Skip to main content

Main menu

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Subscribers
    • Advertisers
    • Editorial Board
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics

User menu

  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Anticancer Research
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics
  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart
Anticancer Research

Advanced Search

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Subscribers
    • Advertisers
    • Editorial Board
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Visit us on Facebook
  • Follow us on Linkedin
Review ArticleReviewsR

Natural Products That Target Cancer Stem Cells

JIM MOSELHY, SOWMYALAKSHMI SRINIVASAN, MURALI K. ANKEM and CHENDIL DAMODARAN
Anticancer Research November 2015, 35 (11) 5773-5788;
JIM MOSELHY
Department of Urology, University of Louisville, Louisville, KY, U.S.A.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
SOWMYALAKSHMI SRINIVASAN
Department of Urology, University of Louisville, Louisville, KY, U.S.A.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
MURALI K. ANKEM
Department of Urology, University of Louisville, Louisville, KY, U.S.A.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
CHENDIL DAMODARAN
Department of Urology, University of Louisville, Louisville, KY, U.S.A.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: chendil.damodaran@louisville.edu
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

The cancer stem cell model suggests that tumor initiation is governed by a small subset of distinct cells with stem-like character termed cancer stem cells (CSCs). CSCs possess properties of self-renewal and intrinsic survival mechanisms that contribute to resistance of tumors to most chemotherapeutic drugs. The failure to eradicate CSCs during the course of therapy is postulated to be the driving force for tumor recurrence and metastasis. Recent studies have focused on understanding the unique phenotypic properties of CSCs from various tumor types, as well as the signaling pathways that underlie self-renewal and drug resistance. Natural products (NPs) such as those derived from botanicals and food sources may modulate vital signaling pathways involved in the maintenance of CSC phenotype. The Wingless/Integrated (WNT), Hedgehog, Notch and PI3K/AKT/mTOR pathways have all been associated with quiescence and self-renewal of CSCs, as well as execution of CSC function including differentiation, multidrug resistance and metastasis. Recent studies evaluating NPs against CSC support the epidemiological evidence linking plant-based diets with reduced malignancy rates. This review covers the key aspects of NPs as modulators of CSC fate.

  • Dietary agents
  • cancer stem cells
  • mechanism of action
  • signal transduction pathways
  • review

In spite of an expansive array of available chemotherapeutic (CT) drugs and vastly improved diagnostic technologies, overall the five-year survival rates for all cancer types in the U.S. have only risen from 50% in the 1970s to about 65% today (1). The therapeutic efficacies of most CT drugs are severely hampered by toxicity profiles that limit their life-extending potential. New technologies aimed at improving the performance of CT drugs such as drug delivery systems and chemosensitizers have been slow to show clinical promise in most types of cancer. The failure to significantly improve cancer survival rates suggested that there are more fundamental ‘upstream’ therapeutic targets during the carcinogenesis process than those that respond to CT drugs in differentiated tumor cells. This led to the cancer stem cell (CSC) hypothesis, which predicted that cancer arises in genetically aberrant cells with tumor-initiating properties and stem-like character akin to normal stem cells (2). The viewpoint of cancer as fundamentally a stem cell disease represents an important paradigm shift in our conceptual understanding of carcinogenesis and tumor biology and has ushered in a new era that challenges the dogmatic approaches to cancer cell destruction. Aberrant gene-expression profiles long considered hallmarks of malignancy has been re-evaluated in the context of the CSC with emphasis on genes that regulate self-renewal processes and differentiation programs which are normally tightly regulated in the non-cancerous somatic stem cell. As a result, there is an urgent need to identify compounds that strike targets involved in CSC self-renewal and differentiation programs, respectively.

A straightforward approach to targeting CSC self-renewal and differentiation programs may be the use of chemopreventive natural product (NP) compounds often found in dietary sources. The potential for dietary NPs to guard against malignant transformation of cells is supported by a plethora of epidemiological evidence that shows a strong correlation between consumption of plant-based diets with reduced cancer risk (3). Efforts are underway to identify and characterize the mechanistic pathways by which various NPs inhibit CSC self-renewal and differentiation programs with the ultimate goal of establishing therapeutic regimens that can prevent tumorigenesis or tumor recurrence and improve patient survival.

This review covers the key properties of CSCs, including self-renewal programs, differentiation programs, survival pathways, detoxification mechanisms and other putative mechanisms involved in cancer relapse, that are potentially amenable to therapeutic intervention using NPs. CSC survival and proliferative pathways that include Wnt/β-catenin, Hedgehog, Notch and PI3K/AKT/mTOR are reviewed to highlight potential key targets for therapeutic intervention. The modulation of detoxification mechanisms including multidrug resistance (MDR) in CSCs by NPs is also addressed. The basis of plant-derived NP activity is considered, highlighting the role of evolutionary pressure in the formation of pharmacologically active compounds. We conclude with a description of select NP compounds grouped by chemical class that have shown activity in regulating CSCs either directly or which have exhibited activity in CSC-associated pathways.

Therapeutic Implications of CSCs in Cancer Treatment

The failure to assess CSC response in patients receiving CT may be a factor contributing to tumor recurrence, as suggested by numerous retrospective studies challenging current Response Evaluation Criteria in Solid Tumors (RECIST) guidelines for evaluating the efficacy of cancer therapeutics in clinical trials (4). Specifically, the time-to-progression, progression-free survival and tumor size criteria for evaluating therapeutic efficacy of drug compounds have been individually and collectively challenged for not accurately reflecting patient outcome. As a result, new guidelines for standardizing and evaluating response criteria in established stem cell-associated malignancies are being proposed (5).

There exist a number of intrinsic drug-resistance mechanisms in CSCs which can inactivate cytotoxic drugs, resulting in tumor recurrence (6). Most CT drugs effect apoptotic response in differentiated tumor cells, thereby conferring CSCs with a survival advantage with even greater proliferative potential (7). The result is that tumor cells repopulate with lineage-dependent genotypic and phenotypic alterations, including MDR, that render CT drugs ineffective and lead to more rapid disease progression and poorer prognoses (8). Furthermore, hypoxic stability of CSCs enables them to survive in poorly vascularized microenvironments that challenge drug accessibility to niche networks and contributes to MDR (9).

If indeed the CSC response is a vital criterion for cancer treatment evaluation, there are still no drugs in clinical use that specifically target CSCs. Developing CSC-specific drugs is complicated by the genotypic variability of CSCs and genomic instability of hyperplastic progeny that makes karyotyping of tumor cell populations enormously challenging (10). Targeting CSC self-renewal programs using agents such as NPs ahead of differentiation programs may prevent the development of MDR-associated mutations during tumor growth that lead to refractory response to treatment (11). NPs have shown promising effects in sensitizing CSCs to CT by targeting molecular signaling pathways that modulate stemness properties in a broad spectrum of cancer types (12). Thus, NPs may be combined with conventional CT drugs to form potent dual-target therapies against CSCs and differentiated tumor cells.

Genetic and Molecular Signatures of CSCs

Specification of the CSC genotype and phenotype assists in the identification of potential molecular targets for screening of NPs for therapeutic activity, and may lead to prognostic markers of tumor recurrence and metastasis (13). Methods for identifying CSCs include analysis of surface Cell Adhesion Molecules (CAM) expression (immunophenotype) profiles including CD133, CD44, CD34, CD24 using Fluorescence Activated Cell Sorting (FACS) (14), immunofluorescent detection by confocal microscopy (15), tumor sphere-forming assays (16), Hoechst dye exclusion in side-population (SP) cells (17), detection of enzymatic activity of Aldehyde Dehydrogenase 1 (ALDH1) (e.g. Aldefluor assay) (18), signaling pathway identification, serial colony-forming unit assays (19), migration assays (20) and label-retention assays (21). Identification of CSCs using these methods can reveal differentially expressed stemness markers which are often associated with MDR. Expression of detoxifying enzymes such as ALDH1 (22), increased expression and activity of drug efflux transporters of the MDR-ATP-Binding Cassette transporters (ABC) family such as ABCG2 (BRCP) (23), expression of other anti-apoptotic factors associated with drug resistance including B-Cell Lymphoma (BCL): BCL2, BCL-xl, survivin and Macrophage Inhibitor Cytokine 1 (MIC1) (24) and activation of transcription factors such as Octamer-binding transcription factor-4 (OCT4), SRY (sex determining region Y)-box 2 (SOX2) and NANOG which drive expression of genes regulating pluripotency (25). Amplification of checkpoint activation and efficient repair of DNA and oxidative damage via constitutive activation of Nuclear Factor-kappa B (NF-κB) and expression of CD133/prominin-1 have also been suggested as CSC biomarkers.

Alkaline phosphatase (ALP) is a ubiquitously expressed hydrolase enzyme that de-phosphorylates a variety of substrates and is a marker of many urological disease states and various types of cancer. One of the most-studied of the ALP isoenzymes is the Regan isoenzyme (placental ALP, ALPP) first described by Fishman (26). ALPP is highly expressed in colon cancer (27), renal carcinoma (28) and metastatic melanoma (29). In stem cells, ALP serves as a useful differentiation marker as the enzyme is significantly down-regulated during maturation of cell lineages (30). Liu et al., compared two ovarian CSC populations from human patient distinguished by differentiation capacity in tumor sphere assays (25). The more aggressive CSC phenotype (with sphere-forming capacity) expressed higher levels of ALP than CSCs with lower sphere-forming potential, whereas ALP was not expressed in differentiated progenies lacking stemness properties. Total serum ALP may also facilitate prediction of cancer course and prognosis. Kim et al., performed a retrospective study of 238 patients with bone metastatic prostate cancer and found that total serum ALP levels exhibited a very strong correlation with likelihood of metastasis whereas falling ALP was a positive prognostic sign (31).

Epithelial Mesenchymal Transition (EMT) is a differentiation program in which epithelial cells transform into more motile and fibroblast-like cells with mesenchymal character and leads to reconstitution of tumors in new vascular niches (32). Recent evidence points to molecular links between genes associated with CSC self-renewal programs and EMT-associated transcription factors (33). Enhanced expression of hypoxia factors Hypoxia-Inducible factor-1α (HIF1α) and HIF2α in CSC/progenitor cells frequently occurs during disease progression and metastasis and a molecular link between hypoxic stability and up-regulated stemness-related gene products and pro-survival elements has been established (34). Indeed many of the altered gene products modulated by HIFs in CSCs also factor in the MDR phenotype including ABCB2, BCL2, BCl-xL, survivin and MIC1, transcription factors OCT3/4, SOX2 and NANOG, pro-angiogenic factors such as Vascular Endothelial Growth Factor (VEGF) (35), EMT markers including Epidermal Growth Factor Receptor (EGFR), C-X-C Chemokine receptor type 4 (CXCR4) (36), Zinc finger protein SNAI1 (SNAIL) and TWIST(37), Glucose Transporter 1/2 (GLUT1/2), cell-cycle regulation (38), altered metabolic pathways such as glycolytic enzymes (39), and microRNAs (miRNAs) (40). Thus, identification of CSC gene-expression patterns associated with EMT may identify the mechanisms and specific CSC development and maintenance programs that have become dysregulated and contribute to the metastatic potential of various types of cancer.

Ultimately, in vivo stemness assays such as serial transplantation in animal models are required to corroborate stemness markers identified through in vitro assays, permit evaluation of the effects of stemness markers on tumor properties and allow for screening of NPs for therapeutic activity in CSC populations (14).

Drug Efflux and Detoxification Mechanisms

MDR-ABC transporters. In the CSC model, drug resistance develops when progenitor cells survive drug exposure and differentiate into lineages bearing assorted mutations exhibiting the MDR phenotype. MDR in CSCs can result from a variety of metabolic and transport-associated resistance mechanisms to cytotoxic drugs that include drug efflux, alteration of cellular targets, reduced drug uptake and transport, enhanced drug metabolism and inactivation by enzymes (41). Inhibition of MDR/ABC efflux pumps is a potential therapeutic approach for targeting CSCs. Certain NPs exhibit high potency and specificity for MDR-associated transport proteins such as lung-resistance proteins (LRPs) and ABC transporters which are often overexpressed in CSCs (42). NPs may re-sensitize MDR cells to CT drugs by inhibiting efflux activity of the drug pump, or alternatively by down-regulation of gene expression (43). ABC targets for NPs can include ABCB1, ABCB5, ABCG2 and ABCC1 genes or their products which are commonly differentially expressed in stem-like versus differentiated tumor cell types (44). Factors such as pluripotency and plasticity of differentiated tumor cell phenotype may contribute to considerable ABC gene expression heterogeneity amongst tumor types and must be considered when evaluating prospective NP compounds for efficacy (45).

P-Glycoprotein is a multidrug efflux transporter product of ABCB1 gene expression. Many NPs have inherent ABCB1 inhibition properties likely as a consequence of evolutionary co-development in the same plant species. Adaptation of P-glycoprotein in herbivores to harmful plant natural products resulted in plant production of secondary metabolites that in turn inhibit P-glycoprotein for self-defense. Hence, it can be speculated that P-glycoprotein inhibitors should be present in plants and belies much of the basis of traditional Chinese medicine (46).

ABCG2 is also an important molecular target as this transporter is commonly overexpressed in many human cancer types (47). ABCG2 has been shown to play a critical role in clinical resistance of tumors to anticancer drugs suggesting CSC involvement (48). ABCG2 is a 72-kDa protein whose gene expression is regulated by numerous growth- and survival-linked cellular transcription factors including NF-κB, HIF1α, EGFR, cAMP Response Element Binding Protein (CREB), Signal Transducer and Activator of Transcription (STAT) and others regulatory elements within the ABCG2 promoter region including have been identified at -312/+362 upstream of the transcriptional start site of the human ABCG2 gene (46). Specific NPs targeting ABCG2 and other important resistance proteins are discussed below.

Detoxifying enzymes. The ALDH family of NAD (P)+-dependent enzymes catalyze the oxidation of aldehydes into carboxylic acids. ALDH1 and other isoforms play a critical functional role in drug detoxification in CSCs that contribute to their survival, differentiation and self-renewal in various cancer types (49). Chemosensitization of tumor cells using the established ALDH inhibitor diethylaminobenzaldehyde (DEAB) was shown to sensitize resistant ALDHhi/CD44+ stem-like breast cancer cell lines to CT drugs and radiotherapy (50). However, DEAB is limited to in vitro assays due to its lack of ALDH isoform-specific activity and high toxicity. NPs have been suggested as an alternative for blockade of ALDH1 activity and to sensitize CSCs to CT drugs (51). NPs with purported ALDH-inhibitory activity include citral, gossypol, daidzin and coprine though screening of these compounds for activity in stem-like cells has not been reported. Cytochrome P450 (CYP450) enzymes are an evolutionary conserved superfamily of hemoproteins whose concomitant expression with P-glycoprotein are believed to be an important evolutionary adaptation against potentially toxic substances. CYP450 may play a pivotal role in chemoprevention of malignancies. CYP enzymes can also metabolically activate carcinogenesis by converting procarcinogens to carcinogens. For example, CYP450 isoforms involved in steroid or retinoic acid metabolism could promote or suppress tumour development through hormonal control (52). Dietary consumption of foods enriched in phytochemicals with CYP450-inhibitory activity has long been associated with anticancer and chemopreventative properties (53). Phytochemicals may also exert action by binding to various functionally diverse cellular targets which epigenetically regulate downstream metastasis suppressing genes (54). Dietary phytochemicals include species such as phytoalexins, flavonoids, terpenes, glycosides, carotenoids, phytosterols, and many others.

Genetic variability may also play a role in tumor development as certain CYP allelic variants can influence the bioactivation of carcinogens and serve as biomarkers for cancer susceptibility. For example, CYP1B1 plays an important role in the bioactivation of carcinogens in different cancer types. CYP1B1 is highly expressed in mammary, ovarian and uterine tissue, where it catalyzes the 4-hydroxylation of estradiol, which can generate free radicals that cause cellular damage and may lead to breast and endometrial carcinogenesis (55). Other CYP1 enzymes such as CYP1A1 and CYP1A2 can also be activated by procarcinogens such as polycyclic aromatic hydrocarbons, nitrosamines and arylamines, which are associated with cancer of the bladder, head and neck where polymorphic variants of these enzymes are often expressed. Phytoalexins are parasite-resisting compounds with antimicrobial and antioxidative properties that comprise part of immune systems of many plants species. Salverastrols are phytoalexins that have been shown to have potent activity against CYP1 enzymes (56). Investigation of plant polyphenols as chemopreventative compounds against new molecular and cellular targets including CYP1 enzymes, epidermal stem cells, cellular senescence, epigenetic enzymes involved in carcinogenesis have been suggested and infer a potential cytoprotective role against malignant transformation of stem cells.

Developmental and Maintenance Signaling Pathways

Our understanding of the development and maintenance signaling pathways used by CSCs is confounded by the fact that the origin of the CSC itself is still under considerable speculation. One view is that the development of genetic instability in normal somatic stem cells results in dysregulation of the self-renewal program and imparts tumorigenicity to the cells. Alternatively, CSCs may be derived from differentiated tumor cells through the acquisition of multiple oncogenic mutations of genes that confer stemness properties on cells by way of a phenomenon referred to as plasticity of phenotype (57). Nevertheless, evidence has linked dysregulation of key regulatory stemness signaling pathways common to embryonic development and tissue homeostasis to CSCs (58). Self-renewal and quiescence are two key hallmark properties of CSCs and somatic stem cells alike. In normal stem cells, self-renewal is under tight regulation by transcription factor-mediated pathways that respond to extrinsic growth factor signals as part of the signal transduction process. On the other hand, dysregulation of transcription factor expression or activity in CSCs can promote abnormal self-renewal response that contributes to tumor progression as the neoplastic cells differentiate into more highly proliferative tumor cells. The major mechanistic routes exploited by CSC for these pro-survival signaling and self-renewal are the WNT/β-catenin, Hedgehog, Notch and PI3K/AKT/mTOR pathways, are presented here.

WNT/β-catenin pathway. The WNT/β-catenin signaling pathway modulates cell proliferation, migration and apoptosis in differentiated cancer cells and has been implicated in the maintenance of CSC self-renewal in various cancers (3). WNT/β-catenin signaling is initiated on binding of WNT to Frizzled receptor resulting in cytoplasmic accumulation of β-catenin. In the non-pathological state, β-catenin is sequestered at the cell membrane by the epithelial cell adhesion protein E-cadherin to maintain cell–cell adhesion (59). In the absence of WNT signaling, β-catenin forms a multi-protein complex with glycogen synthase kinase 3β (GSK3β), adenomatous polyposis coli, casein kinase 1α, and axin. GSK3β plays a central role in controlling the activity of the WNT/β-catenin pathway by regulating β-catenin stability and degradation. GSK3β-dependent phosphorylation of β-catenin at Ser33/Ser37/Thr41 restricts its nuclear translocation by inducing ubiquitin-proteasome degradation (60). Activated β-catenin complexes recruit transcriptional co-activators CREB binding protein (CBP) and p300 that regulate expression of downstream WNT target genes.

β-Catenin nuclear translocation and activation of target genes is also associated with other transcription factor complexes including T-cell factor/lymphoid enhancer factor (TCF/LEF). Thus, GSK3β acts as a tumor suppressor by curbing canonical WNT/β-catenin signaling. The WNT/GSK3β/β-catenin signaling axis has been linked with self-renewal of both normal stem cells and CSCs. Suppression of GSK3β activity was shown to be critical for maintenance of murine pluripotent stem cells (61). Inactivating mutations of GSK3β have been associated with faulty CSC development programs in Breakpoint Cluster Region-Abelson (BCR–ABL) chronic myeloid leukemia (CML), corroborating earlier findings that associated nuclear accumulation of β-catenin in BCR–ABL CSCs with progression of the disease. Mouse xenografts of pre-leukemic and leukemic stem cells of mixed-lineage myeloid leukemia (MML) exhibited differential WNT/GSK3β/β-catenin signaling including elevation of β-catenin levels in the tumorigenic CSCs that resulted in enhanced self-renewal, higher tumor relapse rates and poorer survival outcomes than MLL pre-leukemic CSCs (62). Overexpression of β-catenin has been shown to cause resistance of CSCs to radiation and chemotherapy in mouse and rat xenografts, respectively (63). Yang et al., demonstrated that introduction of constitutively active β-catenin (S37Y) in tumorigenic hepatocellular carcinoma cells imparted cisplatin chemoresistance, whereas elimination of β-catenin virtually abrogated the chemoresistant cell population endowed with progenitor-like features (64).

Hedgehog. The Hedgehog family of signaling molecules normally function tissue development by regulating cellular differentiation and proliferation. The Hedgehog pathway is an important cancer target as dysregulation of Hedgehog is found in a wide variety of cancer types. Although several Hedgehog-inhibitory drugs are approved or presently under clinical development, it has been reported that these drugs may actually promote drug-resistant tumors, potentially due to CSC selection (65).

The transcription factor NF-κB is a downstream factor activated by the Sonic Hedgehog (Shh) pathway in pancreatic cancers (66). In human leukemia, CD34+ sub-population exhibits the preponderance of Hedgehog signaling. Su et al., examined the role of Shh in survival and growth of Chronic myeloid Leukemia (CML) progenitor cells (67). Low level of Shh protein was observed in CML bone marrow stromal cells. This was associated with CD34+ progenitor cells that were less sensitive to exogenous Shh peptide, but more sensitive to cyclopamine than CD34− cells. This implies that activation of Shh signaling can occur autonomously in progenitor cells.

Notch pathway. The Notch signaling pathway is a highly evolutionarily conserved component involved in the maintenance of cell diversity and stem-cell self-renewal (68). Four known Notch proteins, Notch1 to Notch4, reside as transmembrane receptors in various stem and progenitor cells. Activation of Notch signaling occurs via binding of Delta-like and Jagged surface ligands, which triggers cleavage by A Disintegrin and Metalloproteinase (ADAM) proteases and secretase proteolytic enzymes (69). The Notch intracellular domain is released in the process and functions as a transcription factor for various genes promoting proliferation including c-Myc, cyclin D1, p21, NF-κB (70). Functional cross-talk between Notch and NF-κB pathways was shown to be active in hyperproliferative colon cancer (2). In a recent intriguing study, highly regenerative prostate luminal epithelial progenitor cells were shown to exhibit enhanced proliferation via Notch signaling that promoted metastatic effects by inhibition of anoikis (71). Markstein et al. (72) developed a systematic method to screen small molecules against CSC populations using a Drosophila model. Drosophila intestinal stem cells are multipotent and give rise to cell types similar to mammalian CSC. Most importantly, both mammalian CSC and Drosophila stem cells act through evolutionarily conserved pathways including EGFR, HIPPO, AKT and Janus Kinase (JAK)-STAT, which enable us to screen CT agents using drosophila as a model.

PI3K/AKT/mTOR and crosstalk. The PI3K/AKT/mTOR axis is a central intracellular signaling pathway regulating cellular apoptotic function. PI3K/AKT/mTOR plays a key role in many cancers owing to the high frequency of mutation to the tumor suppressor gene phosphatase and tensin homolog (PTEN), which regulates PI3K signaling. Unrepressed PI3K signaling results in constitutive activation of downstream pathway components that include the AKT and mTOR kinases and drive a host of cellular pro-survival adaptations (73). PTEN loss has been shown to mediate AKT activation and increase stemness properties of CSC populations in prostate cancer (74). Furthermore, crosstalk between PI3K/AKT and other pro-survival as well as mitogenic pathways has been shown to drive cancer growth (75). Inactivation of GSK3β by AKT may result in down-regulation of WNT, Hedgehog, and Notch signaling pathways (76). Crosstalk has been demonstrated to regulate activity of mammary stem/progenitor cells through GSK3β abrogation and β-catenin activation of downstream events (77). Crosstalk between tyrosine kinase receptors, GSK3β and Bone Morphogenetic Protein 2 (BMP2) signaling during osteoblastic differentiation of human mesenchymal stem cells was observed (78). It was suggested that PI3K signaling together with nuclear accumulation of β-Catenin is necessary to fully activate canonical WNT signaling in colon cancer and correlated with a high risk of distant metastasis in patients with colon cancer (79).

NPs as Modulators of CSC Pathways

Modulation of ABC transporters. NPs that have been evaluated directly in assays of activity in CSCs, or have shown strong evidence of activity against specific targets in CSC renewal or differentiation-associated pathways are shown in Table I and Figure 1.

Many phytochemicals including carotenoids, capsanthin and capsorubin, lycopene, lutein, antheraxanthin, violaxanthin and flavonoids (rotenone, chrysin, phloretin and sakuranetin) and various traditional chinese medical herbs, exhibit ABCB1- and ABCG2-modulating activity (80). The phytoalexin, allixin, isolated from garlic has shown anti-tumor-promoting effects in vivo, inhibiting skin tumor formation by Tissue Plasminogen Activator (TPA) in 7,12-Dimethylbenz[a] anthracene (DMBA)-initiated mice.

View this table:
  • View inline
  • View popup
Table I.

Natural products with reported activity towards cancer stem cells and related mechanisms.

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

A schematic representation of molecular signaling of cancer stem cell (CSC) and the effect of natural compounds on these molecular targets. ABC; Adenosine triphosphate (ATP)-binding cassette, ABCB1/G2; ABC subfamily B/G member ½, AP1; activator protein 1, BAD; BCL2-Associated Agonist Of Cell Death, APC; Adenomatous Polyposis Coli, BCLXL; B-cell lymphoma-extra-large, BNIP3; BCL2/Adenovirus E1B 19kDa Interacting Protein 3, CSC (s); cancer stem cell (s), DAPK1; death-associated protein kinase 3, DLK1; Delta-Like 1 Homolog, GRB2; Growth Factor Receptor-Bound Protein 2, GSKb; Glycogen Synthase Kinase 3 Beta, Hh; Hedgehog,FRs2: Fibroblast Growth Factor Receptor Substrate 2, FN1; Fibronectin 1, HBP1; HMG-Box Transcription Factor 1, HSP90; heat shocking protein-90, IKB; Inhibitor Of Kappa Light Polypeptide Gene Enhancer In B-Cells, JUN; Jun Proto-Oncogene, LGR4; Leucine-Rich Repeat Containing G Protein-Coupled Receptor 4, MAML1; Mastermind-Like 1, MAPK; Mitogen-Activated Protein Kinase 1, MDR, multi-drug resistance; OCT-1; octamer-binding transcription factor 1,p53: protein 53 PI3K; Phosphatidylinositol-4,5-Bisphosphate 3-Kinase, Catalytic Subunit Alpha, PTEN; Phosphatase And Tensin Homolog RAS; Rat Sarcoma Viral Oncogene Homolog, RIN1; Ras And Rab Interactor 1, RAF1; Raf-1 Proto-Oncogene, Serine/Threonine Kinase, STAT; Signal transducer and activator of transcription; SMO; Smoothened, Frizzled Class Receptor, SUFU; Suppressor Of Fused Homolog, SOS; Son Of Sevenless Homolog,SMO; Smoothened, Frizzled Class Receptor, TNF; Tumor Necrosis Factor, TRAIL; tumor necrosis factor-related apoptosis induced ligand, WNT; Wingless-Type MMTV Integration Site Family.

An array of dietary phytochemicals including phenolic acids, flavonoids, triterpenes and other dietary phytochemicals were tested in cell- and membrane-based transport inhibition assays of ABCG2. The non-flavonoid phytochemicals berberine, celastrol, ellagic acid, limonin, oleanolic acid, sinapic acid and ursolic acid demonstrated significant inhibition of ABCG2-mediated transport. Chrysoeriol, laricitrin, myricetin 3′,4′,5′-trimethylether, pinocembrin, quercitrin, tamarixetin, tricetin and tricetin 3′,4′,5′-trimethylether were also identified as novel flavonoid ABCG2 inhibitors (81). Interestingly, cannabinoids have been shown to be effective inhibitors of ABCG2 with IC50 of 1.7 μM reported for tetrahydrocannabinol (82).

Tian et al., investigated the interaction between ABCG2 and several bisbenzylisoquinoline alkaloid compounds (83). Using the LLC-PK1/BCRP cell model, the authors showed that the alkaloid compounds liensinine and dauricine were substrates of ABCG2, corroborating their results from molecular docking analysis. On the basis of intracellular accumulation of these compounds and substrate interaction at ABCG2 sites of hotspot specificty, the authors concluded that ABCG2 could mediate the excretion of liensinine and dauricine. The ABCG2 antagonist action of these compounds could potentially be used in chemosensitizing CSCs to toxic action of various CTs.

Salinomycin

Salinomycin is a polyether ionophore antibiotic isolated from Streptomyces albus, that has been shown to kill CSCs in different types of human cancer, most likely by interfering with ABC drug transporters, the WNT/β-catenin signaling pathway, and other CSC pathways. It is not clear by which mechanisms salinomycin eliminates CSCs, but it is important to note that salinomycin, in combination with cytotoxic drugs was much more effective in eradicating human cancer in mouse xenograft than CT drug alone (84). This reinforces the notion that efficient cancer therapy should target all cancer cell populations, including CSCs, more differentiated progenitors, and bulk tumor cells that may be achieved by combining CSC-targeting agents against new molecular and cellular targets with conventional cytotoxic modalities such as CT drugs and radiation.

Isothiocyanates. Cruciferous vegetables such as broccoli and sprouts contain isothiocyanates including sulforaphane, that are enzymatically hydrolyzed from glucosinolates (85). Dietary isothiocyanate sulforaphane was shown to alter phosphorylation of several kinases and their substrates including GSK3, JNK and Protein Kinase C (PKC) (86). Sulforaphane was also shown to target breast CSCs and was associated with reduction of AKT, phospho-GSK3β and β-catenin (87). Sulforaphane showed strong chemopreventative activity in challenges posed by chemically induced cancers in animal models (88). Kallifatidis et al., reported that sulforaphane could abrogate the resistance of pancreatic CSCs to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) by interfering with TRAIL-activated NF-κB signaling (89). Hence, they concluded that combination of sulforaphane with TRAIL would be a promising strategy for targeting pancreatic CSCs. Sulforaphane has also been reported to down-regulate NF-κB function in prostate and colon cancer cells (90). Expression of WNT-9a was shown to be significantly suppressed in ApcMin/+ mouse adenomas treated with sulforaphane (91).

Sulforaphane. Sulforaphane was shown to induce down-regulation of β-catenin in human cervical carcinoma HeLa and hepatocarcinoma HepG2 cells, although no direct effects on CSCs have been demonstrated to date (92). Sulforaphane has been reported to down-regulate AKT pathway in ovarian, prostate, and colorectal cancers and it was demonstrated that PI3K/AKT pathway regulates breast stem CSC by promoting β-catenin downstream events through phosphorylation of GSK3β (77). In studies, non-obese diabetic/severe combined immunodeficiency (NOD/SCID) mice inoculated with tumor cells derived from sulforaphane-treated primary xenografts failed to develop tumor re-growth up to 33 days, whereas control tumor cells quickly gave rise to large tumors (87). This was associated with down-regulation of WNT/β-catenin self-renewal pathway in sulforaphane-treated breast cancer cells.

Isoflavones. Consumption of dietary soy isoflavones including genistein has been shown to be associated with reduced risk of breast cancer in recent studies (93). The soy isoflavone genistein has been shown to have a potent antiproliferative effect on various types of cancer (94). Soy isoflavones were found to inhibit the phosphorylation of AKT and Forkhead Box O 3a (FOXO3a), enhance the expression of GSK3β, leading to increased phosphorylation of β-catenin in prostate cancer cells (95). Genistein was reported to attenuate β-catenin-mediated expression of WNT downstream target genes in mammary epithelial cells by up-regulating E-cadherin (96). Montales et al., showed repression of mammosphere formation of human breast cancer cells by soy isoflavone genistein and blueberry polyphenolic acids illustrating the potential of diet-mediated targeting of CSC/progenitor cells (97). Ning et al., investigated the activity of a genistein derivative and observed specific inhibition of ovarian CSCs mediated by down-regulation of FOXM1 (98).

Polyphenols. Polyphenolic catechins including epigallocatechin-3-gallate (EGCG) found in green tea extracts have demonstrated chemopreventative activity against various types of cancers (99). EGCG has been shown to inhibit NF-κB activity, Mitogen-Activated Protein Kinase (MAPK) pathway, activator protein-1 (AP1) activity, and EGFR-mediated downstream signaling pathways, etc. (100). Several mechanisms of EGCG may be operative in WNT inhibition. EGCG was shown to block WNT signaling by stabilizing mRNA of WNT inhibitor HMG-box transcription factor 1 (HBP1), resulting in reduction of breast cancer cell proliferation and invasiveness (101). Other reports have shown EGCG to activate endogenous WNT inhibitor protein Secreted frizzled-related protein 1 (SFRP1) in hepatoblastomas (102). Adenomas isolated from EGCG-treated ApcMin/+ mice, the benchmark transgenic model for recapitulating human colon cancer, showed inhibition of tumorigenesis, as evidenced by decreased nuclear import of β-catenin, lower pAKT levels and reduced adenoma size (103). EGCG was also shown to inhibit the chaperoning function of heat-shock protein 90 in pancreatic cancer cells, thereby down-regulating AKT signaling (104). EGCG was reported to negatively regulate NF-κB activity by inhibiting its ATP or Interleukin 1β (IL1β) activation (105). Shh expression and Hedgehog signaling pathway are still being evaluated as targets of EGCG, however, numerous reports have shown the ability of EGCG to specifically modulate CSCs in a variety of cancer types (106). Eid et al., investigated the cytotoxicity of various phytochemicals, including phenolics (EGCG and thymol), terpenoids (menthol, aromadendrene, β-sitosterol-O-glucoside, and β-carotene) and alkaloids (glaucine, harmine, and sanguinarine), alone or in combination with the cytotoxic monodesmosidic steroidal saponin digitonin in Caco-2, MCF-7, CEM/ADR5000, and CCRF-CEM cells (107). Digitonin was combined together with combinations of phenolics, terpenoids, and alkaloids and exhibited synergistic therapeutic effects even in MDR cells such as CEM/ADR5000 cells expressing high levels of ABCB1.

Saponin. Saponins are glycosides found in abundance in various plant species. Saponins from the roots of Platycodon grandiflorum suppressed Transforming Growth Factor β1 (TGFβ1)-induced epithelial-mesenchymal transition via repression of PI3K/AKT, ERK1/2 and SMAD2/3 pathway in human lung carcinoma A549 cells (35). SB365, a saponin D-derivative obtained from Pulsatilla koreana suppressed tumor sphere formation, reduced HIF1α and VEGF expression, and induced apoptosis of pancreatic cancer cells (108).

Quercetin. Quercetin is a polyphenol flavanoid found in various fruits, vegetables, leaves and grains. Wang et al., investigated the cooperative effects of administration of the querecetin and green tea in human prostate cancer xenografts in mice (109). Inhibition of tumor growth was correlated with effects of administration of the two compounds with numerous effects on important stem cell markers including ABCC1. Quantitative Real time-Polymerase Chain Reaction (qTR-PCR) analysis of catechol-O-methyltransferase and ABCC1 gene expression revealed cooperative down-regulation of these genes by co-administration of querecetin/green tea.

Curcumin. Curcumin derived from the spice turmeric (Curcuma longa) has been reported to modulate multiple signaling pathways in a variety of cancer types. Curcumin blocked the pro-inflammatory transcription factor NF-κB via down-regulation of 26S-driven degradation of IκBα in Human Papilloma Virus (HPV)-associated cervical cancer cells (110). Esophageal squamous carcinoma is an aggressive cancer with poor prognosis due to the presence of CSCs (111). Almanaa et al., investigated the effects of curcumin on CSCs of human esophageal squamous carcinoma cells lines (112). ALDH1A1, CD44+ and NF-κB were used to compare CSC sub-populations within original cell lines surviving up to 60 μM curcumin treatment. Curcumin-surviving cell lines showed significant loss in ALDHA1+ and CD44+ cell populations, indicating selective targeting of CSC population by the natural product. Furthermore, the tumor sphere-forming capability of YES-2 (human squamous carcinoma) cell lines surviving curcumin treatment was significantly lower than that of the untreated parental cell line.

Clearly, the ability of curcumin to target CSC populations within a tumor makes it an attractive candidate for combination therapy with established CTs in which tumor recurrence suggests ineffective eradication of CSC sub-populations. Yu et al., combined curcumin with either 5-fluorouracil or oxaliplatin, and observed a significant inhibition of CSC population which was confirmed by CSC markers such as CD44 and CD166 in colon cancer cell lines that are resistant to CT agents (113). Similarly, curcumin in combination with the CT agent dasatinib down-regulated ALDH, CD44, CD133 and CD166 markers at mRNA level in chemoresistant colon cancer (12).

Other polyphenols. Gingerols are found in abundance in ginger, and differ in their chemical structure by the length of unbranched alkyl chains. 6-Gingerol has been purported to exert cancer chemopreventative effects by influencing various steps of the metastatic process (114). MDA-MB-231 cells treated with 6-gingerol showed reduction in matrix metalloproteases MMP2 and MMP9 and pointed towards favorable response to the drug in processes involved in cell adhesion, migration, invasion and proliferation (115). Growth arrest and apoptosis of human colorectal cancer cells treated with 6-gingerol, was also shown to occur by multiple mechanisms including protein degradation as well as β-catenin, PKC and GSK3β pathways (116). Tests of 6-gingerol on CSC sub-populations appear warranted. Shogaol (6-shogaol), is a constituent of ginger. A derivative of shogaol, 3-Ph-3-SG, was shown to inhibit Phorbol 12-myristate 13-acetate (PMA)-activated MMP9 expression in MDA-MB-231 and MCF-7 breast carcinoma cells (117). Invasion was suppressed by exertion of cytoprotective effects through modulation of NF-κB and Nuclear respiratory Factor 2 - Kelch-like ECH-associated protein 1 (NRF2-KEAP1) signaling pathways.

The terpenoid phenolic aldehyde, gossypol, is generally isolated as a racemic mixture from cottonseed plant (Gossypium). The tumor-inhibitory properties of gossypol in CSC-relevant pathways have been ascribed to blocking of the anti-apoptotic functions of BCL-2 and BCL-xL, (118), p53 induction (119) and even VEGF angiogenesis (120), although it appears its mechanism of action is more consistent with its inhibition of dehydrogenase enzymes, including various ALDH isozymes (51). Gossypol acts as a noncompetitive inhibitor of ALDH and was shown to be more selective for ALDH3 than ALDH1 and ALDH2 isozymes, perhaps reflecting on the paucity of its screening as a prospective CSC inhibitor. (121). Sabutoclax, a selective MCL1 antagonist derived from gossypol inhibited tumorigenesis in transgenic mouse and human xenograft of prostate cancer (122).

Psoralidin, a natural phenolic compound found in the seeds of Psoralea corylifolia, was shown to induce growth arrest of ALDH+ breast CSCs derived from MDA-MB-231 by down-regulation of Notch1 (123).

Vinca alkaloids. Vinorelbine, a potent vinca alkaloid, was suggested as a possible treatment against breast CSCs (124). A screen of colorectal carcinomas obtained from patients treated with vinorelbine, however, indicated remission of tumors and relapse of cancer cells on the basis of high proliferative index, and overexpression of CSC markers NANOG, BM1, CD44, CD133 and Death receptor 5 (DR5). It was suggested that the Notch signaling pathway and mTOR signaling may be responsible for these effects (125).

Sesquiterpene lactones. Parthenolide is a sesquiterpene lactone of the germacranolide class which occurs naturally in the plant feverfew (Tanacetum parthenium). Guzman et al., investigated activity of parthenolide against CSCs derived from AML and CML (126). Parthenolide induced significant apoptosis in primary human AML cells and blast crisis CML cells while sparing normal hematopoietic cells. In NOD/SCID mice, parthenolide exhibited preferential targeting of AML progenitor and stem cell populations when compared to the established chemoptherapeutic drug, cytosine arabinoside. Inhibition of NF-κB, proapoptotic activation of p53, and increased reactive oxygen species were factors cited by the authors in parthenolides activity against these CSCs. (−)-Galiellalactone is a fungal metabolite that can be isolated from ascomycetes Galiella rufa strain. Hellsten et al., showed that galiellalactone inhibited CSC-like ALDH+ DU145 and LNCaP prostate cancer cell proliferation and tumorigenicity in mouse xenografts by targeting JAK/STAT phosphorylation of STAT3 (127).

Polyynes

Falcarinol and falcarindiol are polyacetylenes derived from carrot, parsley and devil's club (Oplopanax horridus) which is related to ginseng. Yoshida et al., demonstrated that falcarindiol can inhibit GSK3β in an ATP noncompetitive manner (128). Falcarinol derivatives were shown by Tan et al., to be potent inhibitors of breast cancer resistance protein (ABCG2) (81). The evaluation of these compounds against CSCs represents an intriguing future direction for CSC research.

Conclusion and Future Perspectives

Conventional therapeutics including chemotherapy and radiation therapy have demonstrated efficacy against many differentiated tumor cell types, but exhibit poor performance against CSC-specific targets, leading to tumor regrowth and metastasis. Many NPs, including those found in common foodstuffs have demonstrated ability to modulate pathways responsible for CSC function and inhibition. As knowledge of molecular biology and properties of CSCs is gleaned for various tumor types, more NP inhibitors of CSCs may be identified and tested in combination with each other and in formulations with conventional CT drugs to form more potent therapeutic treatment strategies than those currently available.

Acknowledgements

This work was supported by the R01CA140605 and R01CA138797.

Footnotes

  • Conflicts of Interest

    The Authors indicate no potential conflict of interest.

  • Received July 16, 2015.
  • Revision received September 11, 2015.
  • Accepted September 21, 2015.
  • Copyright© 2015 International Institute of Anticancer Research (Dr. John G. Delinassios), All rights reserved

References

  1. ↵
    Http://seer.Cancer.Gov/index.Html. NCI, 2011.
  2. ↵
    1. Ahmed I,
    2. Roy B,
    3. Chandrakesan P,
    4. Venugopal A,
    5. Xia L,
    6. Jensen R,
    7. Anant S,
    8. Umar S
    : Evidence of functional cross talk between the notch and nf-kappab pathways in nonneoplastic hyperproliferating colonic epithelium. Am J Physiol Gastrointest Liver Physiol 304(4): G356-370, 2013.
    OpenUrlAbstract/FREE Full Text
  3. ↵
    1. Akiyama T
    : Wnt/beta-catenin signaling. Cytokine Growth Factor Rev 11(4): 273-282, 2000.
    OpenUrlCrossRefPubMed
  4. ↵
    1. Eisenhauer EA,
    2. Therasse P,
    3. Bogaerts J,
    4. Schwartz LH,
    5. Sargent D,
    6. Ford R,
    7. Dancey J,
    8. Arbuck S,
    9. Gwyther S,
    10. Mooney M,
    11. Rubinstein L,
    12. Shankar L,
    13. Dodd L,
    14. Kaplan R,
    15. Lacombe D,
    16. Verweij J
    : New response evaluation criteria in solid tumours: Revised recist guideline (version 1.1). Eur J Cancer 45(2): 228-247, 2009.
    OpenUrlCrossRefPubMed
  5. ↵
    1. Savona MR,
    2. Malcovati L,
    3. Komrokji R,
    4. Tiu RV,
    5. Mughal TI,
    6. Orazi A,
    7. Kiladjian JJ,
    8. Padron E,
    9. Solary E,
    10. Tibes R,
    11. Itzykson R,
    12. Cazzola M,
    13. Mesa R,
    14. Maciejewski J,
    15. Fenaux P,
    16. Garcia-Manero G,
    17. Gerds A,
    18. Sanz G,
    19. Niemeyer CM,
    20. Cervantes F,
    21. Germing U,
    22. Cross NC,
    23. List AF,
    24. Group MMIW
    : An international consortium proposal of uniform response criteria for myelodysplastic/myeloproliferative neoplasms (mds/mpn) in adults. Blood 125(12): 1857-1865, 2015.
    OpenUrlAbstract/FREE Full Text
  6. ↵
    1. Colak S,
    2. Medema JP
    : Cancer stem cells – important players in tumor therapy resistance. FEBS J 281(21): 4779-4791, 2014.
    OpenUrlCrossRefPubMed
  7. ↵
    1. Zheng X,
    2. Cui D,
    3. Xu S,
    4. Brabant G,
    5. Derwahl M
    : Doxorubicin fails to eradicate cancer stem cells derived from anaplastic thyroid carcinoma cells: Characterization of resistant cells. Int J Oncol 37(2): 307-315, 2010.
    OpenUrlPubMed
  8. ↵
    1. Yokoyama Y,
    2. Sato S,
    3. Fukushi Y,
    4. Sakamoto T,
    5. Futagami M,
    6. Saito Y
    : Significance of multi-drug-resistant proteins in predicting chemotherapy response and prognosis in epithelial ovarian cancer. J Obstet Gynaecol Res 25(6): 387-394, 1999.
    OpenUrlPubMed
  9. ↵
    1. Crowder SW,
    2. Balikov DA,
    3. Hwang YS,
    4. Sung HJ
    : Cancer stem cells under hypoxia as a chemoresistance factor in breast and brain. Curr Pathobiol Rep 2(1): 33-40, 2014.
    OpenUrlPubMed
  10. ↵
    1. Grichnik JM
    : Genomic instability and tumor stem cells. J Invest Dermatol 126(6): 1214-1216, 2006.
    OpenUrlCrossRefPubMed
  11. ↵
    1. Angeloni V,
    2. Tiberio P,
    3. Appierto V,
    4. Daidone MG
    : Implications of stemness-related signaling pathways in breast cancer response to therapy. Semin Cancer Biol 31: 43-51, 2015.
    OpenUrlPubMed
  12. ↵
    1. Nautiyal J,
    2. Kanwar SS,
    3. Yu Y,
    4. Majumdar AP
    : Combination of dasatinib and curcumin eliminates chemo-resistant colon cancer cells. J Mol Signal 6(7), 2011.
  13. ↵
    1. Dingli D,
    2. Michor F
    : Successful therapy must eradicate cancer stem cells. Stem Cells 24(12): 2603-2610, 2006.
    OpenUrlCrossRefPubMed
  14. ↵
    1. Dou J,
    2. Pan M,
    3. Wen P,
    4. Li Y,
    5. Tang Q,
    6. Chu L,
    7. Zhao F,
    8. Jiang C,
    9. Hu W,
    10. Hu K,
    11. Gu N
    : Isolation and identification of cancer stem-like cells from murine melanoma cell lines. Cell Mol Immunol 4(6): 467-472, 2007.
    OpenUrlPubMed
  15. ↵
    1. Mu H,
    2. Lin KX,
    3. Zhao H,
    4. Xing S,
    5. Li C,
    6. Liu F,
    7. Lu HZ,
    8. Zhang Z,
    9. Sun YL,
    10. Yan XY,
    11. Cai JQ,
    12. Zhao XH
    : Identification of biomarkers for hepatocellular carcinoma by semiquantitative immunocytochemistry. World J Gastroenterol 20(19): 5826-5838, 2014.
    OpenUrlCrossRefPubMed
  16. ↵
    1. Qiu X,
    2. Wang Z,
    3. Li Y,
    4. Miao Y,
    5. Ren Y,
    6. Luan Y
    : Characterization of sphere-forming cells with stem-like properties from the small cell lung cancer cell line h446. Cancer Lett 323(2): 161-170, 2012.
    OpenUrlCrossRefPubMed
  17. ↵
    1. Mannelli G,
    2. Gallo O
    : Cancer stem cells hypothesis and stem cells in head and neck cancers. Cancer Treat Rev 38(5): 515-539, 2012.
    OpenUrlCrossRefPubMed
  18. ↵
    1. Ueda K,
    2. Ogasawara S,
    3. Akiba J,
    4. Nakayama M,
    5. Todoroki K,
    6. Ueda K,
    7. Sanada S,
    8. Suekane S,
    9. Noguchi M,
    10. Matsuoka K,
    11. Yano H
    : Aldehyde dehydrogenase 1 identifies cells with cancer stem cell-like properties in a human renal cell carcinoma cell line. PLoS One 8(10): e75463, 2013.
    OpenUrlPubMed
  19. ↵
    1. Kitamura H,
    2. Okudela K,
    3. Yazawa T,
    4. Sato H,
    5. Shimoyamada H
    : Cancer stem cell: Implications in cancer biology and therapy with special reference to lung cancer. Lung Cancer 66(3): 275-281, 2009.
    OpenUrlCrossRefPubMed
  20. ↵
    1. Saha A,
    2. Shree Padhi S,
    3. Roy S,
    4. Banerjee B
    : Method of detecting new cancer stem cell-like enrichment in development front assay (dfa). J Stem Cells 9(4): 235-242, 2014.
    OpenUrlPubMed
  21. ↵
    1. Sztiller-Sikorska M,
    2. Koprowska K,
    3. Majchrzak K,
    4. Hartman M,
    5. Czyz M
    : Natural compounds' activity against cancer stem-like or fast-cycling melanoma cells. PLoS One 9(3): e90783, 2014.
    OpenUrlPubMed
  22. ↵
    1. Shervington A,
    2. Lu C
    : Expression of multidrug resistance genes in normal and cancer stem cells. Cancer Invest 26(5): 535-542, 2008.
    OpenUrlCrossRefPubMed
  23. ↵
    1. Pandian V,
    2. Ramraj S,
    3. Khan FH,
    4. Azim T,
    5. Aravindan N
    : Metastatic neuroblastoma cancer stem cells exhibit flexible plasticity and adaptive stemness signaling. Stem Cell Res Ther 6: 2, 2015.
    OpenUrl
  24. ↵
    1. Mimeault M,
    2. Batra SK
    : Hypoxia-inducing factors as master regulators of stemness properties and altered metabolism of cancer- and metastasis-initiating cells. J Cell Mol Med 17(1): 30-54, 2013.
    OpenUrlCrossRefPubMed
  25. ↵
    1. Liu KC,
    2. Yo YT,
    3. Huang RL,
    4. Wang YC,
    5. Liao YP,
    6. Huang TS,
    7. Chao TK,
    8. Lin CK,
    9. Weng SJ,
    10. Ma KH,
    11. Chang CC,
    12. Yu MH,
    13. Lai HC
    : Ovarian cancer stem-like cells show induced translineage-differentiation capacity and are suppressed by alkaline phosphatase inhibitor. Oncotarget 4(12): 2366-2382, 2013.
    OpenUrlPubMed
  26. ↵
    1. Fishman WH,
    2. Inglis NR,
    3. Green S,
    4. Anstiss CL,
    5. Gosh NK,
    6. Reif AE,
    7. Rustigian R,
    8. Krant MJ,
    9. Stolbach LL
    : Immunology and biochemistry of regan isoenzyme of alkaline phosphatase in human cancer. Nature 219(5155): 697-699, 1968.
    OpenUrlCrossRefPubMed
  27. ↵
    1. Miyazaki M,
    2. Tsuboi S,
    3. Mihara K,
    4. Kosaka T,
    5. Fukaya K,
    6. Kino K,
    7. Mori M,
    8. Namba M
    : Establishment and characterization of a human colon cancer cell line, oums-23, from a patient with familial adenomatous polyposis. J Cancer Res Clin Oncol 122(2): 95-101, 1996.
    OpenUrlPubMed
  28. ↵
    1. Bukowczan J,
    2. Pattman S,
    3. Jenkinson F,
    4. Quinton R
    : Regan isoenzyme of alkaline phosphatase as a tumour marker for renal cell carcinoma. Ann Clin Biochem 51(Pt 5): 611-614, 2014.
    OpenUrlAbstract/FREE Full Text
  29. ↵
    1. Frieling GW,
    2. Al-Zaid T,
    3. Prieto VG
    : Placental alkaline phosphatase positivity in metastatic melanoma. Am J Dermatopathol 36(2): 189-190, 2014.
    OpenUrlPubMed
  30. ↵
    1. Stefkova K,
    2. Prochazkova J,
    3. Pachernik J
    : Alkaline phosphatase in stem cells. Stem Cells Int 2015: 628368, 2015.
    OpenUrlPubMed
  31. ↵
    1. Kim KH,
    2. Han KS,
    3. Kim KH,
    4. Kim DK,
    5. Koo KC,
    6. Rha KH,
    7. Choi YD,
    8. Hong SJ
    : The prognostic effect of prostate-specific antigen half-life at the first follow-up visit in newly diagnosed metastatic prostate cancer. Urol Oncol, 2015.
  32. ↵
    1. Findlay VJ,
    2. Wang C,
    3. Watson DK,
    4. Camp ER
    : Epithelial-to-mesenchymal transition and the cancer stem cell phenotype: Insights from cancer biology with therapeutic implications for colorectal cancer. Cancer Gene Ther 21(5): 181-187, 2014.
    OpenUrlPubMed
  33. ↵
    1. Scheel C,
    2. Weinberg RA
    : Cancer stem cells and epithelial-mesenchymal transition: Concepts and molecular links. Semin Cancer Biol 22(5-6): 396-403, 2012.
    OpenUrlCrossRefPubMed
  34. ↵
    1. Das B,
    2. Tsuchida R,
    3. Malkin D,
    4. Koren G,
    5. Baruchel S,
    6. Yeger H
    : Hypoxia enhances tumor stemness by increasing the invasive and tumorigenic side population fraction. Stem Cells 26(7): 1818-1830, 2008.
    OpenUrlCrossRefPubMed
  35. ↵
    1. Choi JH,
    2. Hwang YP,
    3. Kim HG,
    4. Khanal T,
    5. Do MT,
    6. Jin SW,
    7. Han HJ,
    8. Lee HS,
    9. Lee YC,
    10. Chung YC,
    11. Jeong TC,
    12. Jeong HG
    : Saponins from the roots of platycodon grandiflorum suppresses tgfbeta1-induced epithelial-mesenchymal transition via repression of pi3k/akt, erk1/2 and smad2/3 pathway in human lung carcinoma a549 cells. Nutr Cancer 66(1): 140-151, 2014.
    OpenUrlPubMed
  36. ↵
    1. Santoyo-Ramos P,
    2. Likhatcheva M,
    3. Garcia-Zepeda EA,
    4. Castaneda-Patlan MC,
    5. Robles-Flores M
    : Hypoxia-inducible factors modulate the stemness and malignancy of colon cancer cells by playing opposite roles in canonical wnt signaling. PLoS One 9(11): e112580, 2014.
    OpenUrlCrossRefPubMed
  37. ↵
    1. Yang MH,
    2. Wu MZ,
    3. Chiou SH,
    4. Chen PM,
    5. Chang SY,
    6. Liu CJ,
    7. Teng SC,
    8. Wu KJ
    : Direct regulation of twist by hif-1alpha promotes metastasis. Nat Cell Biol 10(3): 295-305, 2008.
    OpenUrlCrossRefPubMed
  38. ↵
    1. Ju X,
    2. Casimiro MC,
    3. Gormley M,
    4. Meng H,
    5. Jiao X,
    6. Katiyar S,
    7. Crosariol M,
    8. Chen K,
    9. Wang M,
    10. Quong AA,
    11. Lisanti MP,
    12. Ertel A,
    13. Pestell RG
    : Identification of a cyclin d1 network in prostate cancer that antagonizes epithelial-mesenchymal restraint. Cancer Res 74(2): 508-519, 2014.
    OpenUrlAbstract/FREE Full Text
  39. ↵
    1. Kawaguchi N,
    2. Machida M,
    3. Hatta K,
    4. Nakanishi T,
    5. Takagaki Y
    : Cell shape and cardiosphere differentiation: A revelation by proteomic profiling. Biochem Res Int 2013: 730874, 2013.
    OpenUrlPubMed
  40. ↵
    1. Grange C,
    2. Collino F,
    3. Tapparo M,
    4. Camussi G
    : Oncogenic micro-rnas and renal cell carcinoma. Front Oncol 4(49, 2014.
    OpenUrlPubMed
  41. ↵
    1. Gottschling S,
    2. Schnabel PA,
    3. Herth FJ,
    4. Herpel E
    : Are we missing the target? Cancer stem cells and drug resistance in non-small cell lung cancer. Cancer Genomics Proteomics 9(5): 275-286, 2012.
    OpenUrlAbstract/FREE Full Text
  42. ↵
    1. Reinecke P,
    2. Steckstor M,
    3. Schmitz M,
    4. Gabbert HE,
    5. Gerharz CD
    : Chemotherapeutic potential of plant alkaloids and multidrug resistance mechanisms in malignant fibrous histiocytoma of the heart. Oncol Rep 11(3): 641-645, 2004.
    OpenUrlPubMed
  43. ↵
    1. Wu CP,
    2. Calcagno AM,
    3. Ambudkar SV
    : Reversal of abc drug transporter-mediated multidrug resistance in cancer cells: Evaluation of current strategies. Curr Mol Pharmacol 1(2): 93-105, 2008.
    OpenUrlCrossRefPubMed
  44. ↵
    1. Nakanishi T,
    2. Bailey-Dell KJ,
    3. Hassel BA,
    4. Shiozawa K,
    5. Sullivan DM,
    6. Turner J,
    7. Ross DD
    : Novel 5’ untranslated region variants of bcrp mrna are differentially expressed in drug-selected cancer cells and in normal human tissues: Implications for drug resistance, tissue-specific expression, and alternative promoter usage. Cancer Res 66(10): 5007-5011, 2006.
    OpenUrlAbstract/FREE Full Text
  45. ↵
    1. Gillet JP,
    2. Calcagno AM,
    3. Varma S,
    4. Marino M,
    5. Green LJ,
    6. Vora MI,
    7. Patel C,
    8. Orina JN,
    9. Eliseeva TA,
    10. Singal V,
    11. Padmanabhan R,
    12. Davidson B,
    13. Ganapathi R,
    14. Sood AK,
    15. Rueda BR,
    16. Ambudkar SV,
    17. Gottesman MM
    : Redefining the relevance of established cancer cell lines to the study of mechanisms of clinical anticancer drug resistance. Proc Natl Acad Sci USA 108(46): 18708-18713, 2011.
    OpenUrlAbstract/FREE Full Text
  46. ↵
    1. Moitra K,
    2. Lou H,
    3. Dean M
    : Multidrug efflux pumps and cancer stem cells: Insights into multidrug resistance and therapeutic development. Clin Pharmacol Ther 89(4): 491-502, 2011.
    OpenUrlCrossRefPubMed
  47. ↵
    1. Doyle L,
    2. Ross DD
    : Multidrug resistance mediated by the breast cancer resistance protein bcrp (abcg2). Oncogene 22(47): 7340-7358, 2003.
    OpenUrlCrossRefPubMed
  48. ↵
    1. Shigeta J,
    2. Katayama K,
    3. Mitsuhashi J,
    4. Noguchi K,
    5. Sugimoto Y
    : Bcrp/abcg2 confers anticancer drug resistance without covalent dimerization. Cancer Sci 101(8): 1813-1821, 2010.
    OpenUrlCrossRefPubMed
  49. ↵
    1. Charafe-Jauffret E,
    2. Ginestier C,
    3. Iovino F,
    4. Tarpin C,
    5. Diebel M,
    6. Esterni B,
    7. Houvenaeghel G,
    8. Extra JM,
    9. Bertucci F,
    10. Jacquemier J,
    11. Xerri L,
    12. Dontu G,
    13. Stassi G,
    14. Xiao Y,
    15. Barsky SH,
    16. Birnbaum D,
    17. Viens P,
    18. Wicha MS
    : Aldehyde dehydrogenase 1-positive cancer stem cells mediate metastasis and poor clinical outcome in inflammatory breast cancer. Clin Cancer Res 16(1): 45-55, 2010.
    OpenUrlAbstract/FREE Full Text
  50. ↵
    1. Croker AK,
    2. Allan AL
    : Inhibition of aldehyde dehydrogenase (aldh) activity reduces chemotherapy and radiation resistance of stem-like aldhhicd44(+) human breast cancer cells. Breast Cancer Res Treat 133(1): 75-87, 2012.
    OpenUrlCrossRefPubMed
  51. ↵
    1. Koppaka V,
    2. Thompson DC,
    3. Chen Y,
    4. Ellermann M,
    5. Nicolaou KC,
    6. Juvonen RO,
    7. Petersen D,
    8. Deitrich RA,
    9. Hurley TD,
    10. Vasiliou V
    : Aldehyde dehydrogenase inhibitors: A comprehensive review of the pharmacology, mechanism of action, substrate specificity, and clinical application. Pharmacol Rev 64(3): 520-539, 2012.
    OpenUrlAbstract/FREE Full Text
  52. ↵
    1. Niwa T,
    2. Murayama N,
    3. Imagawa Y,
    4. Yamazaki H
    : Regioselective hydroxylation of steroid hormones by human cytochromes p450. Drug Metab Rev 47(2): 89-110, 2015.
    OpenUrlPubMed
  53. ↵
    1. Teel RW,
    2. Huynh H
    : Modulation by phytochemicals of cytochrome p450-linked enzyme activity. Cancer Lett 133(2): 135-141, 1998.
    OpenUrlCrossRefPubMed
  54. ↵
    1. Shukla S,
    2. Meeran SM,
    3. Katiyar SK
    : Epigenetic regulation by selected dietary phytochemicals in cancer chemoprevention. Cancer Lett 355(1): 9-17, 2014.
    OpenUrlPubMed
  55. ↵
    1. Go RE,
    2. Hwang KA,
    3. Choi KC
    : Cytochrome p450 1 family and cancers. J Steroid Biochem Mol Biol 147: 24-30, 2015.
    OpenUrlPubMed
  56. ↵
    1. Piotrowska H,
    2. Kucinska M,
    3. Murias M
    : Expression of cyp1a1, cyp1b1 and mnsod in a panel of human cancer cell lines. Mol Cell Biochem 383(1-2): 95-102, 2013.
    OpenUrlPubMed
  57. ↵
    1. Li L,
    2. Borodyansky L,
    3. Yang Y
    : Genomic instability en route to and from cancer stem cells. Cell Cycle 8(7): 1000-1002, 2009.
    OpenUrlPubMed
  58. ↵
    1. Mathieu J,
    2. Zhang Z,
    3. Zhou W,
    4. Wang AJ,
    5. Heddleston JM,
    6. Pinna CM,
    7. Hubaud A,
    8. Stadler B,
    9. Choi M,
    10. Bar M,
    11. Tewari M,
    12. Liu A,
    13. Vessella R,
    14. Rostomily R,
    15. Born D,
    16. Horwitz M,
    17. Ware C,
    18. Blau CA,
    19. Cleary MA,
    20. Rich JN,
    21. Ruohola-Baker H
    : Hif induces human embryonic stem cell markers in cancer cells. Cancer Res 71(13): 4640-4652, 2011.
    OpenUrlAbstract/FREE Full Text
  59. ↵
    1. Liu S,
    2. Dontu G,
    3. Wicha MS
    : Mammary stem cells, self-renewal pathways, and carcinogenesis. Breast Cancer Res 7(3): 86-95, 2005.
    OpenUrlCrossRefPubMed
  60. ↵
    1. Kim SE,
    2. Huang H,
    3. Zhao M,
    4. Zhang X,
    5. Zhang A,
    6. Semonov MV,
    7. MacDonald BT,
    8. Zhang X,
    9. Garcia Abreu J,
    10. Peng L,
    11. He X
    : Wnt stabilization of beta-catenin reveals principles for morphogen receptor-scaffold assemblies. Science 340(6134): 867-870, 2013.
    OpenUrlAbstract/FREE Full Text
  61. ↵
    1. Bechard M,
    2. Trost R,
    3. Singh AM,
    4. Dalton S
    : Frat is a phosphatidylinositol 3-kinase/akt-regulated determinant of glycogen synthase kinase 3beta subcellular localization in pluripotent cells. Mol Cell Biol 32(2): 288-296, 2012.
    OpenUrlAbstract/FREE Full Text
  62. ↵
    1. Yeung J,
    2. Esposito MT,
    3. Gandillet A,
    4. Zeisig BB,
    5. Griessinger E,
    6. Bonnet D,
    7. So CW
    : Beta-catenin mediates the establishment and drug resistance of mll leukemic stem cells. Cancer Cell 18(6): 606-618, 2010.
    OpenUrlCrossRefPubMed
  63. ↵
    1. Woodward WA,
    2. Chen MS,
    3. Behbod F,
    4. Alfaro MP,
    5. Buchholz TA,
    6. Rosen JM
    : Wnt/beta-catenin mediates radiation resistance of mouse mammary progenitor cells. Proc Natl Acad Sci USA 104(2): 618-623, 2007.
    OpenUrlAbstract/FREE Full Text
  64. ↵
    1. Yang W,
    2. Yan HX,
    3. Chen L,
    4. Liu Q,
    5. He YQ,
    6. Yu LX,
    7. Zhang SH,
    8. Huang DD,
    9. Tang L,
    10. Kong XN,
    11. Chen C,
    12. Liu SQ,
    13. Wu MC,
    14. Wang HY
    : Wnt/beta-catenin signaling contributes to activation of normal and tumorigenic liver progenitor cells. Cancer Res 68(11): 4287-4295, 2008.
    OpenUrlAbstract/FREE Full Text
  65. ↵
    1. Drenkhahn SK,
    2. Jackson GA,
    3. Slusarz A,
    4. Starkey NJ,
    5. Lubahn DB
    : Inhibition of hedgehog/gli signaling by botanicals: A review of compounds with potential hedgehog pathway inhibitory activities. Curr Cancer Drug Targets 13(5): 580-595, 2013.
    OpenUrlCrossRefPubMed
  66. ↵
    1. Nakashima H,
    2. Nakamura M,
    3. Yamaguchi H,
    4. Yamanaka N,
    5. Akiyoshi T,
    6. Koga K,
    7. Yamaguchi K,
    8. Tsuneyoshi M,
    9. Tanaka M,
    10. Katano M
    : Nuclear factor-kappab contributes to hedgehog signaling pathway activation through sonic hedgehog induction in pancreatic cancer. Cancer Res 66(14): 7041-7049, 2006.
    OpenUrlAbstract/FREE Full Text
  67. ↵
    1. Su W,
    2. Meng F,
    3. Huang L,
    4. Zheng M,
    5. Liu W,
    6. Sun H
    : Sonic hedgehog maintains survival and growth of chronic myeloid leukemia progenitor cells through beta-catenin signaling. Exp Hematol 40(5): 418-427, 2012.
    OpenUrlCrossRefPubMed
  68. ↵
    1. Farnie G,
    2. Clarke RB
    : Mammary stem cells and breast cancer- -role of notch signalling. Stem Cell Rev 3(2): 169-175, 2007.
    OpenUrlCrossRefPubMed
  69. ↵
    1. Borggrefe T,
    2. Oswald F
    : The notch signaling pathway: Transcriptional regulation at notch target genes. Cell Mol Life Sci 66(10): 1631-1646, 2009.
    OpenUrlCrossRefPubMed
  70. ↵
    1. Espinosa L,
    2. Cathelin S,
    3. D'Altri T,
    4. Trimarchi T,
    5. Statnikov A,
    6. Guiu J,
    7. Rodilla V,
    8. Ingles-Esteve J,
    9. Nomdedeu J,
    10. Bellosillo B,
    11. Besses C,
    12. Abdel-Wahab O,
    13. Kucine N,
    14. Sun SC,
    15. Song G,
    16. Mullighan CC,
    17. Levine RL,
    18. Rajewsky K,
    19. Aifantis I,
    20. Bigas A
    : The notch/hes1 pathway sustains nf-kappab activation through cyld repression in t cell leukemia. Cancer Cell 18(3): 268-281, 2010.
    OpenUrlCrossRefPubMed
  71. ↵
    1. Kwon OJ,
    2. Valdez JM,
    3. Zhang L,
    4. Zhang B,
    5. Wei X,
    6. Su Q,
    7. Ittmann MM,
    8. Creighton CJ,
    9. Xin L
    : Increased notch signalling inhibits anoikis and stimulates proliferation of prostate luminal epithelial cells. Nat Commun 5: 4416, 2014.
    OpenUrlPubMed
  72. ↵
    1. Markstein M,
    2. Dettorre S,
    3. Cho J,
    4. Neumuller RA,
    5. Craig-Muller S,
    6. Perrimon N
    : Systematic screen of chemotherapeutics in drosophila stem cell tumors. Proc Natl Acad Sci USA 111(12): 4530-4535, 2014.
    OpenUrlAbstract/FREE Full Text
  73. ↵
    1. Courtney KD,
    2. Corcoran RB,
    3. Engelman JA
    : The pi3k pathway as drug target in human cancer. Journal of clinical oncology: official journal of the American Society of Clinical Oncology 28(6): 1075-1083, 2010.
    OpenUrlPubMed
  74. ↵
    1. Kim RJ,
    2. Bae E,
    3. Hong YK,
    4. Hong JY,
    5. Kim NK,
    6. Ahn HJ,
    7. Oh JJ,
    8. Park DS
    : Pten loss-mediated akt activation increases the properties of cancer stem-like cell populations in prostate cancer. Oncology 87(5): 270-279, 2014.
    OpenUrlPubMed
  75. ↵
    1. Aksamitiene E,
    2. Kiyatkin A,
    3. Kholodenko BN
    : Cross-talk between mitogenic ras/mapk and survival pi3k/akt pathways: A fine balance. Biochemical Society transactions 40(1): 139-146, 2012.
    OpenUrlAbstract/FREE Full Text
  76. ↵
    1. Voskas D,
    2. Ling LS,
    3. Woodgett JR
    : Does gsk-3 provide a shortcut for pi3k activation of wnt signalling? F1000 biology reports 2: 82, 2010.
    OpenUrl
  77. ↵
    1. Korkaya H,
    2. Paulson A,
    3. Charafe-Jauffret E,
    4. Ginestier C,
    5. Brown M,
    6. Dutcher J,
    7. Clouthier SG,
    8. Wicha MS
    : Regulation of mammary stem/progenitor cells by pten/akt/beta-catenin signaling. PLoS Biol 7(6): e1000121, 2009.
    OpenUrlCrossRefPubMed
  78. ↵
    1. Biver E,
    2. Thouverey C,
    3. Magne D,
    4. Caverzasio J
    : Crosstalk between tyrosine kinase receptors, gsk3 and bmp2 signaling during osteoblastic differentiation of human mesenchymal stem cells. Mol Cell Endocrinol 382(1): 120-130, 2014.
    OpenUrlPubMed
  79. ↵
    1. Ormanns S,
    2. Neumann J,
    3. Horst D,
    4. Kirchner T,
    5. Jung A
    : Wnt signaling and distant metastasis in colon cancer through transcriptional activity of nuclear beta-catenin depend on active pi3k signaling. Oncotarget 5(10): 2999-3011, 2014.
    OpenUrlPubMed
  80. ↵
    1. Eichhorn T,
    2. Efferth T
    : P-glycoprotein and its inhibition in tumors by phytochemicals derived from chinese herbs. J Ethnopharmacol 141(2): 557-570, 2012.
    OpenUrlPubMed
  81. ↵
    1. Tan KW,
    2. Li Y,
    3. Paxton JW,
    4. Birch NP,
    5. Scheepens A
    : Identification of novel dietary phytochemicals inhibiting the efflux transporter breast cancer resistance protein (bcrp/abcg2). Food Chem 138(4): 2267-2274, 2013.
    OpenUrlCrossRefPubMed
  82. ↵
    1. Holland ML,
    2. Lau DT,
    3. Allen JD,
    4. Arnold JC
    : The multidrug transporter abcg2 (bcrp) is inhibited by plant-derived cannabinoids. Br J Pharmacol 152(5): 815-824, 2007.
    OpenUrlCrossRefPubMed
  83. ↵
    1. Tian Y,
    2. Qian S,
    3. Jiang Y,
    4. Shen Q,
    5. Zheng J,
    6. Zhou H,
    7. Zeng S
    : The interaction between human breast cancer resistance protein (bcrp) and five bisbenzylisoquinoline alkaloids. Int J Pharm 453(2): 371-379, 2013.
    OpenUrlPubMed
  84. ↵
    1. Kai M,
    2. Kanaya N,
    3. Wu SV,
    4. Mendez C,
    5. Nguyen D,
    6. Luu T,
    7. Chen S
    : Targeting breast cancer stem cells in triple-negative breast cancer using a combination of lbh589 and salinomycin. Breast Cancer Res Treat 151(2): 281-294, 2015.
    OpenUrlPubMed
  85. ↵
    1. Clarke JD,
    2. Dashwood RH,
    3. Ho E
    : Multi-targeted prevention of cancer by sulforaphane. Cancer Lett 269(2): 291-304, 2008.
    OpenUrlCrossRefPubMed
  86. ↵
    1. Forster T,
    2. Rausch V,
    3. Zhang Y,
    4. Isayev O,
    5. Heilmann K,
    6. Schoensiegel F,
    7. Liu L,
    8. Nessling M,
    9. Richter K,
    10. Labsch S,
    11. Nwaeburu CC,
    12. Mattern J,
    13. Gladkich J,
    14. Giese N,
    15. Werner J,
    16. Schemmer P,
    17. Gross W,
    18. Gebhard MM,
    19. Gerhauser C,
    20. Schaefer M,
    21. Herr I
    : Sulforaphane counteracts aggressiveness of pancreatic cancer driven by dysregulated cx43-mediated gap junctional intercellular communication. Oncotarget 5(6): 1621-1634, 2014.
    OpenUrlPubMed
  87. ↵
    1. Li Y,
    2. Zhang T,
    3. Korkaya H,
    4. Liu S,
    5. Lee HF,
    6. Newman B,
    7. Yu Y,
    8. Clouthier SG,
    9. Schwartz SJ,
    10. Wicha MS,
    11. Sun D
    : Sulforaphane, a dietary component of broccoli/broccoli sprouts, inhibits breast cancer stem cells. Clin Cancer Res 16(9): 2580-2590, 2010.
    OpenUrlAbstract/FREE Full Text
  88. ↵
    1. Chung FL,
    2. Conaway CC,
    3. Rao CV,
    4. Reddy BS
    : Chemoprevention of colonic aberrant crypt foci in fischer rats by sulforaphane and phenethyl isothiocyanate. Carcinogenesis 21(12): 2287-2291, 2000.
    OpenUrlAbstract/FREE Full Text
  89. ↵
    1. Kallifatidis G,
    2. Rausch V,
    3. Baumann B,
    4. Apel A,
    5. Beckermann BM,
    6. Groth A,
    7. Mattern J,
    8. Li Z,
    9. Kolb A,
    10. Moldenhauer G,
    11. Altevogt P,
    12. Wirth T,
    13. Werner J,
    14. Schemmer P,
    15. Buchler MW,
    16. Salnikov AV,
    17. Herr I
    : Sulforaphane targets pancreatic tumour-initiating cells by nf-kappab-induced antiapoptotic signalling. Gut 58(7): 949-963, 2009.
    OpenUrlAbstract/FREE Full Text
  90. ↵
    1. Jeong WS,
    2. Kim IW,
    3. Hu R,
    4. Kong AN
    : Modulatory properties of various natural chemopreventive agents on the activation of nf-kappab signaling pathway. Pharm Res 21(4): 661-670, 2004.
    OpenUrlCrossRefPubMed
  91. ↵
    1. Khor TO,
    2. Hu R,
    3. Shen G,
    4. Jeong WS,
    5. Hebbar V,
    6. Chen C,
    7. Xu C,
    8. Nair S,
    9. Reddy B,
    10. Chada K,
    11. Kong AN
    : Pharmacogenomics of cancer chemopreventive isothiocyanate compound sulforaphane in the intestinal polyps of apcmin/+ mice. Biopharm Drug Dispos 27(9): 407-420, 2006.
    OpenUrlPubMed
  92. ↵
    1. Park SY,
    2. Kim GY,
    3. Bae SJ,
    4. Yoo YH,
    5. Choi YH
    : Induction of apoptosis by isothiocyanate sulforaphane in human cervical carcinoma hela and hepatocarcinoma hepg2 cells through activation of caspase-3. Oncol Rep 18(1): 181-187, 2007.
    OpenUrlPubMed
  93. ↵
    1. Verheus M,
    2. van Gils CH,
    3. Keinan-Boker L,
    4. Grace PB,
    5. Bingham SA,
    6. Peeters PH
    : Plasma phytoestrogens and subsequent breast cancer risk. J Clin Oncol 25(6): 648-655, 2007.
    OpenUrlAbstract/FREE Full Text
  94. ↵
    1. Barnes S
    : Effect of genistein on in vitro and in vivo models of cancer. J Nutr 125(3 Suppl): 777S-783S, 1995.
    OpenUrlAbstract/FREE Full Text
  95. ↵
    1. Sarkar FH,
    2. Li Y,
    3. Wang Z,
    4. Kong D
    : Cellular signaling perturbation by natural products. Cell Signal 21(11): 1541-1547, 2009.
    OpenUrlCrossRefPubMed
  96. ↵
    1. Su Y,
    2. Simmen RC
    : Soy isoflavone genistein upregulates epithelial adhesion molecule e-cadherin expression and attenuates beta-catenin signaling in mammary epithelial cells. Carcinogenesis 30(2): 331-339, 2009.
    OpenUrlAbstract/FREE Full Text
  97. ↵
    1. Montales MT,
    2. Rahal OM,
    3. Kang J,
    4. Rogers TJ,
    5. Prior RL,
    6. Wu X,
    7. Simmen RC
    : Repression of mammosphere formation of human breast cancer cells by soy isoflavone genistein and blueberry polyphenolic acids suggests diet-mediated targeting of cancer stem-like/progenitor cells. Carcinogenesis 33(3): 652-660, 2012.
    OpenUrlAbstract/FREE Full Text
  98. ↵
    1. Ning YX,
    2. Li QX,
    3. Ren KQ,
    4. Quan MF,
    5. Cao JG
    : 7-difluoromethoxyl-5,4′-di-n-octyl genistein inhibits ovarian cancer stem cell characteristics through the downregulation of foxm1. Oncol Lett 8(1): 295-300, 2014.
    OpenUrlPubMed
  99. ↵
    1. Landis-Piwowar KR,
    2. Huo C,
    3. Chen D,
    4. Milacic V,
    5. Shi G,
    6. Chan TH,
    7. Dou QP
    : A novel prodrug of the green tea polyphenol (−)-epigallocatechin-3-gallate as a potential anticancer agent. Cancer Res 67(9): 4303-4310, 2007.
    OpenUrlAbstract/FREE Full Text
  100. ↵
    1. Shimizu M,
    2. Deguchi A,
    3. Lim JT,
    4. Moriwaki H,
    5. Kopelovich L,
    6. Weinstein IB
    : (-)-epigallocatechin gallate and polyphenon e inhibit growth and activation of the epidermal growth factor receptor and human epidermal growth factor receptor-2 signaling pathways in human colon cancer cells. Clin Cancer Res 11(7): 2735-2746, 2005.
    OpenUrlAbstract/FREE Full Text
  101. ↵
    1. Kawasaki BT,
    2. Hurt EM,
    3. Mistree T,
    4. Farrar WL
    : Targeting cancer stem cells with phytochemicals. Mol Interv 8(4): 174-184, 2008.
    OpenUrlCrossRefPubMed
  102. ↵
    1. Godeke J,
    2. Maier S,
    3. Eichenmuller M,
    4. Muller-Hocker J,
    5. von Schweinitz D,
    6. Kappler R
    : Epigallocatechin-3-gallate inhibits hepatoblastoma growth by reactivating the wnt inhibitor sfrp1. Nutr Cancer 65(8): 1200-1207, 2013.
    OpenUrlPubMed
  103. ↵
    1. Hao X,
    2. Sun Y,
    3. Yang CS,
    4. Bose M,
    5. Lambert JD,
    6. Ju J,
    7. Lu G,
    8. Lee MJ,
    9. Park S,
    10. Husain A,
    11. Wang S
    : Inhibition of intestinal tumorigenesis in apc(min/+) mice by green tea polyphenols (polyphenon e) and individual catechins. Nutr Cancer 59(1): 62-69, 2007.
    OpenUrlCrossRefPubMed
  104. ↵
    1. Li Y,
    2. Zhang T,
    3. Jiang Y,
    4. Lee HF,
    5. Schwartz SJ,
    6. Sun D
    : (−)-epigallocatechin-3-gallate inhibits hsp90 function by impairing hsp90 association with cochaperones in pancreatic cancer cell line mia paca-2. Mol Pharm 6(4): 1152-1159, 2009.
    OpenUrlCrossRefPubMed
  105. ↵
    1. Kim SJ,
    2. Jeong HJ,
    3. Lee KM,
    4. Myung NY,
    5. An NH,
    6. Yang WM,
    7. Park SK,
    8. Lee HJ,
    9. Hong SH,
    10. Kim HM,
    11. Um JY
    : Epigallocatechin-3-gallate suppresses nf-kappab activation and phosphorylation of p38 mapk and jnk in human astrocytoma u373mg cells. J Nutr Biochem 18(9): 587-596, 2007.
    OpenUrlCrossRefPubMed
  106. ↵
    1. D'Angelo L,
    2. Piazzi G,
    3. Pacilli A,
    4. Prossomariti A,
    5. Fazio C,
    6. Montanaro L,
    7. Graziani G,
    8. Fogliano V,
    9. Munarini A,
    10. Bianchi F,
    11. Belluzzi A,
    12. Bazzoli F,
    13. Ricciardiello L
    : A combination of eicosapentaenoic acid-free fatty acid, epigallocatechin-3-gallate and proanthocyanidins has a strong effect on mtor signaling in colorectal cancer cells. Carcinogenesis, 2014.
  107. ↵
    1. Eid SY,
    2. El-Readi MZ,
    3. Wink M
    : Digitonin synergistically enhances the cytotoxicity of plant secondary metabolites in cancer cells. Phytomedicine 19(14): 1307-1314, 2012.
    OpenUrlCrossRefPubMed
  108. ↵
    1. Son MK,
    2. Jung KH,
    3. Lee HS,
    4. Lee H,
    5. Kim SJ,
    6. Yan HH,
    7. Ryu YL,
    8. Hong SS
    : Sb365, pulsatilla saponin d suppresses proliferation and induces apoptosis of pancreatic cancer cells. Oncol Rep 30(2): 801-808, 2013.
    OpenUrlPubMed
  109. ↵
    1. Wang P,
    2. Vadgama JV,
    3. Said JW,
    4. Magyar CE,
    5. Doan N,
    6. Heber D,
    7. Henning SM
    : Enhanced inhibition of prostate cancer xenograft tumor growth by combining quercetin and green tea. J Nutr Biochem 25(1): 73-80, 2014.
    OpenUrlPubMed
  110. ↵
    1. Divya CS,
    2. Pillai MR
    : Antitumor action of curcumin in human papillomavirus associated cells involves down-regulation of viral oncogenes, prevention of nfkb and ap-1 translocation, and modulation of apoptosis. Mol Carcinog 45(5): 320-332, 2006.
    OpenUrlCrossRefPubMed
  111. ↵
    1. Duan H,
    2. Zhang X,
    3. Wang FX,
    4. Cai MY,
    5. Ma GW,
    6. Yang H,
    7. Fu JH,
    8. Tan ZH,
    9. Meng YQ,
    10. Fu XY,
    11. Ma QL,
    12. Lin P
    : Prognostic role of neutrophil-lymphocyte ratio in operable esophageal squamous cell carcinoma. World J Gastroenterol 21(18): 5591-5597, 2015.
    OpenUrlPubMed
  112. ↵
    1. Almanaa TN,
    2. Geusz ME,
    3. Jamasbi RJ
    : Effects of curcumin on stem-like cells in human esophageal squamous carcinoma cell lines. BMC Complement Altern Med 12: 195, 2012.
    OpenUrlCrossRefPubMed
  113. ↵
    1. Yu Y,
    2. Kanwar SS,
    3. Patel BB,
    4. Nautiyal J,
    5. Sarkar FH,
    6. Majumdar AP
    : Elimination of colon cancer stem-like cells by the combination of curcumin and folfox. Transl Oncol 2(4): 321-328, 2009.
    OpenUrlCrossRefPubMed
  114. ↵
    1. Poltronieri J,
    2. Becceneri AB,
    3. Fuzer AM,
    4. Filho JC,
    5. Martin AC,
    6. Vieira PC,
    7. Pouliot N,
    8. Cominetti MR
    : [6]-gingerol as a cancer chemopreventive agent: A review of its activity on different steps of the metastatic process. Mini Rev Med Chem 14(4): 313-321, 2014.
    OpenUrlPubMed
  115. ↵
    1. Lee HS,
    2. Seo EY,
    3. Kang NE,
    4. Kim WK
    : [6]-gingerol inhibits metastasis of mda-mb-231 human breast cancer cells. J Nutr Biochem 19(5): 313-319, 2008.
    OpenUrlCrossRefPubMed
  116. ↵
    1. Lee SH,
    2. Cekanova M,
    3. Baek SJ
    : Multiple mechanisms are involved in 6-gingerol-induced cell growth arrest and apoptosis in human colorectal cancer cells. Mol Carcinog 47(3): 197-208, 2008.
    OpenUrlCrossRefPubMed
  117. ↵
    1. Gan FF,
    2. Ling H,
    3. Ang X,
    4. Reddy SA,
    5. Lee SS,
    6. Yang H,
    7. Tan SH,
    8. Hayes JD,
    9. Chui WK,
    10. Chew EH
    : A novel shogaol analog suppresses cancer cell invasion and inflammation, and displays cytoprotective effects through modulation of nf-kappab and nrf2-keap1 signaling pathways. Toxicol Appl Pharmacol 272(3): 852-862, 2013.
    OpenUrlPubMed
  118. ↵
    1. Yan F,
    2. Cao XX,
    3. Jiang HX,
    4. Zhao XL,
    5. Wang JY,
    6. Lin YH,
    7. Liu QL,
    8. Zhang C,
    9. Jiang B,
    10. Guo F
    : A novel water-soluble gossypol derivative increases chemotherapeutic sensitivity and promotes growth inhibition in colon cancer. J Med Chem 53(15): 5502-5510, 2010.
    OpenUrlPubMed
  119. ↵
    1. Volate SR,
    2. Kawasaki BT,
    3. Hurt EM,
    4. Milner JA,
    5. Kim YS,
    6. White J,
    7. Farrar WL
    : Gossypol induces apoptosis by activating p53 in prostate cancer cells and prostate tumor-initiating cells. Mol Cancer Ther 9(2): 461-470, 2010.
    OpenUrlAbstract/FREE Full Text
  120. ↵
    1. Pang X,
    2. Wu Y,
    3. Wu Y,
    4. Lu B,
    5. Chen J,
    6. Wang J,
    7. Yi Z,
    8. Qu W,
    9. Liu M
    : (−)-gossypol suppresses the growth of human prostate cancer xenografts via modulating vegf signaling-mediated angiogenesis. Mol Cancer Ther 10(5): 795-805, 2011.
    OpenUrlAbstract/FREE Full Text
  121. ↵
    1. Rekha GK,
    2. Sladek NE
    : Multienzyme-mediated stable and transient multidrug resistance and collateral sensitivity induced by xenobiotics. Cancer Chemother Pharmacol 40(3): 215-224, 1997.
    OpenUrlCrossRefPubMed
  122. ↵
    1. Jackson RS 2nd.,
    2. Placzek W,
    3. Fernandez A,
    4. Ziaee S,
    5. Chu CY,
    6. Wei J,
    7. Stebbins J,
    8. Kitada S,
    9. Fritz G,
    10. Reed JC,
    11. Chung LW,
    12. Pellecchia M,
    13. Bhowmick NA
    : Sabutoclax, a mcl-1 antagonist, inhibits tumorigenesis in transgenic mouse and human xenograft models of prostate cancer. Neoplasia 14(7): 656-665, 2012.
    OpenUrlPubMed
  123. ↵
    1. Suman S,
    2. Das TP,
    3. Damodaran C
    : Silencing notch signaling causes growth arrest in both breast cancer stem cells and breast cancer cells. Br J Cancer 109(10): 2587-2596, 2013.
    OpenUrlCrossRefPubMed
  124. ↵
    1. Liu Y,
    2. Lu WL,
    3. Guo J,
    4. Du J,
    5. Li T,
    6. Wu JW,
    7. Wang GL,
    8. Wang JC,
    9. Zhang X,
    10. Zhang Q
    : A potential target associated with both cancer and cancer stem cells: A combination therapy for eradication of breast cancer using vinorelbine stealthy liposomes plus parthenolide stealthy liposomes. J Control Release 129(1): 18-25, 2008.
    OpenUrlCrossRefPubMed
  125. ↵
    1. Giannios J ME
    : Effect of vinorelbine on chemoresistant cancer stem cell renewal in colorectal cancer (crc) and on metastasis. Gastrointestinal Cancers Symposium, 2010.
  126. ↵
    1. Guzman ML,
    2. Rossi RM,
    3. Neelakantan S,
    4. Li X,
    5. Corbett CA,
    6. Hassane DC,
    7. Becker MW,
    8. Bennett JM,
    9. Sullivan E,
    10. Lachowicz JL,
    11. Vaughan A,
    12. Sweeney CJ,
    13. Matthews W,
    14. Carroll M,
    15. Liesveld JL,
    16. Crooks PA,
    17. Jordan CT
    : An orally bioavailable parthenolide analog selectively eradicates acute myelogenous leukemia stem and progenitor cells. Blood 110(13): 4427-4435, 2007.
    OpenUrlAbstract/FREE Full Text
  127. ↵
    1. Hellsten R,
    2. Johansson M,
    3. Dahlman A,
    4. Sterner O,
    5. Bjartell A
    : Galiellalactone inhibits stem cell-like aldh-positive prostate cancer cells. PLoS One 6(7): e22118, 2011.
    OpenUrlCrossRefPubMed
  128. ↵
    1. Yoshida J,
    2. Seino H,
    3. Ito Y,
    4. Nakano T,
    5. Satoh T,
    6. Ogane Y,
    7. Suwa S,
    8. Koshino H,
    9. Kimura K
    : Inhibition of glycogen synthase kinase-3beta by falcarindiol isolated from japanese parsley (oenanthe javanica). J Agric Food Chem 61(31): 7515-7521, 2013.
    OpenUrlCrossRef
PreviousNext
Back to top

In this issue

Anticancer Research: 35 (11)
Anticancer Research
Vol. 35, Issue 11
November 2015
  • Table of Contents
  • Table of Contents (PDF)
  • Index by author
  • Back Matter (PDF)
  • Ed Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on Anticancer Research.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Natural Products That Target Cancer Stem Cells
(Your Name) has sent you a message from Anticancer Research
(Your Name) thought you would like to see the Anticancer Research web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
18 + 0 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
Natural Products That Target Cancer Stem Cells
JIM MOSELHY, SOWMYALAKSHMI SRINIVASAN, MURALI K. ANKEM, CHENDIL DAMODARAN
Anticancer Research Nov 2015, 35 (11) 5773-5788;

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
Natural Products That Target Cancer Stem Cells
JIM MOSELHY, SOWMYALAKSHMI SRINIVASAN, MURALI K. ANKEM, CHENDIL DAMODARAN
Anticancer Research Nov 2015, 35 (11) 5773-5788;
Reddit logo Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Therapeutic Implications of CSCs in Cancer Treatment
    • Genetic and Molecular Signatures of CSCs
    • Drug Efflux and Detoxification Mechanisms
    • Developmental and Maintenance Signaling Pathways
    • NPs as Modulators of CSC Pathways
    • Salinomycin
    • Polyynes
    • Conclusion and Future Perspectives
    • Acknowledgements
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • Approbation of the Cancer Treatment Approach Based on the Eradication of TAMRA+ Cancer Stem Cells in a Model of Murine Cyclophosphamide Resistant Lymphosarcoma
  • Lung Cancer Stem Cells and Cancer Stem Cell-targeting Natural Compounds
  • Cancer Stem Cell-Suppressing Activity of Chrysotoxine, a Bibenzyl from Dendrobium pulchellum
  • A Novel mTOR Inhibitor; Anthracimycin for the Treatment of Human Hepatocellular Carcinoma
  • Google Scholar

More in this TOC Section

  • Cytokine-based Cancer Immunotherapy: Challenges and Opportunities for IL-10
  • Proteolytic Enzyme Therapy in Complementary Oncology: A Systematic Review
  • Multimodal Treatment of Primary Advanced Ovarian Cancer
Show more Reviews

Similar Articles

Keywords

  • Dietary agents
  • cancer stem cells
  • mechanism of action
  • signal transduction pathways
  • review
Anticancer Research

© 2023 Anticancer Research

Powered by HighWire