Next Article in Journal
Doxycycline Ameliorates the Severity of Experimental Proliferative Vitreoretinopathy in Mice
Next Article in Special Issue
ATG 4B Serves a Crucial Role in RCE-4-Induced Inhibition of the Bcl-2–Beclin 1 Complex in Cervical Cancer Ca Ski Cells
Previous Article in Journal
Aberrant Upregulation of Indoleamine 2,3-Dioxygenase 1 Promotes Proliferation and Metastasis of Hepatocellular Carcinoma Cells via Coordinated Activation of AhR and β-Catenin Signaling
Previous Article in Special Issue
MUG Mel3 Cell Lines Reflect Heterogeneity in Melanoma and Represent a Robust Model for Melanoma in Pregnancy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeting Cancer Stem Cells by Dietary Agents: An Important Therapeutic Strategy against Human Malignancies

by
Mahshid Deldar Abad Paskeh
1,2,†,
Shafagh Asadi
3,†,
Amirhossein Zabolian
4,
Hossein Saleki
5,
Mohammad Amin Khoshbakht
5,
Sina Sabet
5,
Mohamad Javad Naghdi
5,
Mehrdad Hashemi
1,2,
Kiavash Hushmandi
6,
Milad Ashrafizadeh
7,8,
Sepideh Mirzaei
9,
Ali Zarrabi
8,10 and
Gautam Sethi
11,12,*
1
Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran
2
Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran
3
Asu Vanda Gene Industrial Research Company, Tehran 1533666398, Iran
4
Resident of Orthopedics, Department of Orthopedics, School of Medicine, 5th Azar Hospital, Golestan University of Medical Sciences, Gorgan 4934174515, Iran
5
Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran
6
Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran 1419963114, Iran
7
Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Istanbul 34956, Turkey
8
Sabanci University Nanotechnology Research and Application Center (SUNUM), Istanbul 34956, Turkey
9
Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran 1477893855, Iran
10
Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer, Istanbul 34396, Turkey
11
Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
12
Cancer Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
*
Author to whom correspondence should be addressed.
These authors have participated equally to manuscript preparation.
Int. J. Mol. Sci. 2021, 22(21), 11669; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms222111669
Submission received: 23 September 2021 / Revised: 23 October 2021 / Accepted: 23 October 2021 / Published: 28 October 2021

Abstract

:
As a multifactorial disease, treatment of cancer depends on understanding unique mechanisms involved in its progression. The cancer stem cells (CSCs) are responsible for tumor stemness and by enhancing colony formation, proliferation as well as metastasis, and these cells can also mediate resistance to therapy. Furthermore, the presence of CSCs leads to cancer recurrence and therefore their complete eradication can have immense therapeutic benefits. The present review focuses on targeting CSCs by natural products in cancer therapy. The growth and colony formation capacities of CSCs have been reported can be attenuated by the dietary agents. These compounds can induce apoptosis in CSCs and reduce tumor migration and invasion via EMT inhibition. A variety of molecular pathways including STAT3, Wnt/β-catenin, Sonic Hedgehog, Gli1 and NF-κB undergo down-regulation by dietary agents in suppressing CSC features. Upon exposure to natural agents, a significant decrease occurs in levels of CSC markers including CD44, CD133, ALDH1, Oct4 and Nanog to impair cancer stemness. Furthermore, CSC suppression by dietary agents can enhance sensitivity of tumors to chemotherapy and radiotherapy. In addition to in vitro studies, as well as experiments on the different preclinical models have shown capacity of natural products in suppressing cancer stemness. Furthermore, use of nanostructures for improving therapeutic impact of dietary agents is recommended to rapidly translate preclinical findings for clinical use.

1. Introduction

The cancer is the second leading cause of death worldwide after cardiovascular diseases [1]. Based on the new estimates published by Siegel and co-authors, prostate cancer is the most common cancer in males, while breast cancer is the most common cancer in females. Noteworthy, lung cancer is the most aggressive cancer in both sexes and causes highest death among other tumors [2]. Regardless of cancer incidence rate and cell deaths, there have been efforts in developing novel therapeutics for tumor treatment [3,4,5,6]. Surgery or tumor resection is beneficial in early stages of cancer and when tumor cells diffuse into various tissues in body, it is impossible to eliminate cancer by using surgery [7,8]. Radiotherapy is less invasive compared to surgery, but it has its own problems including side effects and risk of resistance [9]. Immunotherapy is a new emerging therapeutic modality in cancer and uses checkpoint inhibitors in impairing cancer progression [10,11,12]. Another strategy is tumor treatment is chemotherapy that is most common compared to other modalities, but resistance and adverse impacts reduce its potential [13,14]. In order to prevent therapy resistance, combination cancer therapy has been utilized. In this strategy, a combination of chemotherapy and radiotherapy, or chemotherapy and immunotherapy are applied to suppress cancer progression and inhibit resistance [15,16]. Other kinds of treatments including photothermal therapy induced by nanoparticles in ablating tumor progression, inducing DNA damage and preventing cell cycle progression [17,18,19]. Besides, gene therapy using small interfering RNA (siRNA), short hairpin RNA (shRNA) and CRISPR/Cas9 system can be applied in cancer suppression [14,20,21,22]. These strategies have been partially advantageous in improving overall survival and prognosis of cancer patients. However, cancer is still a challenge for healthcare providers and new attempts should be made in this case [23,24,25,26,27,28,29,30].
The plant derived-natural products have been under attention in recent years in field of cancer therapy [31,32,33]. These agents have great therapeutic activities that anti-tumor activity is among them [34,35]. Due to the potential of phytochemicals in apoptosis induction, cell cycle arrest, metastasis inhibition and multitargeting capacity, they are able to inhibit cancer progression [20,36,37,38,39,40,41,42]. Clinical trials have shown that plant derived-natural products are generally well-tolerated in cancer patients [43,44]. Therefore, they can be extracted for developing commercialized drugs in cancer therapy [45]. Experiments have shown that natural occurring compounds are beneficial in sensitizing tumor cells to therapeutic modalities [46,47]. For instance, curcumin can increase sensitivity of cancer cells to cisplatin, docetaxel, paclitaxel and doxorubicin [31,48,49]. A combination of resveratrol and radiotherapy is advantageous in suppressing cancer progression [50]. Hence, application of natural products alone or in combination with other therapeutic strategies can pave the way towards effective tumor treatment [51,52,53]. The present review article focuses on using dietary agents in cancer therapy via targeting cancer stem cells (CSCs). For this purpose, we first provide a summary of CSCs, their analysis, markers and metabolism. Then, we show how CSCs can enhance stemness and progression of tumor cells. Finally, we mechanistically discuss how each phytochemical can be beneficial in cancer suppression via targeting CSCs.

2. Cancer Stem Cells

The stem cells have self-renewal capacity and can develop colonies [54,55]. The stem cells exist in various phases of life from embryonic phase to adulthood and are able to differentiation in forming various organs and tissues of body [56,57]. A kind of cells with characteristics similar to stem cells was found to be involved in carcinogenesis and called CSCs [58,59,60]. The CSCs are abundantly found in the tumor microenvironment and due to their self-renewal capacity, they can preserve population of tumor cells and mediating tumorigenesis. Besides, CSCs can differentiate into different cell kinds, enhancing cancer progression [61,62]. Overall, there are two concepts for carcinogenesis. At the first model, known as stochastic model, tumor cells are similar and have the same potential in tumorigenesis. Based on this model, the accumulation of mutations has resulted in carcinogenesis [61,63]. However, upon discovery of CSCs, a new concept of tumorigenesis was introduced, known as hierarchical model that CSCs are responsible for cancer development, maintaining and tumor seeding [64].
The identification of CSCs occurred in nineteenth century, when it was found that there are dormant cells in adult tissues that can be activated by stimuli and have capacity of proliferation and generating large masses of cells [65]. Although this was a great idea showing a special function for stem cells in cancer progression, it was ignored until in 1994 that Lapidot and colleagues isolated CSCs from leukemia cells and confirmed their presence [58]. The isolated CSCs were injected in mice and they showed potential in tumor initiation and development. After the isolation of CSCs from breast cancer in 2003 [60], more investigation was performed to isolated CSCs from other kinds of tumors including brain tumors, colorectal cancer and liver cancer [66,67,68].
Overall, CSCs have three distinct features from normal cells including differentiation, self-renewal capacity and homeostasis control [69,70]. Regardless of tumor stage, CSCs can be abundantly found in TME and a variety of techniques for isolation and enrichment of CSCs are utilized such as side population detection of cells with ability for Hoechst 33,342 exclusion, sphere forming capacity and aldehyde dehydrogenase (ALDH) measurement [71]. The Oct4, SOX2, Nanog, c-Myc and KLF4 are able to regulate CSC features and related signaling networks are Wnt, Notch, Hedgehog and PI3K/Akt, among others. Furthermore, complicated conditions in TME such as hypoxia, stromal cells, growth factors and extracellular matrix are able to control CSC characteristics in tumor cells [72,73,74,75,76,77].
One of the important aspects of CSCs is their metabolism. Based on experiments, CSCs rely on glycolysis, mitochondrial oxidative phosphorylation and other metabolic pathways that can be targeted therapeutically for suppressing CSCs [78,79,80]. It has been reported that CSCs are able to induce glycolysis via upregulating glucose transporters (GLUTs), hexokinases (HKs), monocarboxylate transporters and pyruvate dehydrogenase kinase 1 [81,82,83,84,85]. The CD133+ cells that exist in pancreatic cancer and glioma, have the ability of oxidative phosphorylation and enhancing expression level of genes involved in tricarboxylic acid (TCA) cycle [86,87]. Noteworthy, the levels of reactive oxygen species (ROS) can also affect CSC metabolism. The CSCs that are in dormant conditions can preserve low levels of ROS via glycolysis or stimulating antioxidant defense system. However, CSCs want to proliferate and differentiate, they induce ROS overgeneration via oxidative phosphorylation [88]. Therefore, metabolism, growth and differentiation of CSCs have a close association that should be considered.

3. Cancer Stem Cells in Oncology

The presence of CSCs is in favor of tumor progression. Thanks to experiments performed recently to shed some light on the role of CSCs. It seems that presence of CSCs in TME results in immune evasion and immunosuppression [89]. The CSCs are able to induce drug resistance feature of tumor cells. It has been reported that STAT3 signaling enhances stemness and CSC features in ovarian tumor cells to mediate their resistance to cisplatin and paclitaxel chemotherapy [90]. An interesting study has provided new insight about process of CSC generation in breast cancer and enhancing carcinogenesis. In this case, adipose-derived stem cells and breast cancer cells (MDA-MB-231 cells) fuse to produce CSCs. This process is mediated by CD44 [91]. As it was mentioned, CSCs involve in triggering drug resistance in tumor cells. Noteworthy, CSCs can also mediate radioresistance features. An experiment on nasopharyngeal cancer demonstrated that hTERT promotes CSCs features in nasopharyngeal cancer and mediates radioresistance. Knock-down of hTERT is correlated with reduced CSC characteristics and enhanced sensitivity to radiotherapy [92].
A variety of molecular pathways can modulate CSC features in tumors. For instance, microRNA (miRNA)-326 is suggested to be a tumor-suppressor in cervical cancer. The miRNA-326 binds to 3′-UTR of transcription factor 4 (TCF4) to diminish its expression. Upon TCF4 down-regulation, a significant decrease occurs in CSC features via down-regulating CD44 and SOX4 expression levels [93]. On the other hand, tumor-promoting factors pave the way for increasing CSC features in tumors. For instance, DUSP9 undergoes overexpression in triple-negative breast cancer and down-regulates ERK1/2 expression to enhance levels of CSC markers including SOX2, Oct4 and ALDH1 [94]. Therefore, recapitulation of CSC niche can promote tumor progression and mediate drug resistance feature [95]. Identification of such factors and reducing their expression can pave the way to drug sensitivity. For instance, silencing RAD51AP1 is associated with impairment in self-renewal capacity of CSCs and inducing drug sensitivity in colorectal cancer [96]. Another example is musashi-1 that enhances glioblastoma progression. Silencing musashi-1 diminishes CSC features in glioblastoma [97].
Each experiment has focused on a certain molecular pathway that leads to cancer progression via enhancing CSC features. The lung cancer stemness and CSC features can be mediated via JAK2/STAT3 axis. As upstream mediator, aryl hydrocarbon receptor induces JAK2/STAT3 axis to promote lung cancer stemness [98]. Another experiment reveals that long non-coding RNA (lncRNA)-WDFY3-AS2 reduces miRNA-139-5p expression to promote SCD4 expression. Then, ovarian cancer stemness enhances via promoting CSC features and resistance to cisplatin is mediated [99]. The NEDD4 expression undergoes upregulation in breast cancer and preserves stemness via promoting CSC features [100].
One of the important aspects of cancer progression is the role of extracellular vesicles (EVs), especially exosomes. Briefly, exosomes can provide cell–cell communication via transferring proteins, lipids and nucleic acids [101,102,103]. A recent study has shown that EVs can stimulate generation of CSCs from stem or progenitor cells [104]. The exosomes containing lncRNA UCA1 enhances SOX2 expression via miRNA-122-5p down-regulation to enhance differentiation and self-renewal capacity of CSCs [105]. It is worth mentioning that CSCs can also secrete exosomes in cancer progression. For instance, exosomes derived from CSCs contain lncRNA DOCK9-AS2 that can enhance growth, metastasis and stemness of thyroid tumor via inducing Wnt/β-catenin axis [106]. Overall, experiments highlight the fact that CSCs play a significant role in progression of tumors and their targeting is of importance in cancer therapy. Furthermore, a variety of molecular pathways including PCGF1, circFAM73A, CXCL1 and NUMB are able to affect CSC features. The growth, metastasis and therapy response are mainly regulated by CSC characteristics [107,108,109,110,111,112,113,114,115].
The interesting point is the role of CSC markers as prognostic factors in tumor [116]. The clinical studies have confirmed this statement. The overexpression of BMI-1 and CD44 as CSC markers occurs in head and neck squamous carcinoma to promote cancer progression and mediate undesirable prognosis [117]. The CD133 and CXCR4 as other CSC markers also demonstrate alterations in osteosarcoma patients. The CD133 upregulation occurs in 26% of patients, while CXCR4 demonstrates overexpression in 36% of cases. The overexpression of aforementioned CSC markers provides undesirable prognosis and survival of osteosarcoma patients [118].

4. Search Strategy

Various databases including Google scholar, Web of Science and Pubmed were used to search and collect articles. The names of dietary agents discussed in this article and other words including “cancer” and “cancer stem cell” were searched to find the relevant articles. Furthermore, there were many phytochemicals found whose impact on CSCs have not been evaluated yet but can be considered in future studies.

5. Dietary Agents and Cancer Stem Cells

5.1. Flavonoids

5.1.1. Flavones

Nobiletin

The nobiletin is a potent anti-tumor agent capable of suppressing tumor migration via EMT inhibition [119]. Nobiletin stimulates apoptosis and DNA damage to impair progression of oral cancer cells [120]. Nobiletin suppresses breast cancer progression in a dose-dependent manner. Nobiletin enhances miRNA-200b expression to elevate apoptosis and pyroptosis in breast cancer cells [121]. Three experiments have shown role of nobiletin in affecting CSCs in tumor therapy. The Wnt/β-catenin signaling is a possible target of nobiletin in impairing CSC characteristics [122]. The invasion and angiogenesis are suppressed by nobiletin via targeting CSCs. Nobiletin (100 and 200 μM) reduces STAT3 expression via binding to CD36 to inhibit NF-κB signaling, leading to a significant decrease in migration and metastasis of CSCs [123]. In order to potentiate efficacy of nobiletin in CSC suppression, its co-administration with xanthohumol is recommended. This combination suppresses migration of CSCs and decreases CD44v6 expression. Furthermore, they induce apoptosis and cycle arrest at G2/M phase. This combination impairs progression of colorectal CSCs and enhances their sensitivity to oxaliplatin and 5-flouroruacil chemotherapy [124].

Chrysin

The chrysin is a new emerging anti-tumor agent capable of suppressing growth and invasion of tumor cells, and promoting their sensitivity to chemotherapy [125]. A combination of chrysin and daidzein inhibits colorectal cancer progression via suppressing ERK and Akt molecular pathways [126]. Chrysin-loaded nanostructures are capable of enhancing apoptosis via triggering p53 expression. Furthermore, chrysin-loaded nanoparticles suppress PI3K/JNK axis and inhibit tumor growth in vivo [127]. Chrysin enhances miRNA-let-7a expression, while it reduces H19 and COPB2 expression levels to impair progression of gastric cancer cells [128].
An experiment has applied micellar nanoparticles for co-delivery of chrysin and docetaxel in cancer therapy. The application of micelles promotes therapeutic effect of both docetaxel and chrysin. The micelles were biodegradable and capable of docetaxel and chrysin co-delivery in synergistic cancer chemotherapy. This combination enhanced ROS levels to induce apoptosis in colon CSCs. Furthermore, docetaxel- and chrysin-loaded micelles inhibit migration and invasion of CSCs, impairing colon cancer metastasis [129]. Another study has focused on a derivative of chrysin, known as CHM-04 that has 3.2-fold higher anti-tumor activity compared to chrysin. The CHM-04 suppresses colony formation capacity and invasion of breast CSCs and induces apoptotic cell death [130].

Apigenin

Similar to chrysin, apigenin is a potent anti-tumor agent against various cancers including breast cancer, lung cancer and gastric cancer [7]. Apigenin impairs progression of multiple myeloma cells in a dose-dependent manner. Apigenin is able to down-regulate STAT1 expression in suppressing COX-2/iNOS axis [131]. A combination of apigenin and hesperidin prevent DNA repair in breast tumor to potentiate DOX activity in cancer suppression [132]. Furthermore, apigenin reduces activity of ABCG2 and ABCC4 as drug efflux transporters to enhance internalization of doxorubicin in breast cancer and mediate apoptosis [133].
The activation of YAP/TAZ axis is responsible for CSC features in triple negative breast cancer. The apigenin administration (0–64 μM) decreases colony formation and self-renewal capacity of CSCs. Furthermore, apigenin reduces number of CD44+ cells in breast cancer. These anti-tumor activities were mediated via suppressing YAP/TAZ axis [134]. The presence of CD133 cells decreases potential of cisplatin in lung cancer suppression. The apigenin administration (10–30 μM) induces apoptosis in CSCs via p53 upregulation and enhances cisplatin cytotoxicity against lung tumors [135]. The stimulation of tumor-promoting factors enhances CSC features in tumors. For instance, activation of PI3K/Akt signaling leads to CSC features in prostate cancer via inducing NF-κB signaling. The apigenin administration (0–100 μM) suppresses PI3K/Akt/NF-κB axis to reduce Oct3/4 levels, as CSC markers in prostate cancer. Apigenin reduces survival of CSCs and enhances p21 and p27 upregulation. For decreasing viability of CSCs in prostate cancer therapy, apigenin induces both intrinsic and extrinsic apoptosis. Furthermore, apigenin disrupts invasion and metastasis of CSCs [136]. When prostate cancer cells obtain stemness and CSC features, they can easily achieve resistance to cisplatin chemotherapy. Similar to previous experiment, apigenin (15 μM) suppressed phosphorylation of PI3K, Akt and NF-κB in impairing CSC features. Furthermore, apigenin inhibited cell cycle progression of CSCs in prostate cancer via upregulating p21, CDK2, CDK4 and CDK6. By reducing Snail expression, apigenin impaired progression and invasion of CSCs. Apigenin enhanced capase-8, Apaf-1 and p53 levels, while it decreased Bcl-2, sharpin and survivin levels in triggering apoptosis in CSCs in prostate cancer. These impacts of apigenin promote sensitivity of prostate cancer cells to cisplatin chemotherapy [137]. For disrupting cancer stemness, apigenin (40 μM) decreases expression levels of CD44, CD105, Nanog, Oct4, VEGF and REX-1 [138]. These studies demonstrate that apigenin is a potent inhibitor of CSCs in tumor treatment that further experiments can focus on revealing more signaling pathways affected by apigenin [139].

Baicalein

The baicalein is another anti-tumor agent that can induce apoptosis in tumors via upregulating caspase-3, -8 and -9 levels. Baicalein reduces MMP-2 and MMP-9 expression levels to impair metastasis of cancer cells [140]. The baicalein prevents SNO-induced ezrin tension and reduces iNOS levels to impair progression of lung tumors [141]. By suppressing Akt and Nrf2 molecular pathways, baicalein induce both apoptosis and autophagy in gastric tumor cells [142]. Furthermore, baicalein mediates proteasomal degradation of MAP4K3 to impair progression of lung tumor cells [143].
The members of Sonic Hedgehog signaling including SHH, SMO and Gli2 undergo upregulation to mediate CSC features in pancreatic cancer. The overexpression of Sonic leads to upregulation of SOX2 and Oct4 as CSC markers in pancreatic cancer. The baicalein administration (0–300 μM) suppresses Sonic signaling to reduce SOX2 expression and impair CSC features in pancreatic cancer [144].

Wogonin

The wogonin has demonstrated great therapeutic impacts in pre-clinical experiments [145]. The wogonin is able to induce senescent in breast tumors via down-regulating TXNRD2 expression [146]. Wogonin diminishes expression levels of Notch1 at mRNA and protein levels to suppress growth and metastasis of skin cancer cells [147]. This section focuses on wogonin impact on CSCs.
As it was mentioned, natural products can enhance ROS levels to induce apoptosis in CSCs [148]. A same strategy is followed by wogonin in osteosarcoma therapy. For this purpose, wogonin (0–80 μM) enhances ROS levels to reduce expression of factors responsible for CSC features such as STAT3, Akt and Notch1 [149]. In addition to triggering apoptosis and reducing survival of CSCs in osteosarcoma, wogonin is able to affect invasion of CSCs. In this way, wogonin administration (0–80 μM) decreases MMP-9 expression to impair migration and invasion of CSCs in osteosarcoma (Figure 1) [150]. Interestingly, studies have only focused on osteosarcoma and more experiments on other tumor models should be performed to shed more light on anti-tumor activities of wogonin via targeting CSCs. Overall, flavones are potential agents in suppressing stemness and CSC features in tumors that have been summarized in Table 1.

5.1.2. Flavanones

Naringenin administration (100 μM) prevents colony formation, metastasis and EMT in breast tumor, while it induces apoptosis via upregulating p53 and ERα at mRNA level, as tumor-suppressor factors [152].
Naringin administration (300 μM) impairs CSC features in esophageal cancer and inhibit viability of CSCs [153].
Hesperetin (50–200 μM) also demonstrated potential in triggering apoptosis in breast CSCs. Hesperetin impairs invasion of breast CSCs and stimulates cell cycle arrest in breast CSCs. The hesperetin enhances p53 expression, while it down-regulates Notch1 expression in impairing stemness in breast CSCs [154].
The experiments evaluating role of flavanones in targeting CSCs in tumor suppression are limited and more studies are required to reveal true potential of these natural products in cancer therapy.

5.1.3. Flavonols

Fisetin

The fisetin is a natural flavonol that has demonstrated high anti-tumor activity and capacity in chemoprevention [155,156]. Fisetin mediates histone demethylation to induce DNA damage and impair progression of pancreatic tumor cells [157]. To date, just one experiment has evaluated role of fisetin in targeting CSCs that is included here. The proliferation, metastasis, angiogenesis and carcinogenesis of renal CSCs undergo inhibition by fisetin. The in vitro and in vivo experiments have shown role of fisetin in suppressing renal CSCs. The fisetin is able to decrease expression levels of cyclin Y and CDK16 via inhibiting 5hmC modification in CpG islands. Furthermore, fisetin can reduce TET1 levels in renal CSCs. Therefore, a significant decrease occurs in growth of renal CSCs and their angiogenesis and migration are suppressed [158].

Epigallocatechin 3-Gallate

The epigallocatechin 3-gallate (EGCG) is another naturally occurring compound that has demonstrated high potential in cancer therapy. The EGCG enhances Beclin and LC3 levels to induce autophagy in bladder cancer cells. Furthermore, EGCG induces apoptosis in bladder cancer via upregulating Bax, caspase-3 and caspase-9 levels [159]. The EGCG reduces Sonic Hedgehog expression to suppress PI3K/Akt axis, leading to apoptosis in colon cancer cells [160]. The EGCG impairs migration and metastasis of cervical tumor cells via down-regulating TGF-β and subsequent inhibition of EMT mechanism [161]. Therefore, EGCG can be considered as a promising agent in cancer therapy [162]. Noteworthy, EGCG targets CSCs in affecting cancer progression. The lung tumor cells demonstrate high expression level of CLOCK to improve their CSC features. Noteworthy, EGCG (0–40 μM) reduces mRNA and protein levels of CLOCK to impair CSCs features and suppress self-renewal capacity of CSCs in lung tumor therapy [163]. The miRNAs are considered as important modulators of CSCs in cancer [164,165,166,167]. The expression level of hsa-miRNA-485-5p undergoes down-regulation in serum of lung tumor patients, showing tumor-suppressor role of this miRNA. Restoring miRNA-485-5p expression impairs lung tumor proliferation and stimulates apoptosis via down-regulating RXRα expression. The EGCG administration (0–40 μM) enhances miRNA-485-5p expression to reduce RXRα expression, leading to a decrease in expression levels of CD133 and CD44 as CSC markers [168]. The CSC features in lung cancer mainly depend on upregulation of β-catenin and its inhibition suppresses lung tumor progression. An experiment has shown that EGCG (0–100 μM) inhibits Wnt/β-catenin axis to impair CSC features in lung cancer [169].
Similar to lung cancer, a number of experiments have focused on anti-tumor activity of EGCG in colorectal cancer via targeting CSCs. In this way, EGCG (0–40 μM) suppresses Wnt/β-catenin axis to reduce expression levels of CD133, CD44, ALDHA1, Nanog and Oct4 in impairing CSC features in colorectal tumor [170]. The colorectal tumor cells demonstrate resistance to various chemotherapeutic agents including oxaliplatin and 5-flourouracil [171,172]. The EGCG (0–400 μM) decreases expression levels of Notch1, Bmi1, Suz12 and EZH2, while it enhances expression levels of miRNA-34a, miRNA-145 and miRNA-200c in impairing CSC features in colorectal cancer and enhancing 5-flourouracil sensitivity [173]. The bladder cancer cells rely on Sonic Hedgehog signaling to enhance their CSC features and mediate their progression. Noteworthy, EGCG suppresses Sonic signaling to decrease CD44, CD133, Nanog, Oct4 and ALDH1 in bladder cancer therapy [174]. Therefore, EGCG is a potent inhibitor of CSCs in various cancers and for this purpose, it targets various molecular pathways including Notch and NF-κB signaling pathways (Table 2) [175,176,177].

5.1.4. Chalcones

Isoliquiritigenin

The isoliquiritigenin (ISL) is derived from licorice root and it is a modulator of molecular pathways in cancer suppression [178]. The ISL suppresses TGF-β signaling to reduce Smad3 expression, resulting in EMT inhibition and decreased endometrial cancer migration [179]. Furthermore, ISL enhances miRNA-200c expression to inhibit β-catenin signaling, leading to EMT suppression in triple-negative breast cancer [180]. A combination of ISL and docosahexaenoic acid induces apoptosis in colorectal cancer via enhancing ROS levels and mediating phosphorylation of JNK and ERK [181]. The ISL-loaded liposomes suppress colorectal tumor progression via inhibiting AMPK-mediated glycolysis [182]. This section focuses on anti-tumor activity of ISL via targeting CSCs. The ISL administration (25 μM) suppresses self-renewal and multidifferential capacities of CSCs in breast cancer. Furthermore, ISL inhibit growth and colony formation of breast CSCs. The ISL suppresses β-catenin signaling and decreases ABCG2 expression to enhance drug sensitivity of breast cancer cells and suppress CSC features [183]. The expression levels of CD44 and ALDH1 undergo down-regulation upon ISL administration (0–50 μM) in oral cancer. The ISL suppresses colony formation and invasion of CSCs in oral tumor. In suppressing CSCs, ISL also decreases expression level of GRP78 at mRNA and protein levels. Furthermore, by reducing ABCG2 expression, ISL enhances sensitivity of oral CSCs to cisplatin chemotherapy [184]. Another experiment reveals that ISL administration (25 and 50 μM) enhances SIF1 expression via demethylation to suppress DNMT1 methyltransferase. Furthermore, ISL simultaneously suppresses β-catenin signaling to eradicate CSCs in breast cancer therapy [185].

5.1.5. Isoflavonoids

Daidzein

An experiment has focused on a derivative of daidzein, known as N-t-boc-Daidzein. This derivative is able to suppress CSC features in ovarian cancer in concentration (0–10 μM)- and time (0–70 h)-dependent manner. This derivative induces apoptosis in CSCs via upregulating caspase-3, -8 and -9 levels. Suppressing growth and survival of CSCs is mediated by N-t-boc-Daidzein. In addition to triggering caspase cascade, N-t-boc-Daidzein mediates mitochondrial depolarization and decreases Akt expression via degradation to impair CSC features in ovarian tumor [186].

Genistein

The genistein is an isoflavonoid compound that has potent anti-tumor activity. The genistein administration decreases chance of mammary cancer recurrence [187]. The glioblastoma cells exposed to radiation, demonstrate enhanced metastasis. The genistein is able to suppress Akt2 pathway in reducing invasion and migration of glioblastoma cells [188]. The genistein administration reduces hsa-circ-0031250 expression to promote miRNA-873-5p expression, leading to lung cancer suppression and decreased proliferation rate [189]. Both inflammation and GSK-3 are suppressed by genistein to inhibit ovarian tumor progression in vivo [190]. In order to improve genistein capacity in cancer suppression, nanoparticles have been developed for its targeted delivery [191,192].
The Sonic Hedgehog signaling enhances CSC features in nasopharyngeal cancer. The administration of genistein (0–100 μM) inhibits sonic signaling and reduces expression level of CSC markers including CD44, ALDH1, Oct4 and Nanog to impair nasopharyngeal progression [193]. A same strategy occurs in renal cancer, so that genistein administration (0–90 μM) suppresses Sonic Hedgehog signaling to impair growth and colony formation of renal CSCs and stimulate apoptotic cell death [194]. The genistein is able to mediate differentiation of CSCs in a paracrine mechanism to suppress cancer progression. An experiment on breast tumor has shown genistein administration (2 μM or 40 nM) stimulates PI3K/Akt and MEK/ERK molecular pathways to induce differentiation of CSCs in breast cancer [195]. Another experiment also reveals that low doses of genistein (15 μM) inhibits self-renewal capacity of gastric CSCs in vitro and in vivo [196]. The capacity of genistein in suppressing CSC features in gastric cancer is attributed to down-regulating FOXM1 expression that subsequently, reduces expression levels of CD133, CD44 and Nanog. Furthermore, by inhibiting FOXM1 expression, genistein reduces Twist1 expression that is in favor of inhibiting EMT in CSCs via reducing N-cadherin levels and enhancing E-cadherin levels [197]. In addition to aforementioned molecular pathways, genistein suppresses Gli1 expression to impair CSC features in gastric cancer [198]. Some of the molecular pathways are similar among various cancers. For instance, previous research demonstrated that genistein suppresses Gli1 pathway in reversing CSC features in gastric cancer. Another experiment in prostate cancer reveals that genistein is able to suppress Gli1 pathway in inhibiting CSC characteristics [199]. Furthermore, it was mentioned that genistein suppresses nasopharyngeal CSCs via inhibiting Sonic signaling. A similar study reveals Hedgehog signaling inhibition and subsequent Gli1 down-regulation by genistein in prostate cancer stemness suppression [199]. Genistein also suppresses Hedgehog signaling in impairing CSC features in breast cancer [200]. Overall, studies highlight the fact that genistein is a potent inhibitor of cancer progression via targeting CSCs (Figure 2, Table 3) [200,201,202,203,204].

5.2. Pomegranate and Its Bioactive Compounds

5.2.1. Ellagic Acid

Ellagic acid is a polyphenolic compound that can suppress cancer progression. The ellagic acid increases sensitivity of bladder cancer cells via inhibiting drug transporters and EMT mechanism [206]. The p21 and p53 levels undergo upregulation by ellagic acid to induce apoptosis in prostate cancer cells [207]. The ellagic acid stimulates autophagy via Akt down-regulation and AMPK upregulation to reduce viability and survival of ovarian cancer cells [208]. The ellagic acid is a potent agent in suppressing drug resistance via down-regulating P-glycoprotein (P-gp_levels [209]. This section focuses on anti-tumor activity of ellagic acid based on targeting CSCs.
Ellagic acid is able to target CSCs in tumor eradication. An experiment has shown role of ellagic acid in breast cancer therapy via targeting CSCs. The β-catenin stabilization is responsible for CSC features in breast cancer and ACTN4 functions as upstream mediator. Silencing ACTN4 impairs growth, migration and colony formation capacities in breast cancer. The ellagic acid administration (0–50 μM) reduces ACTN4 expression to inhibit β-catenin signaling in CSCs, impairing breast cancer progression [210]. The ALDH level as an inducer of drug resistance and CSC marker undergoes down-regulation by a combination of ellagic acid and urolithins in colon CSCs [211]. Although just two experiments have evaluated role of ellagic acid in targeting CSCs, these obviously demonstrate role of ellagic acid in tumor treatment and more studies are required in this case.

5.2.2. Caffeic Acid

Caffeic acid (CA) is a phenolic acid compound that is isolated from natural sources such as tea and has various therapeutic activities. The anti-tumor activity of CA and its derivatives have been under attention, summarized in this review [212]. The CA can mediate ROS overgeneration to induce cell death in cervical cancer cells [213]. Furthermore, CA suppresses P-gp function to mediate chemosensitivity of tumors [214]. Noteworthy, CA derivatives are also able to induce apoptosis via survivin down-regulation and caspase-3 and -9 upregulation [215,216].
The colorectal CSCs that have high ability in mediating radioresistance feature. The CD44+ and CD133+ CSCs in colorectal cancer have self-renewal capacity and carcinogenesis impact. The CA administration inhibits PI3K/Akt axis that is responsible for CSCs features in colorectal cancer, leading to enhanced radiosensitivity and reduced carcinogenesis impact [217]. The keratinocytes demonstrate high expression level of NF-κB that is beneficial for upregulating Snail expression and mediating migration and CSCs features. The CA administration (0–100 μM) enhances p38 expression to reduce potential of NF-κB in binding to Snail. Therefore, invasion and CSC features undergo inhibition [218]. The CA has a naturally occurring derivative, known as caffeic acid phenethyl ester (CAPE) that can suppress self-renewal capacity and progenitor formation in breast cancer in a dose-dependent manner [219]. The expression of miRNAs is mainly affected by CA in cancer therapy. The TGF-β/Smad2 axis mediate CSC features in tumors. The CA administration (20 μM) increases miRNA-184a expression via DNA methylation, as a tumor-suppressor factor. Then, overexpressed miRNA-184a inhibits Smad2 expression by binding to its 3′-UTR, impairing CSC features [220]. Hence, CA and its derivative CAPE are potential anti-tumor agents in cancer treatment via targeting CSCs.

5.2.3. Luteolin

The luteolin is a natural flavonoid that can suppress cancer progression via regulating autophagy [221]. The luteolin suppresses Akt/mTOR axis to reduce MMP-9 expression, impairing metastasis and invasion of breast cancer cells [222]. The luteolin suppresses EMT mechanism via down-regulating YAP/TAZ, resulting in a significant decrease in breast cancer invasion [223]. Furthermore, lutelon enhances death receptor 5 (DR5) expression and mediates mitochondrial fission to promote TRAIL sensitivity of lung tumors [224]. To date, just one experiment has evaluated role of luteolin in targeting CSCs. Mechanistically, IL-6 stimulates STAT3 signaling to promote CSC features in oral cancer and mediate their resistance to radiotherapy. The luteolin administration (0–40 μM) suppresses IL-6/STAT3 axis to impair self-renewal capacity of CSCs and reduce expression level of CSC markers including ALDH1 and CD44 [225].

5.2.4. Quercetin

Quercetin is a flavonol present in fruits and vegetables such as grape, anion, apple, berries and broccoli [226]. The diet intake of quercetin is estimated to be higher than 70% of all flavonol intake [227]. Quercetin has different pharmacological activities including immunomodulatory, hepatoprotective, neuroprotective and nephroprotective that can be mediated via regulating autophagy [228]. Furthermore, quercetin can be considered as a protective agent against ischemic/reperfusion injury [229]. Regardless of its protective impacts, quercetin is suggested to display significant anti-tumor activity. Quercetin reduces CDK6 expression to impair progression of breast and lung cancer cells [230]. Quercetin enhances reactive oxygen species (ROS) levels to induce ferroptosis. Furthermore, quercetin promotes cell death via lysosome activation [231]. Angiogenesis and metastasis of esophageal cancer cells was suppressed by quercetin via reducing expression levels of VEGF-A, MMP-2 and MMP-9 [159]. Quercetin reverses multidrug resistance and using nanoparticles for its delivery enhances its potential in cancer suppression [232]. This section focuses on CSC targeting by quercetin in cancer therapy.
The PI3K/Akt/mTOR axis is responsible for growth and progression of CSCs in breast cancer. The quercetin administration (0–200 μM) suppresses PI3K/Akt/mTOR axis in induce cell cycle arrest in G1 phase in breast cancer cells. The quercetin impairs viability, colony formation and mammosphere formation in CD44+ stem cells in breast cancer and triggers apoptosis [233]. Another experiment also reveals role of quercetin in eradicating CSCs in breast cancer. The quercetin administration (0–200 μM) suppresses growth, metastasis and self-renewal capacity of CSCs in breast cancer. For this purpose, quercetin reduces expression levels of ALDH1, CXCR4, mucin 1 and epithelial cell adhesion molecule (EpCAM) [234]. The nuclear translocation of Y-box binding protein 1 (YB-1) is responsible for CSCs features in breast cancer. For reversing multidrug resistance in breast cancer, quercetin decreases expression level of P-gp as an efflux transporter. More importantly, quercetin inhibits nuclear translocation of YB-1 to suppress CSCs features in breast cancer cells and enhance their sensitivity to doxorubicin, paclitaxel and vincristine [235]. It is worth mentioning that by down-regulating expression levels of P-gp, BCRP and MRP1, quercetin (0–2 μM) increases internalization of doxorubicin in breast cancer cells, resulting in eradication of CSCs [236].
Another cancer that can be affected by quercetin is pancreatic cancer. For enhancing potential of quercetin in suppressing CSCs, its combination therapy has been used. For this purpose, a combination of sulforaphane (SFN) (0–10 μM) and quercetin (20 μM) effectively inhibits self-renewal capacity of CSCs [237]. In pancreatic cancer, CSC divisions tend to be symmetric. The low expression of miRNA-200b-3p induces Notch signaling to make daughter cells be symmetric. However, quercetin administration (50 μM) enhances expression level of miRNA-200b-3p as tumor-suppressor to reduce Notch expression, resulting in asymmetric divisions in CSCs [238]. A combination of SFN, quercetin and catechins reduces ALDH1 expression in pancreatic cancer, as CSC marker. For this purpose, SFN, quercetin and catechins enhances miRNA-let-7 expression to down-regulate K-ras expression [239]. Hence, quercetin and its combination with anti-tumor agents synergistically suppress CSC features in pancreatic cancer [240].
The induction of Notch1 signaling and enhanced CSC features lead to radioresistance in colon cancer. The quercetin administration (20 μM) suppresses Notch1 signaling and reduces expression levels of CSC markers including SOX9, CD133 and CD44 [241]. The CD133+ colorectal cancer cells seem to be responsible for triggering drug resistance in colorectal cancer. The quercetin administration (10–100 μM) induces apoptosis and cycle arrest (G2/M phase) in CD133+ cells to enhance sensitivity of colorectal tumor cells to doxorubicin chemotherapy [242]. Therefore, quercetin targets CSCs to increase therapy response of colorectal cancer cells.
The prostate cancer (PCa) is the most common cancer and second malignant tumor in men [2,243]. The various molecular pathways are responsible for PCa progression and various anti-tumor compounds and genetic tools have been utilized for its treatment [20,244,245,246,247]. The quercetin has shown capacity in suppressing growth and invasion of PCa stem cells (CD44+/CD133+ cells). The midkin (MK) pathway is responsible for CSC features in PCa. Co-application of quercetin and siRNA-MK enhances potential in suppressing CSCs. Furthermore, this combination induces apoptosis and G1 arrest in CSCs. The molecular pathways including PI3K/Akt, ERK1/2, p38, ABCG2 and NF-κB undergo down-regulation by quercetin and siRNA-MK in inhibiting CSCs in PCa [248]. Furthermore, a combination of quercetin and EGCG induces apoptosis (via caspase-3/7 upregulation and down-regulating Bcl-2, survivin and XIAP) in CSCs and prevents their growth and invasion. The reduced metastasis of CSCs by quercetin and EGCG is attributed to suppressing epithelial-to-mesenchymal transition (EMT) via down-regulating Snail, vimentin, Slug and β-catenin [249]. Therefore, quercetin is a well-known compound in eradicating CSCs in preventing cancer progression (Figure 3, Table 4) [250,251].

5.3. Carotenoids

5.3.1. Astaxanthin

The astaxanthin is derived from algae and has anti-tumor activity. Astaxanthin The astaxanthin impairs gastric cancer development and progression via suppressing inflammatory storm [252]. Astaxanthin induces apoptosis and promotes PARPγ expression to reduce esophageal cancer progression [253]. The pontin overexpression is responsible for CSC features in breast cancer. Suppressing pontin expression impairs colony formation and CSC characteristics in breast cancer. The astaxanthin administration (80 and 100 μM) reduces pontin expression and impairs CSC features via down-regulating Oct4, Nanog and mutp53 levels [254].

5.3.2. β-Carotene

The β-carotene is another member of carotenoids that is able to effectively suppress cancer progression. The β-carotene suppresses IL-6/STAT3 axis and inhibits M2 polarization of macrophages to impair colon tumor progression [255]. β-carotene induces apoptosis and reduces antioxidant markers to impair breast tumor progression [256]. A combination of β-carotene and lycopene suppresses esophageal cancer progression via down-regulating COX-2 and cyclin D1 expression levels [257]. For promoting potential of β-carotene in breast cancer suppression, lipid–polymer nanoparticles have been developed for its targeted delivery [258]. Compared to astaxanthin, more experiments have focused on role of β-carotene in targeting CSCs. The β-carotene administration (20 and 40 μM) reduces growth and colony formation capacities of colon CSCs. The β-carotene reduces DNMT3A expression at mRNA levels and prevents DNA methylation. Furthermore, β-carotene promotes histone H3 and H4 acetylation levels in impairing CSC progression [259]. The self-renewal capacity of CSCs undergoes a decrease by β-carotene in neuroblastoma cells and this leads to enhanced sensitivity of tumor cells to cisplatin chemotherapy [260]. Furthermore, in vivo experiment on xenograft model has shown role of β-carotene in suppressing CSC features in neuroblastoma [261]. The Oct3/4 and DLK1 undergo down-regulation by β-carotene in decreasing CSC features and stemness of neuroblastoma cells [262]. The potential of β-carotene in reducing DLK1 expression and impairing CSC features is mediated via retinoic acid receptor β [263].

5.4. Sulforaphane

The sulforaphane (SFN) is an isothiocyanate and is present in various vegetables including cabbage broccoli, cauliflower and Brussels. The SFN is generated upon glucoraphanin hydrolysis. The SNF application in cancer therapy has witnessed a growing increase [264]. A recent experiment demonstrates that SFN reduces expression level of H19, as a lncRNA to suppress growth and invasion of pancreatic cancer cells [265]. The SFN is able to suppress tubulin polymerization in triggering apoptosis and cell cycle arrest in glioblastoma cells [266]. The expression level of FAT-1 undergoes down-regulation by SFN to impair proliferation and invasion of bladder cancer cells [267]. The SFN administration is of interest in increasing sensitivity of breast cancer cells to doxorubicin chemotherapy via inhibiting myeloid-derived suppressor cells [268]. Therefore, SFN is a potent anti-tumor agent [269] and this section focuses on CSC targeting by SFN in cancer treatment.
The activation of Sonic Hedgehog signaling promotes stemness of gastric cancer cells. The SFN administration (0–10 μM) stimulates apoptosis in CSCs and inhibits their growth to suppress gastric cancer proliferation. For this purpose, SFN inhibits Sonic signaling [270]. The upregulation of TAp63α occurs in colorectal tumor spheres with CSC features. Noteworthy, TAp63α enhances self-renewal capacity and CSC markers in colorectal tumor. Investigation of molecular pathways reveals that SFN increases Lgr5 expression by binding to its promoter, leading to β-catenin signaling activation. The SFN administration (0–10 μM) reduces TAp63α expression to inhibit expression of CSC markers such as CD133, CD44, Nanog and Oct4 [224].
Human tumor necrosis factor (TNF)-related apoptosis ligand (TRAIL) is suggested to be involved in triggering apoptosis in tumor cells [271]. However, experiments have shown capacity of tumor cells in obtaining resistance to TRAIL-mediated apoptosis [272,273,274,275]. Furthermore, TRAIL can surprisingly enhance progression of cancer cells via inducing NF-κB signaling [276,277]. An experiment has shown synergistic impact between TRAIL and SFN in PCa therapy. Both TRAIL and SFN are able to suppress CSCs in PCa, and SFN has higher ability compared to TRAIL. However, TRAIL induces NF-κB signaling and promotes PCa progression. The SFN administration (10 μM) decreases expression level of NF-kB, CXCR4, Jagged1, Notch-1, SOX2, Nanog and ALDH1 to impairs CSC features and stemness in PCa [278]. It is worth mentioning that down-regulation of ALDH1, c-Rel and Nothc-1 as CSC markers by SFN, significantly elevates sensitivity of prostate and pancreas cancer cells to cisplatin, gemcitabine and 5-flourouracil chemotherapy [279]. The stimulation of NF-κB signaling is responsible for ALDH1 upregulation and enhanced CSC features in pancreatic cancer. Therefore, NF-κB inhibition by SFN (20 μmol/L) impairs CSC features in pancreatic cancer cells and enhances their sensitivity to sorafenib chemotherapy [280]. Overall, the experiments advocate the fact that SFN is a potent agent in targeting CSC and suppressing stemness of cancers (Figure 4, Table 5) [281,282,283,284,285].

5.5. Curcumin

The curcuma longa is a medicinal plant belonging to family of Zingiberaceae family that has a well-known bioactive compound, curcumin that can be isolated from its rhizome [294,295]. The curcumin and other curcuminoids lead to yellow color of rhizome. Although content of curcuminoids in rhizome of curcuma longa is various, it seems that curcumin is the main component and comprises 77% [296]. The curcumin has demonstrated various therapeutic activities that among them, anti-cancer potential is of importance [297]. The curcumin administration is beneficial in suppressing proliferation and metastasis of cancer cells. A recent experiment has shown role of curcumin in down-regulating NF-κB, ERK, MMP-2 and MMP-9, while it can enhance p38 and JNK levels to induce cell death and prevent invasion of leukemia cells [298]. Recently, attention has been directed towards chemopreventive role of curcumin in cancer therapy [299]. The curcumin- and docetaxel-loaded micellar nanoparticles effectively suppress progression of ovarian cancer cells via suppressing angiogenesis and inducing apoptosis. The curcumin is able to sensitize ovarian cancer cells to docetaxel chemotherapy [300]. Due to poor bioavailability of curcumin, nanostructures are mainly applied for curcumin delivery alone or its combination with other anti-tumor agents to exert synergistic cancer therapy [31,301]. A fibrin matrix has been developed for prolonged release of curcumin and suppressing cancer metastasis. As studies have shown role of the nanoplatforms for promoting therapeutic activity of curcumin, future clinical trials can be performed [302]. The current section focuses on anti-tumor activity of curcumin based on targeting CSCs.
The capacity of curcumin in cancer suppression via targeting CSCs is attributed to affecting various molecular pathways. The activation of Wnt/β-catenin and Sonic Hedgehog molecular pathways results in CSC features in lung cancer and enhancing tumor progression. The curcumin administration (0–40 μM) induces apoptosis in CSCs and diminishes their proliferation. Furthermore, curcumin reduces CSC hallmarks such as Nanog, Oct4, CD133, CD44 and ALDH1A1. Mechanistically, these anti-tumor activities of curcumin are mediated via inhibiting Wnt and Hedgehog signaling pathways [303]. In addition, nuclear factor-kappaB (NF-κB) pathway also participates in enhancing survival of CSCs [304]. The down-regulation of NF-κB signaling impairs CSC features in bladder cancer [305]. In liver cancer cells, curcumin reduces their proliferation and colony formation capacities as well as inhibiting CSCs features. The curcumin capacity in suppressing CSCs features in liver cancer is pertained to inhibiting NF-κB signaling [306]. Hence, identification of molecular pathways affected by curcumin in suppressing CSC features can broaden our understanding towards underlying mechanisms of its anti-tumor activity.
The signal transducer and activator of transcription 3 (STAT3) is another tumor-promoting factor related to cancer progression [307,308,309]. The upregulation of STAT3 enhances growth and invasion of tumor cells and mediates their therapy resistance [310,311,312,313,314,315,316]. The activation of STAT3 signaling promotes CSC features in tumors and its inhibition, for instance by acetaminophen, reduces CSC markers and colony formation capacity [317,318]. The curcumin administration (0–40 µM) decreases levels of CD44, ALDH, SOX2, Nanog and c-Myc as CSC markers to impairs thyroid cancer progression and induce apoptosis. Furthermore, by reducing CSC features, curcumin enhances potential of cisplatin in thyroid cancer suppression [319]. Similar phenomenon occurs in bladder cancer, so that curcumin administration (0–50 µM) decreases CD44, CD133, ALDH1, Nanog and Oct4 to impair CSC characteristics in bladder cancer and induce apoptosis and proliferation inhibition. The investigation of molecular pathways reveals that these anti-tumor activities are mediated via sonic signaling inhibition [320]. The interesting point is the curcumin and CD44 coupling in cancer therapy. An experiment demonstrates that colon cancer cells overexpressing CD44 as CSC marker are more sensitive to curcumin compared to CD44- cells. The curcumin-CD44 coupling stimulates apoptosis in colorectal stem cells via preventing influx of glutamine into cancer cells and reducing its intracellular accumulation [321].
In order to potentiate anti-tumor activity of curcumin, its combination with other tumor-suppressor agents is performed. A combination of curcumin and quinacrine can be beneficial in impairing breast cancer progression via targeting CSCs. The quinacrine is able to induce DNA damage in cancer cells, but its intracellular accumulation should be improved. In this case, curcumin is helpful and by suppressing ABCG2 activity via occupying its ligand-binding site, enhances internalization of quinacrine, resulting in cell death and DNA damage in breast CSCs [322]. Furthermore, curcumin also has capacity of inducing DNA damage in CSCs [323]. The curcumin can negatively affect colony formation capacity of lung cancer cells. It is suggested that curcumin prevents self-renewal capacity of CSCs to suppress colony formation of lung cancer cells [323]. Therefore, curcumin is a potent suppressor of CSCs in lung cancer and for this purpose, it can inhibit JAK2/STAT3 axis to impair CSC features [324]. Based on previously discussed molecular pathways, both STAT3 and NF-κB pathways are suppressed by curcumin in targeting CSCs. Noteworthy, STAT3 can function as upstream mediator of NF-κB and mediates its activation via enhancing IKKα stability [325]. A combination of curcumin (10 μM) and epigallocatechin gallate (10 μM) suppresses STAT3/NF-κB axis to impair CSC features and reduce number of CD44+ cells [326]. In addition to curcumin, its derivatives have been also capable of targeting CSCs. The colon cancer cells positive for ALDH and CD133 demonstrate activation of STAT3 signaling and its phosphorylation. An analogue of curcumin, known as GO-Y030 can suppress STAT3 phosphorylation to impair CSC features in colon cancer and prevent its progression [327]. Therefore, curcumin and its derivatives suppress proliferation and invasion of tumors via targeting CSCs. Furthermore, they can enhance sensitivity of cancer cells to therapies by targeting CSCs [328]. Interestingly, curcumin can be advantageous in enhancing sensitivity of cancer cells to radiotherapy [329]. The breast cancer stem-like cells have the ability of obtaining radioresistance. A combination of curcumin and glucose gold nanoparticles induces apoptosis in CSCs and reduces expression levels of hypoxia inducible factor-1α (HIF-1α) and HSP90 in sensitizing breast cancer cells to radiotherapy [330]. Therefore, curcumin is a versatile agent in suppressing CSC features in cancer and various molecular pathways such as Wnt, Sonic, STAT3 and NF-κB, among others, are affected to reduce CSC markers such as CD44, ALDH and CD133 (Figure 5, Table 6) [331,332,333,334,335,336,337,338].

5.6. Resveratrol

The resveratrol (Res) is a well-known compound in treatment of different ailments including diabetes, cancer and neurological disorders [340,341]. The recent years have witnessed an increase in attention towards Res application in cancer therapy. Res induces apoptosis via impairing mitochondrial function in cancer cells. Furthermore, tumor-promoting factors such as NF-κB, COX-2 and PI3K undergo down-regulation by Res in tumor suppression [342]. A combination of Res and radiation stimulates apoptosis in breast cancer via Bcl-2 down-regulation and Bax upregulation [50]. Res suppresses phosphorylation of STAT3 at tyrosine 705 to impair metastasis of cervical cancer cells [343]. Down-regulation of HPV E6 and E7 by Res leads to apoptosis and cell cycle arrest in cervical cancer [344]. Furthermore, Res inhibits progression and growth of PCa cells via suppressing PI3K/Akt signaling [345]. The c-Myc as downstream target of PI3K/Akt axis is suppressed by Res in lung cancer therapy [346]. Loading Res on nanostructures significantly promotes its potential in thyroid cancer suppression. Furthermore, Res-loaded nanoparticles decrease tumor volume up to 55% in thyroid cancer in vivo [347].
The tumor sphere formation capacity in renal cancer is mediated by CSCs and Sonic signaling plays a significant role in this case. The Res administration (0–30 μM) down-regulates Sonic signaling to inhibit CSCs features in renal cancer, leading to apoptosis and proliferation inhibition [348]. As it was mentioned earlier, Res is loaded on nanoparticles to promote its anti-tumor activity [349,350,351]. Another promising strategy for enhancing bioavailability of Res in synthesizing analogues with superior activities. The pterostilbene is a derivative of Res that has higher bioavailability compared to Res. The pterostilbene is able to reduce expression levels of CD133, Oct4, SOX2, Nanog and STAT3 in impairing CSC features and retarding progression of cervical cancer [352]. Furthermore, Res is able to suppress Wnt/β-catenin in reducing CSC features in breast cancer. It is worth mentioning that Res enhances LC-3II, Beclin-1 and ATG7 expression levels in triggering autophagy in CSCs and reducing breast cancer progression [353]. It is worth mentioning that autophagy has both tumor-promoting and tumor-suppressor roles in cancer [221,354,355] and its induction by Res requires more clarification.
The STAT3 signaling is considered as a tumor-promoting factor in osteosarcoma. The upregulation of STAT3 by VEGFR2 results in metastasis of osteosarcoma cells [356]. Furthermore, induction of JAK2/STAT3 axis by exosomal LCP1 leads to carcinogenesis and migration of osteosarcoma cells [357]. The Res administration (0–3 μM) decreases cytokine generation and suppresses JAK2/STAT3 axis to impair CSCs features in osteosarcoma via CD133 down-regulation [358]. Another molecular pathway that is responsible for CSCs features is nutrient-deprivation autophagy factor-1 (NAF-1). The Res inhibits NAF-1 signaling to reduce CSC features including SOX2, Oct4 and Nanog in pancreatic cancer treatment [359]. The interesting point is the interaction between tumor microenvironment (TME) components and molecular pathways in providing CSCs features. The T lymphocytes and fibroblasts cells present in TME can effectively induce NF-κB signaling and upon nuclear translocation, NF-κB significantly enhances growth, metastasis and survival of CSCs in colorectal cancer. The TNF-β and TGF-β3 are secreted by T lymphocytes and fibroblasts in inducing NF-κB signaling. Noteworthy, Res (0–10 μM) prevents secretion of TNF-β and TGF-β3 by lymphocytes and fibroblasts in reducing CSC features in colorectal cancer [360]. Another experiment reveals that cancer-associated fibroblasts (CAFs) existing in TME are able to reduce SOX2 expression and suppress stemness in breast cancer via reducing CSC features [361].
Another strategy that can be followed by Res, is the induction of endothelial differentiation. For this purpose, Res and sulindac stimulate vascular endothelial cadherin (VE-cadherin) and von Willebrand factor (vWF) in triggering trans-differentiation in CSCs and mediating their transformation into endothelial lineage [362]. Recently, attention has been directed towards using siRNA and natural compounds in synergistic cancer therapy [312]. An experiment has shown that RAD51 has oncogenic role and enhances CSC features in cervical cancer. A combination of Res and RAD51-siRNA induces apoptosis in HeLa cells and prevents cervical cancer progression [363]. Overall, experiments reveal role of Res in suppressing CSCs in cancer progression (Figure 6, Table 7) [364,365,366,367].

5.7. Berberine

The berberine (BBR) is an isoquinoline alkaloid mainly derived from Coptis chinesis with great therapeutic impacts [375]. In addition, BBR can be isolated from other Berberis plants such as Berberis julianae and Scutellaria baicalensis [376]. Although BBR has pharmacological activities including antioxidant, anti-inflammatory, anti-diabetes, hepatoprotective and renoproective, among others, much attention has been directed towards its anti-tumor activities [377,378,379,380]. The BBR induces apoptosis, autophagy and cycle arrest (G1 phase) in colon cancer cells. Mechanistically, BBR enhances PTEN expression, while it reduces expression of PI3K, Akt and mTOR [151]. Furthermore, BBR suppresses fatty acid metabolism and diminishes extracellular vesicle production in favor of cancer growth suppression [381]. The BBR is able to decrease expression level of IGF2BP3 in triggering cycle arrest (G0/G1 phase) in colorectal cancer cells [382]. The current section emphasizes on BBR function in affecting CSCs.
The pancreatic cancer treatment is an increasing challenge for physicians, as the tumor cells have high growth and migration capabilities [383,384]. Furthermore, they can develop drug resistance and risk of recurrence is present in pancreatic cancer patients. The CSCs play a significant role in aforementioned processes [385,386,387,388]. An experiment has shown that BBR (15 µM) reduces expression levels of SOX2, Nanog and POU5F1 as CSC markers to impair resistance of pancreatic cancer cells to gemcitabine and suppress their progression [389]. One of the targets of BBR in cancer therapy is miRNAs. Overall, the expression level of tumor-promoting miRNAs undergoes down-regulation by BBR, while an increase occurs in expression profile of tumor-suppressor miRNAs [390,391,392]. The oral CSCs have self-renewal capacity and demonstrate high colony formation and migration abilities that are in favor of triggering drug resistance. The miRNA-21 overexpression plays a tumor-promoting role in oral CSCs. The BBR (10 μM) reduces expression level of miRNA-21 to impair self-renewal capacity of CSCs and decrease ALDH1 expression. Then, a significant increase occurs in sensitivity of oral CSCs to cisplatin and 5-flourouracil chemotherapy [393].
The glioma-associated oncogene-1 (Gli1) is a tumor-promoting factor that its upregulation enhances growth and invasion of colorectal cancer cells [394]. Noteworthy, Gli1 participates in enhancing CSCs features and mediating tumor progression [395]. Therefore, its inhibition by BBR can exert anti-tumor activities. A recent experiment has shown that chemotherapy enhances ovarian cancer progression via enhancing CSCs features. The Gli1 and its downstream target BMI1 are involved in promoting CSCs features in ovarian cancer. The BBR administration down-regulates Gli1 expression to inhibit BMI1, leading to a decrease in CSC features [396]. Overall, CD133, β-catenin, n-Myc, nestin, SOX2 and Notch2 undergo down-regulation by BBR in suppressing CSC features [397]. Furthermore, BBR co-administration with Dodecyl-TPP (d-TPP) can synergistically suppress CSC features in breast cancer [205]. Moreover, to selectively target CSCs, nanostructures can be utilized [398,399]. In an effort, BBR was loaded on liposomal nanocarriers and results demonstrated potential of these nanocarriers in crossing over CSC membrane. Then, they can reduce expression level of ABCC1, ABCC2, ABCC3 and ABCG2. Furthermore, BBR-loaded liposomes selectively internalize in mitochondria and induce apoptosis via triggering mitochondrial membrane potential loss, enhancing Bax expression and reducing Bcl-2 level. Then, cytochrome C (cyt C) release occurs and promotes caspase-3 and -9 expressions to mediate apoptosis in CSCs and suppress breast cancer progression. Furthermore, in vivo experiment on xenografts in nude mice also has shown potential of BBR-loaded liposomes in suppressing tumor growth (Table 8) [400].

5.8. Ginseng and Its Derivatives

The ginsenosides are derivatives from ginseng that have therapeutics impacts including anti-diabetes, anti-inflammatory and neuroprotective, and among them, anti-tumor activity of ginsenosides is of importance [401,402,403]. The ginsenoside Rg3 is able to suppress progression of colorectal tumor cells and enhance their sensitivity to oxaliplatin and 5-flouroruacil chemotherapy. Mechanistically, ginsenoside Rg3 (200 μM) reduces levels of CD24, CD44 and EpCAM to inhibit stemness and population of CSCs [404]. The ginsenoside Rh2 also suppress skin cancer progression in a concentration-dependent manner (0–1 mg/mL) and induces autophagy to suppress β-catenin signaling and impair CSC features in skin cancer. Autophagy inhibition abrogates capacity of ginsenoside Rh2 in suppressing β-catenin signaling and CSC features in skin cancer. Therefore, autophagy induction by ginsenoside Rh2 is of importance for inhibiting β-catenin-mediated CSC features in skin cancer [405]. However, a special attention should be directed towards autophagy, as it has both tumor-promoting and tumor-suppressor roles in cancer [355]. An experiment demonstrates that ginsenoside F2 (0–120 μM) stimulates apoptosis and autophagy in breast CSCs. Noteworthy, autophagy plays as a pro-survival mechanism and autophagy inhibition increases ginsenoside F2 capacity in mediating apoptosis in CSCs [406]. The ginsenoside Rb1 and compound K are able to suppress self-renewal capacity of CSCs in ovarian cancer. Furthermore, Rb1 and compound K enhance sensitivity of ovarian cancer cells to cisplatin and paclitaxel chemotherapy via suppressing CSCs. The drug sensitivity activity of Rb1 and compound K are mediated via suppressing β-catenin signaling and subsequent down-regulation of ABCG2 and P-gp. Furthermore, Rb1 and compound K suppress CSC migration and invasion via EMT inhibition [407]. For suppressing self-renewal capacity of CSCs, ginsenoside Rg3 (0–100 μM) suppresses Akt signaling. Furthermore, ginsenoside Rg3 reduces HIF-1α expression to down-regulate SOX-2 and Bmi-1 expression levels, leading to decreased stemness and CSC features in breast cancer [408]. Overall, ginsenosides similar to genistein, are able to suppress cancer progression via affecting CSCs [409,410,411]. Besides, ginsenosides are derived from ginseng extract and it has been reported that ginseng extract is also capable of suppressing CSC features in cancer (Figure 7, Table 9) [412,413]. Figure 8 and Figure 9 show the chemical structures of selected phytochemicals discussed in the current review.

6. Conclusions and Remarks

A variety of factors have been found to be responsible for tumor progression. The CSCs role in tumor progression has been confirmed in different experiments. In addition, molecular pathways participating in CSC feature suppression/induction have been shown. The CSCs enhance stemness of tumor cells and can mediate their growth and migration. Furthermore, CSCs are able to reduce sensitivity of tumor cells to chemotherapy and radiotherapy. The CD44, CD33, ALDH1, Nanog, SOX2 and Oct2 are the most well-known CSC markers. The overexpression of these factors significantly enhances stemness of tumors. Various molecular pathways are able to enhance cancer progression and stemness that include STAT3, NF-κB, Wnt/β-catenin signaling and Sonic Hedgehog, among others. Such molecular pathways were discussed in main text and it was shown that their inhibition can reduce expression of CSC markers and mediate tumor suppression.
Different strategies can be employed for suppressing CSCs. One of the potential methods is application of gene therapy. As signaling networks involved in regulating CSC features have been recognized, gene therapy can inactivate/activate molecular pathways and modulate CSC markers. However, there are some drawbacks with gene therapy, such as high cost. Therefore, plant derived-natural products are of importance due to their multitargeting capacity. The present review attempted to reveal the possible role of selected dietary agents in suppressing CSCs. As it was mentioned in main text and Tables, various doses of these anti-cancer compounds have been utilized and most of them are less than 100 μM. However, each natural product has its own IC50 and based on various experiments performed in this case, it is possible to determine optimal dose for using in cancer therapy. One of the limitations of experiments is lack of using nanostructures for delivery of dietary agents and enhancing their capacity in CSC suppression. Hence, future experiments can focus on this aspect and different nanoparticles such as lipid-based nanoparticles, carbon-based nanoparticles and polymeric nanoparticles, among others, can be utilized for targeted delivery. Furthermore, nanostructures overcome poor bioavailability of dietary agents in CSC suppression.
Noteworthy, the molecular pathways inducing CSC marker overexpression, can be targeted by natural products. The PI3K/Akt, Wnt, STAT3, NF-κB and Sonic signaling pathways undergo down-regulation by natural products to reduce expression level of CSC markers including CD44, CD133, Notch, Oct4 and Nanog, impairing cancer stemness and progression. The dietary agents induce apoptosis via both extrinsic and intrinsic pathways to reduce viability of CSCs. These compounds decrease expression level of MMPs and suppress EMT to reduce migration and invasion of CSCs. All these impacts are vital for preventing cancer progression. By suppressing CSC features, natural products enhance sensitivity of tumors to radiotherapy and chemotherapy. However, there are limited experiments in this case. Furthermore, there is lack of study evaluating potential of natural products in targeting CSCs and their interaction with immune system. One of the limitations related that was noted in these studies is that most of them have been performed under in vitro settings and have evaluated the potential impact of dietary agents in targeting CSCs in different cellular models. Hence, additional in vivo studies should be performed to analyze the exact potential of the natural products in targeting CSCs, suppressing tumor progression and investigating possible limitations associated with their use. There are no clinical trials performed so far to evaluate the role of natural products in targeting CSCs in cancer patients. However, there are clinical studies on using dietary agents in treatment of cancer patients (NCT03769766, NCT03980509, NCT01042938; clinicaltrials.gov, accessed on 22 October 2021). Therefore, still enormous work remains to be done to analyze the role of natural products in specifically targeting CSCs and providing effective treatment for cancer patients.

Author Contributions

M.D.A.P., S.A. and A.Z. (Amirhossein Zabolian), H.S., M.A.K., S.S., M.J.N., M.H., K.H. and M.A. prepared first draft and participated in manuscript writing. S.M., A.Z. (Ali Zarrabi) and G.S. collected articles, supervised and edited English language. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Singapore Ministry of Education Tier 1 Grant [R-184-000-301-114] to GS.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

siRNA: Small interfering RNA; shRNA, short hairpin RNA; CSCs, cancer stem cells; ALDH, aldehyde dehydrogenase; GLUTs, glucose transporters; HKs, hexokinases; ROS, reactive oxygen species; TCA, tricarboxylic acid; miRNA, microRNA; TCF4, transcription factor 4; EVs, extracellular vesicles; EGCG, epigallocatechin 3-gallate; ISL, isoliquiritigenin; P-gp, P-glycoprotein; CA, caffeic acid; CAPE, caffeic acid phenyl ester; DR5, death receptor 5; EpCAM, epithelial cell adhesion molecule; YB-1, Y-box binding protein 1; COX-2, cyclooxygenase-2; STAT3, signal transducer and activator of transcription 3; EMT, epithelial-to-mesenchymal transition; NF-κB, nuclear factor-kappaB; SFN, sulforaphane; TGF-β, transforming growth factor-β; TNF, tumor necrosis factor; TRAIL, TNF-related apoptosis ligand; HIF-α, hypoxia inducible factor-α; PCa, prostate cancer; MK, midkin; Res, resveratrol; NAF-1, nutrient-deprivation autophagy factor-1; TME, tumor microenvironment; VE, vascular endothelial; vWF, von Willebrand factor; BBR, berberine; Gli1, glioma-associated oncogene-1; d-TPP, Dodecyl-TPP; cyt C, cytochrome C.

References

  1. Abadi, A.J.; Mirzaei, S.; Mahabady, M.K.; Hashemi, F.; Zabolian, A.; Hashemi, F.; Raee, P.; Aghamiri, S.; Ashrafizadeh, M.; Aref, A.R.; et al. Curcumin and its derivatives in cancer therapy: Potentiating antitumor activity of cisplatin and reducing side effects. Phytother Res. 2021. [Google Scholar] [CrossRef] [PubMed]
  2. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics, 2021. CA Cancer J. Clin. 2021, 71, 7–33. [Google Scholar] [CrossRef]
  3. Ashrafizade, M.; Delfi, M.; Hashemi, F.; Zabolian, A.; Saleki, H.; Bagherian, M.; Azami, N.; Farahani, M.V.; Omid Sharifzadeh, S.; Hamzehlou, S. Biomedical application of chitosan-based nanoscale delivery systems: Potential usefulness in siRNA delivery for cancer therapy. Carbohydr. Polym. 2021, 260, 117809. [Google Scholar] [CrossRef] [PubMed]
  4. Kirtonia, A.; Gala, K.; Fernandes, S.G.; Pandya, G.; Pandey, A.K.; Sethi, G.; Khattar, E.; Garg, M. Repurposing of drugs: An attractive pharmacological strategy for cancer therapeutics. Semin. Cancer Biol. 2021, 68, 258–278. [Google Scholar] [CrossRef]
  5. Merarchi, M.; Sethi, G.; Shanmugam, M.K.; Fan, L.; Arfuso, F.; Ahn, K.S. Role of Natural Products in Modulating Histone Deacetylases in Cancer. Molecules 2019, 24, 1047. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Chopra, P.; Sethi, G.; Dastidar, S.G.; Ray, A. Polo-like kinase inhibitors: An emerging opportunity for cancer therapeutics. Expert Opin. Investig. Drugs 2010, 19, 27–43. [Google Scholar] [CrossRef] [PubMed]
  7. Ashrafizadeh, M.; Bakhoda, M.R.; Bahmanpour, Z.; Ilkhani, K.; Zarrabi, A.; Makvandi, P.; Khan, H.; Mazaheri, S.; Darvish, M.; Mirzaei, H. Apigenin as tumor suppressor in cancers: Biotherapeutic activity, nanodelivery, and mechanisms with emphasis on pancreatic cancer. Front. Chem. 2020, 8, 829. [Google Scholar] [CrossRef] [PubMed]
  8. Antognelli, C.; Mandarano, M.; Prosperi, E.; Sidoni, A.; Talesa, V.N. Glyoxalase-1-Dependent Methylglyoxal Depletion Sustains PD-L1 Expression in Metastatic Prostate Cancer Cells: A Novel Mechanism in Cancer Immunosurveillance Escape and a Potential Novel Target to Overcome PD-L1 Blockade Resistance. Cancers 2021, 13, 2965. [Google Scholar] [CrossRef]
  9. Ashrafizadeh, M.; Farhood, B.; Eleojo Musa, A.; Taeb, S.; Najafi, M. The interactions and communications in tumor resistance to radiotherapy: Therapy perspectives. Int. Immunopharmacol. 2020, 87, 106807. [Google Scholar] [CrossRef] [PubMed]
  10. Cao, J.; Sun, X.; Zhang, X.; Chen, D. 6PGD Upregulation is Associated with Chemo- and Immuno-Resistance of Renal Cell Carcinoma via AMPK Signaling-Dependent NADPH-Mediated Metabolic Reprograming. Am. J. Med. Sci. 2020, 360, 279–286. [Google Scholar] [CrossRef]
  11. Gupta, B.; Sadaria, D.; Warrier, V.U.; Kirtonia, A.; Kant, R.; Awasthi, A.; Baligar, P.; Pal, J.K.; Yuba, E.; Sethi, G.; et al. Plant lectins and their usage in preparing targeted nanovaccines for cancer immunotherapy. Semin. Cancer Biol. 2020. [Google Scholar] [CrossRef] [PubMed]
  12. Warrier, V.U.; Makandar, A.I.; Garg, M.; Sethi, G.; Kant, R.; Pal, J.K.; Yuba, E.; Gupta, R.K. Engineering anti-cancer nanovaccine based on antigen cross-presentation. Biosci. Rep. 2019, 39, BSR20193220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Mirzaei, S.; Zarrabi, A.; Asnaf, S.E.; Hashemi, F.; Zabolian, A.; Hushmandi, K.; Raei, M.; Goharrizi, M.A.S.B.; Makvandi, P.; Samarghandian, S.; et al. The role of microRNA-338-3p in cancer: Growth, invasion, chemoresistance, and mediators. Life Sci. 2021, 368, 119005. [Google Scholar] [CrossRef]
  14. Dehshahri, A.; Ashrafizadeh, M.; Ghasemipour Afshar, E.; Pardakhty, A.; Mandegary, A.; Mohammadinejad, R.; Sethi, G. Topoisomerase inhibitors: Pharmacology and emerging nanoscale delivery systems. Pharmacol. Res. 2020, 151, 104551. [Google Scholar] [CrossRef] [PubMed]
  15. Wang, C.; Tang, H.; Geng, A.; Dai, B.; Zhang, H.; Sun, X.; Chen, Y.; Qiao, Z.; Zhu, H.; Yang, J.; et al. Rational combination therapy for hepatocellular carcinoma with PARP1 and DNA-PK inhibitors. Proc. Natl. Acad. Sci. USA 2020, 117, 26356–26365. [Google Scholar] [CrossRef] [PubMed]
  16. Galván Morales, M.A.; Barrera Rodríguez, R.; Santiago Cruz, J.R.; Teran, L.M. Overview of New Treatments with Immunotherapy for Breast Cancer and a Proposal of a Combination Therapy. Molecules 2020, 25, 5685. [Google Scholar] [CrossRef] [PubMed]
  17. Kong, Q.; Wei, D.; Xie, P.; Wang, B.; Yu, K.; Kang, X.; Wang, Y. Photothermal Therapy via NIR II Light Irradiation Enhances DNA Damage and Endoplasmic Reticulum Stress for Efficient Chemotherapy. Front. Pharmacol. 2021, 12, 670207. [Google Scholar] [CrossRef] [PubMed]
  18. Zhou, H.; Zeng, X.; Li, A.; Zhou, W.; Tang, L.; Hu, W.; Fan, Q.; Meng, X.; Deng, H.; Duan, L.; et al. Upconversion NIR-II fluorophores for mitochondria-targeted cancer imaging and photothermal therapy. Nat. Commun. 2020, 11, 6183. [Google Scholar] [CrossRef] [PubMed]
  19. Jo, G.; Lee, B.Y.; Kim, E.J.; Park, M.H.; Hyun, H. Indocyanine Green and Methyl-β-Cyclodextrin Complex for Enhanced Photothermal Cancer Therapy. Biomedicines 2020, 8, 476. [Google Scholar] [CrossRef] [PubMed]
  20. Ashrafizadeh, M.; Hushmandi, K.; Rahmani Moghadam, E.; Zarrin, V.; Hosseinzadeh Kashani, S.; Bokaie, S.; Najafi, M.; Tavakol, S.; Mohammadinejad, R.; Nabavi, N.; et al. Progress in delivery of siRNA-based therapeutics employing nano-vehicles for treatment of prostate cancer. Bioengineering 2020, 7, 91. [Google Scholar] [CrossRef]
  21. Mohammadinejad, R.; Dehshahri, A.; Sassan, H.; Behnam, B.; Ashrafizadeh, M.; Samareh Gholami, A.; Pardakhty, A.; Mandegary, A. Preparation of carbon dot as a potential CRISPR/Cas9 plasmid delivery system for lung cancer cells. Minerva Biotecnol. 2020, 32, 106–113. [Google Scholar] [CrossRef]
  22. Mohammadinejad, R.; Sassan, H.; Pardakhty, A.; Hashemabadi, M.; Ashrafizadeh, M.; Dehshahri, A.; Mandegary, A. ZEB1 and ZEB2 gene editing mediated by CRISPR/Cas9 in A549 cell line. Bratisl. Lek. Listy 2020, 121, 31–36. [Google Scholar] [CrossRef] [PubMed]
  23. Antognelli, C.; Frosini, R.; Santolla, M.F.; Peirce, M.J.; Talesa, V.N. Oleuropein-Induced Apoptosis Is Mediated by Mitochondrial Glyoxalase 2 in NSCLC A549 Cells: A Mechanistic Inside and a Possible Novel Nonenzymatic Role for an Ancient Enzyme. Oxidative Med. Cell. Longev. 2019, 2019, 8576961. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Mirzaei, S.; Gholami, M.H.; Hashemi, F.; Zabolian, A.; Farahani, M.V.; Hushmandi, K.; Zarrabi, A.; Goldman, A.; Ashrafizadeh, M.; Orive, G. Advances in understanding the role of P-gp in doxorubicin resistance: Molecular pathways, therapeutic strategies, and prospects. Drug Discov. Today 2021. [Google Scholar] [CrossRef] [PubMed]
  25. Kirtonia, A.; Ashrafizadeh, M.; Zarrabi, A.; Hushmandi, K.; Zabolian, A.; Bejandi, A.K.; Rani, R.; Pandey, A.K.; Baligar, P.; Kumar, V.; et al. Long noncoding RNAs: A novel insight in the leukemogenesis and drug resistance in acute myeloid leukemia. J. Cell. Physiol. 2021. [Google Scholar] [CrossRef]
  26. Ashrafizadeh, M.; Zarrabi, A.; Mirzaei, S.; Hashemi, F.; Samarghandian, S.; Zabolian, A.; Hushmandi, K.; Ang, H.L.; Sethi, G.; Kumar, A.P.; et al. Gallic acid for cancer therapy: Molecular mechanisms and boosting efficacy by nanoscopical delivery. Food Chem. Toxicol. 2021, 157, 112576. [Google Scholar] [CrossRef]
  27. Ashrafizadeh, M.; Mirzaei, S.; Gholami, M.H.; Hashemi, F.; Zabolian, A.; Raei, M.; Hushmandi, K.; Zarrabi, A.; Voelcker, N.H.; Aref, A.R.; et al. Hyaluronic acid-based nanoplatforms for Doxorubicin: A review of stimuli-responsive carriers, co-delivery and resistance suppression. Carbohydr. Polym. 2021, 272, 118491. [Google Scholar] [CrossRef]
  28. Paskeh, M.D.A.; Mirzaei, S.; Orouei, S.; Zabolian, A.; Saleki, H.; Azami, N.; Hushmandi, K.; Baradaran, B.; Hashmi, M.; Aref, A.R.; et al. The role of miRNA-489 as a new onco-suppressor factor in different cancers based on pre-clinical and clinical evidence. Int. J. Biol. Macromol. 2021, 191, 727–732. [Google Scholar] [CrossRef]
  29. Mirzaei, S.; Gholami, M.H.; Hashemi, F.; Zabolian, A.; Hushmandi, K.; Rahmanian, V.; Entezari, M.; Girish, Y.R.; Kumar, K.S.S.; Aref, A.; et al. Employing siRNA tool and its delivery platforms in suppressing cisplatin resistance: Approaching to a new era of cancer chemotherapy. Life Sci. 2021, 277, 119430. [Google Scholar] [CrossRef]
  30. Mirzaei, S.; Mahabady, M.K.; Zabolian, A.; Abbaspour, A.; Fallahzadeh, P.; Noori, M.; Hashemi, F.; Hushmandi, K.; Daneshi, S.; Kumar, A.P.; et al. Small interfering RNA (siRNA) to target genes and molecular pathways in glioblastoma therapy: Current status with an emphasis on delivery systems. Life Sci. 2021, 275, 119368. [Google Scholar] [CrossRef]
  31. Ashrafizadeh, M.; Najafi, M.; Makvandi, P.; Zarrabi, A.; Farkhondeh, T.; Samarghandian, S. Versatile role of curcumin and its derivatives in lung cancer therapy. J. Cell. Physiol. 2020, 235, 9241–9268. [Google Scholar] [CrossRef] [PubMed]
  32. Sethi, G.; Shanmugam, M.K.; Warrier, S.; Merarchi, M.; Arfuso, F.; Kumar, A.P.; Bishayee, A. Pro-Apoptotic and Anti-Cancer Properties of Diosgenin: A Comprehensive and Critical Review. Nutrients 2018, 10, 645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Kashyap, D.; Tuli, H.S.; Yerer, M.B.; Sharma, A.; Sak, K.; Srivastava, S.; Pandey, A.; Garg, V.K.; Sethi, G.; Bishayee, A. Natural product-based nanoformulations for cancer therapy: Opportunities and challenges. Semin. Cancer Biol. 2021, 69, 5–23. [Google Scholar] [CrossRef]
  34. Patel, S.M.; Nagulapalli Venkata, K.C.; Bhattacharyya, P.; Sethi, G.; Bishayee, A. Potential of neem (Azadirachta indica L.) for prevention and treatment of oncologic diseases. Semin. Cancer Biol. 2016, 40–41, 100–115. [Google Scholar] [CrossRef]
  35. Liu, C.; Ho, P.C.; Wong, F.C.; Sethi, G.; Wang, L.Z.; Goh, B.C. Garcinol: Current status of its anti-oxidative, anti-inflammatory and anti-cancer effects. Cancer Lett. 2015, 362, 8–14. [Google Scholar] [CrossRef]
  36. Samec, M.; Liskova, A.; Koklesova, L.; Mersakova, S.; Strnadel, J.; Kajo, K.; Pec, M.; Zhai, K.; Smejkal, K.; Mirzaei, S. Flavonoids targeting HIF-1: Implications on cancer metabolism. Cancers 2021, 13, 130. [Google Scholar] [CrossRef]
  37. Koklesova, L.; Liskova, A.; Samec, M.; Zhai, K.; Abotaleb, M.; Ashrafizadeh, M.; Brockmueller, A.; Shakibaei, M.; Biringer, K.; Bugos, O.; et al. Carotenoids in Cancer Metastasis—Status Quo and Outlook. Biomolecules 2020, 10, 1653. [Google Scholar] [CrossRef] [PubMed]
  38. Koklesova, L.; Liskova, A.; Samec, M.; Buhrmann, C.; Samuel, S.M.; Varghese, E.; Ashrafizadeh, M.; Najafi, M.; Shakibaei, M.; Büsselberg, D.; et al. Carotenoids in cancer apoptosis—the road from bench to bedside and back. Cancers 2020, 12, 2425. [Google Scholar] [CrossRef]
  39. Mishra, S.; Verma, S.S.; Rai, V.; Awasthee, N.; Chava, S.; Hui, K.M.; Kumar, A.P.; Challagundla, K.B.; Sethi, G.; Gupta, S.C. Long non-coding RNAs are emerging targets of phytochemicals for cancer and other chronic diseases. Cell. Mol. Life Sci. 2019, 76, 1947–1966. [Google Scholar] [CrossRef]
  40. Tewari, D.; Nabavi, S.F.; Nabavi, S.M.; Sureda, A.; Farooqi, A.A.; Atanasov, A.G.; Vacca, R.A.; Sethi, G.; Bishayee, A. Targeting activator protein 1 signaling pathway by bioactive natural agents: Possible therapeutic strategy for cancer prevention and intervention. Pharmacol. Res. 2018, 128, 366–375. [Google Scholar] [CrossRef]
  41. Shanmugam, M.K.; Ong, T.H.; Kumar, A.P.; Lun, C.K.; Ho, P.C.; Wong, P.T.; Hui, K.M.; Sethi, G. Ursolic acid inhibits the initiation, progression of prostate cancer and prolongs the survival of TRAMP mice by modulating pro-inflammatory pathways. PLoS ONE 2012, 7, e32476. [Google Scholar] [CrossRef]
  42. Li, F.; Shanmugam, M.K.; Chen, L.; Chatterjee, S.; Basha, J.; Kumar, A.P.; Kundu, T.K.; Sethi, G. Garcinol, a polyisoprenylated benzophenone modulates multiple proinflammatory signaling cascades leading to the suppression of growth and survival of head and neck carcinoma. Cancer Prev. Res. 2013, 6, 843–854. [Google Scholar] [CrossRef] [Green Version]
  43. Choi, Y.H.; Han, D.H.; Kim, S.W.; Kim, M.J.; Sung, H.H.; Jeon, H.G.; Jeong, B.C.; Seo, S.I.; Jeon, S.S.; Lee, H.M.; et al. A randomized, double-blind, placebo-controlled trial to evaluate the role of curcumin in prostate cancer patients with intermittent androgen deprivation. Prostate 2019, 79, 614–621. [Google Scholar] [CrossRef]
  44. Howells, L.M.; Iwuji, C.O.O.; Irving, G.R.B.; Barber, S.; Walter, H.; Sidat, Z.; Griffin-Teall, N.; Singh, R.; Foreman, N.; Patel, S.R.; et al. Curcumin Combined with FOLFOX Chemotherapy Is Safe and Tolerable in Patients with Metastatic Colorectal Cancer in a Randomized Phase IIa Trial. J. Nutr. 2019, 149, 1133–1139. [Google Scholar] [CrossRef] [Green Version]
  45. Hsieh, Y.S.; Yang, S.F.; Sethi, G.; Hu, D.N. Natural bioactives in cancer treatment and prevention. BioMed Res. Int. 2015, 2015, 182835. [Google Scholar] [CrossRef] [Green Version]
  46. Manu, K.A.; Shanmugam, M.K.; Ramachandran, L.; Li, F.; Siveen, K.S.; Chinnathambi, A.; Zayed, M.E.; Alharbi, S.A.; Arfuso, F.; Kumar, A.P.; et al. Isorhamnetin augments the anti-tumor effect of capecitabine through the negative regulation of NF-κB signaling cascade in gastric cancer. Cancer Lett. 2015, 363, 28–36. [Google Scholar] [CrossRef]
  47. Manu, K.A.; Shanmugam, M.K.; Li, F.; Chen, L.; Siveen, K.S.; Ahn, K.S.; Kumar, A.P.; Sethi, G. Simvastatin sensitizes human gastric cancer xenograft in nude mice to capecitabine by suppressing nuclear factor-kappa B-regulated gene products. J. Mol. Med. 2014, 92, 267–276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Hussain, Y.; Islam, L.; Khan, H.; Filosa, R.; Aschner, M.; Javed, S. Curcumin-cisplatin chemotherapy: A novel strategy in promoting chemotherapy efficacy and reducing side effects. Phytother. Res. 2021. [Google Scholar] [CrossRef] [PubMed]
  49. Ashrafizadeh, M.; Zarrabi, A.; Hashemi, F.; Moghadam, E.R.; Hashemi, F.; Entezari, M.; Hushmandi, K.; Mohammadinejad, R.; Najafi, M. Curcumin in cancer therapy: A novel adjunct for combination chemotherapy with paclitaxel and alleviation of its adverse effects. Life Sci. 2020, 256, 117984. [Google Scholar] [CrossRef]
  50. Amini, P.; Nodooshan, S.J.; Ashrafizadeh, M.; Eftekhari, S.-M.; Aryafar, T.; Khalafi, L.; Musa, A.E.; Mahdavi, S.R.; Najafi, M.; Farhood, B. Resveratrol induces apoptosis and attenuates proliferation of MCF-7 cells in combination with radiation and hyperthermia. Curr. Mol. Med. 2021, 21, 142–150. [Google Scholar] [CrossRef] [PubMed]
  51. Moballegh Nasery, M.; Abadi, B.; Poormoghadam, D.; Zarrabi, A.; Keyhanvar, P.; Khanbabaei, H.; Ashrafizadeh, M.; Mohammadinejad, R.; Tavakol, S.; Sethi, G. Curcumin Delivery Mediated by Bio-Based Nanoparticles: A Review. Molecules 2020, 25, 689. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Dai, X.; Ahn, K.S.; Wang, L.Z.; Kim, C.; Deivasigamni, A.; Arfuso, F.; Um, J.Y.; Kumar, A.P.; Chang, Y.C.; Kumar, D.; et al. Ascochlorin Enhances the Sensitivity of Doxorubicin Leading to the Reversal of Epithelial-to-Mesenchymal Transition in Hepatocellular Carcinoma. Mol. Cancer Ther. 2016, 15, 2966–2976. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Ong, S.K.L.; Shanmugam, M.K.; Fan, L.; Fraser, S.E.; Arfuso, F.; Ahn, K.S.; Sethi, G.; Bishayee, A. Focus on Formononetin: Anticancer Potential and Molecular Targets. Cancers 2019, 11, 611. [Google Scholar] [CrossRef] [Green Version]
  54. Bhuvanalakshmi, G.; Gamit, N.; Patil, M.; Arfuso, F.; Sethi, G.; Dharmarajan, A.; Kumar, A.P.; Warrier, S. Stemness, Pluripotentiality, and Wnt Antagonism: sFRP4, a Wnt antagonist Mediates Pluripotency and Stemness in Glioblastoma. Cancers 2018, 11, 25. [Google Scholar] [CrossRef] [Green Version]
  55. Mandal, S.; Arfuso, F.; Sethi, G.; Dharmarajan, A.; Warrier, S. Encapsulated human mesenchymal stem cells (eMSCs) as a novel anti-cancer agent targeting breast cancer stem cells: Development of 3D primed therapeutic MSCs. Int. J. Biochem. Cell Biol. 2019, 110, 59–69. [Google Scholar] [CrossRef] [PubMed]
  56. Carvalho, L.S.; Gonçalves, N.; Fonseca, N.A.; Moreira, J.N. Cancer Stem Cells and Nucleolin as Drivers of Carcinogenesis. Pharmaceuticals 2021, 14, 60. [Google Scholar] [CrossRef]
  57. Ma, Z.; Wang, Y.Y.; Xin, H.W.; Wang, L.; Arfuso, F.; Dharmarajan, A.; Kumar, A.P.; Wang, H.; Tang, F.R.; Warrier, S.; et al. The expanding roles of long non-coding RNAs in the regulation of cancer stem cells. Int. J. Biochem. Cell Biol. 2019, 108, 17–20. [Google Scholar] [CrossRef] [PubMed]
  58. Lapidot, T.; Sirard, C.; Vormoor, J.; Murdoch, B.; Hoang, T.; Caceres-Cortes, J.; Minden, M.; Paterson, B.; Caligiuri, M.A.; Dick, J.E. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 1994, 367, 645–648. [Google Scholar] [CrossRef] [PubMed]
  59. Bonnet, D.; Dick, J.E. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat. Med. 1997, 3, 730–737. [Google Scholar] [CrossRef] [PubMed]
  60. Al-Hajj, M.; Wicha, M.S.; Benito-Hernandez, A.; Morrison, S.J.; Clarke, M.F. Prospective identification of tumorigenic breast cancer cells. Proc. Natl. Acad. Sci. USA 2003, 100, 3983–3988. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Koren, E.; Fuchs, Y. The bad seed: Cancer stem cells in tumor development and resistance. Drug Resist. Updates 2016, 28, 1–12. [Google Scholar] [CrossRef] [PubMed]
  62. Warrier, S.; Patil, M.; Bhansali, S.; Varier, L.; Sethi, G. Designing precision medicine panels for drug refractory cancers targeting cancer stemness traits. Biochim. Biophys. Acta. Rev. Cancer 2021, 1875, 188475. [Google Scholar] [CrossRef] [PubMed]
  63. Plaks, V.; Kong, N.; Werb, Z. The cancer stem cell niche: How essential is the niche in regulating stemness of tumor cells? Cell Stem Cell 2015, 16, 225–238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Cao, J.; Bhatnagar, S.; Wang, J.; Qi, X.; Prabha, S.; Panyam, J. Cancer stem cells and strategies for targeted drug delivery. Drug Deliv. Transl. Res. 2020, 11, 1779–1805. [Google Scholar] [CrossRef] [PubMed]
  65. Capp, J.-P. Cancer stem cells: From historical roots to a new perspective. J. Oncol. 2019, 2019, 5189232. [Google Scholar] [CrossRef] [PubMed]
  66. Singh, S.K.; Hawkins, C.; Clarke, I.D.; Squire, J.A.; Bayani, J.; Hide, T.; Henkelman, R.M.; Cusimano, M.D.; Dirks, P.B. Identification of human brain tumour initiating cells. Nature 2004, 432, 396–401. [Google Scholar] [CrossRef] [PubMed]
  67. Ricci-Vitiani, L.; Lombardi, D.G.; Pilozzi, E.; Biffoni, M.; Todaro, M.; Peschle, C.; De Maria, R. Identification and expansion of human colon-cancer-initiating cells. Nature 2007, 445, 111–115. [Google Scholar] [CrossRef] [PubMed]
  68. Ma, S.; Chan, K.W.; Hu, L.; Lee, T.K.W.; Wo, J.Y.H.; Ng, I.O.L.; Zheng, B.J.; Guan, X.Y. Identification and characterization of tumorigenic liver cancer stem/progenitor cells. Gastroenterology 2007, 132, 2542–2556. [Google Scholar] [CrossRef] [PubMed]
  69. Elgendy, S.M.; Alyammahi, S.K.; Alhamad, D.W.; Abdin, S.M.; Omar, H.A. Ferroptosis: An emerging approach for targeting cancer stem cells and drug resistance. Crit. Rev. Oncol./Hematol. 2020, 155, 103095. [Google Scholar] [CrossRef] [PubMed]
  70. Hiremath, I.S.; Goel, A.; Warrier, S.; Kumar, A.P.; Sethi, G.; Garg, M. The multidimensional role of the Wnt/β-catenin signaling pathway in human malignancies. J. Cell. Physiol. 2021. [Google Scholar] [CrossRef]
  71. Duan, J.-J.; Qiu, W.; Xu, S.-L.; Wang, B.; Ye, X.-Z.; Ping, Y.-F.; Zhang, X.; Bian, X.-W.; Yu, S.-C. development. Strategies for isolating and enriching cancer stem cells: Well begun is half done. Stem Cells Dev. 2013, 22, 2221–2239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Batlle, E.; Clevers, H. Cancer stem cells revisited. Nat. Med. 2017, 23, 1124–1134. [Google Scholar] [CrossRef] [PubMed]
  73. Prager, B.C.; Xie, Q.; Bao, S.; Rich, J.N. Cancer Stem Cells: The Architects of the Tumor Ecosystem. Cell Stem Cell 2019, 24, 41–53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Yang, L.; Shi, P.; Zhao, G.; Xu, J.; Peng, W.; Zhang, J.; Zhang, G.; Wang, X.; Dong, Z.; Chen, F.; et al. Targeting cancer stem cell pathways for cancer therapy. Signal Transduct. Target. Ther. 2020, 5, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Nallanthighal, S.; Heiserman, J.P.; Cheon, D.J. The Role of the Extracellular Matrix in Cancer Stemness. Front. Cell Dev. Biol. 2019, 7, 86. [Google Scholar] [CrossRef]
  76. Ma, Z.; Wang, L.Z.; Cheng, J.T.; Lam, W.S.T.; Ma, X.; Xiang, X.; Wong, A.L.; Goh, B.C.; Gong, Q.; Sethi, G.; et al. Targeting Hypoxia-Inducible Factor-1-Mediated Metastasis for Cancer Therapy. Antioxid. Redox Signal. 2021, 34, 1484–1497. [Google Scholar] [CrossRef] [PubMed]
  77. Ong, P.S.; Wang, L.Z.; Dai, X.; Tseng, S.H.; Loo, S.J.; Sethi, G. Judicious Toggling of mTOR Activity to Combat Insulin Resistance and Cancer: Current Evidence and Perspectives. Front. Pharmacol. 2016, 7, 395. [Google Scholar] [CrossRef] [PubMed]
  78. De Francesco, E.M.; Sotgia, F.; Lisanti, M.P. Cancer stem cells (CSCs): Metabolic strategies for their identification and eradication. Biochem. J. 2018, 475, 1611–1634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Jagust, P.; de Luxán-Delgado, B.; Parejo-Alonso, B.; Sancho, P. Metabolism-Based Therapeutic Strategies Targeting Cancer Stem Cells. Front. Pharmacol. 2019, 10, 203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Snyder, V.; Reed-Newman, T.C.; Arnold, L.; Thomas, S.M.; Anant, S. Cancer Stem Cell Metabolism and Potential Therapeutic Targets. Front. Oncol. 2018, 8, 203. [Google Scholar] [CrossRef] [PubMed]
  81. Peng, F.; Wang, J.H.; Fan, W.J.; Meng, Y.T.; Li, M.M.; Li, T.T.; Cui, B.; Wang, H.F.; Zhao, Y.; An, F.; et al. Glycolysis gatekeeper PDK1 reprograms breast cancer stem cells under hypoxia. Oncogene 2018, 37, 1062–1074. [Google Scholar] [CrossRef] [Green Version]
  82. Ciavardelli, D.; Rossi, C.; Barcaroli, D.; Volpe, S.; Consalvo, A.; Zucchelli, M.; De Cola, A.; Scavo, E.; Carollo, R.; D’Agostino, D.; et al. Breast cancer stem cells rely on fermentative glycolysis and are sensitive to 2-deoxyglucose treatment. Cell Death Dis. 2014, 5, e1336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Liu, P.P.; Liao, J.; Tang, Z.J.; Wu, W.J.; Yang, J.; Zeng, Z.L.; Hu, Y.; Wang, P.; Ju, H.Q.; Xu, R.H.; et al. Metabolic regulation of cancer cell side population by glucose through activation of the Akt pathway. Cell Death Differ. 2014, 21, 124–135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Hur, W.; Ryu, J.Y.; Kim, H.U.; Hong, S.W.; Lee, E.B.; Lee, S.Y.; Yoon, S.K. Systems approach to characterize the metabolism of liver cancer stem cells expressing CD133. Sci. Rep. 2017, 7, 45557. [Google Scholar] [CrossRef] [Green Version]
  85. Shen, Y.A.; Wang, C.Y.; Hsieh, Y.T.; Chen, Y.J.; Wei, Y.H. Metabolic reprogramming orchestrates cancer stem cell properties in nasopharyngeal carcinoma. Cell Cycle 2015, 14, 86–98. [Google Scholar] [CrossRef] [Green Version]
  86. Janiszewska, M.; Suvà, M.L.; Riggi, N.; Houtkooper, R.H.; Auwerx, J.; Clément-Schatlo, V.; Radovanovic, I.; Rheinbay, E.; Provero, P.; Stamenkovic, I. Imp2 controls oxidative phosphorylation and is crucial for preserving glioblastoma cancer stem cells. Genes Dev. 2012, 26, 1926–1944. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Sancho, P.; Burgos-Ramos, E.; Tavera, A.; Bou Kheir, T.; Jagust, P.; Schoenhals, M.; Barneda, D.; Sellers, K.; Campos-Olivas, R.; Graña, O.; et al. MYC/PGC-1α Balance Determines the Metabolic Phenotype and Plasticity of Pancreatic Cancer Stem Cells. Cell Metab. 2015, 22, 590–605. [Google Scholar] [CrossRef] [Green Version]
  88. Tanabe, A.; Sahara, H. The metabolic heterogeneity and flexibility of cancer stem cells. Cancers 2020, 12, 2780. [Google Scholar] [CrossRef]
  89. Jain, S.; Annett, S.L.; Morgan, M.P.; Robson, T. The Cancer Stem Cell Niche in Ovarian Cancer and Its Impact on Immune Surveillance. Int. J. Mol. Sci. 2021, 22, 4091. [Google Scholar] [CrossRef]
  90. Escalona, R.M.; Bilandzic, M.; Western, P.; Kadife, E.; Kannourakis, G.; Findlay, J.K.; Ahmed, N. TIMP-2 regulates proliferation, invasion and STAT3-mediated cancer stem cell-dependent chemoresistance in ovarian cancer cells. BMC Cancer 2020, 20, 960. [Google Scholar] [CrossRef]
  91. Chan, Y.W.; So, C.; Yau, K.L.; Chiu, K.C.; Wang, X.; Chan, F.L.; Tsang, S.Y. Adipose-derived stem cells and cancer cells fuse to generate cancer stem cell-like cells with increased tumorigenicity. J. Cell. Physiol. 2020, 235, 6794–6807. [Google Scholar] [CrossRef]
  92. Chen, K.; Li, L.; Qu, S.; Pan, X.; Sun, Y.; Wan, F.; Yu, B.; Zhou, L.; Zhu, X. Silencing hTERT attenuates cancer stem cell-like characteristics and radioresistance in the radioresistant nasopharyngeal carcinoma cell line CNE-2R. Aging 2020, 12, 25599–25613. [Google Scholar] [CrossRef] [PubMed]
  93. Zhang, J.; He, H.; Wang, K.; Xie, Y.; Yang, Z.; Qie, M.; Liao, Z.; Zheng, Z. miR-326 inhibits the cell proliferation and cancer stem cell-like property of cervical cancer in vitro and oncogenesis in vivo via targeting TCF4. Ann. Transl. Med. 2020, 8, 1638. [Google Scholar] [CrossRef]
  94. Jimenez, T.; Barrios, A.; Tucker, A.; Collazo, J.; Arias, N.; Fazel, S.; Baker, M.; Halim, M.; Huynh, T.; Singh, R.; et al. DUSP9-mediated reduction of pERK1/2 supports cancer stem cell-like traits and promotes triple negative breast cancer. Am. J. Cancer Res. 2020, 10, 3487–3506. [Google Scholar] [PubMed]
  95. Moon, S.; Ok, Y.; Hwang, S.; Lim, Y.S.; Kim, H.Y.; Na, Y.J.; Yoon, S. A Marine Collagen-Based Biomimetic Hydrogel Recapitulates Cancer Stem Cell Niche and Enhances Progression and Chemoresistance in Human Ovarian Cancer. Mar. Drugs 2020, 18, 498. [Google Scholar] [CrossRef] [PubMed]
  96. Bridges, A.E.; Ramachandran, S.; Tamizhmani, K.; Parwal, U.; Lester, A.; Rajpurohit, P.; Morera, D.S.; Hasanali, S.L.; Arjunan, P.; Jedeja, R.N.; et al. RAD51AP1 Loss Attenuates Colorectal Cancer Stem Cell Renewal and Sensitizes to Chemotherapy. Mol. Cancer Res. 2021, 19. [Google Scholar] [CrossRef]
  97. Yarmishyn, A.A.; Yang, Y.P.; Lu, K.H.; Chen, Y.C.; Chien, Y.; Chou, S.J.; Tsai, P.H.; Ma, H.I.; Chien, C.S.; Chen, M.T.; et al. Musashi-1 promotes cancer stem cell properties of glioblastoma cells via upregulation of YTHDF1. Cancer Cell Int. 2020, 20, 597. [Google Scholar] [CrossRef]
  98. Xiong, J.; Zhang, X.; Zhang, Y.; Wu, B.; Fang, L.; Wang, N.; Yi, H.; Chang, N.; Chen, L.; Zhang, J. Aryl hydrocarbon receptor mediates Jak2/STAT3 signaling for non-small cell lung cancer stem cell maintenance. Exp. Cell Res. 2020, 396, 112288. [Google Scholar] [CrossRef]
  99. Wu, Y.; Wang, T.; Xia, L.; Zhang, M. LncRNA WDFY3-AS2 promotes cisplatin resistance and the cancer stem cell in ovarian cancer by regulating hsa-miR-139-5p/SDC4 axis. Cancer Cell Int. 2021, 21, 284. [Google Scholar] [CrossRef]
  100. Jeon, S.A.; Kim, D.W.; Lee, D.B.; Cho, J.Y. NEDD4 Plays Roles in the Maintenance of Breast Cancer Stem Cell Characteristics. Front. Oncol. 2020, 10, 1680. [Google Scholar] [CrossRef]
  101. Ashrafizaveh, S.; Ashrafizadeh, M.; Zarrabi, A.; Husmandi, K.; Zabolian, A.; Shahinozzaman, M.; Aref, A.R.; Hamblin, M.R.; Nabavi, N.; Crea, F. Long non-coding RNA in the doxorubicin resistance of cancer cells. Cancer Lett. 2021, 508, 104–114. [Google Scholar] [CrossRef] [PubMed]
  102. Wee, I.; Syn, N.; Sethi, G.; Goh, B.C.; Wang, L. Role of tumor-derived exosomes in cancer metastasis. Biochim. Biophys. Acta Rev. Cancer 2019, 1871, 12–19. [Google Scholar] [CrossRef] [PubMed]
  103. Weng, J.; Xiang, X.; Ding, L.; Wong, A.L.; Zeng, Q.; Sethi, G.; Wang, L.; Lee, S.C.; Goh, B.C. Extracellular vesicles, the cornerstone of next-generation cancer diagnosis? Semin. Cancer Biol. 2021, 74, 105–120. [Google Scholar] [CrossRef] [PubMed]
  104. Afify, S.M.; Hassan, G.; Yan, T.; Seno, A.; Seno, M. Cancer Stem Cell Initiation by Tumor-Derived Extracellular Vesicles. In Methods in Molecular Biology; Springer: New York, NY, USA, 2021. [Google Scholar] [CrossRef]
  105. Gao, Z.; Wang, Q.; Ji, M.; Guo, X.; Li, L.; Su, X. Exosomal lncRNA UCA1 modulates cervical cancer stem cell self-renewal and differentiation through microRNA-122-5p/SOX2 axis. J. Transl. Med. 2021, 19, 229. [Google Scholar] [CrossRef] [PubMed]
  106. Dai, W.; Jin, X.; Han, L.; Huang, H.; Ji, Z.; Xu, X.; Tang, M.; Jiang, B.; Chen, W. Exosomal lncRNA DOCK9-AS2 derived from cancer stem cell-like cells activated Wnt/β-catenin pathway to aggravate stemness, proliferation, migration, and invasion in papillary thyroid carcinoma. Cell Death Dis. 2020, 11, 743. [Google Scholar] [CrossRef] [PubMed]
  107. Sun, L.; Huang, C.; Zhu, M.; Guo, S.; Gao, Q.; Wang, Q.; Chen, B.; Li, R.; Zhao, Y.; Wang, M.; et al. Gastric cancer mesenchymal stem cells regulate PD-L1-CTCF enhancing cancer stem cell-like properties and tumorigenesis. Theranostics 2020, 10, 11950–11962. [Google Scholar] [CrossRef] [PubMed]
  108. Ji, G.; Zhou, W.; Du, J.; Zhou, J.; Wu, D.; Zhao, M.; Yang, L.; Hao, A. PCGF1 promotes epigenetic activation of stemness markers and colorectal cancer stem cell enrichment. Cell Death Dis. 2021, 12, 633. [Google Scholar] [CrossRef]
  109. Xia, Y.; Lv, J.; Jiang, T.; Li, B.; Li, Y.; He, Z.; Xuan, Z.; Sun, G.; Wang, S.; Li, Z.; et al. CircFAM73A promotes the cancer stem cell-like properties of gastric cancer through the miR-490-3p/HMGA2 positive feedback loop and HNRNPK-mediated β-catenin stabilization. J. Exp. Clin. Cancer Res. 2021, 40, 103. [Google Scholar] [CrossRef]
  110. Ciummo, S.L.; D’Antonio, L.; Sorrentino, C.; Fieni, C.; Lanuti, P.; Stassi, G.; Todaro, M.; Di Carlo, E. The C-X-C Motif Chemokine Ligand 1 Sustains Breast Cancer Stem Cell Self-Renewal and Promotes Tumor Progression and Immune Escape Programs. Front. Cell Dev. Biol. 2021, 9, 689286. [Google Scholar] [CrossRef]
  111. Wang, X.; Cai, J.; Zhao, L.; Zhang, D.; Xu, G.; Hu, J.; Zhang, T.; Jin, M. NUMB suppression by miR-9-5P enhances CD44(+) prostate cancer stem cell growth and metastasis. Sci. Rep. 2021, 11, 11210. [Google Scholar] [CrossRef]
  112. Pan, X.W.; Zhang, H.; Xu, D.; Chen, J.X.; Chen, W.J.; Gan, S.S.; Qu, F.J.; Chu, C.M.; Cao, J.W.; Fan, Y.H.; et al. Identification of a novel cancer stem cell subpopulation that promotes progression of human fatal renal cell carcinoma by single-cell RNA-seq analysis. Int. J. Biol. Sci. 2020, 16, 3149–3162. [Google Scholar] [CrossRef] [PubMed]
  113. Lin, X.; Chen, W.; Wei, F.; Xie, X. TV-circRGPD6 Nanoparticle Suppresses Breast Cancer Stem Cell-Mediated Metastasis via the miR-26b/YAF2 Axis. Mol. Ther. J. Am. Soc. Gene Ther. 2021, 29, 244–262. [Google Scholar] [CrossRef] [PubMed]
  114. Iwamoto, K.; Takahashi, H.; Okuzaki, D.; Osawa, H.; Ogino, T.; Miyoshi, N.; Uemura, M.; Matsuda, C.; Yamamoto, H.; Mizushima, T.; et al. Syntenin-1 promotes colorectal cancer stem cell expansion and chemoresistance by regulating prostaglandin E2 receptor. Br. J. Cancer 2020, 123, 955–964. [Google Scholar] [CrossRef] [PubMed]
  115. Li, M.; Pan, M.; Wang, J.; You, C.; Zhao, F.; Zheng, D.; Guo, M.; Xu, H.; Wu, D.; Wang, L.; et al. miR-7 Reduces Breast Cancer Stem Cell Metastasis via Inhibiting RELA to Decrease ESAM Expression. Mol. Ther. Oncolytics 2020, 18, 70–82. [Google Scholar] [CrossRef]
  116. Ko, C.C.H.; Chia, W.K.; Selvarajah, G.T.; Cheah, Y.K.; Wong, Y.P.; Tan, G.C. The Role of Breast Cancer Stem Cell-Related Biomarkers as Prognostic Factors. Diagnostics 2020, 10, 721. [Google Scholar] [CrossRef] [PubMed]
  117. Jakob, M.; Sharaf, K.; Schirmer, M.; Leu, M.; Küffer, S.; Bertlich, M.; Ihler, F.; Haubner, F.; Canis, M.; Kitz, J. Role of cancer stem cell markers ALDH1, BCL11B, BMI-1, and CD44 in the prognosis of advanced HNSCC. Strahlenther. Onkol. 2021, 197, 231–245. [Google Scholar] [CrossRef] [PubMed]
  118. Mardani, A.; Gheytanchi, E.; Mousavie, S.H.; Madjd Jabari, Z.; Shooshtarizadeh, T. Clinical Significance of Cancer Stem Cell Markers CD133 and CXCR4 in Osteosarcomas. Asian Pac. J. Cancer Prev. 2020, 21, 67–73. [Google Scholar] [CrossRef] [Green Version]
  119. Ashrafizadeh, M.; Zarrabi, A.; Saberifar, S.; Hashemi, F.; Hushmandi, K.; Hashemi, F.; Moghadam, E.R.; Mohammadinejad, R.; Najafi, M.; Garg, M. Nobiletin in cancer therapy: How this plant derived-natural compound targets various oncogene and onco-suppressor pathways. Biomedicines 2020, 8, 110. [Google Scholar] [CrossRef]
  120. Yang, J.; Yang, Y.; Wang, L.; Jin, Q.; Pan, M. Nobiletin selectively inhibits oral cancer cell growth by promoting apoptosis and DNA damage in vitro. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2020, 130, 419–427. [Google Scholar] [CrossRef] [PubMed]
  121. Wang, J.G.; Jian, W.J.; Li, Y.; Zhang, J. Nobiletin promotes the pyroptosis of breast cancer via regulation of miR-200b/JAZF1 axis. Kaohsiung J. Med. Sci. 2021, 37, 572–582. [Google Scholar] [CrossRef] [PubMed]
  122. Hermawan, A.; Putri, H. Bioinformatics Studies Provide Insight into Possible Target and Mechanisms of Action of Nobiletin against Cancer Stem Cells. Asian Pac. J. Cancer Prev. 2020, 21, 611–620. [Google Scholar] [CrossRef]
  123. Sp, N.; Kang, D.Y.; Kim, D.H.; Park, J.H.; Lee, H.G.; Kim, H.J.; Darvin, P.; Park, Y.M.; Yang, Y.M. Nobiletin Inhibits CD36-Dependent Tumor Angiogenesis, Migration, Invasion, and Sphere Formation Through the Cd36/Stat3/Nf-Κb Signaling Axis. Nutrients 2018, 10, 772. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Turdo, A.; Glaviano, A.; Pepe, G.; Calapà, F.; Raimondo, S.; Fiori, M.E.; Carbone, D.; Basilicata, M.G.; Di Sarno, V.; Ostacolo, C.; et al. Nobiletin and Xanthohumol Sensitize Colorectal Cancer Stem Cells to Standard Chemotherapy. Cancers 2021, 13, 3927. [Google Scholar] [CrossRef]
  125. Moghadam, E.R.; Ang, H.L.; Asnaf, S.E.; Zabolian, A.; Saleki, H.; Yavari, M.; Esmaeili, H.; Zarrabi, A.; Ashrafizadeh, M.; Kumar, A.P. Broad-Spectrum Preclinical Antitumor Activity of Chrysin: Current Trends and Future Perspectives. Biomolecules 2020, 10, 1374. [Google Scholar] [CrossRef] [PubMed]
  126. Salama, A.A.A.; Allam, R.M. Promising targets of chrysin and daidzein in colorectal cancer: Amphiregulin, CXCL1, and MMP-9. Eur. J. Pharmacol. 2021, 892, 173763. [Google Scholar] [CrossRef]
  127. Kim, K.M.; Jung, J. Upregulation of G Protein-Coupled Estrogen Receptor by Chrysin-Nanoparticles Inhibits Tumor Proliferation and Metastasis in Triple Negative Breast Cancer Xenograft Model. Front. Endocrinol. 2020, 11, 560605. [Google Scholar] [CrossRef]
  128. Chen, L.; Li, Q.; Jiang, Z.; Li, C.; Hu, H.; Wang, T.; Gao, Y.; Wang, D. Chrysin Induced Cell Apoptosis Through H19/let-7a/COPB2 Axis in Gastric Cancer Cells and Inhibited Tumor Growth. Front. Oncol. 2021, 11, 651644. [Google Scholar] [CrossRef]
  129. Ghamkhari, A.; Pouyafar, A.; Salehi, R.; Rahbarghazi, R. Chrysin and Docetaxel Loaded Biodegradable Micelle for Combination Chemotherapy of Cancer Stem Cell. Pharm. Res. 2019, 36, 165. [Google Scholar] [CrossRef] [PubMed]
  130. Debnath, S.; Kanakaraju, M.; Islam, M.; Yeeravalli, R.; Sen, D.; Das, A. In silico design, synthesis and activity of potential drug-like chrysin scaffold-derived selective EGFR inhibitors as anticancer agents. Comput. Biol. Chem. 2019, 83, 107156. [Google Scholar] [CrossRef]
  131. Adham, A.N.; Abdelfatah, S.; Naqishbandi, A.M.; Mahmoud, N.; Efferth, T. Cytotoxicity of apigenin toward multiple myeloma cell lines and suppression of iNOS and COX-2 expression in STAT1-transfected HEK293 cells. Phytomed. Int. J. Phytother. Phytopharm. 2021, 80, 153371. [Google Scholar] [CrossRef]
  132. Korga-Plewko, A.; Michalczyk, M.; Adamczuk, G.; Humeniuk, E.; Ostrowska-Lesko, M.; Jozefczyk, A.; Iwan, M.; Wojcik, M.; Dudka, J. Apigenin and Hesperidin Downregulate DNA Repair Genes in MCF-7 Breast Cancer Cells and Augment Doxorubicin Toxicity. Molecules 2020, 25, 4421. [Google Scholar] [CrossRef] [PubMed]
  133. Sudhakaran, M.; Parra, M.R.; Stoub, H.; Gallo, K.A.; Doseff, A.I. Apigenin by targeting hnRNPA2 sensitizes triple-negative breast cancer spheroids to doxorubicin-induced apoptosis and regulates expression of ABCC4 and ABCG2 drug efflux transporters. Biochem. Pharmacol. 2020, 182, 114259. [Google Scholar] [CrossRef]
  134. Li, Y.W.; Xu, J.; Zhu, G.Y.; Huang, Z.J.; Lu, Y.; Li, X.Q.; Wang, N.; Zhang, F.X. Apigenin suppresses the stem cell-like properties of triple-negative breast cancer cells by inhibiting YAP/TAZ activity. Cell Death Discov. 2018, 4, 105. [Google Scholar] [CrossRef] [PubMed]
  135. Li, Y.; Chen, X.; He, W.; Xia, S.; Jiang, X.; Li, X.; Bai, J.; Li, N.; Chen, L.; Yang, B. Apigenin Enhanced Antitumor Effect of Cisplatin in Lung Cancer via Inhibition of Cancer Stem Cells. Nutr. Cancer 2021, 73, 1489–1497. [Google Scholar] [CrossRef] [PubMed]
  136. Erdogan, S.; Doganlar, O.; Doganlar, Z.B.; Serttas, R.; Turkekul, K.; Dibirdik, I.; Bilir, A. The flavonoid apigenin reduces prostate cancer CD44(+) stem cell survival and migration through PI3K/Akt/NF-κB signaling. Life Sci. 2016, 162, 77–86. [Google Scholar] [CrossRef] [PubMed]
  137. Erdogan, S.; Turkekul, K.; Serttas, R.; Erdogan, Z. The natural flavonoid apigenin sensitizes human CD44(+) prostate cancer stem cells to cisplatin therapy. Biomed. Pharmacother. 2017, 88, 210–217. [Google Scholar] [CrossRef]
  138. Ketkaew, Y.; Osathanon, T.; Pavasant, P.; Sooampon, S. Apigenin inhibited hypoxia induced stem cell marker expression in a head and neck squamous cell carcinoma cell line. Arch. Oral Biol. 2017, 74, 69–74. [Google Scholar] [CrossRef] [PubMed]
  139. Erdogan, S.; Turkekul, K.; Dibirdik, I.; Doganlar, Z.B.; Doganlar, O.; Bilir, A. Midkine silencing enhances the anti-prostate cancer stem cell activity of the flavone apigenin: Cooperation on signaling pathways regulated by ERK, p38, PTEN, PARP, and NF-κB. Investig. New Drugs 2020, 38, 246–263. [Google Scholar] [CrossRef] [PubMed]
  140. Tuli, H.S.; Aggarwal, V.; Kaur, J.; Aggarwal, D.; Parashar, G.; Parashar, N.C.; Tuorkey, M.; Kaur, G.; Savla, R.; Sak, K.; et al. Baicalein: A metabolite with promising antineoplastic activity. Life Sci. 2020, 259, 118183. [Google Scholar] [CrossRef]
  141. Zhang, X.; Ruan, Q.; Zhai, Y.; Lu, D.; Li, C.; Fu, Y.; Zheng, Z.; Song, Y.; Guo, J. Baicalein inhibits non-small-cell lung cancer invasion and metastasis by reducing ezrin tension in inflammation microenvironment. Cancer Sci. 2020, 111, 3802–3812. [Google Scholar] [CrossRef]
  142. Li, P.; Hu, J.; Shi, B.; Tie, J. Baicalein enhanced cisplatin sensitivity of gastric cancer cells by inducing cell apoptosis and autophagy via Akt/mTOR and Nrf2/Keap 1 pathway. Biochem. Biophys. Res. Commun. 2020, 531, 320–327. [Google Scholar] [CrossRef] [PubMed]
  143. Li, J.; Yan, L.; Luo, J.; Tong, L.; Gao, Y.; Feng, W.; Wang, F.; Cui, W.; Li, S.; Sun, Z. Baicalein suppresses growth of non-small cell lung carcinoma by targeting MAP4K3. Biomed. Pharmacother. 2021, 133, 110965. [Google Scholar] [CrossRef]
  144. Song, L.; Chen, X.; Wang, P.; Gao, S.; Qu, C.; Liu, L. Effects of baicalein on pancreatic cancer stem cells via modulation of sonic Hedgehog pathway. Acta Biochim. Biophys. Sin. 2018, 50, 586–596. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Sharifi-Rad, J.; Herrera-Bravo, J.; Salazar, L.A.; Shaheen, S.; Abdulmajid Ayatollahi, S.; Kobarfard, F.; Imran, M.; Imran, A.; Custódio, L.; Dolores López, M.; et al. The Therapeutic Potential of Wogonin Observed in Preclinical Studies. Evid.-Based Complement. Altern. Med. 2021, 2021, 9935451. [Google Scholar] [CrossRef]
  146. Yang, D.; Guo, Q.; Liang, Y.; Zhao, Y.; Tian, X.; Ye, Y.; Tian, J.; Wu, T.; Lu, N. Wogonin induces cellular senescence in breast cancer via suppressing TXNRD2 expression. Arch. Toxicol. 2020, 94, 3433–3447. [Google Scholar] [CrossRef] [PubMed]
  147. Xin, N.J.; Han, M.; Gao, C.; Fan, T.T.; Shi, W. Wogonin suppresses proliferation and invasion of skin epithelioid carcinoma cells through Notch1. Cell. Mol. Biol. 2020, 66, 29–33. [Google Scholar] [CrossRef] [PubMed]
  148. Kirtonia, A.; Sethi, G.; Garg, M. The multifaceted role of reactive oxygen species in tumorigenesis. Cell. Mol. Life Sci. 2020, 77, 4459–4483. [Google Scholar] [CrossRef]
  149. Koh, H.; Sun, H.N.; Xing, Z.; Liu, R.; Chandimali, N.; Kwon, T.; Lee, D.S. Wogonin Influences Osteosarcoma Stem Cell Stemness Through ROS-dependent Signaling. In Vivo 2020, 34, 1077–1084. [Google Scholar] [CrossRef]
  150. Huynh, D.L.; Kwon, T.; Zhang, J.J.; Sharma, N.; Gera, M.; Ghosh, M.; Kim, N.; Kim Cho, S.; Lee, D.S.; Park, Y.H.; et al. Wogonin suppresses stem cell-like traits of CD133 positive osteosarcoma cell via inhibiting matrix metallopeptidase-9 expression. BMC Complement. Altern. Med. 2017, 17, 304. [Google Scholar] [CrossRef] [Green Version]
  151. Li, G.; Zhang, C.; Liang, W.; Zhang, Y.; Shen, Y.; Tian, X. Berberine regulates the Notch1/PTEN/PI3K/AKT/mTOR pathway and acts synergistically with 17-AAG and SAHA in SW480 colon cancer cells. Pharm. Biol. 2021, 59, 21–30. [Google Scholar] [CrossRef]
  152. Hermawan, A.; Ikawati, M.; Jenie, R.I.; Khumaira, A.; Putri, H.; Nurhayati, I.P.; Angraini, S.M.; Muflikhasari, H.A. Identification of potential therapeutic target of naringenin in breast cancer stem cells inhibition by bioinformatics and in vitro studies. Saudi Pharm. J. Off. Publ. Saudi Pharm. Soc. 2021, 29, 12–26. [Google Scholar] [CrossRef]
  153. Tajaldini, M.; Samadi, F.; Khosravi, A.; Ghasemnejad, A.; Asadi, J. Protective and anticancer effects of orange peel extract and naringin in doxorubicin treated esophageal cancer stem cell xenograft tumor mouse model. Biomed. Pharmacother. 2020, 121, 109594. [Google Scholar] [CrossRef] [PubMed]
  154. Hermawan, A.; Ikawati, M.; Khumaira, A.; Putri, H.; Jenie, R.I.; Angraini, S.M.; Muflikhasari, H.A. Bioinformatics and In Vitro Studies Reveal the Importance of p53, PPARG and Notch Signaling Pathway in Inhibition of Breast Cancer Stem Cells by Hesperetin. Adv. Pharm. Bull. 2021, 11, 351–360. [Google Scholar] [CrossRef]
  155. Farooqi, A.A.; Naureen, H.; Zahid, R.; Youssef, L.; Attar, R.; Xu, B. Cancer chemopreventive role of fisetin: Regulation of cell signaling pathways in different cancers. Pharmacol. Res. 2021, 172, 105784. [Google Scholar] [CrossRef]
  156. Imran, M.; Saeed, F.; Gilani, S.A.; Shariati, M.A.; Imran, A.; Afzaal, M.; Atif, M.; Tufail, T.; Anjum, F.M. Fisetin: An anticancer perspective. Food Sci. Nutr. 2021, 9, 3–16. [Google Scholar] [CrossRef]
  157. Ding, G.; Xu, X.; Li, D.; Chen, Y.; Wang, W.; Ping, D.; Jia, S.; Cao, L. Fisetin inhibits proliferation of pancreatic adenocarcinoma by inducing DNA damage via RFXAP/KDM4A-dependent histone H3K36 demethylation. Cell Death Dis. 2020, 11, 893. [Google Scholar] [CrossRef] [PubMed]
  158. Si, Y.; Liu, J.; Shen, H.; Zhang, C.; Wu, Y.; Huang, Y.; Gong, Z.; Xue, J.; Liu, T. Fisetin decreases TET1 activity and CCNY/CDK16 promoter 5hmC levels to inhibit the proliferation and invasion of renal cancer stem cell. J. Cell. Mol. Med. 2019, 23, 1095–1105. [Google Scholar] [CrossRef]
  159. Yin, Z.; Li, J.; Kang, L.; Liu, X.; Luo, J.; Zhang, L.; Li, Y.; Cai, J. Epigallocatechin-3-gallate induces autophagy-related apoptosis associated with LC3B II and Beclin expression of bladder cancer cells. J. Food Biochem. 2021, 45, e13758. [Google Scholar] [CrossRef] [PubMed]
  160. Ding, F.; Yang, S. Epigallocatechin-3-gallate inhibits proliferation and triggers apoptosis in colon cancer via the hedgehog/phosphoinositide 3-kinase pathways. Can. J. Physiol. Pharmacol. 2021, 99. [Google Scholar] [CrossRef] [PubMed]
  161. Panji, M.; Behmard, V.; Zare, Z.; Malekpour, M.; Nejadbiglari, H.; Yavari, S.; Nayerpour Dizaj, T.; Safaeian, A.; Maleki, N.; Abbasi, M.; et al. Suppressing effects of green tea extract and Epigallocatechin-3-gallate (EGCG) on TGF-β- induced Epithelial-to-mesenchymal transition via ROS/Smad signaling in human cervical cancer cells. Gene 2021, 794, 145774. [Google Scholar] [CrossRef]
  162. Mokhtari, H.; Yaghmaei, B.; Sirati-Sabet, M.; Jafari, N.; Mardomi, A.; Abediankenari, S.; Mahrooz, A. Epigallocatechin-3-gallate Enhances the Efficacy of MicroRNA-34a Mimic and MicroRNA-93 Inhibitor Co-transfection in Prostate Cancer Cell Line. Iran. J. Allergy Asthma Immunol. 2020, 19, 612–623. [Google Scholar] [CrossRef] [PubMed]
  163. Jiang, P.; Xu, C.; Zhang, P.; Ren, J.; Mageed, F.; Wu, X.; Chen, L.; Zeb, F.; Feng, Q.; Li, S. Epigallocatechin-3-gallate inhibits self-renewal ability of lung cancer stem-like cells through inhibition of CLOCK. Int. J. Mol. Med. 2020, 46, 2216–2224. [Google Scholar] [CrossRef]
  164. Xia, L.; Li, F.; Qiu, J.; Feng, Z.; Xu, Z.; Chen, Z.; Sun, J. Oncogenic miR-20b-5p contributes to malignant behaviors of breast cancer stem cells by bidirectionally regulating CCND1 and E2F1. BMC Cancer 2020, 20, 949. [Google Scholar] [CrossRef] [PubMed]
  165. Wei, Y.; Li, H.; Qu, Q. miR-484 suppresses endocrine therapy-resistant cells by inhibiting KLF4-induced cancer stem cells in estrogen receptor-positive cancers. Breast Cancer 2021, 28, 175–186. [Google Scholar] [CrossRef] [PubMed]
  166. Zhou, Q.; Cui, F.; Lei, C.; Ma, S.; Huang, J.; Wang, X.; Qian, H.; Zhang, D.; Yang, Y. ATG7-mediated autophagy involves in miR-138-5p regulated self-renewal and invasion of lung cancer stem-like cells derived from A549 cells. Anti-Cancer Drugs 2021, 32, 376–385. [Google Scholar] [CrossRef]
  167. Razi, S.; Sadeghi, A.; Asadi-Lari, Z.; Tam, K.J.; Kalantari, E.; Madjd, Z. DCLK1, a promising colorectal cancer stem cell marker, regulates tumor progression and invasion through miR-137 and miR-15a dependent manner. Clin. Exp. Med. 2021, 21, 139–147. [Google Scholar] [CrossRef]
  168. Jiang, P.; Xu, C.; Chen, L.; Chen, A.; Wu, X.; Zhou, M.; Haq, I.U.; Mariyam, Z.; Feng, Q. Epigallocatechin-3-gallate inhibited cancer stem cell-like properties by targeting hsa-mir-485-5p/RXRα in lung cancer. J. Cell. Biochem. 2018, 119, 8623–8635. [Google Scholar] [CrossRef] [PubMed]
  169. Zhu, J.; Jiang, Y.; Yang, X.; Wang, S.; Xie, C.; Li, X.; Li, Y.; Chen, Y.; Wang, X.; Meng, Y.; et al. Wnt/β-catenin pathway mediates (-)-Epigallocatechin-3-gallate (EGCG) inhibition of lung cancer stem cells. Biochem. Biophys. Res. Commun. 2017, 482, 15–21. [Google Scholar] [CrossRef] [PubMed]
  170. Chen, Y.; Wang, X.Q.; Zhang, Q.; Zhu, J.Y.; Li, Y.; Xie, C.F.; Li, X.T.; Wu, J.S.; Geng, S.S.; Zhong, C.Y.; et al. (-)-Epigallocatechin-3-Gallate Inhibits Colorectal Cancer Stem Cells by Suppressing Wnt/β-Catenin Pathway. Nutrients 2017, 9, 572. [Google Scholar] [CrossRef] [Green Version]
  171. Park, M.; Sundaramoorthy, P.; Sim, J.J.; Jeong, K.Y.; Kim, H.M. Synergistically Anti-metastatic Effect of 5-Flourouracil on Colorectal Cancer Cells via Calcium-mediated Focal Adhesion Kinase Proteolysis. Anticancer Res. 2017, 37, 103–114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Ashrafizadeh, M.; Zarrabi, A.; Hushmandi, K.; Hashemi, F.; Hashemi, F.; Samarghandian, S.; Najafi, M. MicroRNAs in cancer therapy: Their involvement in oxaliplatin sensitivity/resistance of cancer cells with a focus on colorectal cancer. Life Sci. 2020, 256, 117973. [Google Scholar] [CrossRef] [PubMed]
  173. Toden, S.; Tran, H.M.; Tovar-Camargo, O.A.; Okugawa, Y.; Goel, A. Epigallocatechin-3-gallate targets cancer stem-like cells and enhances 5-fluorouracil chemosensitivity in colorectal cancer. Oncotarget 2016, 7, 16158–16171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Sun, X.; Song, J.; Li, E.; Geng, H.; Li, Y.; Yu, D.; Zhong, C. (-)-Epigallocatechin-3-gallate inhibits bladder cancer stem cells via suppression of sonic hedgehog pathway. Oncol. Rep. 2019, 42, 425–435. [Google Scholar] [CrossRef] [PubMed]
  175. Abd El-Rahman, S.S.; Shehab, G.; Nashaat, H. Epigallocatechin-3-Gallate: The Prospective Targeting of Cancer Stem Cells and Preventing Metastasis of Chemically-Induced Mammary Cancer in Rats. Am. J. Med. Sci. 2017, 354, 54–63. [Google Scholar] [CrossRef]
  176. Lee, S.H.; Nam, H.J.; Kang, H.J.; Kwon, H.W.; Lim, Y.C. Epigallocatechin-3-gallate attenuates head and neck cancer stem cell traits through suppression of Notch pathway. Eur. J. Cancer 2013, 49, 3210–3218. [Google Scholar] [CrossRef] [PubMed]
  177. Li, Y.J.; Wu, S.L.; Lu, S.M.; Chen, F.; Guo, Y.; Gan, S.M.; Shi, Y.L.; Liu, S.; Li, S.L. (-)-Epigallocatechin-3-gallate inhibits nasopharyngeal cancer stem cell self-renewal and migration and reverses the epithelial-mesenchymal transition via NF-κB p65 inactivation. Tumour Biol. Int. Soc. Oncodev. Biol. Med. 2015, 36, 2747–2761. [Google Scholar] [CrossRef]
  178. Wang, K.L.; Yu, Y.C.; Hsia, S.M. Perspectives on the Role of Isoliquiritigenin in Cancer. Cancers 2021, 13, 115. [Google Scholar] [CrossRef]
  179. Chen, H.Y.; Chiang, Y.F.; Huang, J.S.; Huang, T.C.; Shih, Y.H.; Wang, K.L.; Ali, M.; Hong, Y.H.; Shieh, T.M.; Hsia, S.M. Isoliquiritigenin Reverses Epithelial-Mesenchymal Transition Through Modulation of the TGF-β/Smad Signaling Pathway in Endometrial Cancer. Cancers 2021, 13, 1236. [Google Scholar] [CrossRef]
  180. Peng, F.; Tang, H.; Du, J.; Chen, J.; Peng, C. Isoliquiritigenin Suppresses EMT-Induced Metastasis in Triple-Negative Breast Cancer through miR-200c/C-JUN/[Formula: See text]-Catenin. Am. J. Chin. Med. 2021, 49, 505–523. [Google Scholar] [CrossRef]
  181. Jin, H.; Kim, H.S.; Yu, S.T.; Shin, S.R.; Lee, S.H.; Seo, G.S. Synergistic anticancer effect of docosahexaenoic acid and isoliquiritigenin on human colorectal cancer cells through ROS-mediated regulation of the JNK and cytochrome c release. Mol. Biol. Rep. 2021, 48, 1171–1180. [Google Scholar] [CrossRef]
  182. Wang, G.; Yu, Y.; Wang, Y.Z.; Yin, P.H.; Xu, K.; Zhang, H. The effects and mechanisms of isoliquiritigenin loaded nanoliposomes regulated AMPK/mTOR mediated glycolysis in colorectal cancer. Artif. Cells Nanomed. Biotechnol. 2020, 48, 1231–1249. [Google Scholar] [CrossRef]
  183. Wang, N.; Wang, Z.; Peng, C.; You, J.; Shen, J.; Han, S.; Chen, J. Dietary compound isoliquiritigenin targets GRP78 to chemosensitize breast cancer stem cells via β-catenin/ABCG2 signaling. Carcinogenesis 2014, 35, 2544–2554. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Hu, F.W.; Yu, C.C.; Hsieh, P.L.; Liao, Y.W.; Lu, M.Y.; Chu, P.M. Targeting oral cancer stemness and chemoresistance by isoliquiritigenin-mediated GRP78 regulation. Oncotarget 2017, 8, 93912–93923. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Wang, N.; Wang, Z.; Wang, Y.; Xie, X.; Shen, J.; Peng, C.; You, J.; Peng, F.; Tang, H.; Guan, X.; et al. Dietary compound isoliquiritigenin prevents mammary carcinogenesis by inhibiting breast cancer stem cells through WIF1 demethylation. Oncotarget 2015, 6, 9854–9876. [Google Scholar] [CrossRef] [Green Version]
  186. Green, J.M.; Alvero, A.B.; Kohen, F.; Mor, G. 7-(O)-Carboxymethyl daidzein conjugated to N-t-Boc-hexylenediamine: A novel compound capable of inducing cell death in epithelial ovarian cancer stem cells. Cancer Biol. Ther. 2009, 8, 1747–1753. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Andrade, F.O.; Liu, F.; Zhang, X.; Rosim, M.P.; Dani, C.; Cruz, I.; Wang, T.T.Y.; Helferich, W.; Li, R.W.; Hilakivi-Clarke, L. Genistein Reduces the Risk of Local Mammary Cancer Recurrence and Ameliorates Alterations in the Gut Microbiota in the Offspring of Obese Dams. Nutrients 2021, 13, 201. [Google Scholar] [CrossRef] [PubMed]
  188. Liu, X.; Wang, Q.; Liu, B.; Zheng, X.; Li, P.; Zhao, T.; Jin, X.; Ye, F.; Zhang, P.; Chen, W.; et al. Genistein inhibits radiation-induced invasion and migration of glioblastoma cells by blocking the DNA-PKcs/Akt2/Rac1 signaling pathway. Radiother. Oncol. J. Eur. Soc. Ther. Radiol. Oncol. 2021, 155, 93–104. [Google Scholar] [CrossRef]
  189. Yu, Y.; Xing, Y.; Zhang, Q.; Zhang, Q.; Huang, S.; Li, X.; Gao, C. Soy isoflavone genistein inhibits hsa_circ_0031250/miR-873-5p/FOXM1 axis to suppress non-small-cell lung cancer progression. IUBMB Life 2021, 73, 92–107. [Google Scholar] [CrossRef] [PubMed]
  190. Erten, F.; Yenice, E.; Orhan, C.; Er, B.; Demirel Öner, P.; Defo Deeh, P.B.; Şahin, K. Genistein suppresses the inflammation and GSK-3 pathway in an animal model of spontaneous ovarian cancer. Turk. J. Med. Sci. 2021, 51, 1465–1471. [Google Scholar] [CrossRef] [PubMed]
  191. Dev, A.; Sardoiwala, M.N.; Kushwaha, A.C.; Karmakar, S.; Choudhury, S.R. Genistein nanoformulation promotes selective apoptosis in oral squamous cell carcinoma through repression of 3PK-EZH2 signalling pathway. Phytomed. Int. J. Phytother. Phytopharm. 2021, 80, 153386. [Google Scholar] [CrossRef]
  192. Vodnik, V.V.; Mojić, M.; Stamenović, U.; Otoničar, M.; Ajdžanović, V.; Maksimović-Ivanić, D.; Mijatović, S.; Marković, M.M.; Barudžija, T.; Filipović, B.; et al. Development of genistein-loaded gold nanoparticles and their antitumor potential against prostate cancer cell lines. Mater. Sci. Eng. C Mater. Biol. Appl. 2021, 124, 112078. [Google Scholar] [CrossRef] [PubMed]
  193. Zhang, Q.; Cao, W.S.; Wang, X.Q.; Zhang, M.; Lu, X.M.; Chen, J.Q.; Chen, Y.; Ge, M.M.; Zhong, C.Y.; Han, H.Y. Genistein inhibits nasopharyngeal cancer stem cells through sonic hedgehog signaling. Phytother. Res. 2019, 33, 2783–2791. [Google Scholar] [CrossRef]
  194. Xie, C.; Zhu, J.; Jiang, Y.; Chen, J.; Wang, X.; Geng, S.; Wu, J.; Zhong, C.; Li, X.; Meng, Z. Sulforaphane Inhibits the Acquisition of Tobacco Smoke-Induced Lung Cancer Stem Cell-Like Properties via the IL-6/ΔNp63α/Notch Axis. Theranostics 2019, 9, 4827–4840. [Google Scholar] [CrossRef] [PubMed]
  195. Liu, Y.; Zou, T.; Wang, S.; Chen, H.; Su, D.; Fu, X.; Zhang, Q.; Kang, X. Genistein-induced differentiation of breast cancer stem/progenitor cells through a paracrine mechanism. Int. J. Oncol. 2016, 48, 1063–1072. [Google Scholar] [CrossRef] [Green Version]
  196. Huang, W.; Wan, C.; Luo, Q.; Huang, Z.; Luo, Q. Genistein-inhibited cancer stem cell-like properties and reduced chemoresistance of gastric cancer. Int. J. Mol. Sci. 2014, 15, 3432–3443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Cao, X.; Ren, K.; Song, Z.; Li, D.; Quan, M.; Zheng, Y.; Cao, J.; Zeng, W.; Zou, H. 7-Difluoromethoxyl-5,4′-di-n-octyl genistein inhibits the stem-like characteristics of gastric cancer stem-like cells and reverses the phenotype of epithelial-mesenchymal transition in gastric cancer cells. Oncol. Rep. 2016, 36, 1157–1165. [Google Scholar] [CrossRef] [PubMed]
  198. Yu, D.; Shin, H.S.; Lee, Y.S.; Lee, D.; Kim, S.; Lee, Y.C. Genistein attenuates cancer stem cell characteristics in gastric cancer through the downregulation of Gli1. Oncol. Rep. 2014, 31, 673–678. [Google Scholar] [CrossRef] [Green Version]
  199. Zhang, L.; Li, L.; Jiao, M.; Wu, D.; Wu, K.; Li, X.; Zhu, G.; Yang, L.; Wang, X.; Hsieh, J.T.; et al. Genistein inhibits the stemness properties of prostate cancer cells through targeting Hedgehog-Gli1 pathway. Cancer Lett. 2012, 323, 48–57. [Google Scholar] [CrossRef] [PubMed]
  200. Fan, P.; Fan, S.; Wang, H.; Mao, J.; Shi, Y.; Ibrahim, M.M.; Ma, W.; Yu, X.; Hou, Z.; Wang, B.; et al. Genistein decreases the breast cancer stem-like cell population through Hedgehog pathway. Stem Cell Res. Ther. 2013, 4, 146. [Google Scholar] [CrossRef] [Green Version]
  201. Ning, Y.X.; Li, Q.X.; Ren, K.Q.; Quan, M.F.; Cao, J.G. 7-difluoromethoxyl-5,4′-di-n-octyl genistein inhibits ovarian cancer stem cell characteristics through the downregulation of FOXM1. Oncol. Lett. 2014, 8, 295–300. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Sekar, V.; Anandasadagopan, S.K.; Ganapasam, S. Genistein regulates tumor microenvironment and exhibits anticancer effect in dimethyl hydrazine-induced experimental colon carcinogenesis. BioFactors 2016, 42, 623–637. [Google Scholar] [CrossRef] [PubMed]
  203. Ning, Y.; Xu, M.; Cao, X.; Chen, X.; Luo, X. Inactivation of AKT, ERK and NF-κB by genistein derivative, 7-difluoromethoxyl-5,4′-di-n-octylygenistein, reduces ovarian carcinoma oncogenicity. Oncol. Rep. 2017, 38, 949–958. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Ning, Y.; Luo, C.; Ren, K.; Quan, M.; Cao, J. FOXO3a-mediated suppression of the self-renewal capacity of sphere-forming cells derived from the ovarian cancer SKOV3 cell line by 7-difluoromethoxyl-5,4′-di-n-octyl genistein. Mol. Med. Rep. 2014, 9, 1982–1988. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. De Francesco, E.M.; Ózsvári, B.; Sotgia, F.; Lisanti, M.P. Dodecyl-TPP Targets Mitochondria and Potently Eradicates Cancer Stem Cells (CSCs): Synergy With FDA-Approved Drugs and Natural Compounds (Vitamin C and Berberine). Front. Oncol. 2019, 9, 615. [Google Scholar] [CrossRef] [Green Version]
  206. Wu, Y.S.; Ho, J.Y.; Yu, C.P.; Cho, C.J.; Wu, C.L.; Huang, C.S.; Gao, H.W.; Yu, D.S. Ellagic Acid Resensitizes Gemcitabine-Resistant Bladder Cancer Cells by Inhibiting Epithelial-Mesenchymal Transition and Gemcitabine Transporters. Cancers 2021, 13, 2032. [Google Scholar] [CrossRef]
  207. Mohammed Saleem, Y.I.; Selim, M.I. MDM2 as a target for ellagic acid-mediated suppression of prostate cancer cells in vitro. Oncol. Rep. 2020, 44, 1255–1265. [Google Scholar] [CrossRef]
  208. Elsaid, F.G.; Alshehri, M.A.; Shati, A.A.; Al-Kahtani, M.A.; Alsheri, A.S.; Massoud, E.E.; El-Kott, A.F.; El-Mekkawy, H.I.; Al-Ramlawy, A.M.; Abdraboh, M.E. The anti-tumourigenic effect of ellagic acid in SKOV-3 ovarian cancer cells entails activation of autophagy mediated by inhibiting Akt and activating AMPK. Clin. Exp. Pharmacol. Physiol. 2020, 47, 1611–1621. [Google Scholar] [CrossRef]
  209. Yoganathan, S.; Alagaratnam, A.; Acharekar, N.; Kong, J. Ellagic Acid and Schisandrins: Natural Biaryl Polyphenols with Therapeutic Potential to Overcome Multidrug Resistance in Cancer. Cells 2021, 10, 458. [Google Scholar] [CrossRef]
  210. Wang, N.; Wang, Q.; Tang, H.; Zhang, F.; Zheng, Y.; Wang, S.; Zhang, J.; Wang, Z.; Xie, X. Direct inhibition of ACTN4 by ellagic acid limits breast cancer metastasis via regulation of β-catenin stabilization in cancer stem cells. J. Exp. Clin. Cancer Res. 2017, 36, 172. [Google Scholar] [CrossRef] [Green Version]
  211. Núñez-Sánchez, M.; Karmokar, A.; González-Sarrías, A.; García-Villalba, R.; Tomás-Barberán, F.A.; García-Conesa, M.T.; Brown, K.; Espín, J.C. In vivo relevant mixed urolithins and ellagic acid inhibit phenotypic and molecular colon cancer stem cell features: A new potentiality for ellagitannin metabolites against cancer. Food Chem. Toxicol. 2016, 92, 8–16. [Google Scholar] [CrossRef]
  212. Mirzaei, S.; Gholami, M.H.; Zabolian, A.; Saleki, H.; Farahani, M.V.; Hamzehlou, S.; Far, F.B.; Sharifzadeh, S.O.; Samarghandian, S.; Khan, H.; et al. Caffeic acid and its derivatives as potential modulators of oncogenic molecular pathways: New hope in the fight against cancer. Pharmacol. Res. 2021, 171, 105759. [Google Scholar] [CrossRef] [PubMed]
  213. Tyszka-Czochara, M.; Bukowska-Strakova, K.; Kocemba-Pilarczyk, K.A.; Majka, M. Caffeic Acid Targets AMPK Signaling and Regulates Tricarboxylic Acid Cycle Anaplerosis while Metformin Downregulates HIF-1α-Induced Glycolytic Enzymes in Human Cervical Squamous Cell Carcinoma Lines. Nutrients 2018, 10, 841. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Teng, Y.N.; Wang, C.C.N.; Liao, W.C.; Lan, Y.H.; Hung, C.C. Caffeic Acid Attenuates Multi-Drug Resistance in Cancer Cells by Inhibiting Efflux Function of Human P-glycoprotein. Molecules 2020, 25, 247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Colpan, R.D.; Erdemir, A. Co-delivery of quercetin and caffeic-acid phenethyl ester by polymeric nanoparticles for improved antitumor efficacy in colon cancer cells. J. Microencapsul. 2021, 38, 381–393. [Google Scholar] [CrossRef] [PubMed]
  216. Sari, C.; SÜmer, C.; Celep EyÜpoĞlu, F. Caffeic acid phenethyl ester induces apoptosis in colorectal cancer cells via inhibition of survivin. Turk. J. Biol. 2020, 44, 264–274. [Google Scholar] [CrossRef]
  217. Park, S.R.; Kim, S.R.; Hong, I.S.; Lee, H.Y. A Novel Therapeutic Approach for Colorectal Cancer Stem Cells: Blocking the PI3K/Akt Signaling Axis with Caffeic Acid. Front. Cell Dev. Biol. 2020, 8, 585987. [Google Scholar] [CrossRef]
  218. Yang, Y.; Li, Y.; Wang, K.; Wang, Y.; Yin, W.; Li, L. P38/NF-κB/snail pathway is involved in caffeic acid-induced inhibition of cancer stem cells-like properties and migratory capacity in malignant human keratinocyte. PLoS ONE 2013, 8, e58915. [Google Scholar] [CrossRef] [Green Version]
  219. Omene, C.O.; Wu, J.; Frenkel, K. Caffeic Acid Phenethyl Ester (CAPE) derived from propolis, a honeybee product, inhibits growth of breast cancer stem cells. Investig. New Drugs 2012, 30, 1279–1288. [Google Scholar] [CrossRef] [Green Version]
  220. Li, Y.; Jiang, F.; Chen, L.; Yang, Y.; Cao, S.; Ye, Y.; Wang, X.; Mu, J.; Li, Z.; Li, L. Blockage of TGFβ-SMAD2 by demethylation-activated miR-148a is involved in caffeic acid-induced inhibition of cancer stem cell-like properties in vitro and in vivo. FEBS Open Bio 2015, 5, 466–475. [Google Scholar] [CrossRef] [Green Version]
  221. Ashrafizadeh, M.; Ahmadi, Z.; Farkhondeh, T.; Samarghandian, S. Autophagy regulation using luteolin: New insight into its anti-tumor activity. Cancer Cell Int. 2020, 20, 537. [Google Scholar] [CrossRef] [PubMed]
  222. Wu, H.T.; Lin, J.; Liu, Y.E.; Chen, H.F.; Hsu, K.W.; Lin, S.H.; Peng, K.Y.; Lin, K.J.; Hsieh, C.C.; Chen, D.R. Luteolin suppresses androgen receptor-positive triple-negative breast cancer cell proliferation and metastasis by epigenetic regulation of MMP9 expression via the AKT/mTOR signaling pathway. Phytomed. Int. J. Phytother. Phytopharm. 2021, 81, 153437. [Google Scholar] [CrossRef]
  223. Cao, D.; Zhu, G.Y.; Lu, Y.; Yang, A.; Chen, D.; Huang, H.J.; Peng, S.X.; Chen, L.W.; Li, Y.W. Luteolin suppresses epithelial-mesenchymal transition and migration of triple-negative breast cancer cells by inhibiting YAP/TAZ activity. Biomed. Pharmacother. 2020, 129, 110462. [Google Scholar] [CrossRef] [PubMed]
  224. Chen, Y.; Wang, M.H.; Zhu, J.Y.; Xie, C.F.; Li, X.T.; Wu, J.S.; Geng, S.S.; Han, H.Y.; Zhong, C.Y. TAp63α targeting of Lgr5 mediates colorectal cancer stem cell properties and sulforaphane inhibition. Oncogenesis 2020, 9, 89. [Google Scholar] [CrossRef] [PubMed]
  225. Tu, D.G.; Lin, W.T.; Yu, C.C.; Lee, S.S.; Peng, C.Y.; Lin, T.; Yu, C.H. Chemotherapeutic effects of luteolin on radio-sensitivity enhancement and interleukin-6/signal transducer and activator of transcription 3 signaling repression of oral cancer stem cells. J. Formos. Med. Assoc. 2016, 115, 1032–1038. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Almeida, A.F.; Borge, G.I.A.; Piskula, M.; Tudose, A.; Tudoreanu, L.; Valentová, K.; Williamson, G.; Santos, C.N. Bioavailability of quercetin in humans with a focus on interindividual variation. Compr. Rev. Food Sci. Food Saf. 2018, 17, 714–731. [Google Scholar] [CrossRef] [Green Version]
  227. Xiao, L.; Luo, G.; Tang, Y.; Yao, P. Quercetin and iron metabolism: What we know and what we need to know. Food Chem. Toxicol. 2018, 114, 190–203. [Google Scholar] [CrossRef] [PubMed]
  228. Ashrafizadeh, M.; Ahmadi, Z.; Farkhondeh, T.; Samarghandian, S. Autophagy as a molecular target of quercetin underlying its protective effects in human diseases. Arch. Physiol. Biochem. 2019. [Google Scholar] [CrossRef] [PubMed]
  229. Ashrafizadeh, M.; Samarghandian, S.; Hushmandi, K.; Zabolian, A.; Shahinozzaman, M.; Saleki, H.; Esmaeili, H.; Raei, M.; Entezari, M.; Zarrabi, A.; et al. Quercetin in attenuation of ischemic/reperfusion injury: A review. Curr. Mol. Pharmacol. 2020. [Google Scholar] [CrossRef]
  230. Yousuf, M.; Khan, P.; Shamsi, A.; Shahbaaz, M.; Hasan, G.M.; Haque, Q.M.R.; Christoffels, A.; Islam, A.; Hassan, M.I. Inhibiting CDK6 Activity by Quercetin Is an Attractive Strategy for Cancer Therapy. ACS Omega 2020, 5, 27480–27491. [Google Scholar] [CrossRef] [PubMed]
  231. Wang, Z.X.; Ma, J.; Li, X.Y.; Wu, Y.; Shi, H.; Chen, Y.; Lu, G.; Shen, H.M.; Lu, G.D.; Zhou, J. Quercetin induces p53-independent cancer cell death through lysosome activation by the transcription factor EB and Reactive Oxygen Species-dependent ferroptosis. Br. J. Pharmacol. 2021, 178, 1133–1148. [Google Scholar] [CrossRef]
  232. Liu, M.; Fu, M.; Yang, X.; Jia, G.; Shi, X.; Ji, J.; Liu, X.; Zhai, G. Paclitaxel and quercetin co-loaded functional mesoporous silica nanoparticles overcoming multidrug resistance in breast cancer. Colloids Surf. B Biointerfaces 2020, 196, 111284. [Google Scholar] [CrossRef]
  233. Li, X.; Zhou, N.; Wang, J.; Liu, Z.; Wang, X.; Zhang, Q.; Liu, Q.; Gao, L.; Wang, R. Quercetin suppresses breast cancer stem cells (CD44(+)/CD24(-)) by inhibiting the PI3K/Akt/mTOR-signaling pathway. Life Sci. 2018, 196, 56–62. [Google Scholar] [CrossRef]
  234. Wang, R.; Yang, L.; Li, S.; Ye, D.; Yang, L.; Liu, Q.; Zhao, Z.; Cai, Q.; Tan, J.; Li, X. Quercetin Inhibits Breast Cancer Stem Cells via Downregulation of Aldehyde Dehydrogenase 1A1 (ALDH1A1), Chemokine Receptor Type 4 (CXCR4), Mucin 1 (MUC1), and Epithelial Cell Adhesion Molecule (EpCAM). Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2018, 24, 412–420. [Google Scholar] [CrossRef] [Green Version]
  235. Li, S.; Zhao, Q.; Wang, B.; Yuan, S.; Wang, X.; Li, K. Quercetin reversed MDR in breast cancer cells through down-regulating P-gp expression and eliminating cancer stem cells mediated by YB-1 nuclear translocation. Phytother. Res. 2018, 32, 1530–1536. [Google Scholar] [CrossRef] [PubMed]
  236. Li, S.; Yuan, S.; Zhao, Q.; Wang, B.; Wang, X.; Li, K. Quercetin enhances chemotherapeutic effect of doxorubicin against human breast cancer cells while reducing toxic side effects of it. Biomed. Pharmacother. 2018, 100, 441–447. [Google Scholar] [CrossRef]
  237. Srivastava, R.K.; Tang, S.N.; Zhu, W.; Meeker, D.; Shankar, S. Sulforaphane synergizes with quercetin to inhibit self-renewal capacity of pancreatic cancer stem cells. Front. Biosci. (Elite Ed.) 2011, 3, 515–528. [Google Scholar] [CrossRef] [Green Version]
  238. Nwaeburu, C.C.; Abukiwan, A.; Zhao, Z.; Herr, I. Quercetin-induced miR-200b-3p regulates the mode of self-renewing divisions in pancreatic cancer. Mol. Cancer 2017, 16, 23. [Google Scholar] [CrossRef] [Green Version]
  239. Appari, M.; Babu, K.R.; Kaczorowski, A.; Gross, W.; Herr, I. Sulforaphane, quercetin and catechins complement each other in elimination of advanced pancreatic cancer by miR-let-7 induction and K-ras inhibition. Int. J. Oncol. 2014, 45, 1391–1400. [Google Scholar] [CrossRef] [Green Version]
  240. Zhou, W.; Kallifatidis, G.; Baumann, B.; Rausch, V.; Mattern, J.; Gladkich, J.; Giese, N.; Moldenhauer, G.; Wirth, T.; Büchler, M.W.; et al. Dietary polyphenol quercetin targets pancreatic cancer stem cells. Int. J. Oncol. 2010, 37, 551–561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  241. Li, Y.; Wang, Z.; Jin, J.; Zhu, S.X.; He, G.Q.; Li, S.H.; Wang, J.; Cai, Y. Quercetin pretreatment enhances the radiosensitivity of colon cancer cells by targeting Notch-1 pathway. Biochem. Biophys. Res. Commun. 2020, 523, 947–953. [Google Scholar] [CrossRef] [PubMed]
  242. Atashpour, S.; Fouladdel, S.; Movahhed, T.K.; Barzegar, E.; Ghahremani, M.H.; Ostad, S.N.; Azizi, E. Quercetin induces cell cycle arrest and apoptosis in CD133(+) cancer stem cells of human colorectal HT29 cancer cell line and enhances anticancer effects of doxorubicin. Iran. J. Basic Med. Sci. 2015, 18, 635–643. [Google Scholar]
  243. Sikka, S.; Chen, L.; Sethi, G.; Kumar, A.P. Targeting PPARγ Signaling Cascade for the Prevention and Treatment of Prostate Cancer. PPAR Res. 2012, 2012, 968040. [Google Scholar] [CrossRef] [Green Version]
  244. Hussain, Y.; Mirzaei, S.; Ashrafizadeh, M.; Zarrabi, A.; Hushmandi, K.; Khan, H.; Daglia, M. Quercetin and Its Nano-Scale Delivery Systems in Prostate Cancer Therapy: Paving the Way for Cancer Elimination and Reversing Chemoresistance. Cancers 2021, 13, 1602. [Google Scholar] [CrossRef] [PubMed]
  245. Soleymani, L.; Zarrabi, A.; Hashemi, F.; Zabolian, A.; Banihashemi, S.; Moghadam, S.; Hushmandi, K.; Samarghandian, S.; Ashrafizadeh, M.; Khan, H. Role of ZEB family members in proliferation, metastasis and chemoresistance of prostate cancer cells: Revealing signaling networks. Curr. Cancer Drug Targets 2021. [Google Scholar] [CrossRef]
  246. Zhang, J.; Ahn, K.S.; Kim, C.; Shanmugam, M.K.; Siveen, K.S.; Arfuso, F.; Samym, R.P.; Deivasigamanim, A.; Lim, L.H.; Wang, L.; et al. Nimbolide-Induced Oxidative Stress Abrogates STAT3 Signaling Cascade and Inhibits Tumor Growth in Transgenic Adenocarcinoma of Mouse Prostate Model. Antioxid. Redox Signal. 2016, 24, 575–589. [Google Scholar] [CrossRef] [PubMed]
  247. Lee, J.H.; Kim, C.; Baek, S.H.; Ko, J.H.; Lee, S.G.; Yang, W.M.; Um, J.Y.; Sethi, G.; Ahn, K.S. Capsazepine inhibits JAK/STAT3 signaling, tumor growth, and cell survival in prostate cancer. Oncotarget 2017, 8, 17700–17711. [Google Scholar] [CrossRef]
  248. Erdogan, S.; Turkekul, K.; Dibirdik, I.; Doganlar, O.; Doganlar, Z.B.; Bilir, A.; Oktem, G. Midkine downregulation increases the efficacy of quercetin on prostate cancer stem cell survival and migration through PI3K/AKT and MAPK/ERK pathway. Biomed. Pharmacother. 2018, 107, 793–805. [Google Scholar] [CrossRef]
  249. Tang, S.N.; Singh, C.; Nall, D.; Meeker, D.; Shankar, S.; Srivastava, R.K. The dietary bioflavonoid quercetin synergizes with epigallocathechin gallate (EGCG) to inhibit prostate cancer stem cell characteristics, invasion, migration and epithelial-mesenchymal transition. J. Mol. Signal. 2010, 5, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  250. Chen, S.F.; Nieh, S.; Jao, S.W.; Liu, C.L.; Wu, C.H.; Chang, Y.C.; Yang, C.Y.; Lin, Y.S. Quercetin suppresses drug-resistant spheres via the p38 MAPK-Hsp27 apoptotic pathway in oral cancer cells. PLoS ONE 2012, 7, e49275. [Google Scholar] [CrossRef] [Green Version]
  251. Shen, X.; Si, Y.; Wang, Z.; Wang, J.; Guo, Y.; Zhang, X. Quercetin inhibits the growth of human gastric cancer stem cells by inducing mitochondrial-dependent apoptosis through the inhibition of PI3K/Akt signaling. Int. J. Mol. Med. 2016, 38, 619–626. [Google Scholar] [CrossRef] [PubMed]
  252. Han, H.; Lim, J.W.; Kim, H. Astaxanthin Inhibits Helicobacter pylori-induced Inflammatory and Oncogenic Responses in Gastric Mucosal Tissues of Mice. J. Cancer Prev. 2020, 25, 244–251. [Google Scholar] [CrossRef] [PubMed]
  253. Cui, L.; Li, Z.; Xu, F.; Tian, Y.; Chen, T.; Li, J.; Guo, Y.; Lyu, Q. Antitumor Effects of Astaxanthin on Esophageal Squamous Cell Carcinoma by up-Regulation of PPARγ. Nutr. Cancer 2021. [Google Scholar] [CrossRef]
  254. Ahn, Y.T.; Kim, M.S.; Kim, Y.S.; An, W.G. Astaxanthin Reduces Stemness Markers in BT20 and T47D Breast Cancer Stem Cells by Inhibiting Expression of Pontin and Mutant p53. Mar. Drugs 2020, 18, 577. [Google Scholar] [CrossRef]
  255. Lee, N.Y.; Kim, Y.; Kim, Y.S.; Shin, J.H.; Rubin, L.P.; Kim, Y. β-Carotene exerts anti-colon cancer effects by regulating M2 macrophages and activated fibroblasts. J. Nutr. Biochem. 2020, 82, 108402. [Google Scholar] [CrossRef] [PubMed]
  256. Sowmya Shree, G.; Yogendra Prasad, K.; Arpitha, H.S.; Deepika, U.R.; Nawneet Kumar, K.; Mondal, P.; Ganesan, P. β-carotene at physiologically attainable concentration induces apoptosis and down-regulates cell survival and antioxidant markers in human breast cancer (MCF-7) cells. Mol. Cell. Biochem. 2017, 436, 1–12. [Google Scholar] [CrossRef]
  257. Ngoc, N.B.; Lv, P.; Zhao, W.E. Suppressive effects of lycopene and β-carotene on the viability of the human esophageal squamous carcinoma cell line EC109. Oncol. Lett. 2018, 15, 6727–6732. [Google Scholar] [CrossRef] [PubMed]
  258. Jain, A.; Sharma, G.; Kushwah, V.; Garg, N.K.; Kesharwani, P.; Ghoshal, G.; Singh, B.; Shivhare, U.S.; Jain, S.; Katare, O.P. Methotrexate and beta-carotene loaded-lipid polymer hybrid nanoparticles: A preclinical study for breast cancer. Nanomedicine 2017, 12, 1851–1872. [Google Scholar] [CrossRef] [PubMed]
  259. Kim, D.; Kim, Y.; Kim, Y. Effects of β-carotene on Expression of Selected MicroRNAs, Histone Acetylation, and DNA Methylation in Colon Cancer Stem Cells. J. Cancer Prev. 2019, 24, 224–232. [Google Scholar] [CrossRef] [Green Version]
  260. Lee, H.A.; Park, S.; Kim, Y. Effect of β-carotene on cancer cell stemness and differentiation in SK-N-BE(2)C neuroblastoma cells. Oncol. Rep. 2013, 30, 1869–1877. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  261. Kim, Y.S.; Gong, X.; Rubin, L.P.; Choi, S.W.; Kim, Y. β-Carotene 15,15′-oxygenase inhibits cancer cell stemness and metastasis by regulating differentiation-related miRNAs in human neuroblastoma. J. Nutr. Biochem. 2019, 69, 31–43. [Google Scholar] [CrossRef]
  262. Lim, J.Y.; Kim, Y.S.; Kim, K.M.; Min, S.J.; Kim, Y. Β-carotene inhibits neuroblastoma tumorigenesis by regulating cell differentiation and cancer cell stemness. Biochem. Biophys. Res. Commun. 2014, 450, 1475–1480. [Google Scholar] [CrossRef]
  263. Kim, Y.S.; Kim, E.; Park, Y.J.; Kim, Y. Retinoic acid receptor β enhanced the anti-cancer stem cells effect of β-carotene by down-regulating expression of delta-like 1 homologue in human neuroblastoma cells. Biochem. Biophys. Res. Commun. 2016, 480, 254–260. [Google Scholar] [CrossRef]
  264. Khan, S.; Awan, K.A.; Iqbal, M.J. Sulforaphane as a potential remedy against cancer: Comprehensive mechanistic review. J. Food Biochem. 2021, e13886. [Google Scholar] [CrossRef]
  265. Luo, Y.; Yan, B.; Liu, L.; Yin, L.; Ji, H.; An, X.; Gladkich, J.; Qi, Z.; De La Torre, C.; Herr, I. Sulforaphane Inhibits the Expression of Long Noncoding RNA H19 and Its Target APOBEC3G and Thereby Pancreatic Cancer Progression. Cancers 2021, 13, 827. [Google Scholar] [CrossRef]
  266. Li, J.; Zhou, Y.; Yan, Y.; Zheng, Z.; Hu, Y.; Wu, W. Sulforaphane-cysteine downregulates CDK4/CDK6 and inhibits tubulin polymerization contributing to cell cycle arrest and apoptosis in human glioblastoma cells. Aging 2020, 12, 16837–16851. [Google Scholar] [CrossRef] [PubMed]
  267. Wang, F.; Liu, P.; An, H.; Zhang, Y. Sulforaphane suppresses the viability and metastasis, and promotes the apoptosis of bladder cancer cells by inhibiting the expression of FAT-1. Int. J. Mol. Med. 2020, 46, 1085–1095. [Google Scholar] [CrossRef] [PubMed]
  268. Rong, Y.; Huang, L.; Yi, K.; Chen, H.; Liu, S.; Zhang, W.; Yuan, C.; Song, X.; Wang, F. Co-administration of sulforaphane and doxorubicin attenuates breast cancer growth by preventing the accumulation of myeloid-derived suppressor cells. Cancer Lett. 2020, 493, 189–196. [Google Scholar] [CrossRef] [PubMed]
  269. Ezeka, G.; Adhikary, G.; Kandasamy, S.; Friedberg, J.S.; Eckert, R.L. Sulforaphane inhibits PRMT5 and MEP50 function to suppress the mesothelioma cancer cell phenotype. Mol. Carcinog. 2021, 60, 429–439. [Google Scholar] [CrossRef]
  270. Ge, M.; Zhang, L.; Cao, L.; Xie, C.; Li, X.; Li, Y.; Meng, Y.; Chen, Y.; Wang, X.; Chen, J.; et al. Sulforaphane inhibits gastric cancer stem cells via suppressing sonic hedgehog pathway. Int. J. Food Sci. Nutr. 2019, 70, 570–578. [Google Scholar] [CrossRef] [PubMed]
  271. Dai, X.; Zhang, J.; Arfuso, F.; Chinnathambi, A.; Zayed, M.E.; Alharbi, S.A.; Kumar, A.P.; Ahn, K.S.; Sethi, G. Targeting TNF-related apoptosis-inducing ligand (TRAIL) receptor by natural products as a potential therapeutic approach for cancer therapy. Exp. Biol. Med. 2015, 240, 760–773. [Google Scholar] [CrossRef] [Green Version]
  272. Stöhr, D.; Schmid, J.O.; Beigl, T.B.; Mack, A.; Maichl, D.S.; Cao, K.; Budai, B.; Fullstone, G.; Kontermann, R.E.; Mürdter, T.E.; et al. Stress-induced TRAILR2 expression overcomes TRAIL resistance in cancer cell spheroids. Cell Death Differ. 2020, 27, 3037–3052. [Google Scholar] [CrossRef]
  273. Bauer, J.A.; Lupica, J.A.; Didonato, J.A.; Lindner, D.J. Nitric Oxide Inhibits NF-κB-mediated Survival Signaling: Possible Role in Overcoming TRAIL Resistance. Anticancer Res. 2020, 40, 6751–6763. [Google Scholar] [CrossRef]
  274. Watanabe, A.; Miyake, K.; Akahane, K.; Goi, K.; Kagami, K.; Yagita, H.; Inukai, T. Epigenetic Modification of Death Receptor Genes for TRAIL and TRAIL Resistance in Childhood B-Cell Precursor Acute Lymphoblastic Leukemia. Genes 2021, 12, 864. [Google Scholar] [CrossRef] [PubMed]
  275. She, T.; Shi, Q.; Li, Z.; Feng, Y.; Yang, H.; Tao, Z.; Li, H.; Chen, J.; Wang, S.; Liang, Y.; et al. Combination of long-acting TRAIL and tumor cell-targeted photodynamic therapy as a novel strategy to overcome chemotherapeutic multidrug resistance and TRAIL resistance of colorectal cancer. Theranostics 2021, 11, 4281–4297. [Google Scholar] [CrossRef]
  276. Stuckey, D.W.; Shah, K. TRAIL on trial: Preclinical advances in cancer therapy. Trends Mol. Med. 2013, 19, 685–694. [Google Scholar] [CrossRef] [Green Version]
  277. Puar, Y.R.; Shanmugam, M.K.; Fan, L.; Arfuso, F.; Sethi, G.; Tergaonkar, V. Evidence for the Involvement of the Master Transcription Factor NF-κB in Cancer Initiation and Progression. Biomedicines 2018, 6, 82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  278. Labsch, S.; Liu, L.; Bauer, N.; Zhang, Y.; Aleksandrowicz, E.; Gladkich, J.; Schönsiegel, F.; Herr, I. Sulforaphane and TRAIL induce a synergistic elimination of advanced prostate cancer stem-like cells. Int. J. Oncol. 2014, 44, 1470–1480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  279. Kallifatidis, G.; Labsch, S.; Rausch, V.; Mattern, J.; Gladkich, J.; Moldenhauer, G.; Büchler, M.W.; Salnikov, A.V.; Herr, I. Sulforaphane increases drug-mediated cytotoxicity toward cancer stem-like cells of pancreas and prostate. Mol. Ther. J. Am. Soc. Gene Ther. 2011, 19, 188–195. [Google Scholar] [CrossRef] [PubMed]
  280. Rausch, V.; Liu, L.; Kallifatidis, G.; Baumann, B.; Mattern, J.; Gladkich, J.; Wirth, T.; Schemmer, P.; Büchler, M.W.; Zöller, M.; et al. Synergistic activity of sorafenib and sulforaphane abolishes pancreatic cancer stem cell characteristics. Cancer Res. 2010, 70, 5004–5013. [Google Scholar] [CrossRef] [Green Version]
  281. Huang, J.; Tao, C.; Yu, Y.; Yu, F.; Zhang, H.; Gao, J.; Wang, D.; Chen, Y.; Gao, J.; Zhang, G.; et al. Simultaneous Targeting of Differentiated Breast Cancer Cells and Breast Cancer Stem Cells by Combination of Docetaxel- and Sulforaphane-Loaded Self-Assembled Poly(D, L-lactide-co-glycolide)/Hyaluronic Acid Block Copolymer-Based Nanoparticles. J. Biomed. Nanotechnol. 2016, 12, 1463–1477. [Google Scholar] [CrossRef]
  282. Zhu, J.; Wang, S.; Chen, Y.; Li, X.; Jiang, Y.; Yang, X.; Li, Y.; Wang, X.; Meng, Y.; Zhu, M.; et al. miR-19 targeting of GSK3β mediates sulforaphane suppression of lung cancer stem cells. J. Nutr. Biochem. 2017, 44, 80–91. [Google Scholar] [CrossRef] [PubMed]
  283. Li, Y.; Zhang, T.; Korkaya, H.; Liu, S.; Lee, H.F.; Newman, B.; Yu, Y.; Clouthier, S.G.; Schwartz, S.J.; Wicha, M.S.; et al. Sulforaphane, a dietary component of broccoli/broccoli sprouts, inhibits breast cancer stem cells. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2010, 16, 2580–2590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  284. Gu, H.F.; Ren, F.; Mao, X.Y.; Du, M. Mineralized and GSH-responsive hyaluronic acid based nano-carriers for potentiating repressive effects of sulforaphane on breast cancer stem cells-like properties. Carbohydr. Polym. 2021, 269, 118294. [Google Scholar] [CrossRef]
  285. Rodova, M.; Fu, J.; Watkins, D.N.; Srivastava, R.K.; Shankar, S. Sonic hedgehog signaling inhibition provides opportunities for targeted therapy by sulforaphane in regulating pancreatic cancer stem cell self-renewal. PLoS ONE 2012, 7, e46083. [Google Scholar] [CrossRef]
  286. Burnett, J.P.; Lim, G.; Li, Y.; Shah, R.B.; Lim, R.; Paholak, H.J.; McDermott, S.P.; Sun, L.; Tsume, Y.; Bai, S.; et al. Sulforaphane enhances the anticancer activity of taxanes against triple negative breast cancer by killing cancer stem cells. Cancer Lett. 2017, 394, 52–64. [Google Scholar] [CrossRef]
  287. Fisher, M.L.; Adhikary, G.; Grun, D.; Kaetzel, D.M.; Eckert, R.L. The Ezh2 polycomb group protein drives an aggressive phenotype in melanoma cancer stem cells and is a target of diet derived sulforaphane. Mol. Carcinog. 2016, 55, 2024–2036. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  288. Li, S.H.; Fu, J.; Watkins, D.N.; Srivastava, R.K.; Shankar, S. Sulforaphane regulates self-renewal of pancreatic cancer stem cells through the modulation of Sonic hedgehog-GLI pathway. Mol. Cell. Biochem. 2013, 373, 217–227. [Google Scholar] [CrossRef]
  289. Bijangi-Vishehsaraei, K.; Reza Saadatzadeh, M.; Wang, H.; Nguyen, A.; Kamocka, M.M.; Cai, W.; Cohen-Gadol, A.A.; Halum, S.L.; Sarkaria, J.N.; Pollok, K.E.; et al. Sulforaphane suppresses the growth of glioblastoma cells, glioblastoma stem cell-like spheroids, and tumor xenografts through multiple cell signaling pathways. J. Neurosurg. 2017, 127, 1219–1230. [Google Scholar] [CrossRef]
  290. Lin, L.C.; Yeh, C.T.; Kuo, C.C.; Lee, C.M.; Yen, G.C.; Wang, L.S.; Wu, C.H.; Yang, W.C.; Wu, A.T. Sulforaphane potentiates the efficacy of imatinib against chronic leukemia cancer stem cells through enhanced abrogation of Wnt/β-catenin function. J. Agric. Food Chem. 2012, 60, 7031–7039. [Google Scholar] [CrossRef] [PubMed]
  291. Wang, F.; Wang, W.; Li, J.; Zhang, J.; Wang, X.; Wang, M. Sulforaphane reverses gefitinib tolerance in human lung cancer cells via modulation of sonic hedgehog signaling. Oncol. Lett. 2018, 15, 109–114. [Google Scholar] [CrossRef] [Green Version]
  292. Liu, C.M.; Peng, C.Y.; Liao, Y.W.; Lu, M.Y.; Tsai, M.L.; Yeh, J.C.; Yu, C.H.; Yu, C.C. Sulforaphane targets cancer stemness and tumor initiating properties in oral squamous cell carcinomas via miR-200c induction. J. Formos. Med. Assoc. 2017, 116, 41–48. [Google Scholar] [CrossRef] [Green Version]
  293. Kerr, C.; Adhikary, G.; Grun, D.; George, N.; Eckert, R.L. Combination cisplatin and sulforaphane treatment reduces proliferation, invasion, and tumor formation in epidermal squamous cell carcinoma. Mol. Carcinog. 2018, 57, 3–11. [Google Scholar] [CrossRef]
  294. Trošelj, K.G.; Samaržija, I.; Tomljanović, M.; Kujundžić, R.N.; Đaković, N.; Mojzeš, A. Implementing Curcumin in Translational Oncology Research. Molecules 2020, 25, 5240. [Google Scholar] [CrossRef]
  295. Deng, S.; Shanmugam, M.K.; Kumar, A.P.; Yap, C.T.; Sethi, G.; Bishayee, A. Targeting autophagy using natural compounds for cancer prevention and therapy. Cancer 2019, 125, 1228–1246. [Google Scholar] [CrossRef]
  296. Goel, A.; Kunnumakkara, A.B.; Aggarwal, B.B. Curcumin as “Curecumin”: From kitchen to clinic. Biochem. Pharmacol. 2008, 75, 787–809. [Google Scholar] [CrossRef] [Green Version]
  297. Termini, D.; Den Hartogh, D.J.; Jaglanian, A.; Tsiani, E. Curcumin against Prostate Cancer: Current Evidence. Biomolecules 2020, 10, 1536. [Google Scholar] [CrossRef]
  298. Zhu, G.; Shen, Q.; Jiang, H.; Ji, O.; Zhu, L.; Zhang, L. Curcumin inhibited the growth and invasion of human monocytic leukaemia SHI-1 cells in vivo by altering MAPK and MMP signalling. Pharm. Biol. 2020, 58, 25–34. [Google Scholar] [CrossRef] [Green Version]
  299. Patra, S.; Pradhan, B.; Nayak, R.; Behera, C.; Rout, L.; Jena, M.; Efferth, T.; Bhutia, S.K. Chemotherapeutic efficacy of curcumin and resveratrol against cancer: Chemoprevention, chemoprotection, drug synergism and clinical pharmacokinetics. Semin. Cancer Biol. 2021, 73, 310–320. [Google Scholar] [CrossRef]
  300. Hu, Y.; Ran, M.; Wang, B.; Lin, Y.; Cheng, Y.; Zheng, S. Co-Delivery of Docetaxel and Curcumin via Nanomicelles for Enhancing Anti-Ovarian Cancer Treatment. Int. J. Nanomed. 2020, 15, 9703–9715. [Google Scholar] [CrossRef]
  301. Ashrafizadeh, M.; Zarrabi, A.; Hashemi, F.; Zabolian, A.; Saleki, H.; Bagherian, M.; Azami, N.; Bejandi, A.K.; Hushmandi, K.; Ang, H.L. Polychemotherapy with curcumin and doxorubicin via biological nanoplatforms: Enhancing antitumor activity. Pharmaceutics 2020, 12, 1084. [Google Scholar] [CrossRef]
  302. Aravind, S.R.; Lakshmi, S.; Ranjith, S.; Krishnan, L.K. Sustained release of curcumin from fibrin matrix induces cancer cell death and immunomodulation. Biomed. Pharmacother. 2021, 133, 110967. [Google Scholar] [CrossRef] [PubMed]
  303. Zhu, J.Y.; Yang, X.; Chen, Y.; Jiang, Y.; Wang, S.J.; Li, Y.; Wang, X.Q.; Meng, Y.; Zhu, M.M.; Ma, X.; et al. Curcumin Suppresses Lung Cancer Stem Cells via Inhibiting Wnt/β-catenin and Sonic Hedgehog Pathways. Phytother. Res. 2017, 31, 680–688. [Google Scholar] [CrossRef] [PubMed]
  304. Windmöller, B.A.; Beshay, M.; Helweg, L.P.; Flottmann, C.; Beermann, M.; Förster, C.; Wilkens, L.; Greiner, J.F.W.; Kaltschmidt, C.; Kaltschmidt, B. Novel Primary Human Cancer Stem-Like Cell Populations from Non-Small Cell Lung Cancer: Inhibition of Cell Survival by Targeting NF-κB and MYC Signaling. Cells 2021, 10, 1024. [Google Scholar] [CrossRef] [PubMed]
  305. Geng, H.; Guo, W.; Feng, L.; Xie, D.; Bi, L.; Wang, Y.; Zhang, T.; Liang, Z.; Yu, D. Diallyl trisulfide inhibited tobacco smoke-mediated bladder EMT and cancer stem cell marker expression via the NF-κB pathway in vivo. J. Int. Med. Res. 2021, 49, 0300060521992900. [Google Scholar] [CrossRef] [PubMed]
  306. Marquardt, J.U.; Gomez-Quiroz, L.; Arreguin Camacho, L.O.; Pinna, F.; Lee, Y.H.; Kitade, M.; Domínguez, M.P.; Castven, D.; Breuhahn, K.; Conner, E.A.; et al. Curcumin effectively inhibits oncogenic NF-κB signaling and restrains stemness features in liver cancer. J. Hepatol. 2015, 63, 661–669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  307. Garg, M.; Shanmugam, M.K.; Bhardwaj, V.; Goel, A.; Gupta, R.; Sharma, A.; Baligar, P.; Kumar, A.P.; Goh, B.C.; Wang, L.; et al. The pleiotropic role of transcription factor STAT3 in oncogenesis and its targeting through natural products for cancer prevention and therapy. Med. Res. Rev. 2020, 41, 1291–1336. [Google Scholar] [CrossRef]
  308. Lee, J.H.; Chiang, S.Y.; Nam, D.; Chung, W.S.; Lee, J.; Na, Y.S.; Sethi, G.; Ahn, K.S. Capillarisin inhibits constitutive and inducible STAT3 activation through induction of SHP-1 and SHP-2 tyrosine phosphatases. Cancer Lett. 2014, 345, 140–148. [Google Scholar] [CrossRef]
  309. Mohan, C.D.; Bharathkumar, H.; Bulusu, K.C.; Pandey, V.; Rangappa, S.; Fuchs, J.E.; Shanmugam, M.K.; Dai, X.; Li, F.; Deivasigamani, A.; et al. Development of a novel azaspirane that targets the Janus kinase-signal transducer and activator of transcription (STAT) pathway in hepatocellular carcinoma in vitro and in vivo. J. Biol. Chem. 2014, 289, 34296–34307. [Google Scholar] [CrossRef] [Green Version]
  310. Ashrafizadeh, M.; Gholami, M.H.; Mirzaei, S.; Zabolian, A.; Haddadi, A.; Farahani, M.V.; Kashani, S.H.; Hushmandi, K.; Najafi, M.; Zarrabi, A.; et al. Dual relationship between long non-coding RNAs and STAT3 signaling in different cancers: New insight to proliferation and metastasis. Life Sci. 2021, 270, 119006. [Google Scholar] [CrossRef]
  311. Mirzaei, S.; Gholami, M.H.; Mahabady, M.K.; Nabavi, N.; Zabolian, A.; Banihashemi, S.M.; Haddadi, A.; Entezari, M.; Hushmandi, K.; Makvandi, P.; et al. Pre-clinical investigation of STAT3 pathway in bladder cancer: Paving the way for clinical translation. Biomed. Pharmacother. 2021, 133, 111077. [Google Scholar] [CrossRef]
  312. Ashrafizadeh, M.; Zarrabi, A.; Orouei, S.; Zarrin, V.; Rahmani Moghadam, E.; Zabolian, A.; Mohammadi, S.; Hushmandi, K.; Gharehaghajlou, Y.; Makvandi, P.; et al. STAT3 pathway in gastric cancer: Signaling, therapeutic targeting and future prospects. Biology 2020, 9, 126. [Google Scholar] [CrossRef]
  313. Arora, L.; Kumar, A.P.; Arfuso, F.; Chng, W.J.; Sethi, G. The Role of Signal Transducer and Activator of Transcription 3 (STAT3) and Its Targeted Inhibition in Hematological Malignancies. Cancers 2018, 10, 327. [Google Scholar] [CrossRef] [Green Version]
  314. Kim, S.M.; Lee, J.H.; Sethi, G.; Kim, C.; Baek, S.H.; Nam, D.; Chung, W.S.; Kim, S.H.; Shim, B.S.; Ahn, K.S. Bergamottin, a natural furanocoumarin obtained from grapefruit juice induces chemosensitization and apoptosis through the inhibition of STAT3 signaling pathway in tumor cells. Cancer Lett. 2014, 354, 153–163. [Google Scholar] [CrossRef]
  315. Kim, C.; Lee, S.G.; Yang, W.M.; Arfuso, F.; Um, J.Y.; Kumar, A.P.; Bian, J.; Sethi, G.; Ahn, K.S. Formononetin-induced oxidative stress abrogates the activation of STAT3/5 signaling axis and suppresses the tumor growth in multiple myeloma preclinical model. Cancer Lett. 2018, 431, 123–141. [Google Scholar] [CrossRef]
  316. Lee, J.H.; Kim, C.; Kim, S.H.; Sethi, G.; Ahn, K.S. Farnesol inhibits tumor growth and enhances the anticancer effects of bortezomib in multiple myeloma xenograft mouse model through the modulation of STAT3 signaling pathway. Cancer Lett. 2015, 360, 280–293. [Google Scholar] [CrossRef] [PubMed]
  317. Chen, Y.; Shao, Z.; Jiang, E.; Zhou, X.; Wang, L.; Wang, H.; Luo, X.; Chen, Q.; Liu, K.; Shang, Z. CCL21/CCR7 interaction promotes EMT and enhances the stemness of OSCC via a JAK2/STAT3 signaling pathway. J. Cell. Physiol. 2020, 235, 5995–6009. [Google Scholar] [CrossRef] [PubMed]
  318. Pingali, P.; Wu, Y.J.; Boothello, R.; Sharon, C.; Li, H.; Sistla, S.; Sankaranarayanan, N.V.; Desai, U.R.; Le, A.T.; Doebele, R.C.; et al. High dose acetaminophen inhibits STAT3 and has free radical independent anti-cancer stem cell activity. Neoplasia 2021, 23, 348–359. [Google Scholar] [CrossRef] [PubMed]
  319. Khan, A.Q.; Ahmed, E.I.; Elareer, N.; Fathima, H.; Prabhu, K.S.; Siveen, K.S.; Kulinski, M.; Azizi, F.; Dermime, S.; Ahmad, A.; et al. Curcumin-Mediated Apoptotic Cell Death in Papillary Thyroid Cancer and Cancer Stem-Like Cells through Targeting of the JAK/STAT3 Signaling Pathway. Int. J. Mol. Sci. 2020, 21, 438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  320. Wang, D.; Kong, X.; Li, Y.; Qian, W.; Ma, J.; Wang, D.; Yu, D.; Zhong, C. Curcumin inhibits bladder cancer stem cells by suppressing Sonic Hedgehog pathway. Biochem. Biophys. Res. Commun. 2017, 493, 521–527. [Google Scholar] [CrossRef] [PubMed]
  321. Huang, Y.T.; Lin, Y.W.; Chiu, H.M.; Chiang, B.H. Curcumin Induces Apoptosis of Colorectal Cancer Stem Cells by Coupling with CD44 Marker. J. Agric. Food Chem. 2016, 64, 2247–2253. [Google Scholar] [CrossRef] [PubMed]
  322. Nayak, D.; Tripathi, N.; Kathuria, D.; Siddharth, S.; Nayak, A.; Bharatam, P.V.; Kundu, C. Quinacrine and curcumin synergistically increased the breast cancer stem cells death by inhibiting ABCG2 and modulating DNA damage repair pathway. Int. J. Biochem. Cell Biol. 2020, 119, 105682. [Google Scholar] [CrossRef]
  323. Mirza, S.; Vasaiya, A.; Vora, H.; Jain, N.; Rawal, R. Curcumin Targets Circulating Cancer Stem Cells by Inhibiting Self-Renewal Efficacy in Non-Small Cell Lung Carcinoma. Anti-Cancer Agents Med. Chem. 2017, 17, 859–864. [Google Scholar] [CrossRef] [PubMed]
  324. Wu, L.; Guo, L.; Liang, Y.; Liu, X.; Jiang, L.; Wang, L. Curcumin suppresses stem-like traits of lung cancer cells via inhibiting the JAK2/STAT3 signaling pathway. Oncol. Rep. 2015, 34, 3311–3317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  325. Hahn, Y.I.; Saeidi, S.; Kim, S.J.; Park, S.Y.; Song, N.Y.; Zheng, J.; Kim, D.H.; Lee, H.B.; Han, W.; Noh, D.Y.; et al. STAT3 Stabilizes IKKα Protein through Direct Interaction in Transformed and Cancerous Human Breast Epithelial Cells. Cancers 2020, 13, 82. [Google Scholar] [CrossRef] [PubMed]
  326. Chung, S.S.; Vadgama, J.V. Curcumin and epigallocatechin gallate inhibit the cancer stem cell phenotype via down-regulation of STAT3-NFκB signaling. Anticancer Res. 2015, 35, 39–46. [Google Scholar]
  327. Lin, L.; Liu, Y.; Li, H.; Li, P.K.; Fuchs, J.; Shibata, H.; Iwabuchi, Y.; Lin, J. Targeting colon cancer stem cells using a new curcumin analogue, GO-Y030. Br. J. Cancer 2011, 105, 212–220. [Google Scholar] [CrossRef] [Green Version]
  328. Zhou, Q.M.; Sun, Y.; Lu, Y.Y.; Zhang, H.; Chen, Q.L.; Su, S.B. Curcumin reduces mitomycin C resistance in breast cancer stem cells by regulating Bcl-2 family-mediated apoptosis. Cancer Cell Int. 2017, 17, 84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  329. Mansouri, K.; Rasoulpoor, S.; Daneshkhah, A.; Abolfathi, S.; Salari, N.; Mohammadi, M.; Rasoulpoor, S.; Shabani, S. Clinical effects of curcumin in enhancing cancer therapy: A systematic review. BMC Cancer 2020, 20, 791. [Google Scholar] [CrossRef] [PubMed]
  330. Yang, K.; Liao, Z.; Wu, Y.; Li, M.; Guo, T.; Lin, J.; Li, Y.; Hu, C. Curcumin and Glu-GNPs Induce Radiosensitivity against Breast Cancer Stem-Like Cells. BioMed Res. Int. 2020, 2020, 3189217. [Google Scholar] [CrossRef] [PubMed]
  331. Bano, N.; Yadav, M.; Das, B.C. Differential Inhibitory Effects of Curcumin Between HPV+ve and HPV-ve Oral Cancer Stem Cells. Front. Oncol. 2018, 8, 412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  332. Kuo, Y.C.; Wang, L.J.; Rajesh, R. Targeting human brain cancer stem cells by curcumin-loaded nanoparticles grafted with anti-aldehyde dehydrogenase and sialic acid: Colocalization of ALDH and CD44. Mater. Sci. Eng. C Mater. Biol. Appl. 2019, 102, 362–372. [Google Scholar] [CrossRef]
  333. Li, X.; Wang, X.; Xie, C.; Zhu, J.; Meng, Y.; Chen, Y.; Li, Y.; Jiang, Y.; Yang, X.; Wang, S.; et al. Sonic hedgehog and Wnt/β-catenin pathways mediate curcumin inhibition of breast cancer stem cells. Anti-Cancer Drugs 2018, 29, 208–215. [Google Scholar] [CrossRef] [PubMed]
  334. Buhrmann, C.; Kraehe, P.; Lueders, C.; Shayan, P.; Goel, A.; Shakibaei, M. Curcumin suppresses crosstalk between colon cancer stem cells and stromal fibroblasts in the tumor microenvironment: Potential role of EMT. PLoS ONE 2014, 9, e107514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  335. Siddappa, G.; Kulsum, S.; Ravindra, D.R.; Kumar, V.V.; Raju, N.; Raghavan, N.; Sudheendra, H.V.; Sharma, A.; Sunny, S.P.; Jacob, T.; et al. Curcumin and metformin-mediated chemoprevention of oral cancer is associated with inhibition of cancer stem cells. Mol. Carcinog. 2017, 56, 2446–2460. [Google Scholar] [CrossRef] [PubMed]
  336. Yu, Y.; Kanwar, S.S.; Patel, B.B.; Nautiyal, J.; Sarkar, F.H.; Majumdar, A.P. Elimination of Colon Cancer Stem-Like Cells by the Combination of Curcumin and FOLFOX. Transl. Oncol. 2009, 2, 321–328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  337. Zhang, H.; Zheng, J.; Shen, H.; Huang, Y.; Liu, T.; Xi, H.; Chen, C. Curcumin Suppresses In Vitro Proliferation and Invasion of Human Prostate Cancer Stem Cells by Modulating DLK1-DIO3 Imprinted Gene Cluster MicroRNAs. Genet. Test. Mol. Biomark. 2018, 22, 43–50. [Google Scholar] [CrossRef] [PubMed]
  338. Chung, S.S.; Dutta, P.; Chard, N.; Wu, Y.; Chen, Q.H.; Chen, G.; Vadgama, J. A novel curcumin analog inhibits canonical and non-canonical functions of telomerase through STAT3 and NF-κB inactivation in colorectal cancer cells. Oncotarget 2019, 10, 4516–4531. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  339. Yoshida, K.; Toden, S.; Ravindranathan, P.; Han, H.; Goel, A. Curcumin sensitizes pancreatic cancer cells to gemcitabine by attenuating PRC2 subunit EZH2, and the lncRNA PVT1 expression. Carcinogenesis 2017, 38, 1036–1046. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  340. Ashrafizadeh, M.; Najafi, M.; Orouei, S.; Zabolian, A.; Saleki, H.; Azami, N.; Sharifi, N.; Hushmandi, K.; Zarrabi, A.; Ahn, K.S. Resveratrol modulates transforming growth factor-beta (tgf-β) signaling pathway for disease therapy: A new insight into its pharmacological activities. Biomedicines 2020, 8, 261. [Google Scholar] [CrossRef] [PubMed]
  341. Ashrafizadeh, M.; Zarrabi, A.; Najafi, M.; Samarghandian, S.; Mohammadinejad, R.; Ahn, K.S. Resveratrol targeting tau proteins, amyloid-beta aggregations, and their adverse effects: An updated review. Phytother. Res. 2020, 34, 2867–2888. [Google Scholar] [CrossRef] [PubMed]
  342. Ashrafizadeh, M.; Taeb, S.; Haghi-Aminjan, H.; Afrashi, S.; Moloudi, K.; Musa, A.E.; Najafi, M.; Farhood, B. Resveratrol as an Enhancer of Apoptosis in Cancer: A Mechanistic Review. Anti-Cancer Agents Med. Chem. 2020. [Google Scholar] [CrossRef]
  343. Sun, X.; Xu, Q.; Zeng, L.; Xie, L.; Zhao, Q.; Xu, H.; Wang, X.; Jiang, N.; Fu, P.; Sang, M. Resveratrol suppresses the growth and metastatic potential of cervical cancer by inhibiting STAT3(Tyr705) phosphorylation. Cancer Med. 2020, 9, 8685–8700. [Google Scholar] [CrossRef] [PubMed]
  344. Sun, X.; Fu, P.; Xie, L.; Chai, S.; Xu, Q.; Zeng, L.; Wang, X.; Jiang, N.; Sang, M. Resveratrol inhibits the progression of cervical cancer by suppressing the transcription and expression of HPV E6 and E7 genes. Int. J. Mol. Med. 2021, 47, 335–345. [Google Scholar] [CrossRef] [PubMed]
  345. Ye, M.; Tian, H.; Lin, S.; Mo, J.; Li, Z.; Chen, X.; Liu, J. Resveratrol inhibits proliferation and promotes apoptosis via the androgen receptor splicing variant 7 and PI3K/AKT signaling pathway in LNCaP prostate cancer cells. Oncol. Lett. 2020, 20, 169. [Google Scholar] [CrossRef]
  346. Li, W.; Li, C.; Ma, L.; Jin, F. Resveratrol inhibits viability and induces apoptosis in the small-cell lung cancer H446 cell line via the PI3K/Akt/c-Myc pathway. Oncol. Rep. 2020, 44, 1821–1830. [Google Scholar] [CrossRef] [PubMed]
  347. Xiong, L.; Lin, X.M.; Nie, J.H.; Ye, H.S.; Liu, J. Resveratrol and its Nanoparticle suppress Doxorubicin/Docetaxel-resistant anaplastic Thyroid Cancer Cells in vitro and in vivo. Nanotheranostics 2021, 5, 143–154. [Google Scholar] [CrossRef] [PubMed]
  348. Sun, H.; Zhang, T.; Liu, R.; Cao, W.; Zhang, Z.; Liu, Z.; Qian, W.; Wang, D.; Yu, D.; Zhong, C. Resveratrol Inhibition of Renal Cancer Stem Cell Characteristics and Modulation of the Sonic Hedgehog Pathway. Nutr. Cancer 2021, 73, 1157–1167. [Google Scholar] [CrossRef] [PubMed]
  349. Ahmadi, Z.; Mohammadinejad, R.; Ashrafizadeh, M. Drug delivery systems for resveratrol, a non-flavonoid polyphenol: Emerging evidence in last decades. J. Drug Deliv. Sci. Technol. 2019, 51, 591–604. [Google Scholar] [CrossRef]
  350. Ashrafizadeh, M.; Javanmardi, S.; Moradi-Ozarlou, M.; Mohammadinejad, R.; Farkhondeh, T.; Samarghandian, S.; Garg, M. Natural products and phytochemical nanoformulations targeting mitochondria in oncotherapy: An updated review on resveratrol. Biosci. Rep. 2020, 40, BSR20200257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  351. Ashrafizadeh, M.; Rafiei, H.; Mohammadinejad, R.; Farkhondeh, T.; Samarghandian, S. Anti-tumor activity of resveratrol against gastric cancer: A review of recent advances with an emphasis on molecular pathways. Cancer Cell Int. 2021, 21, 66. [Google Scholar] [CrossRef] [PubMed]
  352. Shin, H.J.; Han, J.M.; Choi, Y.S.; Jung, H.J. Pterostilbene Suppresses both Cancer Cells and Cancer Stem-Like Cells in Cervical Cancer with Superior Bioavailability to Resveratrol. Molecules 2020, 25, 228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  353. 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] [Green Version]
  354. Ashrafizadeh, M.; Ahmadi, Z.; Farkhondeh, T.; Samarghandian, S. Modulatory effects of statins on the autophagy: A therapeutic perspective. J. Cell. Physiol. 2020, 235, 3157–3168. [Google Scholar] [CrossRef] [PubMed]
  355. Ashrafizadeh, M.; Zarrabi, A.; Orouei, S.; Hushmandi, K.; Hakimi, A.; Zabolian, A.; Daneshi, S.; Samarghandian, S.; Baradaran, B.; Najafi, M. MicroRNA-mediated autophagy regulation in cancer therapy: The role in chemoresistance/chemosensitivity. Eur. J. Pharmacol. 2020, 892, 173660. [Google Scholar] [CrossRef] [PubMed]
  356. Zheng, B.; Zhou, C.; Qu, G.; Ren, C.; Yan, P.; Guo, W.; Yue, B. VEGFR2 Promotes Metastasis and PD-L2 Expression of Human Osteosarcoma Cells by Activating the STAT3 and RhoA-ROCK-LIMK2 Pathways. Front. Oncol. 2020, 10, 543562. [Google Scholar] [CrossRef] [PubMed]
  357. Ge, X.; Liu, W.; Zhao, W.; Feng, S.; Duan, A.; Ji, C.; Shen, K.; Liu, W.; Zhou, J.; Jiang, D.; et al. Exosomal Transfer of LCP1 Promotes Osteosarcoma Cell Tumorigenesis and Metastasis by Activating the JAK2/STAT3 Signaling Pathway. Mol. Ther. Nucleic Acids 2020, 21, 900–915. [Google Scholar] [CrossRef] [PubMed]
  358. Peng, L.; Jiang, D. Resveratrol eliminates cancer stem cells of osteosarcoma by STAT3 pathway inhibition. PLoS ONE 2018, 13, e0205918. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  359. Qin, T.; Cheng, L.; Xiao, Y.; Qian, W.; Li, J.; Wu, Z.; Wang, Z.; Xu, Q.; Duan, W.; Wong, L.; et al. NAF-1 Inhibition by Resveratrol Suppresses Cancer Stem Cell-Like Properties and the Invasion of Pancreatic Cancer. Front. Oncol. 2020, 10, 1038. [Google Scholar] [CrossRef]
  360. Buhrmann, C.; Shayan, P.; Brockmueller, A.; Shakibaei, M. Resveratrol Suppresses Cross-Talk between Colorectal Cancer Cells and Stromal Cells in Multicellular Tumor Microenvironment: A Bridge between In Vitro and In Vivo Tumor Microenvironment Study. Molecules 2020, 25, 4292. [Google Scholar] [CrossRef]
  361. Suh, J.; Kim, D.H.; Surh, Y.J. Resveratrol suppresses migration, invasion and stemness of human breast cancer cells by interfering with tumor-stromal cross-talk. Arch. Biochem. Biophys. 2018, 643, 62–71. [Google Scholar] [CrossRef]
  362. Pouyafar, A.; Rezabakhsh, A.; Rahbarghazi, R.; Heydarabad, M.Z.; Shokrollahi, E.; Sokullu, E.; Khaksar, M.; Nourazarian, A.; Avci, Ç.B. Treatment of cancer stem cells from human colon adenocarcinoma cell line HT-29 with resveratrol and sulindac induced mesenchymal-endothelial transition rate. Cell Tissue Res. 2019, 376, 377–388. [Google Scholar] [CrossRef]
  363. Ruíz, G.; Valencia-González, H.A.; León-Galicia, I.; García-Villa, E.; García-Carrancá, A.; Gariglio, P. Inhibition of RAD51 by siRNA and Resveratrol Sensitizes Cancer Stem Cells Derived from HeLa Cell Cultures to Apoptosis. Stem Cells Int. 2018, 2018, 2493869. [Google Scholar] [CrossRef] [PubMed]
  364. Pradhan, R.; Chatterjee, S.; Hembram, K.C.; Sethy, C.; Mandal, M.; Kundu, C.N. Nano formulated Resveratrol inhibits metastasis and angiogenesis by reducing inflammatory cytokines in oral cancer cells by targeting tumor associated macrophages. J. Nutr. Biochem. 2021, 92, 108624. [Google Scholar] [CrossRef] [PubMed]
  365. Jhaveri, A.; Luther, E.; Torchilin, V. The effect of transferrin-targeted, resveratrol-loaded liposomes on neurosphere cultures of glioblastoma: Implications for targeting tumour-initiating cells. J. Drug Target. 2019, 27, 601–613. [Google Scholar] [CrossRef] [PubMed]
  366. Buhrmann, C.; Yazdi, M.; Popper, B.; Kunnumakkara, A.B.; Aggarwal, B.B.; Shakibaei, M. Induction of the Epithelial-to-Mesenchymal Transition of Human Colorectal Cancer by Human TNF-β (Lymphotoxin) and its Reversal by Resveratrol. Nutrients 2019, 11, 704. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  367. Su, Y.C.; Li, S.C.; Wu, Y.C.; Wang, L.M.; Chao, K.S.; Liao, H.F. Resveratrol downregulates interleukin-6-stimulated sonic hedgehog signaling in human acute myeloid leukemia. Evid.-Based Complement. Altern. Med. 2013, 2013, 547430. [Google Scholar] [CrossRef]
  368. Zhou, C.; Qian, W.; Ma, J.; Cheng, L.; Jiang, Z.; Yan, B.; Li, J.; Duan, W.; Sun, L.; Cao, J.; et al. Resveratrol enhances the chemotherapeutic response and reverses the stemness induced by gemcitabine in pancreatic cancer cells via targeting SREBP1. Cell Prolif. 2019, 52, e12514. [Google Scholar] [CrossRef]
  369. Shankar, S.; Nall, D.; Tang, S.N.; Meeker, D.; Passarini, J.; Sharma, J.; Srivastava, R.K. Resveratrol inhibits pancreatic cancer stem cell characteristics in human and KrasG12D transgenic mice by inhibiting pluripotency maintaining factors and epithelial-mesenchymal transition. PLoS ONE 2011, 6, e16530. [Google Scholar] [CrossRef] [Green Version]
  370. Lu, K.H.; Chen, Y.W.; Tsai, P.H.; Tsai, M.L.; Lee, Y.Y.; Chiang, C.Y.; Kao, C.L.; Chiou, S.H.; Ku, H.H.; Lin, C.H.; et al. Evaluation of radiotherapy effect in resveratrol-treated medulloblastoma cancer stem-like cells. Childs Nerv. Syst. 2009, 25, 543–550. [Google Scholar] [CrossRef]
  371. Seino, M.; Okada, M.; Shibuya, K.; Seino, S.; Suzuki, S.; Takeda, H.; Ohta, T.; Kurachi, H.; Kitanaka, C. Differential contribution of ROS to resveratrol-induced cell death and loss of self-renewal capacity of ovarian cancer stem cells. Anticancer Res. 2015, 35, 85–96. [Google Scholar]
  372. Dewangan, J.; Tandon, D.; Srivastava, S.; Verma, A.K.; Yapuri, A.; Rath, S.K. Novel combination of salinomycin and resveratrol synergistically enhances the anti-proliferative and pro-apoptotic effects on human breast cancer cells. Apoptosis Int. J. Program. Cell Death 2017, 22, 1246–1259. [Google Scholar] [CrossRef]
  373. Shen, Y.A.; Lin, C.H.; Chi, W.H.; Wang, C.Y.; Hsieh, Y.T.; Wei, Y.H.; Chen, Y.J. Resveratrol Impedes the Stemness, Epithelial-Mesenchymal Transition, and Metabolic Reprogramming of Cancer Stem Cells in Nasopharyngeal Carcinoma through p53 Activation. Evid.-Based Complement. Altern. Med. 2013, 2013, 590393. [Google Scholar] [CrossRef]
  374. Buhrmann, C.; Yazdi, M.; Popper, B.; Shayan, P.; Goel, A.; Aggarwal, B.B.; Shakibaei, M. Resveratrol Chemosensitizes TNF-β-Induced Survival of 5-FU-Treated Colorectal Cancer Cells. Nutrients 2018, 10, 888. [Google Scholar] [CrossRef] [Green Version]
  375. Song, D.; Hao, J.; Fan, D. Biological properties and clinical applications of berberine. Front. Med. 2020, 14, 564–582. [Google Scholar] [CrossRef]
  376. Zhou, M.; Deng, Y.; Liu, M.; Liao, L.; Dai, X.; Guo, C.; Zhao, X.; He, L.; Peng, C.; Li, Y. The pharmacological activity of berberine, a review for liver protection. Eur. J. Pharmacol. 2020, 890, 173655. [Google Scholar] [CrossRef]
  377. Chuang, T.C.; Wu, K.; Lin, Y.Y.; Kuo, H.P.; Kao, M.C.; Wang, V.; Hsu, S.C.; Lee, S.L. Dual down-regulation of EGFR and ErbB2 by berberine contributes to suppression of migration and invasion of human ovarian cancer cells. Environ. Toxicol. 2021, 36, 737–747. [Google Scholar] [CrossRef] [PubMed]
  378. Liu, J.; Luo, X.; Guo, R.; Jing, W.; Lu, H. Cell Metabolomics Reveals Berberine-Inhibited Pancreatic Cancer Cell Viability and Metastasis by Regulating Citrate Metabolism. J. Proteome Res. 2020, 19, 3825–3836. [Google Scholar] [CrossRef] [PubMed]
  379. Mohammadinejad, R.; Ahmadi, Z.; Tavakol, S.; Ashrafizadeh, M. Berberine as a potential autophagy modulator. J. Cell. Physiol. 2019, 234, 14914–14926. [Google Scholar] [CrossRef] [PubMed]
  380. Ashrafizadeh, M.; Najafi, M.; Mohammadinejad, R.; Farkhondeh, T.; Samarghandian, S. Berberine Administration in Treatment of Colitis: A Review. Curr. Drug Targets 2020, 21, 1385–1393. [Google Scholar] [CrossRef] [PubMed]
  381. Gu, S.; Song, X.; Xie, R.; Ouyang, C.; Xie, L.; Li, Q.; Su, T.; Xu, M.; Xu, T.; Huang, D.; et al. Berberine inhibits cancer cells growth by suppressing fatty acid synthesis and biogenesis of extracellular vesicles. Life Sci. 2020, 257, 118122. [Google Scholar] [CrossRef]
  382. Zhang, Y.; Liu, X.; Yu, M.; Xu, M.; Xiao, Y.; Ma, W.; Huang, L.; Li, X.; Ye, X. Berberine inhibits proliferation and induces G0/G1 phase arrest in colorectal cancer cells by downregulating IGF2BP3. Life Sci. 2020, 260, 118413. [Google Scholar] [CrossRef] [PubMed]
  383. Jung, Y.Y.; Ko, J.H.; Um, J.Y.; Chinnathambi, A.; Alharbi, S.A.; Sethi, G.; Ahn, K.S. LDL cholesterol promotes the proliferation of prostate and pancreatic cancer cells by activating the STAT3 pathway. J. Cell. Physiol. 2021, 236, 5253–5264. [Google Scholar] [CrossRef] [PubMed]
  384. Pandya, G.; Kirtonia, A.; Sethi, G.; Pandey, A.K.; Garg, M. The implication of long non-coding RNAs in the diagnosis, pathogenesis and drug resistance of pancreatic ductal adenocarcinoma and their possible therapeutic potential. Biochim. Biophys. Acta Rev. Cancer 2020, 1874, 188423. [Google Scholar] [CrossRef]
  385. Suzuki, S.; Okada, M.; Sanomachi, T.; Togashi, K.; Seino, S.; Sato, A.; Yamamoto, M.; Kitanaka, C. Therapeutic targeting of pancreatic cancer stem cells by dexamethasone modulation of the MKP-1-JNK axis. J. Biol. Chem. 2020, 295, 18328–18342. [Google Scholar] [CrossRef] [PubMed]
  386. Jagust, P.; Alcalá, S.; Sainz, B., Jr.; Heeschen, C.; Sancho, P. Glutathione metabolism is essential for self-renewal and chemoresistance of pancreatic cancer stem cells. World J. Stem Cells 2020, 12, 1410–1428. [Google Scholar] [CrossRef] [PubMed]
  387. Valle, S.; Alcalá, S.; Martin-Hijano, L.; Cabezas-Sáinz, P.; Navarro, D.; Muñoz, E.R.; Yuste, L.; Tiwary, K.; Walter, K.; Ruiz-Cañas, L.; et al. Exploiting oxidative phosphorylation to promote the stem and immunoevasive properties of pancreatic cancer stem cells. Nat. Commun. 2020, 11, 5265. [Google Scholar] [CrossRef] [PubMed]
  388. Alcalá, S.; Mayoral-Varo, V.; Ruiz-Cañas, L.; López-Gil, J.C.; Heeschen, C.; Martín-Pérez, J.; Sainz, B., Jr. Targeting SRC Kinase Signaling in Pancreatic Cancer Stem Cells. Int. J. Mol. Sci. 2020, 21, 7437. [Google Scholar] [CrossRef] [PubMed]
  389. Park, S.H.; Sung, J.H.; Chung, N. Berberine diminishes side population and down-regulates stem cell-associated genes in the pancreatic cancer cell lines PANC-1 and MIA PaCa-2. Mol. Cell. Biochem. 2014, 394, 209–215. [Google Scholar] [CrossRef]
  390. Zhu, C.; Li, J.; Hua, Y.; Wang, J.; Wang, K.; Sun, J. Berberine Inhibits the Expression of SCT through miR-214-3p Stimulation in Breast Cancer Cells. Evid.-Based Complement. Altern. Med. 2020, 2020, 2817147. [Google Scholar] [CrossRef] [PubMed]
  391. Li, J.; Zhang, S.; Wu, L.; Pei, M.; Jiang, Y. Berberine inhibited metastasis through miR-145/MMP16 axis in vitro. J. Ovarian Res. 2021, 14, 4. [Google Scholar] [CrossRef] [PubMed]
  392. Zhan, Y.; Han, J.; Xia, J.; Wang, X. Berberine Suppresses Mice Depression Behaviors and Promotes Hippocampal Neurons Growth Through Regulating the miR-34b-5p/miR-470-5p/BDNF Axis. Neuropsychiatr. Dis. Treat. 2021, 17, 613–626. [Google Scholar] [CrossRef] [PubMed]
  393. Lin, C.Y.; Hsieh, P.L.; Liao, Y.W.; Peng, C.Y.; Lu, M.Y.; Yang, C.H.; Yu, C.C.; Liu, C.M. Berberine-targeted miR-21 chemosensitizes oral carcinomas stem cells. Oncotarget 2017, 8, 80900–80908. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  394. Xu, M.; Wang, J.; Li, H.; Zhang, Z.; Cheng, Z. AIM2 inhibits colorectal cancer cell proliferation and migration through suppression of Gli1. Aging 2020, 13, 1017–1031. [Google Scholar] [CrossRef] [PubMed]
  395. Doheny, D.; Sirkisoon, S.; Carpenter, R.L.; Aguayo, N.R.; Regua, A.T.; Anguelov, M.; Manore, S.G.; Arrigo, A.; Jalboush, S.A.; Wong, G.L.; et al. Combined inhibition of JAK2-STAT3 and SMO-GLI1/tGLI1 pathways suppresses breast cancer stem cells, tumor growth, and metastasis. Oncogene 2020, 39, 6589–6605. [Google Scholar] [CrossRef] [PubMed]
  396. Zhao, Y.; Yang, X.; Zhao, J.; Gao, M.; Zhang, M.; Shi, T.; Zhang, F.; Zheng, X.; Pan, Y.; Shao, D.; et al. Berberine inhibits chemotherapy-exacerbated ovarian cancer stem cell-like characteristics and metastasis through GLI1. Eur. J. Pharmacol. 2021, 895, 173887. [Google Scholar] [CrossRef] [PubMed]
  397. Naveen, C.R.; Gaikwad, S.; Agrawal-Rajput, R. Berberine induces neuronal differentiation through inhibition of cancer stemness and epithelial-mesenchymal transition in neuroblastoma cells. Phytomed. Int. J. Phytother. Phytopharm. 2016, 23, 736–744. [Google Scholar] [CrossRef]
  398. Liufu, C.; Li, Y.; Lin, Y.; Yu, J.; Du, M.; Chen, Y.; Yang, Y.; Gong, X.; Chen, Z. Synergistic ultrasonic biophysical effect-responsive nanoparticles for enhanced gene delivery to ovarian cancer stem cells. Drug Deliv. 2020, 27, 1018–1033. [Google Scholar] [CrossRef]
  399. Ertas, Y.N.; Abedi Dorcheh, K.; Akbari, A.; Jabbari, E. Nanoparticles for Targeted Drug Delivery to Cancer Stem Cells: A Review of Recent Advances. Nanomaterials 2021, 11, 1755. [Google Scholar] [CrossRef] [PubMed]
  400. Ma, X.; Zhou, J.; Zhang, C.X.; Li, X.Y.; Li, N.; Ju, R.J.; Shi, J.F.; Sun, M.G.; Zhao, W.Y.; Mu, L.M.; et al. Modulation of drug-resistant membrane and apoptosis proteins of breast cancer stem cells by targeting berberine liposomes. Biomaterials 2013, 34, 4452–4465. [Google Scholar] [CrossRef] [PubMed]
  401. Ashrafizadeh, M.; Ahmadi, Z.; Mohammadinejad, R.; Farkhondeh, T.; Samarghandian, S. MicroRNAs mediate the anti-tumor and protective effects of ginsenosides. Nutr. Cancer 2020, 72, 1264–1275. [Google Scholar] [CrossRef]
  402. Ashrafizadeh, M.; Tavakol, S.; Mohammadinejad, R.; Ahmadi, Z.; Yaribeygi, H.; Jamialahmadi, T.; Johnston, T.P.; Sahebkar, A. Paving the Road toward Exploiting the Therapeutic Effects of Ginsenosides: An Emphasis on Autophagy and Endoplasmic Reticulum Stress. Adv. Exp. Med. Biol. 2021, 1308, 137–160. [Google Scholar] [PubMed]
  403. Hashemi, F.; Zarrabi, A.; Zabolian, A.; Saleki, H.; Farahani, M.V.; Sharifzadeh, S.O.; Ghahremaniyeh, Z.; Bejandi, A.K.; Hushmandi, K.; Ashrafizadeh, M. Novel strategy in breast cancer therapy: Revealing the bright side of ginsenosides. Curr. Mol. Pharmacol. 2021. [Google Scholar] [CrossRef] [PubMed]
  404. Tang, Y.C.; Zhang, Y.; Zhou, J.; Zhi, Q.; Wu, M.Y.; Gong, F.R.; Shen, M.; Liu, L.; Tao, M.; Shen, B.; et al. Ginsenoside Rg3 targets cancer stem cells and tumor angiogenesis to inhibit colorectal cancer progression in vivo. Int. J. Oncol. 2018, 52, 127–138. [Google Scholar] [CrossRef] [PubMed]
  405. Liu, S.; Chen, M.; Li, P.; Wu, Y.; Chang, C.; Qiu, Y.; Cao, L.; Liu, Z.; Jia, C. Ginsenoside rh2 inhibits cancer stem-like cells in skin squamous cell carcinoma. Cell. Physiol. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 2015, 36, 499–508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  406. Mai, T.T.; Moon, J.; Song, Y.; Viet, P.Q.; Phuc, P.V.; Lee, J.M.; Yi, T.H.; Cho, M.; Cho, S.K. Ginsenoside F2 induces apoptosis accompanied by protective autophagy in breast cancer stem cells. Cancer Lett. 2012, 321, 144–153. [Google Scholar] [CrossRef]
  407. Deng, S.; Wong, C.K.C.; Lai, H.C.; Wong, A.S.T. Ginsenoside-Rb1 targets chemotherapy-resistant ovarian cancer stem cells via simultaneous inhibition of Wnt/β-catenin signaling and epithelial-to-mesenchymal transition. Oncotarget 2017, 8, 25897–25914. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  408. Oh, J.; Yoon, H.J.; Jang, J.H.; Kim, D.H.; Surh, Y.J. The standardized Korean Red Ginseng extract and its ingredient ginsenoside Rg3 inhibit manifestation of breast cancer stem cell-like properties through modulation of self-renewal signaling. J. Ginseng Res. 2019, 43, 421–430. [Google Scholar] [CrossRef]
  409. Yang, Z.; Zhao, T.; Liu, H.; Zhang, L. Ginsenoside Rh2 inhibits hepatocellular carcinoma through β-catenin and autophagy. Sci. Rep. 2016, 6, 19383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  410. Lu, S.L.; Wang, Y.H.; Liu, G.F.; Wang, L.; Li, Y.; Guo, Z.Y.; Cheng, C. Graphene Oxide Nanoparticle-Loaded Ginsenoside Rg3 Improves Photodynamic Therapy in Inhibiting Malignant Progression and Stemness of Osteosarcoma. Front. Mol. Biosci. 2021, 8, 663089. [Google Scholar] [CrossRef]
  411. Phi, L.T.H.; Wijaya, Y.T.; Sari, I.N.; Kim, K.S.; Yang, Y.G.; Lee, M.W.; Kwon, H.Y. 20(R)-Ginsenoside Rg3 Influences Cancer Stem Cell Properties and the Epithelial-Mesenchymal Transition in Colorectal Cancer via the SNAIL Signaling Axis. OncoTargets Ther. 2019, 12, 10885–10895. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  412. Park, J.W.; Park, J.H.; Han, J.W. Fermented Ginseng Extract, BST204, Suppresses Tumorigenesis and Migration of Embryonic Carcinoma through Inhibition of Cancer Stem Cell Properties. Molecules 2020, 25, 3128. [Google Scholar] [CrossRef] [PubMed]
  413. Oh, J.; Jeon, S.B.; Lee, Y.; Lee, H.; Kim, J.; Kwon, B.R.; Yu, K.Y.; Cha, J.D.; Hwang, S.M.; Choi, K.M.; et al. Fermented red ginseng extract inhibits cancer cell proliferation and viability. J. Med. Food 2015, 18, 421–428. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The potential of flavones in suppressing CSCs. Abbreviations: CSCs, cancer stem cells; NF-κB, nuclear factor-kappaB; CDK, cyclin-dependent kinase; Oct4, octamer-4; YAP, Yes-associated protein, SOX2, sex determining region Y-box 2; TAZ, Transcriptional coactivator with PDZ-binding motif; CHM-04, a chrysin derivative; STAT3, signal transducer and activator of transcription 3; Akt, protein kinase-B; PI3K, phosphoinositide 3-kinase; ROS, reactive oxygen species.
Figure 1. The potential of flavones in suppressing CSCs. Abbreviations: CSCs, cancer stem cells; NF-κB, nuclear factor-kappaB; CDK, cyclin-dependent kinase; Oct4, octamer-4; YAP, Yes-associated protein, SOX2, sex determining region Y-box 2; TAZ, Transcriptional coactivator with PDZ-binding motif; CHM-04, a chrysin derivative; STAT3, signal transducer and activator of transcription 3; Akt, protein kinase-B; PI3K, phosphoinositide 3-kinase; ROS, reactive oxygen species.
Ijms 22 11669 g001
Figure 2. The potential of isoflavonoids in suppressing CSCs.
Figure 2. The potential of isoflavonoids in suppressing CSCs.
Ijms 22 11669 g002
Figure 3. The potential of bioactive constituents of pomegranate in suppressing CSCs.
Figure 3. The potential of bioactive constituents of pomegranate in suppressing CSCs.
Ijms 22 11669 g003
Figure 4. The potential of carotenoids in suppressing CSCs.
Figure 4. The potential of carotenoids in suppressing CSCs.
Ijms 22 11669 g004
Figure 5. Curcumin in inhibiting CSCs.
Figure 5. Curcumin in inhibiting CSCs.
Ijms 22 11669 g005
Figure 6. Res in suppressing CSCs in tumor therapy.
Figure 6. Res in suppressing CSCs in tumor therapy.
Ijms 22 11669 g006
Figure 7. The berberine and ginsenosides as modulators of CSCs in tumor eradication.
Figure 7. The berberine and ginsenosides as modulators of CSCs in tumor eradication.
Ijms 22 11669 g007
Figure 8. The chemical structures of selected phytochemicals discussed in the review.
Figure 8. The chemical structures of selected phytochemicals discussed in the review.
Ijms 22 11669 g008
Figure 9. Chemical structures of ginsenosides.
Figure 9. Chemical structures of ginsenosides.
Ijms 22 11669 g009
Table 1. Flavones in targeting CSCs for tumor suppression.
Table 1. Flavones in targeting CSCs for tumor suppression.
Anti-Tumor AgentCancer TypeIn Vitro/In VivoCell Line/Animal ModelStudy DesignRemarksRefs
ChrysinColorectal cancerIn vitroHT-29 cells-The chrysin- and docetaxel-loaded micelles exert synergistic therapy in suppressing growth and invasion of CSCs
Enhancing ROS production to mediate cell death
[129]
CHM-04 (chrysin derivative)Breast cancerIn vitroMCF-7 and MDA-MB-231 cells10 μMHigher anti-tumor potential (3.2-fold increase) compared to chrysin
Inducing apoptosis and reducing migration
Suppressing colony formation of CSCs
[130]
ApigeninBreast cancerIn vitro
In vivo
MDA-MB-231 cells
Nude mice
0–64 μMSuppressing CSC features in breast cancer via inhibiting YAP/TAZ axis[134]
ApigeninLung cancerIn vitroA549 and H1299 cells10, 20 and 30 μMEnhancing p53 expression and impairing CSC features
Promoting sensitivity of cancer cells to cisplatin chemotherapy
[151]
ApigeninProstate cancerIn vitroPC3 cells25 μMReducing viability of CSCs and triggering apoptosis via p21 and p27 upregulation
Triggering extrinsic pathway of apoptosis
Suppressing PI3K/Akt/NF-κB axis
[136]
ApigeninProstate cancerIn vitroPC3 cells15 μMInducing apoptosis and cell cycle arrest in CSCs
Suppressing migration and invasion of CSCs
Potentiating anti-tumor activity of cisplatin
Inhibiting PI3K/Akt and NF-κB pathways
[137]
ApigeninHead and neck cancerIn vitroHN-8, HN-30, and HSC-3 cells0–100 μMDecreasing viability of cancer cells in a dose-dependent manner
Suppressing CSC features via reducing expression levels of CD44, Nanog and CD105
[138]
ApigeninProstate cancerIn vitroPC3, LNCaP, or isolated CD44+ CD133+ and CD44+ stem cells0–2 μMInducing apoptosis in CSCs
Suppressing proliferation and viability of CSCs
Upregulating expression levels of p21, p27, Bax, Bid, caspase-3 and caspase-8
Inhibiting ERK, PARP and NF-κB expression levels
[139]
BaicaleinPancreatic cancerIn vitroPANC-1, BxPC-3, and SW1990 cells0–300 μMReducing SOX-2 expression and impairing CSC features via down-regulating Sonic expression[144]
WogoninOsteosarcomaIn vitroCD133+ Cal72 cells0–80 μMTriggering cell death in cancer cells via enhancing ROS levels[149]
WogoninOsteosarcomaIn vitroCD133+ CAL72 cells0–80 μMTriggering apoptosis in CSCs
Inhibiting self-renewal capacity
Reducing migration and invasion of CSCs via reducing MMP-9 expression
[150]
Table 2. Gallate in suppressing CSCs.
Table 2. Gallate in suppressing CSCs.
Cancer TypeIn Vitro/In VivoCell Line/Animal ModelStudy DesignRemarksRefs
Lung cancerIn vitro
In vivo
A549 and H1299 cell lines
Xenograft model
0–40 μM
20 mg/kg
Suppressing self-renewal capacity of tumor cells
CLOCK down-regulation
Suppressing tumor growth in vivo
[163]
Lung cancerIn vitroA549, H460, H1299, and HEK-293T cells0–40 μMSuppressing CSC features in lung cancer
Promoting miRNA-485-5p expression
Down-regulating PXRα expression
[168]
Lung cancerIn vitroA549 and H1299 cells0–100 μMThe enrichment of CSC features in lung cancer
Wnt/β-catenin inhibition prevents CSC characteristics
[169]
Colorectal cancerIn vitroDLD-1 and SW480 cells0–60 μMProliferation inhibition
Apoptosis induction
Impairing CSC features via inhibiting Wnt/β-catenin axis
[170]
Colorectal cancerIn vitroHCT116 cells0–200 μMInhibiting self-renewal capacity of CSCs
Reducing expression levels of Notch1, Suz12 and EZH2
Increasing sensitivity of tumor cells to 5-flourouracil chemotherapy
[173]
Bladder cancerIn vitroEJ and UM-UC-3 cells0–90 μMInhibiting Sonic Hedgehog signaling
Reducing CSC features
Apoptosis induction in CSCs
Impairing proliferation of CSCs
[174]
Head and neck cancerIn vitro
In vivo
K3, K4 and K5 cells
Xenograft model
0–10 μMImpairing CSC features via suppressing Notch signaling[176]
Nasopharyngeal cancerIn vitro
In vivo
CNE2 and C666-1 cells
Xenograft model
0–50 μMInhibiting self-renewal capacity of CSCs
Suppressing migration via EMT inhibition
Down-regulating NF-κB expression
[177]
Table 3. The potential of genistein in cancer suppression via targeting CSCs.
Table 3. The potential of genistein in cancer suppression via targeting CSCs.
Cancer TypeIn Vitro/In VivoCell Line/Animal ModelStudy DesignRemarksRefs
Nasopharyngeal cancerIn vitroCNE2 and HONE1 cells0–100 μMReducing expression level of CSC markers including CD44 and CD133 via suppressing Sonic signaling[193]
Renal cancerIn vitro786-O and ACHN cell lines0–90 μMSuppressing growth and colony formation capacities of CSCs
Triggering apoptosis and reducing CSC marker expression
Inhibiting Sonic Hedgehog signaling
[205]
Breast cancerIn vitroMCF-7 and MDA-MB-231 cells2 μM or 40 nMDecreasing number of CSCs
Inducing PI3K/Akt and MEK/ERK pathways in a paracrine manner and mediating differentiation of CSCs
[195]
Gastric cancerIn vitroMGC-803 and SGC-7901 cells15 μMSuppressing colony formation and self-renewal capacities
Inhibiting chemoresistance via down-regulating ERK1/2 and ABCG2
[196]
Gastric cancerIn vitroSGC-7901 cells0–10 μmol/LDecreasing FoxM1 expression to prevent self-renewal capacity and migration of CSCs
Decreasing expression levels of CSC markers including CD44, CD133 and ALDH1
[197]
Gastric cancerIn vitroAGS and MKN45 cells10 μg/mlDecreasing expression level of CD44 via suppressing Sonic Hedgehog signaling[198]
Prostate cancerIn vitro
In vivo
22RV1, DU145 cells
Xenograft animal model
15 and 30 μM
10 mg/kg
Suppressing Hedgehog/Gli1 axis to impair stemness and CSC features in prostate cancer[199]
Table 4. The CSCs as promising targets of quercetin in tumor therapy.
Table 4. The CSCs as promising targets of quercetin in tumor therapy.
Cancer TypeIn Vitro/In VivoCell Line/Animal ModelStudy DesignRemarksRefs
Breast cancerIn vitro
In vivo
MCF-7 cells
Nude mice
0–200 μMReducing number of CSCs
Impairing CSC features
Suppressing PI3K/Akt/mTOR axis
[233]
Breast cancerIn vitroMDA-MB-231 cells0–200 μMDisrupting tumor progression and CSC features via down-regulating ALDH1A1, CXCR4, MUC1 and EpCAM[234]
Breast cancerIn vitroMCF-7 cells0–70 μMSuppressing CSCs in breast cancer via preventing nuclear translocation of YB-1[235]
Breast cancerIn vitroMCF-10A, MCF-7, MDA-MB-231 and AC16 cells0–2 μMEnhancing intracellular accumulation of doxorubicin via down-regulating P-gp, MRP1 and BCRP
Eliminating CSCs and potentiating doxorubicin’s anti-tumor activity
[236]
Pancreatic cancerIn vitroAsPC1 and PANC1 cells50 μMSuppressing self-renewal capacity of tumor cells via enhancing miRNA-200b expression[238]
Pancreatic cancerIn vitro
In vivo
BxPc-3 and MIA-PaCa2 cells
Nude mice and xenografts
100, 200 and 400 μM
50 mg/kg
Synergistic impact between quercetin and sulforaphane
Down-regulating NF-κB signaling
Impairing tumor growth in vitro and in vivo
[240]
Colon cancerIn vitroDLD-1 and HT-29 cells0–50 μMSuppressing Notch signaling, impairing CSCs features and increasing sensitivity of tumor cells to radiotherapy[241]
Colorectal cancerIn vitroHT29 cells0–100 μMTriggering apoptosis and cell cycle arrest in CSCs
Promoting sensitivity of tumor cells to doxorubicin chemotherapy
[242]
Prostate cancerIn vitroPC3, LNCaP and ARPE-19 cells40 μMQueretin and midkine-siRNA co-application suppresses CSC features via dual inhibition of PI3K/Akt and MAPK/ERK molecular pathways[248]
Table 5. The sulforaphane as a potential anti-tumor agent in cancer therapy.
Table 5. The sulforaphane as a potential anti-tumor agent in cancer therapy.
Cancer TypeIn vitro/In VivoCell Line/Animal ModelStudy DesignRemarksRefs
Triple-negative breast cancerIn vitroSUM149 and SUM159 cells2.5 and 5 μMInducing apoptosis in CSCs and reducing ALDH expression as CSC marker[286]
MelanomaIn vitro
In vivo
A375 cells
Xenograft
20 μM
10 μM/kg
The EZH2 promotes CSC features in melanoma and is suppressed by sulforaphane[287]
Pancreatic cancerIn vivoMice20 mg/kgSuppressing Sonic/Gli1 axis to impair CSC features and self-renewal capacity in pancreatic cancer[288]
GlioblastomaIn vitro
In vivo
U87, U373, U118, and SF767 cells
Mice
0–50 μM
100 mg/kg
Suppressing stem cell-like spheroids
Inducing apoptosis and eliminating CSCs
Promoting drug sensitivity
[289]
LeukemiaIn vitroKU812 cells0–30 μMEnhancing ROS levels
Suppressing β-catenin signaling to reduce GSH levels
Promoting potential of imatinib in tumor suppression
[290]
Lung cancerIn vitroNSCLC PC9 cells0–12 μMSuppressing Sonic Hedgehog signaling
Decreasing levels of CD133 and CD44
Inhibiting gefitinib resistance
[291]
Lung cancerIn vitro
In vivo
A549 cells
Nude mice
0–40 μM
25 and 50 mg/kg
Preventing tobacco-mediated CSC feature acquisition via suppressing IL-6/ΔNp63α/Notch axis[194]
Oral cancerIn vitroSAS or GNM cells0–50 μMUpregulating miRNA-200c expression to impair CSC features and stemness in oral cancer[292]
Epidermal squamous cell carcinomaIn vitroSCC-13 cells0–20 μMThe SCC-3 cells derived from CSCs have high sensitivity to a combination of sulforaphane and cisplatin
This combination reduces viability of tumor cells and their capacity in colony formation
[293]
Table 6. Curcumin potential of suppressing CSCs in tumor treatment.
Table 6. Curcumin potential of suppressing CSCs in tumor treatment.
Cancer TypeIn Vitro/In VivoCell Line/Animal ModelStudy DesignRemarksRefs
Lung cancerIn vitroA549 and H1299 cells0–40 μMInducing apoptosis
Reducing levels of CSC markers
Inhibiting Wnt/β-catenin and Sonic signaling pathways
[303]
Bladder cancerIn vitroUM-UC-3 and EJ cells0–50 μMApoptosis induction
Decreasing expression levels of CD44, CD133, Oct4, Nanog and ALDH1
Inhibiting Sonic Hedgehog signaling
[320]
Breast cancerIn vitroMCF-10A and MCF-7 cells8 μMA combination of curcumin and quinacrine induces DNA damage and reduces ABCG2 expression to impair cancer progression[322]
Breast cancerIn vitroSUM159 and MCF7 cell lines0–40 μMReducing colony formation capacity of CSCs
Down-regulating expression levels of CD44, ALDH1, Nanog and Oct4
Inhibiting Wnt/β-catenin axis
[333]
Breast cancerIn vitroMDA-MB-231 and MCF-7 cells5 μmol/LInducing apoptosis via Bcl-2 down-regulation
Enhancing sensitivity of breast CSCs to mitomycin C
[328]
Brain cancerIn vitroU87MG cells-The curcumin-loaded nanoparticles efficiently penetrate into BBB to induce apoptosis in CSCs and reduce tumor progression[332]
Pancreatic cancerIn vitro
In vivo
BxPC3, MiaPaCa2 and Panc1 PDAC cells
Nude mice
0–20 μM
100 mg/kg
Suppressing self-renewal capacity of tumor cells
Retarding tumor growth in vivo
Enhancing sensitivity to gemcitabine chemotherapy
[339]
Prostate cancerIn vitroDU145 cells-Inhibiting growth and invasion of prostate CSCs
Overexpression of miRNA-770-5p and miRNA-1247
[337]
Colorectal cancerIn vitroHCT116 and DLD1 cells0–20 μMReducing expression level of CSC markers including CD44, Oct4 and ALDH1
Inhibiting STAT3 signaling
[338]
Table 7. Determining resveratrol potential in targeting CSCs and suppressing tumor progression.
Table 7. Determining resveratrol potential in targeting CSCs and suppressing tumor progression.
Cancer TypeIn Vitro/In VivoCell Line/Animal ModelStudy DesignRemarksRefs
Pancreatic cancerIn vitroMiaPaCa-2 and Panc-1 cells50 μmol/LPromoting gemcitabine sensitivity
Suppressing lipid metabolism
Down-regulation of SREBP1
Inhibiting stemness
Suppressing colony formation and CSC features
[368]
Pancreatic cancerIn vitro
In vivo
CSCs and mice0–30 μMInhibiting tumor growth and development in vivo
Inducing apoptosis in CSCs via upregulating caspase-3 and -7, and down-regulating Bcl-2 and XIAP
Decreasing expression levels of Nanog, SOX2, c-Myc and Oct4 as CSC markers
Reducing ABCG2 expression as an inducer of drug resistance in CSCs
[369]
MedulloblastomaIn vitroCSCs derived from medulloblastoma150 μMSuppressing proliferation of CSCs and increasing sensitivity to radiotherapy[370]
Ovarian cancerIn vitroA2780 cells0–50 μMSuppressing self-renewal capacity of CSCs
Triggering cell death in CSCs via mediating ROS overgeneration
[371]
Breast cancerIn vitroMCF-7 cells0–500 μMPreventing proliferation of CSCs
Triggering apoptosis
Mediating oxidative damage
Caspase cascade activation and inducing PARP cleavage
Reducing SOD, MnSOD and catalase levels
[372]
Nasopharyngeal carcinomaIn vitroTW01, TW06, and HONE-1 cells0–100 μMSuppressing self-renewal capacity and migration of CSCs
P53 overexpression
EMT inhibition
[373]
Colorectal cancerIn vitroHCT116 cells5 μMUpregulation of CD133, CD44 and ALDH1 as CSC markers by TGF-β
Suppressing CSC features by resveratrol and triggering apoptosis in CSCs by caspase-3 upregulation
Promoting drug sensitivity
[374]
Table 8. The BBR in targeting CSCs for tumor suppression.
Table 8. The BBR in targeting CSCs for tumor suppression.
Cancer TypeIn Vitro/In VivoCell Line/Animal ModelStudy DesignRemarksRefs
Pancreatic cancerIn vitroPANC-1 and MIA PaCa-2 cells15 μMReducing population of CSCs
Decreasing expression level of CSC markers including SOX2, ALDH1, Nanog and POU5F1
Promoting sensitivity of tumor cells to gemcitabine chemotherapy
[389]
Oral cancerIn vitroSAS and OECM-1 cells0–40 μMSuppressing tumor progression in a dose-dependent manner
Reducing colony formation, migration and self-renewal capacity of tumor cells
Reducing miRNA-21 expression to impair CSC features
[393]
Ovarian cancerIn vitroSKOV3 and A2780 cells5 μMChemotherapy promotes Gli1 expression and facilitates CSC features
Suppressing Gli1 pathway by berberine
Inhibiting EMT-mediated metastasis and reducing CSC features
[396]
NeuroblastomaIn vitroN2a cells10 and 20 μg/mLInducing apoptosis and cell cycle arrest in tumor cells
Suppressing metastasis via EMT inhibition
Inhibiting CSCs features
[397]
Breast cancerIn vitro
In vivo
MCF-7 cells
Xenografts
40 μM of liposomal berberineThe berberine-loaded liposomes selectively target CSCs and induce apoptosis
Enhancing berberine accumulation in tumor cells
Retarding tumor growth in vivo
[400]
Table 9. Ginsenosides as potential inhibitors of CSCs in different tumors.
Table 9. Ginsenosides as potential inhibitors of CSCs in different tumors.
Anti-Tumor AgentCancer TypeIn Vitro/In VivoCell Line/Animal ModelStudy DesignRemarksRefs
Ginsenoside Rg3Colorectal cancerIn vitro
In vivo
LoVo, SW620 and HCT116 cells
Nude mice
-Promoting anti-tumor potential of oxaliplatin and 5-flourouracil
Suppressing stemness and CSC features
[404]
Ginsenoside Rh2Skin cancerIn vitroA431 cells0–1 mg/mLInhibiting viability of tumor cells
Autophagy induction and β-catenin signaling inhibition
[405]
Ginsenoside Rb1Ovarian cancerIn vitro
In vivo
SKOV-3 and HEYA8 cells
Mice
0–500 nM
50 mg/kg
Suppressing Wnt signaling and reversing EMT to impair cancer stemness and inhibit drug resistance[407]
Gisnenoside Rg3Breast cancerIn vitroMCF-7 and MDA-MB-231 cells0–100 μMInhibiting Akt/HIF-1α axis
Reducing expression levels of SOX2 and Bmi-1
Inhibiting CSC features and self-renewal capacities
[408]
Ginsenoside F2Breast cancerIn vitroMCF-7 cells0–140 μMApoptosis induction in CSCs
Inhibiting autophagy promotes potential of ginsenoside F2 in tumor suppression
[406]
Ginsenoside Rh2Hepatocellular carcinomaIn vitro
In vivo
HepG2 and Huh7 cells
Mice
0–1 mg/mL
1 mg/kg
Inhibiting CSC features in a dose-dependent manner
Autophagy induction and suppressing β-catenin signaling
[409]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Deldar Abad Paskeh, M.; Asadi, S.; Zabolian, A.; Saleki, H.; Khoshbakht, M.A.; Sabet, S.; Naghdi, M.J.; Hashemi, M.; Hushmandi, K.; Ashrafizadeh, M.; et al. Targeting Cancer Stem Cells by Dietary Agents: An Important Therapeutic Strategy against Human Malignancies. Int. J. Mol. Sci. 2021, 22, 11669. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms222111669

AMA Style

Deldar Abad Paskeh M, Asadi S, Zabolian A, Saleki H, Khoshbakht MA, Sabet S, Naghdi MJ, Hashemi M, Hushmandi K, Ashrafizadeh M, et al. Targeting Cancer Stem Cells by Dietary Agents: An Important Therapeutic Strategy against Human Malignancies. International Journal of Molecular Sciences. 2021; 22(21):11669. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms222111669

Chicago/Turabian Style

Deldar Abad Paskeh, Mahshid, Shafagh Asadi, Amirhossein Zabolian, Hossein Saleki, Mohammad Amin Khoshbakht, Sina Sabet, Mohamad Javad Naghdi, Mehrdad Hashemi, Kiavash Hushmandi, Milad Ashrafizadeh, and et al. 2021. "Targeting Cancer Stem Cells by Dietary Agents: An Important Therapeutic Strategy against Human Malignancies" International Journal of Molecular Sciences 22, no. 21: 11669. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms222111669

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