelain and N-acetylcysteine inhibit proliferation and survival of gastrointestinal cancer cells in vitro: Significance of combination therapy. J. Exp. Clin. Cancer Res. 2014, 33, 92–107. [Google Scholar] [CrossRef] [PubMed]
  66. Gao, S.; Sun, D.; Wang, G.; Zhang, J.; Jiang, Y.; Li, G.; Zhang, K.; Wang, L.; Huang, J.; Chen, L. Growth inhibitory effect of paratocarpin E, a prenylated chalcone isolated from Euphorbia humifusa wild., by induction of autophagy and apoptosis in human breast cancer cells. Bioorg. Chem. 2016, 69, 121–128. [Google Scholar] [CrossRef] [PubMed]
  67. Rasul, A.; Yu, B.; Zhong, L.; Khan, M.; Yang, H.; Ma, T. Cytotoxic effect of evodiamine in SGC-7901 human gastric adenocarcinoma cells via simultaneous induction of apoptosis and autophagy. Oncol. Rep. 2012, 27, 1481–1487. [Google Scholar] [PubMed]
  68. Li, B.; Takeda, T.; Tsuiji, K.; Wong, T.F.; Tadakawa, M.; Kondo, A.; Nagase, S.; Yaegashi, N. Curcumin induces cross-regulation between autophagy and apoptosis in uterine leiomyosarcoma cells. Int. J. Gynecol. Cancer 2013, 23, 803–808. [Google Scholar] [CrossRef] [PubMed]
  69. She, T.; Qu, L.; Wang, L.; Yang, X.; Xu, S.; Feng, J.; Gao, Y.; Zhao, C.; Han, Y.; Cai, S.; et al. Sarsaparilla (Smilax glabra rhizome) extract inhibits cancer cell growth by S phase arrest, apoptosis, and autophagy via redox-dependent ERK1/2 pathway. Cancer Prev. Res. (Phila) 2015, 8, 464–474. [Google Scholar] [CrossRef] [PubMed]
  70. Yamamoto, M.; Suzuki, S.O.; Himeno, M. Resveratrol-induced autophagy in human U373 glioma cells. Oncol. Lett. 2010, 1, 489–493. [Google Scholar] [CrossRef] [PubMed]
  71. Chun, J.; Kang, M.; Kim, Y.S. A triterpenoid saponin from Adenophora triphylla var. Japonica suppresses the growth of human gastric cancer cells via regulation of apoptosis and autophagy. Tumour Biol. 2014, 35, 12021–12030. [Google Scholar] [CrossRef] [PubMed]
  72. Wang, Z.; Wang, N.; Liu, P.; Chen, Q.; Situ, H.; Xie, T.; Zhang, J.; Peng, C.; Lin, Y.; Chen, J. Microrna-25 regulates chemoresistance-associated autophagy in breast cancer cells, a process modulated by the natural autophagy inducer isoliquiritigenin. Oncotarget 2014, 5, 7013–7026. [Google Scholar] [CrossRef] [PubMed]
  73. Lim, C.B.; Fu, P.Y.; Ky, N.; Zhu, H.S.; Feng, X.; Li, J.; Srinivasan, K.G.; Hamza, M.S.; Zhao, Y. NF-κB p65 repression by the sesquiterpene lactone, helenalin, contributes to the induction of autophagy cell death. BMC Complement Altern. Med. 2012, 12, 93–105. [Google Scholar] [CrossRef] [PubMed]
  74. Garufi, A.; Pistritto, G.; Cirone, M.; D’Orazi, G. Reactivation of mutant p53 by capsaicin, the major constituent of peppers. J. Exp. Clin. Cancer Res. 2016, 35, 136–145. [Google Scholar] [CrossRef] [PubMed]
  75. Afiyatullov, S.S.; Leshchenko, E.V.; Berdyshev, D.V.; Sobolevskaya, M.P.; Antonov, A.S.; Denisenko, V.A.; Popov, R.S.; Pivkin, M.V.; Udovenko, A.A.; Pislyagin, E.A.; et al. Zosteropenillines: Polyketides from the marinederived fungus Penicillium thomii. Mar. Drugs 2017, 15, 46. [Google Scholar] [CrossRef] [PubMed]
  76. Zhang, L.D.; Liu, Z.; Liu, H.; Ran, D.M.; Guo, J.H.; Jiang, B.; Wu, Y.L.; Gao, F.H. Oridonin enhances the anticancer activity of NVP-BEZ235 against neuroblastoma cells in vitro and in vivo through autophagy. Int. J. Oncol. 2016, 49, 657–665. [Google Scholar] [CrossRef] [PubMed]
  77. Filippi-Chiela, E.C.; Villodre, E.S.; Zamin, L.L.; Lenz, G. Autophagy interplay with apoptosis and cell cycle regulation in the growth inhibiting effect of resveratrol in glioma cells. PLoS ONE 2011, 6, e20849. [Google Scholar] [CrossRef] [PubMed]
  78. Rafatian, G.; Khodagholi, F.; Farimani, M.M.; Abraki, S.B.; Gardaneh, M. Increase of autophagy and attenuation of apoptosis by Salvigenin promote survival of SH-SY5Y cells following treatment with H2O2. Mol. Cell. Biochem. 2012, 371, 9–22. [Google Scholar] [CrossRef] [PubMed]
  79. Dimasi, P.; Quintiero, A.; Shelkovnikova, T.A.; Buchman, V.L. Modulation of p-eIF2α cellular levels and stress granule assembly/disassembly by trehalose. Sci. Rep. 2017, 7, 44088–44098. [Google Scholar] [CrossRef] [PubMed]
  80. Zhang, Q.C.; Pan, Z.H.; Liu, B.N.; Meng, Z.W.; Wu, X.; Zhou, Q.H.; Xu, K. Benzyl isothiocyanate induces protective autophagy in human lung cancer cells through an endoplasmic reticulum stress-mediated mechanism. Acta. Pharmacol. Sin. 2017, 38, 539–550. [Google Scholar] [CrossRef] [PubMed]
  81. Yang, J.C.; Lu, M.C.; Lee, C.L.; Chen, G.Y.; Lin, Y.Y.; Chang, F.R.; Wu, Y.C. Selective targeting of breast cancer cells through ROS-mediated mechanisms potentiates the lethality of paclitaxel by a novel diterpene, gelomulide k. Free Radic. Biol. Med. 2011, 51, 641–657. [Google Scholar] [CrossRef] [PubMed]
  82. Sun, Z.L.; Dong, J.L.; Wu, J. Juglanin induces apoptosis and autophagy in human breast cancer progression via ROS/JNK promotion. Biomed Pharmacother. 2017, 85, 303–312. [Google Scholar] [CrossRef] [PubMed]
  83. Chen, L.; Meng, Y.; Sun, Q.; Zhang, Z.; Guo, X.; Sheng, X.; Tai, G.; Cheng, H.; Zhou, Y. Ginsenoside compound K sensitizes human colon cancer cells to TRAIL-induced apoptosis via autophagy-dependent and -independent DR5 upregulation. Cell Death Dis. 2016, 7, e2334. [Google Scholar] [CrossRef] [PubMed]
  84. Xavier, C.P.; Lima, C.F.; Pedro, D.F.; Wilson, J.M.; Kristiansen, K.; Pereira-Wilson, C. Ursolic acid induces cell death and modulates autophagy through JNK pathway in apoptosis-resistant colorectal cancer cells. J. Nutr. Biochem. 2013, 24, 706–712. [Google Scholar] [CrossRef] [PubMed]
  85. Hung, A.C.; Tsai, C.H.; Hou, M.F.; Chang, W.L.; Wang, C.H.; Lee, Y.C.; Ko, A.; Hu, S.C.; Chang, F.R.; Hsieh, P.W.; et al. The synthetic β-nitrostyrene derivative CYT-Rx20 induces breast cancer cell death and autophagy via ROS-mediated MEK/ERK pathway. Cancer Lett. 2016, 371, 251–261. [Google Scholar] [CrossRef] [PubMed]
  86. Fu, Y.; Chang, H.; Peng, X.; Bai, Q.; Yi, L.; Zhou, Y.; Zhu, J.; Mi, M. Resveratrol inhibits breast cancer stem-like cells and induces autophagy via suppressing Wnt/β-catenin signaling pathway. PLoS ONE 2014, 9, e102535. [Google Scholar] [CrossRef] [PubMed]
  87. Huang, H.; Weng, H.; Dong, B.; Zhao, P.; Zhou, H.; Qu, L. Oridonin triggers chaperon-mediated proteasomal degradation of BCR-ABL in leukemia. Sci. Rep. 2017, 7, 41525. [Google Scholar] [CrossRef] [PubMed]
  88. Tan, H.Y.; Wang, N.; Man, K.; Tsao, S.W.; Che, C.M.; Feng, Y. Autophagy-induced RelB/p52 activation mediates tumour-associated macrophage repolarisation and suppression of hepatocellular carcinoma by natural compound baicalin. Cell Death Dis. 2015, 6, e1942. [Google Scholar] [CrossRef] [PubMed]
  89. Amalraj, A.; Pius, A.; Gopi, S.; Gopi, S. Biological activities of curcuminoids, other biomolecules from turmeric and their derivatives—A review. J. Tradit. Complement Med. 2017, 7, 205–233. [Google Scholar] [CrossRef] [PubMed]
  90. Mirzaei, H.; Shakeri, A.; Rashidi, B.; Jalili, A.; Banikazemi, Z.; Sahebkar, A. Phytosomal curcumin: A review of pharmacokinetic, experimental and clinical studies. Biomed. Pharmacother. 2017, 85, 102–112. [Google Scholar] [CrossRef] [PubMed]
  91. Klinger, N.V.; Mittal, S. Therapeutic potential of curcumin for the treatment of brain tumors. Oxid. Med. Cell. Longev. 2016, 2016, 9324085. [Google Scholar] [CrossRef] [PubMed]
  92. Zhuang, W.; Long, L.; Zheng, B.; Ji, W.; Yang, N.; Zhang, Q.; Liang, Z. Curcumin promotes differentiation of glioma-initiating cells by inducing autophagy. Cancer Sci. 2012, 103, 684–690. [Google Scholar] [CrossRef] [PubMed]
  93. Zhou, G.Z.; Xu, S.L.; Sun, G.C.; Chen, X.B. Novel curcumin analogue IHCH exhibits potent antiproliferative effects by inducing autophagy in A549 lung cancer cells. Mol. Med. Rep. 2014, 10, 441–446. [Google Scholar] [PubMed]
  94. Katkar, K.; Suthar, A.; Chauhan, V. The chemistry, pharmacologic, and therapeutic applications of polyalthia longifolia. Pharmacogn. Rev. 2010, 4, 62–68. [Google Scholar] [PubMed]
  95. Chang, F.R.; Hwang, T.L.; Yang, Y.L.; Li, C.E.; Wu, C.C.; Issa, H.H.; Hsieh, W.B.; Wu, Y.C. Anti-inflammatory and cytotoxic diterpenes from formosan Polyalthia longifolia var. Pendula. Planta Med. 2006, 72, 1344–1347. [Google Scholar] [CrossRef] [PubMed]
  96. Mukherjee, P.K.; Wahile, A. Integrated approaches towards drug development from Ayurveda and other indian system of medicines. J. Ethnopharmacol. 2006, 103, 25–35. [Google Scholar] [CrossRef] [PubMed]
  97. Murphy, M.M.; Subramanyam, M.; Bindu, M.H.; Annapurna, J. Antimicrobial activity of clerodane diterpenoids from polyalthia longifolia seeds. Fitoterapia 2005, 76, 336–339. [Google Scholar]
  98. Sari, D.P.; Ninomiya, M.; Efdi, M.; Santoni, A.; Ibrahim, S.; Tanaka, K.; Koketsu, M. Clerodane diterpenes isolated from polyalthia longifolia induce apoptosis in human leukemia HL-60 cells. J. Oleo. Sci. 2013, 62, 843–848. [Google Scholar] [CrossRef] [PubMed]
  99. Wu, T.H.; Cheng, Y.Y.; Chen, C.J.; Ng, L.T.; Chou, L.C.; Huang, L.J.; Chen, Y.H.; Kuo, S.C.; El-Shazly, M.; Wu, Y.C.; et al. Three new clerodane diterpenes from Polyalthia longifolia var. Pendula. Molecules 2014, 19, 2049–2060. [Google Scholar] [CrossRef] [PubMed]
  100. Shih, Y.T.; Hsu, Y.Y.; Chang, F.R.; Wu, Y.C.; Lo, Y.C. 6-hydroxycleroda-3,13-dien-15,16-olide protects neuronal cells from lipopolysaccharide-induced neurotoxicity through the inhibition of microglia-mediated inflammation. Planta Med. 2010, 76, 120–127. [Google Scholar] [CrossRef] [PubMed]
  101. Lin, Y.H.; Lee, C.C.; Chang, F.R.; Chang, W.H.; Wu, Y.C.; Chang, J.G. 16-hydroxycleroda-3,13-dien-15,16-olide regulates the expression of histone-modifying enzymes PRC2 complex and induces apoptosis in CML K562 cells. Life Sci. 2011, 89, 886–895. [Google Scholar] [CrossRef] [PubMed]
  102. Lin, Y.H.; Lee, C.C.; Chan, W.L.; Chang, W.H.; Wu, Y.C.; Chang, J.G. 16-hydroxycleroda-3,13-dien-15,16-olide deregulates PI3K and Aurora B activities that involve in cancer cell apoptosis. Toxicology 2011, 285, 72–80. [Google Scholar] [CrossRef] [PubMed]
  103. Thiyagarajan, V.; Lin, S.H.; Chia, Y.C.; Weng, C.F. A novel inhibitor, 16-hydroxy-cleroda-3,13-dien-16,15-olide, blocks the autophosphorylation site of focal adhesion kinase (Y397) by molecular docking. Biochim. Biophys. Acta. 2013, 1830, 4091–4101. [Google Scholar] [CrossRef] [PubMed]
  104. Thiyagarajan, V.; Sivalingam, K.S.; Viswanadha, V.P.; Weng, C.F. 16-hydroxy-cleroda-3,13-dien-16,15-olide induced glioma cell autophagy via ROS generation and activation of p38 MAPK and ERK-1/2. Environ. Toxicol. Pharmacol. 2016, 45, 202–211. [Google Scholar] [CrossRef] [PubMed]
  105. Cheng, M.F.; Lin, S.R.; Tseng, F.J.; Huang, Y.C.; Tsai, M.J.; Fu, Y.S.; Weng, C.F. The autophagic inhibition oral squamous cell carcinoma cancer growth of 16-hydroxy-cleroda-3,14-dine-15,16-olide. Oncotarget 2017, in press. [Google Scholar]
  106. Laatsch, H.; Kellner, M.; Weyland, H. Butyl-meta-cycloheptylprodiginine—A revision of the structure of the former ortho-isomer. J. Antibiot. (Tokyo) 1991, 44, 187–191. [Google Scholar] [CrossRef] [PubMed]
  107. Chang, C.C.; Chen, W.C.; Ho, T.F.; Wu, H.S.; Wei, Y.H. Development of natural anti-tumor drugs by microorganisms. J. Biosci. Bioeng. 2011, 111, 501–511. [Google Scholar] [CrossRef] [PubMed]
  108. Marchal, E.; Smithen, D.A.; Uddin, M.I.; Robertson, A.W.; Jakeman, D.L.; Mollard, V.; Goodman, C.D.; MacDougall, K.S.; McFarland, S.A.; McFadden, G.I.; et al. Synthesis and antimalarial activity of prodigiosenes. Org. Biomol. Chem. 2014, 12, 4132–4142. [Google Scholar] [CrossRef] [PubMed]
  109. Lapenda, J.C.; Silva, P.A.; Vicalvi, M.C.; Sena, K.X.; Nascimento, S.C. Antimicrobial activity of prodigiosin isolated from Serratia marcescens UFPEDA 398. World J. Microbiol. Biotechnol. 2015, 31, 399–406. [Google Scholar] [CrossRef] [PubMed]
  110. Wang, Y.; Nakajima, A.; Hosokawa, K.; Soliev, A.B.; Osaka, I.; Arakawa, R.; Enomoto, K. Cytotoxic prodigiosin family pigments from pseudoalteromonas sp. 1020R isolated from the pacific coast of Japan. Biosci. Biotechnol. Biochem. 2012, 76, 1229–1232. [Google Scholar] [CrossRef] [PubMed]
  111. Kimyon, O.; Das, T.; Ibugo, A.I.; Kutty, S.K.; Ho, K.K.; Tebben, J.; Kumar, N.; Manefield, M. Serratia secondary metabolite prodigiosin inhibits pseudomonas aeruginosa biofilm development by producing reactive oxygen species that damage biological molecules. Front Microbiol. 2016, 7, 972–987. [Google Scholar] [CrossRef] [PubMed]
  112. Song, Y.; Liu, G.; Li, J.; Huang, H.; Zhang, X.; Zhang, H.; Ju, J. Cytotoxic and antibacterial angucycline- and prodigiosin-analogues from the deep-sea derived streptomyces sp. Scsio 11594. Mar. Drugs 2015, 13, 1304–1316. [Google Scholar] [CrossRef] [PubMed]
  113. Kancharla, P.; Lu, W.; Salem, S.M.; Kelly, J.X.; Reynolds, K.A. Stereospecific synthesis of 23-hydroxyundecylprodiginines and analogues and conversion to antimalarial premarineosins via a rieske oxygenase catalyzed bicyclization. J. Org. Chem. 2014, 79, 11674–11689. [Google Scholar] [CrossRef] [PubMed]
  114. Perez-Tomas, R.; Vinas, M. New insights on the antitumoral properties of prodiginines. Curr. Med. Chem. 2010, 17, 2222–2231. [Google Scholar] [CrossRef] [PubMed]
  115. Sam, S.; Sam, M.R.; Esmaeillou, M.; Safaralizadeh, R. Effective targeting survivin, caspase-3 and microrna-16-1 expression by methyl-3-pentyl-6-methoxyprodigiosene triggers apoptosis in colorectal cancer stem-like cells. Pathol. Oncol. Res. 2016, 22, 715–723. [Google Scholar] [CrossRef] [PubMed]
  116. Yu, C.J.; Ou, J.H.; Wang, M.L.; Jialielihan, N.; Liu, Y.H. Elevated survivin mediated multidrug resistance and reduced apoptosis in breast cancer stem cells. J. BUON 2015, 20, 1287–1294. [Google Scholar] [PubMed]
  117. Papireddy, K.; Smilkstein, M.; Kelly, J.X.; Salem, S.M.; Alhamadsheh, M.; Haynes, S.W.; Challis, G.L.; Reynolds, K.A. Antimalarial activity of natural and synthetic prodiginines. J. Med. Chem. 2011, 54, 5296–5306. [Google Scholar] [CrossRef] [PubMed]
  118. Chang, C.C.; Wang, Y.H.; Chern, C.M.; Liou, K.T.; Hou, Y.C.; Peng, Y.T.; Shen, Y.C. Prodigiosin inhibits gp91phox and iNOS expression to protect mice against the oxidative/nitrosative brain injury induced by hypoxia-ischemia. Toxicol. Appl. Pharmacol. 2011, 257, 137–147. [Google Scholar] [CrossRef] [PubMed]
  119. Dalili, D.; Fouladdel, S.; Rastkari, N.; Samadi, N.; Ahmadkhaniha, R.; Ardavan, A.; Azizi, E. Prodigiosin, the red pigment of Serratia marcescens, shows cytotoxic effects and apoptosis induction in HT-29 and T47D cancer cell lines. Nat. Prod. Res. 2012, 26, 2078–2083. [Google Scholar] [PubMed]
  120. Hassankhani, R.; Sam, M.R.; Esmaeilou, M.; Ahangar, P. Prodigiosin isolated from cell wall of Serratia marcescens alters expression of apoptosis-related genes and increases apoptosis in colorectal cancer cells. Med. Oncol. 2015, 32, 366–374. [Google Scholar] [CrossRef] [PubMed]
  121. Hong, B.; Prabhu, V.V.; Zhang, S.; van den Heuvel, A.P.; Dicker, D.T.; Kopelovich, L.; El-Deiry, W.S. Prodigiosin rescues deficient p53 signaling and antitumor effects via upregulating p73 and disrupting its interaction with mutant p53. Cancer Res. 2014, 74, 1153–1165. [Google Scholar] [CrossRef] [PubMed]
  122. Krishna, P.S.; Vani, K.; Prasad, M.R.; Samatha, B.; Bindu, N.S.; Charya, M.A.; Shetty, P.R. In-silico molecular docking analysis of prodigiosin and cycloprodigiosin as COX-2 inhibitors. Springerplus 2013, 2, 172–178. [Google Scholar] [CrossRef] [PubMed]
  123. Hosseini, A.; Espona-Fiedler, M.; Soto-Cerrato, V.; Quesada, R.; Perez-Tomas, R.; Guallar, V. Molecular interactions of prodiginines with the BH3 domain of anti-apoptotic Bcl-2 family members. PLoS ONE 2013, 8, e57562. [Google Scholar] [CrossRef] [PubMed]
  124. Elahian, F.; Moghimi, B.; Dinmohammadi, F.; Ghamghami, M.; Hamidi, M.; Mirzaei, S.A. The anticancer agent prodigiosin is not a multidrug resistance protein substrate. DNA Cell Biol. 2013, 32, 90–97. [Google Scholar] [CrossRef] [PubMed]
  125. Pan, M.Y.; Shen, Y.C.; Lu, C.H.; Yang, S.Y.; Ho, T.F.; Peng, Y.T.; Chang, C.C. Prodigiosin activates endoplasmic reticulum stress cell death pathway in human breast carcinoma cell lines. Toxicol. Appl. Pharmacol. 2012, 265, 325–334. [Google Scholar] [CrossRef] [PubMed]
Table 1. Natural autophagic inducers from compound names and herb sources evaluated for various types of cancer.
Table 1. Natural autophagic inducers from compound names and herb sources evaluated for various types of cancer.
CompoundsSourcesCancer TypeReference
Rdipusilloside IArdisia pusilla A. DCGlioblastoma multiforme[32]
GossypolCottonGlioblastoma multiforme[33]
Breast cancer[21]
Monanchocidin AMonanchora pulchraGenitourinary malignancies[34]
ZerumboneZingiber zerumbet SmithProstate cancer[35]
Ery5MagnololProstate cancer[36]
Cotylenin A + phenethyl isothiocyanateGarcinia yunnanensis HuPancreatic cancer[37]
Oblongifolin C Cholangiocarcinoma[38]
Spicatoside ALiriope platyphyllaColorectal cancer[39]
Compound 4hColchicum autumnaleColorectal cancer[40]
Dimethyl cardamoninSyzygium samarangense (Blume) Merr. & L.M. Perry (Myrtaceae)Colorectal cancer[41]
JCTH-4pancratistatinColorectal cancer[23]
IsocryptotanshinoneSalvia miltiorrhizaLung cancer[42]
HonokiolMagnolia officinalisMelanoma[24]
IsobavachalconePsoralea corylifoliaMyeloma[25]
CelastrolTripterygium wilfordiiCervical cancer[43]
GoniothalaminGoniothalamus macrophyllus (Blume) Hook. f. & Thomson var. macrophyllusRenal cancer[44]
Table 2. Natural autophagy inducers and their effect on signal transduction pathways.
Table 2. Natural autophagy inducers and their effect on signal transduction pathways.
PathwaysCompoundsCancer TypeReference
Akt(+)-GrandifloracinPancreatic Carcinoma[49]
PI3K/AktMagnololGastric Adenocarcinoma[50]
ApigeninLeukemia[51]
PI3K/Akt/HK2NeoalbaconolNasopharynx cancer[52]
Akt/mTORSalvianolic Acid BColorectal cancer[53]
Guttiferone KUterus Carcinoma[54]
ResveratrolMelanoma[55]
HonokiolMelanoma[56]
Akt/mTOR/p70S6KCurcuminGlioma[57]
TR3/Akt21-(3,4,5-Trihydroxyphenyl) nonan-1-oneMelanoma[58]
AMPK(2R)-kazinol B + Bafilomycin A1Hepatocarcinoma[59]
OridoninColorectal cancer[60]
AMPK/mTORKazinol ABladder cancer[61]
CryptotanshinoneHepatocarcinoma[62]
Tanshinone IIALeukemia[63]
CaMKK/AMPK/mTORAlisol BNasopharynx cancer
Prostate cancer
Beclin 1Bromelain + N-acetylcysteineGastric Adenocarcinoma[64,65]
Paratocarpin EBreast cancer[66]
EvodiamineGastric Adenocarcinoma[67]
Alma extractOvarian cancer[19]
ERK1/2CurcuminUterine cancer Leiomyosarcoma[68]
CurcuminGlioma[57]
Sarsaparilla extractGastric Adenocarcinoma[69]
Breast cancer
Colorectal cancer
p38/ERK1/2ResveratrolGlioma[70]
p38/JNKCompound 1 from Adenophora triphylla var. japonicaGastric Adenocarcinoma[71]
Raf/ERK/p90RSKTanshinone IIALeukemia[63]
miR-25/ULK1IsoliquiritigeninBreast cancer[72]
NF-κBHelenalinOvarian cancer[73]
p53CapsaicinLung cancer[74]
p62Zosteropenillines 7Prostate cancer[75]
p62/LC3-IIOridonin + NVP-BEZ235Neuroblastoma[76]
LC3-II/Atg5/Beclin-1ResveratrolGlioblastoma multiforme[77]
LC3/Atg7/Atg12SalvigeninNeuroblastoma[78]
p-eIF2α TrehaloseNeuroblastoma[79]
PERK/eIF2αBenzyl isothiocyanateLung cancer[80]
ROSGelomulide KBreast cancer[81]
ROS/JNKJuglaninBreast cancer[82]
Ginsenoside KColorectal cancer[83]
Ursolic acidColorectal cancer[84]
ROS/MEK/ERKCYT-Rx20Breast cancer[85]
Wnt/β-cateninResveratrolBreast cancer[86]
HSF1/Hsp70/ubiquintinOridoninLeukemia[87]
RelB/p52BaicalinHepatocarcinoma[88]
AMP-activated protein kinase (AMPK); Calcium/calmodulin-dependent protein kinase kinase (CaMKK); c-Jun N-terminal kinases (JNK); Eukaryotic translation initiation factor 2 subunit 1 (eIF2α); Extracellular signal–regulated kinases (ERK); Heat shock factor 1 (HSF1); Heat shock protein 70 (Hsp70); Hexokinase 2 (HK2); Mammalian target of rapamycin (mTOR); Microtubule-associated protein 1 light chain 3 α (LC3); Mitogen-activated protein kinase kinase (MEK); Nerve growth factor IB (TR3); Nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB); p70 ribosomal protein S6 kinase β-1 (p70S6K); Phosphoinositide 3-kinase (PI3K); Protein kinase B (Akt, PKB); Protein kinase RNA-like endoplasmic reticulum kinase (PERK); Reactive oxygen species (ROS); Sequestosome-1 (SQSTM1); Uncoordinated-51-like kinase (ULK); Wingless/Integrated (Wnt).

Share and Cite

MDPI and ACS Style

Lin, S.-R.; Fu, Y.-S.; Tsai, M.-J.; Cheng, H.; Weng, C.-F. Natural Compounds from Herbs that can Potentially Execute as Autophagy Inducers for Cancer Therapy. Int. J. Mol. Sci. 2017, 18, 1412. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18071412

AMA Style

Lin S-R, Fu Y-S, Tsai M-J, Cheng H, Weng C-F. Natural Compounds from Herbs that can Potentially Execute as Autophagy Inducers for Cancer Therapy. International Journal of Molecular Sciences. 2017; 18(7):1412. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18071412

Chicago/Turabian Style

Lin, Shian-Ren, Yaw-Syan Fu, May-Jywan Tsai, Henrich Cheng, and Ching-Feng Weng. 2017. "Natural Compounds from Herbs that can Potentially Execute as Autophagy Inducers for Cancer Therapy" International Journal of Molecular Sciences 18, no. 7: 1412. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18071412

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop