Next Article in Journal
The N-Terminus Makes the Difference: Impact of Genotype-Specific Disparities in the N-Terminal Part of The Hepatitis B Virus Large Surface Protein on Morphogenesis of Viral and Subviral Particles
Previous Article in Journal
The Hippo Pathway Effector YAP1 Regulates Intestinal Epithelial Cell Differentiation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advances in Therapeutic Targeting of Cancer Stem Cells within the Tumor Microenvironment: An Updated Review

by
Kevin Dzobo
1,2,*,†,
Dimakatso Alice Senthebane
1,2,†,
Chelene Ganz
1,2,†,
Nicholas Ekow Thomford
3,4,
Ambroise Wonkam
3 and
Collet Dandara
3
1
International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Wernher and Beit Building (South), UCT Medical Campus, Anzio Road, Observatory, Cape Town 7925, South Africa
2
Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
3
Division of Human Genetics, Department of Pathology and Institute for Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa
4
Department of Medical Biochemistry, School of Medical Sciences, College of Health Sciences, University of Cape Coast, PMB, Cape Coast, Ghana
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cells 2020, 9(8), 1896; https://doi.org/10.3390/cells9081896
Submission received: 7 July 2020 / Revised: 4 August 2020 / Accepted: 11 August 2020 / Published: 13 August 2020
(This article belongs to the Section Stem Cells)

Abstract

:
Despite great strides being achieved in improving cancer patients’ outcomes through better therapies and combinatorial treatment, several hurdles still remain due to therapy resistance, cancer recurrence and metastasis. Drug resistance culminating in relapse continues to be associated with fatal disease. The cancer stem cell theory posits that tumors are driven by specialized cancer cells called cancer stem cells (CSCs). CSCs are a subpopulation of cancer cells known to be resistant to therapy and cause metastasis. Whilst the debate on whether CSCs are the origins of the primary tumor rages on, CSCs have been further characterized in many cancers with data illustrating that CSCs display great abilities to self-renew, resist therapies due to enhanced epithelial to mesenchymal (EMT) properties, enhanced expression of ATP-binding cassette (ABC) membrane transporters, activation of several survival signaling pathways and increased immune evasion as well as DNA repair mechanisms. CSCs also display great heterogeneity with the consequential lack of specific CSC markers presenting a great challenge to their targeting. In this updated review we revisit CSCs within the tumor microenvironment (TME) and present novel treatment strategies targeting CSCs. These promising strategies include targeting CSCs-specific properties using small molecule inhibitors, immunotherapy, microRNA mediated inhibitors, epigenetic methods as well as targeting CSC niche-microenvironmental factors and differentiation. Lastly, we present recent clinical trials undertaken to try to turn the tide against cancer by targeting CSC-associated drug resistance and metastasis.

Graphical Abstract

1. Introduction

Cancer remains one of the major causes of mortality globally, with many recent studies showing significant increases in its incidence [1,2]. Recent advances in cancer diagnosis and treatment have resulted in improvements in patients’ outcomes, however, several hurdles remain including drug resistance, cancer relapse and metastasis [3]. Drug resistance which can lead to relapse continues to be associated with fatal disease [3]. Data from several studies reveal that therapy resistance and chemoresistance in particular limits the therapeutic value of many drugs, resulting in relapse and metastasis [4]. Senthebane and colleagues revealed that tumor microenvironment (TME) components including cancer-associated fibroblasts (CAFs) and the extracellular matrix (ECM) are major contributors to chemoresistance [3]. Recent data also points to cancer stem cells (CSCs) as responsible for therapy resistance and metastasis [5,6,7].
CSCs have been defined as a subset of cancer cells with the ability to self-renew and to differentiate into non-CSC cancer cells within the tumor mass [6,8]. The CSC field was shaped by great research done on hematopoietic stem cells (HSCs). HSCs are hierarchically arranged with HSCs being the founder cells that undergo asymmetric cell division giving rise to differentiated daughter cells and one quiescent stem cell with self-renewal abilities [9]. The dividing daughter cells will over time become restricted in terms of lineages it can form. The studies on HSCs ignited research on mammalian tissue and cell renewal as well as in cancer. In addition, cancer patients with chronic myeloid leukemia (CML) were shown to have rare quiescent cells also referred to as Philadelphia chromosome-positive and BCR-ABL-positive cells and these cells were able to resist drug treatment [10,11]. The above-mentioned studies and revelations allowed further research on self-renewal and eventually gave birth to the CSC field as it is today. CSCs are able to reproduce primary tumor heterogeneity as well as metastases in distant tissues and organs [12]. As postulated by Paget, cancer cells can escape the primary tumor site and spread to other tissues and organs where they can proliferate and therefore act as “seeds” for the growth of secondary tumors [12]. It is possible that cancer cells can detach from the primary tumor and enter circulation, however, they are likely not to survive the arduous journey to other organs and cannot “seed” metastases at secondary sites. With their demonstrable survival abilities, enhanced expression of transmembrane transporters and tumorigenic abilities, CSCs on the other hand are likely to survive in circulation and be able to “seed” new tumors at secondary sites [13,14]. CSCs are also responsible for the development of therapy resistance, with many studies demonstrating that CSCs are able to withstand conventional therapies such as chemotherapy and radiotherapy [15]. The ability to resist conventional therapies has been attributed to many properties including increased expression of drug transporters, maintenance of a slow dividing state (quiescence) as well as efficient DNA repair mechanisms [16,17,18]. To overcome CSC resistance, new therapies are under development including epigenetic therapies, immunotherapy as well as drugs targeting angiogenesis [19].
From the early days of their discovery, many studies have shown that CSCs are undifferentiated tumor cells able to generate tumors [20,21,22]. To date, several studies have been able to prove the existence of CSCs in cancers such as CML, ovarian, lung and breast cancer [23,24]. Methods used to identify CSCs range from antibody-based isolation, enzyme activity of ALDH, tumorsphere formation, use of dyes such as PKH26 and side population sorting [25,26]. Side population cells display enhanced abilities to efflux dyes and drugs at a higher rate than the main cell population due to increased expression of ATP-binding cassette (ABC) transporter proteins. These methods are all not specific, and in most cases, scientists combine these methods to get a cell population with high numbers of CSCs. The gold standard method to study whether cancer cells have tumor-initiating capabilities is the use of limiting dilution in xenograft animals. A detailed review of CSCs definition and terminology is provided by Valent and colleagues [15]. Recently introduced “humanized” animal models are better models than traditional animal models as they can recapitulate some human cancers better [27,28].
Due to their ability to resist therapy, CSCs can travel to distant sites and form new tumors. Whilst the process of metastasis appears disorganized, metastatic lesions are the main cause of cancer deaths and therapy resistance [29]. Signaling pathways upregulated and dysregulated in CSCs and CSC-cell interactions are therefore some of the targets of new drugs under development. Conventional cancer treatment strategies mainly target rapidly proliferating cancer cells and can reduce tumor mass, tumor relapse can result from a few remaining cancer cells including CSCs (Figure 1) [30]. Our ability to target CSCs largely depends on new evidence and in-depth characterization of these cells. It is plausible to postulate that long-lasting cancer treatment efficacy can only come from both shrinkage of the primary tumor as well as the prevention of cells such as CSCs from metastasizing to new sites throughout the body.
This review is an updated critical analysis and distillation of available information on CSCs and their involvement in cancer therapy resistance and metastasis. By targeting inherent CSCs properties that allow CSCs to be tumorigenic, resistant and also metastatic, new drugs being developed offer a better promise at curing cancer.

2. Properties of Cancer Stem Cells

2.1. Cancer Stem Cell Markers and Therapy Resistance

Current therapies are unable to eliminate cancer partly due to CSCs’ enhanced ability to withstand treatment regimens [15,30]. CSCs are thought to account for a small percentage of the total number of cancer cells within a tumor but have self-renewal and differentiation capabilities [21]. A major hurdle faced by scientists working with CSCs has been the isolation and characterization of these cells. Antibodies against several CSC markers have been used to isolate CSCs from solid tumors [26]. Commonly used CSC markers and methods for isolation and characterization include CD24, CD44, CD133 and ALDH enzymatic assay (Figure 2; Table 1) [31]. These CSC markers are either used alone or in different combinations in different cancers. For example, gastric CSCs display high CD44, CD133 as well as Lgr5 [32]. Lung CSCs express several markers including CD133+, ALDH1+ and CD44+ [33]. Whilst the same CSC markers can be found in different cancers, some cancers have distinct markers for example melanoma CSCs are ABCB5+ whilst medulloblastoma CSCs are CD15+ (Table 1).
Several studies demonstrated an increase in CSCs in tumors after cancer treatment, clearly illustrating their persistence during treatment [75,76,77]. CSCs are able to resist therapeutic interventions due to several reasons including their cellular plasticity, enhanced expression of ABC drug transporters, ability to detoxify of drugs and compounds, increased adaptation to stressful conditions such as hypoxia, attaining quiescence and activation of survival pathways [77,78,79].
CSCs ability to resist therapy is widespread and referred to as multidrug resistance. This capability stems from the ability of CSCs to express increased detoxifying enzymes, increased activation of survival signaling pathways, DNA repair mechanisms as well as drug efflux pumps [30,78]. In addition, CSCs have been noted for their immune evasion capabilities, their ability to undergo epithelial to EMT as well as to adapt their metabolism to survive low nutrient conditions [77,78]. Thus, the hallmarks of CSCs include quiescence, increased expression of drug metabolizing and detoxifying enzymes, enhanced DNA reparability, the ability to undergo EMT and overexpression of ABC membrane transporters. Lately, CSCs have also been shown to undergo epigenetic reprogramming, making them very difficult to eradicate in cancers [80].
The ALDH superfamily is a large family of proteins and several members including aldehyde dehydrogenase 1 (ALDH1) have been implicated in drug detoxifying activities [81,82]. In its entirety, the ALDH superfamily is composed of 19 enzymes with ALDH1 being the main isoform [81,82,83]. This family of detoxifying enzymes is involved in the oxidation of aldehydes to carboxylic acids as well as retinol to retinoic acid [84,85]. Besides being expressed by normal cells, ALDH1 is expressed highly in CSCs [86,87]. As a result, ALDH1 expression and activity can be used reliably to identify CSCs in some cancers. Vogler and colleagues demonstrated that ALDH1 expression can be used as an independent prognostic marker for low survival in colorectal patients [88]. In addition, van den Hoogen and coworkers also showed that enhanced ALDH1 activity can be used to identify tumor-forming cells as well as cells with the propensity to form prostate cancer metastases [89]. Ueda and colleagues also showed that ALDH1 activity can be used to identify cancer cells with CSC-like properties in human renal cell carcinoma cell line [90]. Ginestier and colleagues demonstrated that ALDH1 is highly expressed in breast CSCs and is a predictor of poor clinical outcome [91]. In addition, ALDH1-expressing cells were able to form xenograft tumors easily [91]. Several other studies demonstrated the successful transplantation of ALDH1-expressing cells into mice [92,93]. The expression of ALDH1 by normal stem cells may explain the aberrant expression of this enzyme in CSCs as normal stem cells are a potential source of CSCs, among other cells [94]. Furthermore, ALDH1 expression has been shown to allow CSCs to resist conventional therapy including commonly used drugs such as paclitaxel, gemcitabine and cisplatin [95,96]. In agreement with the above, several studies demonstrated that inhibition of ALDH1 activity in CSCs sensitizes these cells to several drugs, linking ALDH1 with therapy resistance [97,98].
In addition, CSCs demonstrate increased expression of drug effluxing proteins such as the ABC transporters (Figure 3) [99,100,101]. The ABC family of transporters consists of 49 molecules using ATP as an energy source during the trafficking of proteins across the cell membrane. Many studies have been performed on the characterization of members of this family including ABCB1 (multidrug resistance 1 (MDR1)), ABCG2, ABCC1 and ABCB5 [102,103]. Through elaborate experiments, several research groups demonstrated that CSCs aberrantly express ABC transporters and are able to withstand toxic levels of drugs and other toxins [104,105]. In elaborate experiments performed by Wright and colleagues, the researchers demonstrated that ABCB1 was aberrantly overexpressed in breast CSCs causing resistance to conventional chemotherapy such as paclitaxel and doxorubicin [106]. Frank and coworkers demonstrated that ABCB5 was overexpressed and caused resistance to doxorubicin in CD133+ circulating melanoma cells [107]. Through the use of a monoclonal antibody against ABCB5, the authors were able to induce cancer cell sensitivity to drugs such as doxorubicin [107]. Shi and colleagues demonstrated that ABCG2-expressing CSCs isolated from hepatocellular carcinoma cell lines via the side population technique are able to resist cisplatin and 5-fluorouracil [108]. The above studies and others demonstrated that inhibition of ABC transporters is a potential mechanism of overcoming CSC chemoresistance [109,110]. Several studies have been performed on the inhibition of ABC transporters and have shown remarkable success in sensitizing both cancer cells and CSCs to several drugs [111,112]. For example, Marcelletti and colleagues utilized zosuquidar, an inhibitor of P-gp (ABCB1) to sensitize cancer cells in acute myeloid leukemia [111,112].
Several other proteins associated with apoptosis are also involved in the survival of cancer cells and CSCs [113,114]. For example, several pro-survival proteins including BCL-2, B-cell lymphoma extra-large (Bcl-xL) and BCL-2-like-2 (BCL-W) have been found to be overexpressed in several cancer types including lymphoid cancer [115,116]. The overexpression of these pro-survival proteins has also been linked with carcinogenesis, with the blocking of these proteins and their associated pathways resulting in reduced tumor growth and enhanced response to chemotherapy [116,117,118].
Cytotoxic drugs target rapidly growing cancer cells, making them ineffective against slow dividing or dormant CSCs [30]. Viale and colleagues demonstrated that leukemia CSCs proliferate at a much lower rate than other cancer cells [119]. Therapies that target cancer cell cycling would therefore be ineffective against CSCs. Therapeutic agents such as paclitaxel would be unable to be less effective against slow dividing CSCs [120]. In addition, several studies demonstrate that CSCs show enhanced DNA damage repair capacity, with phosphorylation of repair enzymes observed in cancers such as breast and gliomas [121,122]. CSCs including glioma stem cells demonstrate great abilities at ROS scavenging thus protecting themselves against oxidative DNA damage [123,124,125]. Therapy itself has been shown to selectively increase CSCs in tumors. For example, Rizzo and colleagues demonstrated that CSCs are enriched in ovarian tumors after chemotherapy [126]. In addition, Levina and coworkers showed that chemotherapy can lead to the propagation of CSCs in lung cancer [127]. Thus, chemotherapy only targets the rapidly proliferating cancer cells leaving the CSCs to propagate the tumor after therapy. Chen and colleagues demonstrated that the drug temozolomide (TMZ) activates CSCs to produce cancer cells after therapy [128]. Qiu and colleagues demonstrated that elevated O6-methylguanine DNA methyltransferase (MGMT) expression and activity in glioma stem-like cells were responsible for temozolomide resistance [129]. Kurtova and coworkers also demonstrated that blockage of tumor repopulation by CSCs is effective at attenuating therapy resistance in bladder cancer [130]. Saito and colleagues demonstrated that inducing cell cycle re-entry through treatment with granulocyte colony-stimulating factor (G-CSF) allows normal chemotherapy to eliminate cancer cells effectively [131]. In addition, the induction of CSCs differentiation has been used successfully to increase CSCs sensitivity to commonly used cancer drugs. Lombardo and coworkers induced colorectal CSCs terminal differentiation via the use of bone morphogenic protein 4 (BMP4) and observed increased CSCs sensitization to standard chemotherapy [132]. Wang and coworkers used silibinin, which blocks colon CSCs self-renewal, resulting in reduced CSC population leading to reduced cancer cell proliferation [133]. Whilst several strategies have been developed to induce CSCs differentiation, all-trans retinoic acid (ATRA) is one of the common drugs used for this purpose [134,135].

2.2. Cancer Stem Cells and Angiogenesis

Many biological processes are dependent on the formation of new blood vessels, a process referred to as angiogenesis. Normal development and tissue repair and regeneration are especially dependent on new blood vessels for the supply of nutrients as well as the removal of toxic material [136,137]. Besides normal biological activities, angiogenesis is a requirement for tumor formation beyond a certain diameter [138,139]. During tumor formation, the usual delicate balance between pro-angiogenesis and anti-angiogenesis is altered, with pro-angiogenesis factors dominating [138]. New blood vessels sprout from pre-existing vessels within and around the tumor, fueling the rapid growth of the tumor [140,141]. The rapid growth of a tumor results in hypoxic conditions within the tumor. CSCs are known to release factors such as hypoxia-inducible factor 1 which induces the release of proangiogenic factors (Figure 4) [142,143]. Hypoxia has also been shown to fuel CSCs [144]. Hypoxia promotes CSC growth in several cancers via the upregulation of adaptive transcriptional programs, allowing CSCs to survive, invade and metastasize [144,145]. Soeda and colleagues demonstrated that hypoxia promotes the self-renewal capacity of CD133-positive human glioma-derived cancer stem cells [144]. Heddleston and colleagues showed that hypoxia promotes CSC self-renewal capabilities and stem-like phenotype even in non-stem cancer cell populations [146]. As reviewed by Heddleston and colleagues, CSC plasticity is influenced by hypoxia and new drugs must target such microenvironmental conditions for durable cancer treatment [147].
Endothelial cells are also recruited to the tumor site. Endothelial cells express VEGFR and the binding of VEGF-A results in the activation of several signaling cascades involved in migration and ECM remodeling [143,148]. Survival pathways including the PI3K-Akt and the MEK-ERK cascades are activated and play key roles in the activation of endothelial cells to form new blood vessels [148]. Several cytokines are also known to be secreted by CSCs within the TME and these include IL-6 and TNF-α [149,150]. The secreted cytokines are involved in the recruitment of immune cells such as myeloid cells to further promote tumorigenesis [149,150]. Dysregulation of the Notch pathway has also been associated with tumor growth in general and survival of CSCs [151,152,153]. Several reports demonstrated that cells showing high expression of Notch signaling have elevated tumor-forming abilities and self-renewal capacity than those with less Notch activation [154,155,156]. Activation of the Notch pathway has also been associated with proangiogenic activity, with Notch ligand Jagged-1 promoting blood vessel formation [157,158,159].
In addition, matrix metalloproteases (MMPs) secreted by both cancer cells and stromal cells remodel the TME, allowing the creation of space for blood vessels formation as well as recruitment of different cells [160,161]. Due to the plasticity of CSCs, suggestions have been made to the effect that CSCs can give rise to endothelial cells and pericytes within and around the tumor [162,163]. Blood vessels within the TME are convoluted and “leaky”, resulting in fewer drugs able to reach cancer cells and CSCs deep within the TME. Tumor-derived cells are also able to intravasate and travel to distance sites, promoting metastasis in the process. Several studies have demonstrated the presence of circulating tumor-derived cells that are able to act as “seeds” for new tumors in distant sites [164,165]. Once the circulating cancer cells reach distant sites, they are able to extravasate and form new tumors in favorable microenvironments [19,166]. The formation of new tumors is dependent on CSCs successfully inducing angiogenesis to allow the exchange of nutrients and metabolic byproducts. Whilst it has been shown that stromal cells play a key role in inducing angiogenesis within tumors, CSCs are also involved in releasing angiogenic factors [167]. For example, Bao and colleagues demonstrated that glioma CSCs release VEGF resulting in increased microvascular density in malignant glioma [168]. Monzani and colleagues also showed that melanoma CSCs co-expressed CD133 and VEGF [169]. Maeda and colleagues also showed that pancreatic CSCs co-express CD133 and VEGF-C resulting in increased microvascular density [170]. It is also possible that cancer cells may enter a state of dormancy in which they remain until induced to proliferate and form new tumors [171,172].

2.3. Cancer Stem Cells and Epithelial to Mesenchymal Transition

Besides the influence of genetic and epigenetic mechanisms on the CSC phenotype, the TME within which CSCs are located plays a huge role in the CSC behavior [30]. As more data emerges the CSC field continues to change and be refined [117,173]. Overall, the CSC phenotype is dynamic and never constant. When CSCs undergo EMT, they acquire characteristics allowing them to migrate, invade surrounding tissues and metastasize [174]. EMT and CSC characteristics appear to share similar molecular pathways that are involved in invasion and migration of cancer cells from the primary tumor. In addition, transcriptional analysis of EMT and those associated with CSCs reveal significant overlap in gene expression including TGF-β, Hedgehog signaling and microRNAs [26]. EMT has been associated with poor prognosis in several cancers including esophageal and colon cancers [175,176]. Several signaling pathways have been identified to be key in modulating CSCs behavior including invasiveness and metastatic ability [177,178]. In addition, several markers identifying CSCs with invasive and metastatic abilities have been revealed including CD44v6 [179,180]. CD44 is specifically expressed by breast epithelial cells undergoing EMT [181]. EMT is characterized by the loss of cell to cell adhesion with cells becoming mesenchymal and markers such as E-cadherin lacking in such cells [182,183]. The loss of E-cadherin from the cell surface is accompanied by the expression of N-cadherin [184]. Histone deacetylation of the CDH1 promoter through the actions of DNMT and HDACs leads to gene silencing [185,186]. Histone methylation within the CDH1 promoter via the EZH2 and PRC2 complex is known to silence its expression [187].
EMT is influenced by several protein factors as well as microRNAs. For example, TGF-β has been regarded as a master regulator of EMT in certain cancers including breast and colorectal cancers [188]. Besides influencing cancer cells, TGF-β can also regulate CAFs with a net effect of promoting metastasis [189]. Furthermore, microRNA-200 family members have been shown to suppress EMT via binding to two transcription factors, zinc finger E-box-binding homeobox 1 (ZEB1) and ZEB2 [190,191]. Tellez and colleagues demonstrated that EMT can be induced by epigenetic mechanisms including chromatin remodeling through H3K27me3 enrichment as well as DNA methylation to sustain silencing of tumor-suppressive microRNAs, microRNA-200b, microRNA-200c and microRNA-205 [192]. Thus, silencing these microRNAs through tri-methylation of DNMT and H3K27 can induce EMT-like and CSC characteristics [192].

2.4. Cancer Stem Cells and Metabolic Activity

Recently, metabolic alterations have been identified to cause cells to acquire stem-cell-like characteristics [193]. These alterations and the subsequent acquisition of stem-cell-like characteristics are thought to be caused by epigenetic changes in adult stem cells as well as cancer cells. Based on the CSC theory, acquisition of stem-cell-like characteristics makes these cells achieve a higher status within the hierarchy through the expression of self-renewal and pluripotent genes [30,78]. According to Menendez and Alarcon, products of mutated metabolic enzymes can behave as oncometabolites, inducing epigenetic changes in genetic material and thus drive tumor initiation and progression [193]. This and more pieces of evidence point to the need for a full view of tumor initiation and progression and not just focus on cancer cells. Metabolic processes can thus be targeted to stop tumor initiation and progression. Specifically, the TME is characterized by low oxygen and glucose levels and thus tends to favor oxidative phosphorylation as the main supplier of energy [194]. Hypoxia has been shown to induce metabolic alterations resulting in acidosis in several cancers [195,196]. Lee and colleagues demonstrated that chemoresistance and enhanced oxidative phosphorylation are correlated [194]. Recent studies demonstrated that indeed, the targeting of oxidative phosphorylation has shown some success in inhibiting CSCs metabolic processes and proliferation in some cancers [197,198].
Inhibition of the mitochondrial complex III resulted in decreased breast CSCs [199]. When relapse occurs, CSCs have been shown to increase oxidative phosphorylation levels to pretreatment levels, demonstrating the importance of oxidative phosphorylation in chemoresistance [200]. The adipose tissue and adipose-derived cells are able to interact with CSCs and have been shown to promote fatty acid oxidation in CSCs and chemoresistance [201]. The mitochondria are also known to play a role in CSC chemoresistance [202]. This is unsurprising as the mitochondria are central to many cellular processes such as metabolism, signaling and apoptosis. Mitochondria have recently been shown to play key roles in CSC behavior [203]. Sancho and colleagues concluded that the removal of CSCs through targeting mitochondrial function might prevent cancer disease from recurring and thus prevent fatal disease [204]. In colon CSCs, tumorigenic ability was associated with enhanced mitochondrial functions [205]. Atovaquone has been used to inhibit the mitochondrial complex II resulting in decreased breast CSCs [199]. Isayev and colleagues demonstrated that inhibition of glucose metabolism through the use of 3-bromopyruvate inhibited pancreatic CSCs growth and resistance to gemcitabine [206]. Several other studies also showed that inhibition of mitochondrial function affect CSC proliferation and self-renewal capabilities [207,208].

2.5. Cancer Stem Cells and Epigenetic Reprogramming

A contributing factor to the complex intra- and inter-tumor heterogeneity and the resulting failure of many anticancer therapies comes from CSC epigenetic alterations. The heritable non-genetic changes to CSCs phenotypes are what are called epigenetic reprogramming of CSCs [209,210]. Most of the proteins and enzymes involved in epigenetic reprogramming of cells including histone modifications and DNA methylations have been well-characterized [211,212]. For example, histone methyltransferases (HMTs) are responsible for methylation of histones whilst histone acetyltransferases are responsible for the acetylation of histones [213]. Demethylation and deacetylation of histones are carried out by histone demethylases (HDMs) and histone deacetylases (HDACs) respectively [213]. When acetylated, histones are more loosely packed and can be accessed by RNA polymerases, allowing transcription of genes around a specific location. On the other hand, methylation can activate or repress gene transcription. For example, the acetylation of histone H3/H4 is linked to the transcription of genes [214,215]. In addition, H3 lysine 4 methylation is also linked to transcription of several genes [216,217]. In contrast, the methylation of H3 lysine 9 and 27 is linked to gene repression [218,219,220]. It has been observed that different patterns of histone modification produce variable transcriptional outcomes, with some giving rise to activation of genes and others to repression [221,222]. Various mechanisms are known to be involved in epigenetic gene regulation, from modifications of cytosines in DNA, covalent modifications of histones, the involvement of noncoding RNAs to chromatin remodeling [223,224,225].
CpG islands are regions of the genome containing a large number of CpG dinucleotide repeats and usually extend for 300–3000 base pairs [226]. In most cases, CpG islands are located close to gene promoters in humans [227]. DNMT in addition to histone modification determines whether transcription occurs or not. When CpG islands are unmethylated, transcription can take place. When CpG islands are methylated the chromatin becomes transcription-suppressive. Methylation of CpG islands is catalyzed by DNMT1, DNMT3A and DNMT3B. Several tumor suppressor genes are silenced via CpG island methylation [228]. Transcription can also be repressed via the Polycomb repressive complexes 1 and 2 (PRC1 and PRC2) [229,230]. Polycomb repressors are able to catalyze the trimethylation of histone 3 lysine 27 (H3K27me3) giving rise to repression of genes associated with many cellular processes such as differentiation, development and choice of lineage [229,231]. Collinson and colleagues demonstrated that Polycomb complex PRC2 mediates H3K27me3 via the histone methyltransferase EZH2, leading to transcriptional repression of several genes [232].
CSCs and their subsets display epigenetic alterations including histone modifications and this eventually contributes to the intratumor heterogeneity observed in many tumors [233]. Several epigenetic regulators have mutations leading to tumor formation and progression as a result of epigenetic dysregulation [234,235]. Several CSC markers including CD133 are known to be regulated by epigenetic alterations [236]. Tabu and colleagues demonstrated that the hypomethylation of the CD133 promoter influences its expression in gliomas [237]. Yi and colleagues observed abnormal DNA methylation of CD133, a CSC marker, in colorectal and glioblastoma tumors [236]. Gorodetska and colleagues observed that EZH2/BRCA1 signaling mechanisms play an important role in the maintenance of prostate CSCs properties [238]. EMT aid in the generation of cells with stem cell characteristics and is modulated by epigenetic mechanisms [239,240]. The involvement of epigenetic mechanisms from CSC formation to maintenance makes epigenetics a therapeutic target in CSCs. Small compound inhibitors with the ability to induce differentiation in CSCs are therefore promising drugs targeting this population of tumor cells.
Several signaling pathways are crucial in facilitating the growth of CSCs and the maintenance of the CSC phenotype. Such signaling pathways include Hedgehog, Notch, JAK-STAT and Wnt-β- catenin signaling [241,242]. It is important to note that these same pathways are also important in regulating self-renewal in normal stem cells [243,244]. Several mutations have been observed in genes along these pathways in many human cancers. Signaling pathways such as Wnt and Notch have been observed in breast cancers for example [245] and in vitro work demonstrated that the overexpression of these pathways is associated with tumorigenicity [155,156]. Triple-negative breast cancer cells demonstrate increased Notch signaling and Notch signaling is associated with CD44 expression in colon cancer cells [158,246]. On the other hand, Wnt-β-catenin signaling has been observed to be associated with cancer stemness and heterogeneity [247]. Several members of the Wnt-β–catenin pathway have been linked to the induction of EMT in several cancers [248]. The hedgehog signaling pathway has been associated with self-renewal in many cancers including breast cancer and gliomas [249,250]. The hedgehog pathway has also been associated with EMT and invasion and migration [251,252].
Most of the above-mentioned signaling pathways are modulated by epigenetic mechanisms [253]. Under normal conditions, most of these pathways are involved in the propagation of CSCs, maintenance of the CSC phenotype as well as in embryonic development [254]. Several regulators of the above-mentioned pathways have been shown to have epigenetic alterations in CSCs. For example, decreased acetylation of H3K16 as well as enhanced H3K27 trimethylation is associated with DKK1 promoter silencing [255]. High levels of histone acetylation are observed at the promoter region of Notch receptor–ligand JAGGED2, resulting in Notch signaling activation in multiple myeloma cells [256]. In colorectal cancer, two Notch signaling targets, HES1 and HES2, show decreased promoter H3K27 methylation, resulting in gene activation [257,258,259]. Rhabdoid tumors show decreased or inactivation of SNF5, a member of chromatin remodeler complex SWI/SNF, leading to activation of Hedgehog signaling [260,261]. Furthermore, the activation of Gli1 and Gli2, downstream effectors of the Hedgehog signaling pathway, require HDAC1 [262,263,264]. As a result of the integration of genetic, epigenetic mechanisms and other factors, CSCs survival and maintenance are promoted.
The KMT2/MLL gene is known to encode for an HMT that influences many cellular processes [265,266]. MLL fusion proteins are present in several CSCs and have been shown to be involved in carcinogenesis in several cancers [267,268]. For example, Krivtsov and colleagues demonstrated that leukemia stem cells, with the MLL-AF9 fusion protein, can maintain the identity of progenitors from which they arose while at the same time activating stem-cell- or self-renewal-associated program [269]. Somervaille and colleagues also demonstrated that the hierarchical maintenance of MLL-myeloid leukemia stem cells utilizes a transcriptional program involving transcription/chromatin regulatory factors Myb, Hmgb3 and Cbx5 [270]. Several mutations have also been identified in histone-encoding genes. Lewis and colleagues showed that the blockage of PRC2 activity via the gain-of-function H3 mutation was prevalent in pediatric glioblastoma [271]. Furthermore, several DNMTs are mutated in acute myeloid leukemia and have been suggested to result in the formation of leukemia stem cells [272,273,274].
CSCs have been shown to play important roles in the propagation, growth and metastasis of colorectal cancer (CRC). Several genetic and epigenetic changes have been observed in CSCs in CRC. For example, the hypermethylation of several tumor suppressor gene promoters including p16, retinoblastoma, SFRP and MLH1, has been widely reported in many studies [7,275,276]. One of the driver mutations in CRC is the APC mutation, which influences the activities of DNMTs [277]. Increased levels of DNMT1 are thought to suppress the transcription of APC, a tumor suppressor gene in CRC [278,279]. The levels of DNMT1 in CSCs have been shown to be involved in CRC initiation and progression, directly linking epigenetic mechanisms to CSC-directed tumorigenesis [280]. Pathania and colleagues showed that DNMT1 is important for mammary and CSC maintenance and tumorigenesis [281].

3. Targeting Cancer Stem Cells in Tumor Microenvironment

Conventional anticancer therapies including chemotherapy and radiotherapy target rapidly proliferating cancer cells and can successfully debulk a tumor. However, several studies have shown that conventional therapies cannot prevent resistance, tumor relapse and metastasis [3,4]. It has been postulated that this is due to the presence of CSCs [30]. Several studies have shown that CSCs can easily undergo EMT, allowing these cells to promote tumor formation and progression [282,283]. The enhanced plasticity and heterogeneity observed within the CSC population, however, make targeting these cells daunting [196,284]. Currently, several strategies are employed to target CSCs in different cancers (Table 2). Combinations of surface markers have been used to isolate and characterize CSCs from different tissues. For example, CD44 and CD24 are used to isolate breast CSCs [285,286]. Combinations of drugs and antibodies have been used to target CSC surface markers successfully in different cancers [287,288]. In addition, the prevention of CSC surface markers from interacting with other proteins via the use of antibodies can result in CSCs being engulfed by immune cells, leading to tumor growth inhibition [289,290,291]. One such example is the use of a monoclonal antibody against CD47 named Hu5F9-G4 [292,293].

3.1. Targeting Cancer Stem Cell Signaling

Several CSC-specific signaling cascades have also been targeted in many cancers (Table 2). The Wnt-β-catenin has been observed to be dysregulated in CSCs in addition to several members of the pathway being mutated [294,295]. Several chemotherapeutic agents ranging from CWP232228, NCB-0846 and PRI-724 are either under clinical trial or being tested in in vitro research. PRI-724 targets CSCs by targeting their rapid cell division [296,297]. Jang and colleagues showed that CWP232228 preferentially targets breast CSCs in in vitro and animal cancer models [298,299].
Another important signaling cascade targeted in CSCs is the Notch pathway. Xu and colleagues demonstrated that inhibition of Notch signaling via the use of RO4929097 combined with chemotherapy has a beneficial effect in glioma patients with an observed reduction in CSCs [300]. Zhao and colleagues showed that chemotherapy together with another Notch inhibitor, DAPT (N-[N-(3, 5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester), targeted CSCs in head and neck cancer [301]. Another signaling pathway that has been inhibited in CSCs is the Hedgehog pathway. The inhibition of the Hedgehog pathway through the use of nitidine chloride was shown to reduce CSC formation and abrogated the EMT process [302]. Hedgehog signaling inhibition coupled to the inhibition of the PI3K-Akt pathway was shown to reduce CSC self-renewal abilities [303]. Miyazaki and colleagues demonstrated that the combined inhibition of Hedgehog signaling and mTOR in pancreatic cancer cell lines suppressed CD133 expression and the ability of CSCs to form tumorspheres [304]. Clinical trials have been performed using gemcitabine and Smoothened inhibitor, Vismodegib [305,306].
Furthermore, the inhibition of the STAT3 pathway through the use of napabucasin was shown to reduce the viability of hematopoietic CSCs as well as their tumorigenic capabilities [307]. Napabucasin was also able to prevent relapse in pancreatic cancer after chemotherapy, demonstrating its effect on tumorigenic cancer cells [308]. Several clinical trials of napabucasin in combination with chemotherapy or immunotherapy are underway for the treatment of many cancers including colorectal carcinoma [309,310,311]. An inhibitor of the PI3K-Akt pathway, VS5584, has been shown to reduce CSCs in breast cancer and has shown its effectiveness at preventing relapse after chemotherapy [312,313]. Several therapeutic agents have been shown to affect many CSC signaling cascades and are therefore appealing. For example, salinomycin has been shown to selectively kill CSCs via inhibition of potassium flux as well as targeting the self-renewal properties of CSCs [314]. CSCs self-renewal is inhibited through the action of salinomycin on pathways such as Wnt and STAT3 signaling [181]. Salinomycin nanoparticles alone or in combination with chemotherapy when used on a breast cancer model were able to enhance mice survival [315].

3.2. Targeting Cancer-Stem-Cell-Associated Tumor Angiogenesis and Metastasis

Given the dependence of tumor formation and growth on the formation of blood vessels within the TME, inhibition of angiogenesis has been touted as having clinical value and could improve cancer patients’ outcomes (Table 2). One of the earliest anti-VEGF approved treatments involved the use of Bevacizumab, a monoclonal antibody that blocks the binding of VEGF to its receptor VEGFR [316]. Bevacizumab is mostly used together with commonly used drugs such as 5-fluorouracil as well as together with panitumumab [316,317]. Bevacizumab is currently being used for the treatment of colorectal, cervical and gastric adenocarcinoma [316,317,318,319]. Bevacizumab has not been successful in the treatment of other cancers such as breast cancer, with results showing poor patients’ overall survival [320,321]. Several tyrosine kinase inhibitors have been used to block angiogenesis including sunitinib and sorafenib. Importantly, sorafenib inhibits VEGFR in addition to PDGFR-β, thus can also affect the pro-tumorigenic behavior of stromal cells such as CAFs [322,323]. Sorafenib is currently being used to treat hepatocellular carcinoma, thyroid cancer and advanced renal cell carcinoma [324,325,326]. Sunitinib is also used for the treatment of renal cell carcinoma, thyroid cancer as well as advanced breast cancer [327,328,329,330]. Reports of resistance against antiangiogenic therapies have been published in addition to decreased amounts of therapy actually reaching cancer cells [331,332]. In addition, antiangiogenic therapies may increase CSCs through formation of hypoxic regions within tumors. Thus, it is important that thorough investigations are done for each drug to understand the mechanism of action.
The key to the formation of new blood vessels is the creation of space for cells to burrow through. Thus, matrix metalloproteinases (MMPs) and their inhibitors, the tissue inhibitors of metalloproteinases (TIMPs), play important roles in enabling the formation of new blood vessels [333,334,335]. Measured through the gold standard, which is the overall improvement of patients’ survival, MMPs inhibitors have produced disappointing results [334,336,337,338]. Many studies have shown that inhibition of MMPs has negative overall effects on normal cellular processes and thus detrimental to the human body [339,340,341]. Whilst MMPs are involved in tumor initiation and development, these enzymes are also important in normal cellular processes, making their inhibition a challenge. Selective and specific inhibitors of MMPs perceived to be involved in tumorigenesis have not been forthcoming or are still under investigation [342,343,344,345]. RO4929097, a gamma-secretase inhibitor, has been shown to reduce CSCs in glioma patients but the use of this inhibitor also resulted in the development of resistance [300,346]. One of the major disadvantages of inhibitors targeting signaling and enzymes (including MMPs) aberrantly expressed in CSCs is the negative side effects and the potential of therapy resistance. Several known protein factors such as cytokines and growth factors promote angiogenesis as well as the migration of CSCs [184,347,348]. Ginestier and colleagues demonstrated that blocking CXCR1 affects mostly breast CSCs in elaborate experiments involving the use of cells and xenografts [349]. The authors used Reparixin in elaborate experiments and showed that it has anti-CSCs activity in breast cancer cell lines [349]. Reparixin has been in several clinical trials with mixed results from such studies (Table 2) [350,351]. Further research involving blocking CXCR1/2 by Singh and colleagues also demonstrated decreased CSC activity in breast cancer [352]. Interactions between CSCs and the stromal component of the tumor microenvironment are mediated via chemokines and their receptors. For example, stromal-derived factor 1 and its receptor CXCR4 are both involved in the interactions between CSCs and cells such as CAFs and CAMs [347]. Stromal-derived factor has been implicated in cancer cell migration as well as invasion of nearby tissues for example [347]. Elaborate experiments by Gassenmaier and colleagues demonstrated that CXCR4 was upregulated in CSCs and that the inhibition of CXCR4 in renal cell carcinoma through the use of AMD3100 would hamper CSCs ability to proliferate and formation of tumorspheres [353]. As reviewed by Trautmann and colleagues, the use of combination therapy through targeting CXCR4-expressing CSCs in addition to radiotherapy can result in a durable cure for cancers [354].

3.3. Targeting the Immune System to Eradicate Cancer Stem Cells

Whilst research on the development of new drugs is an ongoing endeavor, new strategies being developed to eradicate cancer include targeting the stromal cells, CSCs and immune cells within the TME [4,30]. Importantly, the induction of an immune reaction to tumor cells as well as strengthening the immune system is some of the various methods being implemented (Table 2). Immunotherapy has been at the forefront of new strategies to boost the immune system of cancer patients, with the hope that it will lead to better patients’ outcomes. Several studies have demonstrated that the immune system can be used to fight cancer [355,356]. As immunotherapy works by inducing an immune response to cancer cells, it is possible to work for all cancers although results show varying patients response rates, with only a fraction of patients benefiting from such a treatment strategy [356]. Many candidate drugs that can inhibit the immune checkpoints are now in use or undergoing different levels of clinical trials [357,358,359,360]. Although several therapies have been developed, notable success came from antibodies targeting the programmed cell death 1 (PD-1) pathway alone or in combination with others [361,362,363]. PD-1 expression is triggered when the T cell receptor binds to cancer cells. In turn, PD-1 binds to PD-1 ligand (PD-L1) on cancer cells leading to exhaustion of T cells. Exhaustion of T cells dampens the anticancer cytotoxic T cell responses [364]. Another promising therapy involves the use of antibodies against the cytotoxic T lymphocyte-associated protein 4 (CTLA-4) [365,366,367]. CTLA-4 causes T cell inhibition via competing with stimulatory molecules for T cells. Binding of CTLA-4 to receptors on T cells causes inhibition of T cell proliferation, dampening cancer cell recognition and killing [368]. By blocking this immune checkpoint through the use of antibodies, allows T cells to proliferate and be able to recognize antigens on the cancer cell surface.
Several antibodies have been developed to induce an anticancer immune response. For example, Ipilimumab is an anti-CTLA-4 inhibitor that was developed and used in patients with advanced melanoma [369]. Patients displayed improved and durable responses but side effects including inflammation of endocrine glands were observed. In combination with others such as Nivolumab, Ipilimumab has been used for several cancers including renal cell carcinoma, melanoma, metastatic colorectal cancer, small cell lung cancer and metastatic esophagogastric cancer [369,370,371]. Promising results from these antibodies led to the approval of several others including avelumab, pembrolizumab, durvalumab and atezolizumab [372,373,374,375]. When used in different cancers, these checkpoint inhibitors display varying response rates with some showing very high responses such as in Hodgkin’s disease where the response rate was around 90 percent [376,377,378]. Reports of cardiotoxicity and pneumonitis show that further research is still required to reduce the side effects associated with these antibodies [379,380]. Interestingly some reports show that checkpoint inhibitors may work synergistically with antibodies against other markers such as HER2 in breast cancer [381,382,383,384].
Resistance to immune checkpoint therapy has been suggested to be caused by CSCs. Stemness as well as increased angiogenesis has been associated with reduced recognition of T cells [385,386,387]. Several studies have demonstrated that CSCs have the ability to evade the immune system [387,388]. Wu and colleagues showed that the overexpression of PD-1 may be the reason CSCs are able to evade the immune system [389]. Bruttel and Wischhusen on the other hand showed that CSCs evade the immune system via lack of molecules needed for T cell recognition [390]. Several other studies showed that CSCs evade the immune system due to their creation of an immune suppressive microenvironment [391,392]. Despite the above, the high expression of PD-L1 on the CSCs’s surfaces makes CSCs targets of checkpoint inhibitors. Standard therapies can be applied first followed by immunotherapy to wipe out the remaining CSCs [393,394,395].
One of the best immunotherapies under trial and available for cancer patients is the chimeric antigen receptor (CAR) T cell transfer. CAR T cell transfer can be used for both solid and liquid malignancies [396,397]. CAR T cells can potentially identify any marker or antigen on the surface of CSCs and thus makes them an appealing substrate for the development of CSC-specific therapies. As reviewed by Guo and colleagues, CAR T cells offer a curable approach for the treatment of cancer and the avoidance of fatal disease [151]. Several studies have investigated the use of CAR T cells together with standard and other therapies for the treatment of different cancer types. For example, Feng and colleagues investigated the potential of combining two CAR T therapies in patients with advanced Cholangiocarcinoma (CCA) [398]. The authors observed that CAR T therapy may be feasible for the treatment of Cholangiocarcinoma (CCA) but cautioned its use before further studies were done due to possible toxicities [398]. In their study, the authors used CAR T anti-EGFR and anti-CD133 in order to specifically target CSCs [398]. Several clinical trials utilizing CAR T therapy are underway for different cancers. Both CAR T anti-EGFR and anti-CD133 are under clinical trials (NCT02541370 and NCT01869166). In another study, Guo and colleagues observed that CAR T anti-EGFR cell immunotherapy was a safe way to treat EGFR-positive advanced biliary tract cancers [399]. In yet another study, a combination of haploidentical CD19-CAR T cells and stem cells achieved full donor engraftment in refractory acute lymphoblastic leukemia [400]. Utilizing well-characterized CSC markers it is possible therefore to use CAR T cells to eliminate CSCs in many cancers. The use of CAR T cells can also solve the problem of non-universal expression of some markers. Whether used alone or in combination with checkpoint inhibitors or standard therapy, CAR T cells are a promising strategy for the treatment of many cancers. As CAR T cells and also checkpoint inhibitors target markers on CSCs and the immune system respectively, both treatment strategies can result in improved treatment outcomes by not being targeted at specific cancer cells.
The above-described strategies require moderation as the overstimulation of the immune system can be detrimental. Done properly with proper control, immunotherapy can become a very good and natural way to respond to the presence of cancer cells in the body. Challenges remain in terms of immunotherapy for cancer treatment. For example, Noh and colleagues demonstrated that CTL-mediated immune selection drives tumor cell evolution toward the CSC phenotype, with the resulting CSCs demonstrating great heterogeneity [401].

3.4. Targeting Epigenetic Modifications in Cancer Stem Cells

Recent data point to possible manipulation of epigenetic states or mechanisms in cancer cells by altering molecular factors that are involved. Major hurdles remain including the identification of compounds and agents able to selectively target epigenetic mechanisms in cancer cells at low concentrations. HDAC inhibitors for example are mostly considered as pan-inhibitors and display many side effects. Many pan-HDAC inhibitors have been approved by the FDA or are under trials. One well studied HDAC inhibitor is Vorinostat, which targets HDAC-1-3 and HDAC 6. Several clinical trials using Vorinostat as a standalone drug or in combination with others are underway for cancers that relapsed or other solid tumors [402,403,404]. Other HDAC inhibitors under clinical trial include Romidepsin which is being studied for both pediatric and adult cancers [405,406]. In addition, DNMTs inhibitors including Azacitidine and Decitabine are also under different stages of clinical trial for several cancers.
The BET family, which are chromatin readers, interact with chromatin modifiers as well as enzymes to affect chromatin modification. Proteins containing bromodomains dock on acetylated histones [407,408]. Consequently, the histone code will influence not only the DNA sequence but also the transcription factors involved [153]. BET inhibitors including JQ1 and I-BET762 have shown efficacy in clinical trials against CSCs in several cancers such as neuroblastoma, acute myeloid leukemia and NUT midline carcinoma [409,410,411]. Major hurdles remain on the use of BET inhibitors with side effects including toxicity to normal cells development of resistance [412,413]. To overcome possible resistance to standard therapy and treatment with epigenetic drugs including BET inhibitors, combination therapy is usually done during treatment. Chemotherapy and radiotherapy can be combined with pan-HDAC and BET inhibitors or with immunotherapy for durable cancer treatment. In most cases, combining epigenetic drugs such as BET inhibitors with chemotherapy and immunotherapy demonstrate synergistic effects in both cells and animal models [414,415]. Li and colleagues demonstrated that drugs targeting cancer cell epigenetics potentiate chemotherapy effects in solid tumors [416,417]. HDAC inhibitors and DNMTs inhibitors have been shown to have synergistic effects in leukemia cells [418,419].

4. Conclusions

Great improvements and success in cancer treatments has been recorded in the past few years mainly due to prevention campaigns, early diagnosis and better therapies. Overall, better and improved cancer patient outcomes have been observed. Even with these observations, millions of cancer patients die each year. One major hurdle to improved cancer patient outcomes is the development of resistance to therapies and disease relapse [3]. Important in cancer relapse is the presence of CSCs, a subpopulation of cancer cells with self-renewal and tumorigenic properties. Whilst many studies and drugs still target all cancer cells within a tumor in order to debulk the tumor, more research is targeted against CSCs. The identification and characterization of CSCs within different tumors can reveal their characteristics and markers that can be used in their elimination. To this end several markers including CD44, ALDH1 and CD133 have been identified in different cancers. Novel strategies including the use of nanotechnology aim to detect, characterize and eliminate CSCs with enhanced efficacy than current methods [420,421]. Currently, several chemotherapeutic agents targeting CSCs are under investigation and some included in clinical trials. CSCs being a small subpopulation of cancer cells may prove to be easily eradicated if targeted properly. Combining CSC surface marker targeting using drugs loaded onto nanomaterials can effectively be used against CSCs [422,423]. Ligands of CSC surface markers allow increased specificity in terms of targeting CSCs and have already been shown to be effective [5,424]. Further research into the solubility and other features of these carriers is underway and is likely to yield better molecules leading to better treatments. To this end, the use of cancer organoids as models of tumors may aid in preclinical studies [30,34]. These new treatment strategies must target the different strategies used by CSCs to survive and promote cancer relapse such as activation of survival signaling pathways, immunosuppression and enhanced metabolic adaptation.

Author Contributions

Conceptualization, K.D., D.A.S. and C.G.; writing—original draft preparation, K.D.; writing—review and editing, K.D., D.A.S., C.G., N.E.T., A.W., and C.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research is supported by funding from the International Centre for Genetic Engineering and Biotechnology (ICGEB), the National Research Foundation (NRF) of South Africa, the Medical Research Council (MRC) of South Africa and the University of Cape Town (UCT). The funders had no role in the conduct of the research or the preparation of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ABCATP-binding cassette
ALDH1aldehyde dehydrogenase 1
ATRAAll-trans retinoic acid
BMI1B-cell-specific Moloney murine leukemia virus integration site 1
BMP-4Bone morphogenic protein 4
CAR TChimeric antigen receptor
CCACholangiocarcinoma
CKcytokeratin
CNScentral nervous system
CRCColorectal Cancer
CSCsCancer stem cells
CTLCytotoxic T lymphocyte
DCLK1Double cortin-like kinase 1;
EMAepithelial membrane antigen;
EMT Epithelial to mesenchymal transition
EpCAMepithelial cell adhesion molecule;
GCSFGranulocyte colony stimulation factor
H3K27me3Histone 3 lysine 27
HDACsHistone deacetylases
HDMsHistone demethylase
HMTsHistone methyltransferase
HNSCChead and neck squamous cell carcinoma
K17Keratin 17
Lgr5Leucine-rich repeat-containing G-protein coupled receptor 5
MDR1Multidrug resistance 1
MGMTO6-methylguanine DNA methyltransferase
MMPsMatrix metalloproteases
PD-1Programmed cell death 1
PD-L1PD-1 ligand
PSAprostate-specific antigen
TMETumor microenvironment
TIMPsTissue inhibitors of metalloproteinases
TMZTemozolomide
ZEB1Zinc finger E-box-binding homeobox 1

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA A Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Torre, L.A.; Siegel, R.L.; Ward, E.M.; Jemal, A. Global Cancer Incidence and Mortality Rates and Trends—An Update. Cancer Epidemiol. Biomark. Prev. 2015, 25, 16–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Senthebane, D.A.; Rowe, A.; Thomford, N.E.; Shipanga, H.; Munro, D.; Al Mazeedi, M.A.M.; Almazyadi, H.A.M.; Kallmeyer, K.; Dandara, C.; Pepper, M.S.; et al. The Role of Tumor Microenvironment in Chemoresistance: To Survive, Keep Your Enemies Closer. Int. J. Mol. Sci. 2017, 18, 1586. [Google Scholar] [CrossRef]
  4. Senthebane, D.A.; Jonker, T.; Rowe, A.; Thomford, N.E.; Munro, D.; Dandara, C.; Wonkam, A.; Govender, D.; Calder, B.; Soares, N.C.; et al. The Role of Tumor Microenvironment in Chemoresistance: 3D Extracellular Matrices as Accomplices. Int. J. Mol. Sci. 2018, 19, 2861. [Google Scholar] [CrossRef] [Green Version]
  5. Al Faraj, A.; Shaik, A.S.; Al Sayed, B.; Halwani, R.; Al Jammaz, I. Specific targeting and noninvasive imaging of breast cancer stem cells using single-walled carbon nanotubes as novel multimodality nanoprobes. Nanomedicine 2016, 11, 31–46. [Google Scholar] [CrossRef] [PubMed]
  6. Fu, Y.; Li, H.; Hao, X. The self-renewal signaling pathways utilized by gastric cancer stem cells. Tumor Boil. 2017, 39. [Google Scholar] [CrossRef] [Green Version]
  7. Humphries, H.N.; Wickremesekera, S.K.; Marsh, R.W.; Brasch, H.D.; Mehrotra, S.; Tan, S.T.; Itinteang, T. Characterization of Cancer Stem Cells in Colon Adenocarcinoma Metastasis to the Liver. Front. Surg. 2018, 4, 76. [Google Scholar] [CrossRef]
  8. Geng, S.-Q.; Alexandrou, A.T.; Li, J.J. Breast cancer stem cells: Multiple capacities in tumor metastasis. Cancer Lett. 2014, 349, 1–7. [Google Scholar] [CrossRef] [Green Version]
  9. Doulatov, S.; Notta, F.; Laurenti, E.; Dick, J.E. Hematopoiesis: A Human Perspective. Cell Stem Cell 2012, 10, 120–136. [Google Scholar] [CrossRef] [Green Version]
  10. Graham, S.M.; Jorgensen, H.G.; Allan, E.; Pearson, C.; Alcorn, M.J.; Richmond, L.; Holyoake, T.L. Primitive, quiescent, Philadelphia-positive stem cells from patients with chronic myeloid leukemia are insensitive to STI571 in vitro. Blood 2002, 99, 319–325. [Google Scholar] [CrossRef]
  11. Holyoake, T.L.; Jiang, X.; Jorgensen, H.G.; Graham, S.; Alcorn, M.J.; Laird, C.; Eaves, A.C.; Eaves, C.J. Primitive quiescent leukemic cells from patients with chronic myeloid leukemia spontaneously initiate factor-independent growth in vitro in association with up-regulation of expression of interleukin-3. Blood 2001, 97, 720–728. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Paget, S. The Distribution of Secondary Growths in Cancer of the Breast. Lancet 1889, 133, 571–573. [Google Scholar] [CrossRef] [Green Version]
  13. Oskarsson, T.; Batlle, E.; Massagué, J. Metastatic stem cells: Sources, niches, and vital pathways. Cell Stem Cell 2014, 14, 306–321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Massagué, J.; Obenauf, A.C. Metastatic colonization by circulating tumour cells. Nature 2016, 529, 298–306. [Google Scholar] [CrossRef] [PubMed]
  15. Valent, P.; Bonnet, D.; De Maria, R.; Lapidot, T.; Copland, M.; Melo, J.V.; Chomienne, C.; Ishikawa, F.; Schuringa, J.J.; Stassi, G.; et al. Cancer stem cell definitions and terminology: The devil is in the details. Nat. Rev. Cancer 2012, 12, 767–775. [Google Scholar] [CrossRef]
  16. Leon, G.; MacDonagh, L.; Finn, S.P.; Cuffe, S.; Barr, M.P. Cancer stem cells in drug resistant lung cancer: Targeting cell surface markers and signaling pathways. Pharmacol. Ther. 2016, 158, 71–90. [Google Scholar] [CrossRef]
  17. Maugeri-Saccà, M.; Bartucci, M.; De Maria, R. DNA Damage Repair Pathways in Cancer Stem Cells. Mol. Cancer Ther. 2012, 11, 1627–1636. [Google Scholar] [CrossRef] [Green Version]
  18. Maugeri-Saccà, M.; Vigneri, P.; De Maria, R. Cancer stem cells and chemosensitivity. Clin. Cancer Res. 2011, 17, 4942–4947. [Google Scholar] [CrossRef] [Green Version]
  19. Turdo, A.; Veschi, V.; Gaggianesi, M.; Chinnici, A.; Bianca, P.; Todaro, M.; Stassi, G. Meeting the Challenge of Targeting Cancer Stem Cells. Front. Cell Dev. Boil. 2019, 7, 16. [Google Scholar] [CrossRef] [Green Version]
  20. 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]
  21. 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]
  22. 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] [Green Version]
  23. Charafe-Jauffret, E.; Ginestier, C.; Iovino, F.; Wicinski, J.; Cervera, N.; Finetti, P.; Hur, M.-H.; Diebel, M.E.; Monville, F.; Dutcher, J.; et al. Breast cancer cell lines contain functional cancer stem cells with metastatic capacity and a distinct molecular signature. Cancer Res. 2009, 69, 1302–1313. [Google Scholar] [CrossRef] [Green Version]
  24. Eramo, A.; Lotti, F.; Sette, G.; Pilozzi, E.; Biffoni, M.; Di Virgilio, A.; Conticello, C.; Ruco, L.; Peschle, C.; De Maria, R. Identification and expansion of the tumorigenic lung cancer stem cell population. Cell Death Differ. 2007, 15, 504–514. [Google Scholar] [CrossRef] [PubMed]
  25. Agliano, A.; Calvo, A.; Box, C. The challenge of targeting cancer stem cells to halt metastasis. Semin. Cancer Boil. 2017, 44, 25–42. [Google Scholar] [CrossRef]
  26. Nguyen, L.V.; Vanner, R.; Dirks, P.; Eaves, C.J. Cancer stem cells: An evolving concept. Nat. Rev. Cancer 2012, 12, 133–143. [Google Scholar] [CrossRef] [PubMed]
  27. Aartsma-Rus, A.; Van Putten, M. The use of genetically humanized animal models for personalized medicine approaches. Dis. Model. Mech. 2019, 13, dmm041673. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Agarwal, Y.; Beatty, C.; Biradar, S.; Castronova, I.; Ho, S.; Melody, K.; Bility, M.T. Moving beyond the mousetrap: Current and emerging humanized mouse and rat models for investigating prevention and cure strategies against HIV infection and associated pathologies. Retrovirology 2020, 17, 1–11. [Google Scholar] [CrossRef] [Green Version]
  29. Mehlen, P.; Puisieux, A. Metastasis: A question of life or death. Nat. Rev. Cancer 2006, 6, 449–458. [Google Scholar] [CrossRef]
  30. Dzobo, K.; Senthebane, D.A.; Rowe, A.; Thomford, N.E.; Mwapagha, L.M.; Al-Awwad, N.; Dandara, C.; Parker, M.I. Cancer Stem Cell Hypothesis for Therapeutic Innovation in Clinical Oncology? Taking the Root Out, Not Chopping the Leaf. OMICS A J. Integr. Boil. 2016, 20, 681–691. [Google Scholar] [CrossRef]
  31. Bozorgi, A.; Khazaei, M.; Khazaei, M.R. New Findings on Breast Cancer Stem Cells: A Review. J. Breast Cancer 2015, 18, 303–312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Singh, S.R. Gastric cancer stem cells: A novel therapeutic target. Cancer Lett. 2013, 338, 110–119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. MacDonagh, L.; Gray, S.G.; Breen, E.; Cuffe, S.; Finn, S.P.; O’Byrne, K.J.; Barr, M.P. Lung cancer stem cells: The root of resistance. Cancer Lett. 2016, 372, 147–156. [Google Scholar] [CrossRef]
  34. Dzobo, K.; Rowe, A.; Senthebane, D.A.; Almazyadi, M.A.; Patten, V.; Parker, M.I. Three-Dimensional Organoids in Cancer Research: The Search for the Holy Grail of Preclinical Cancer Modeling. OMICS J. Integr. Boil. 2018, 22, 733–748. [Google Scholar] [CrossRef] [PubMed]
  35. Ortiz-Sánchez, E.; Santiago-López, L.; Cruz-Domínguez, V.B.; Toledo-Guzmán, M.E.; Hernández-Cueto, D.; Muñiz-Hernández, S.; Garrido, E.; De León, D.C.; García-Carrancá, A. Characterization of cervical cancer stem cell-like cells: Phenotyping, stemness, and human papilloma virus co-receptor expression. Oncotarget 2016, 7, 31943–31954. [Google Scholar] [CrossRef] [Green Version]
  36. Hou, T.; Zhang, W.; Tong, C.; Kazobinka, G.; Huang, X.; Huang, Y.; Zhang, Y. Putative stem cell markers in cervical squamous cell carcinoma are correlated with poor clinical outcome. BMC Cancer 2015, 15, 785. [Google Scholar] [CrossRef] [Green Version]
  37. Tyagi, A.; Vishnoi, K.; Mahata, S.; Verma, G.; Srivastava, Y.; Masaldan, S.; Roy, B.G.; Bharti, A.C.; Das, B.C. Cervical Cancer Stem Cells Selectively Overexpress HPV Oncoprotein E6 that Controls Stemness and Self-Renewal through Upregulation of HES1. Clin. Cancer Res. 2016, 22, 4170–4184. [Google Scholar] [CrossRef] [Green Version]
  38. Ming, X.-Y.; Fu, L.; Zhang, L.-Y.; Qin, Y.-R.; Cao, T.-T.; Chan, K.W.; Ma, S.K.Y.; Xie, D.; Guan, X.-Y. Integrin α7 is a functional cancer stem cell surface marker in oesophageal squamous cell carcinoma. Nat. Commun. 2016, 7, 13568. [Google Scholar] [CrossRef] [Green Version]
  39. Zhao, J.-S.; Li, W.-J.; Ge, D.; Zhang, P.-J.; Li, J.-J.; Lu, C.-L.; Ji, X.-D.; Guan, D.-X.; Gao, H.; Xu, L.-Y.; et al. Tumor Initiating Cells in Esophageal Squamous Cell Carcinomas Express High Levels of CD44. PLoS ONE 2011, 6, e21419. [Google Scholar] [CrossRef] [Green Version]
  40. Yuan, Z.-X.; Mo, J.; Zhao, G.; Shu, G.; Fu, H.-L.; Zhao, W. Targeting Strategies for Renal Cell Carcinoma: From Renal Cancer Cells to Renal Cancer Stem Cells. Front. Pharmacol. 2016, 7, 423. [Google Scholar] [CrossRef] [Green Version]
  41. Peired, A.; Sisti, A.; Romagnani, P. Renal Cancer Stem Cells: Characterization and Targeted Therapies. Stem Cells Int. 2016, 2016, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Cheng, B.; Yang, G.; Jiang, R.; Cheng, Y.; Yang, H.; Pei, L.; Qiu, X. Cancer stem cell markers predict a poor prognosis in renal cell carcinoma: A meta-analysis. Oncotarget 2016, 7, 65862–65875. [Google Scholar] [CrossRef] [PubMed]
  43. Fan, F.; Bellister, S.; Lü, J.; Ye, X.; Boulbès, D.R.; Tozzi, F.; Sceusi, E.; Kopetz, S.; Tian, F.; Xia, L.; et al. The requirement for freshly isolated human colorectal cancer (CRC) cells in isolating CRC stem cells. Br. J. Cancer 2014, 112, 539–546. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Dalerba, P.; Kalisky, T.; Sahoo, D.; Rajendran, P.S.; Rothenberg, M.E.; Leyrat, A.A.; Sim, S.; Okamoto, J.; Johnston, D.M.; Qian, D.; et al. Single-cell dissection of transcriptional heterogeneity in human colon tumors. Nat. Biotechnol. 2011, 29, 1120–1127. [Google Scholar] [CrossRef] [PubMed]
  45. Yeung, T.M.; Gandhi, S.C.; Wilding, J.L.; Muschel, R.; Bodmer, W.F.B.J. Cancer stem cells from colorectal cancer-derived cell lines. Proc. Nat. Acad. Sci. USA 2010, 107, 3722–3727. [Google Scholar] [CrossRef] [Green Version]
  46. Kemper, K.; Prasetyanti, P.R.; De Lau, W.; Rodermond, H.; Clevers, H.; Medema, J.P. Monoclonal Antibodies Against Lgr5 Identify Human Colorectal Cancer Stem Cells. Stem Cells 2012, 30, 2378–2386. [Google Scholar] [CrossRef]
  47. Yang, Z.F.; Ho, D.W.; Ng, M.N.; Lau, C.K.; Yu, W.C.; Ngai, P.; Chu, P.W.; Lam, C.T.; Poon, R.T.; Fan, S.T. Significance of CD90+ Cancer Stem Cells in Human Liver Cancer. Cancer Cell 2008, 13, 153–166. [Google Scholar] [CrossRef] [Green Version]
  48. Kimura, O.; Takahashi, T.; Ishii, N.; Inoue, Y.; Ueno, Y.; Kogure, T.; Fukushima, K.; Shiina, M.; Yamagiwa, Y.; Kondo, Y.; et al. Characterization of the epithelial cell adhesion molecule (EpCAM) + cell population in hepatocellular carcinoma cell lines. Cancer Sci. 2010, 101, 2145–2155. [Google Scholar] [CrossRef]
  49. Nie, S.; McDermott, S.P.; Deol, Y.; Tan, Z.; Wicha, M.S.; Lubman, D.M. A quantitative proteomics analysis of MCF7 breast cancer stem and progenitor cell populations. Proteomics. 2015, 15, 3772–3783. [Google Scholar] [CrossRef] [Green Version]
  50. A Theodoropoulos, P.; Chiotaki, R.; Polioudaki, H. Stem cell technology in breast cancer: Current status and potential applications. Stem Cells Cloning Adv. Appl. 2016, 9, 17–29. [Google Scholar] [CrossRef] [Green Version]
  51. Takaishi, S.; Okumura, T.; Tu, S.; Wang, S.S.; Shibata, W.; Vigneshwaran, R.; Gordon, S.A.; Shimada, Y.; Wang, T.C. Identification of gastric cancer stem cells using the cell surface marker CD44. Stem Cells 2009, 27, 1006–1020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Zhang, C.; Li, C.; He, F.; Cai, Y.; Yang, H. Identification of CD44+CD24+ gastric cancer stem cells. J. Cancer Res. Clin. Oncol. 2011, 137, 1679–1686. [Google Scholar] [CrossRef] [PubMed]
  53. Mao, J.; Fan, S.; Ma, W.; Fan, P.; Wang, B.; Zhang, J.; Wang, H.; Tang, B.; Zhang, Q.; Yu, X.; et al. Roles of Wnt/β-catenin signaling in the gastric cancer stem cells proliferation and salinomycin treatment. Cell Death Dis. 2014, 5, e1039. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Beier, D.; Hau, P.; Proescholdt, M.; Lohmeier, A.; Wischhusen, J.; Oefner, P.J.; Aigner, L.; Brawanski, A.; Bogdahn, U.; Beier, C.P. CD133+ and CD133− glioblastoma-derived cancer stem cells show differential growth characteristics and molecular profiles. Cancer Res. 2007, 67, 4010–4015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Yan, X.; Ma, L.; Yi, D.; Yoon, J.-G.; Diercks, A.; Foltz, G.; Price, N.D.; Hood, L.E.; Tian, Q. A CD133-related gene expression signature identifies an aggressive glioblastoma subtype with excessive mutations. Proc. Nat. Acad. Sci. USA 2011, 108, 1591–1596. [Google Scholar] [CrossRef] [Green Version]
  56. Wang, J.C.Y.; Dick, J.E. Cancer stem cells: Lessons from leukemia. Trends Cell Boil. 2005, 15, 494–501. [Google Scholar] [CrossRef]
  57. Afify, S.M.M.; Hassan, G.; Osman, A.; Calle, A.S.; Nawara, H.M.; Zahra, M.H.; El-Ghlban, S.; Mansour, H.; Alam, J.; Abu Quora, H.A.; et al. Metastasis of Cancer Stem Cells Developed in the Microenvironment of Hepatocellular Carcinoma. Bioengineering 2019, 6, 73. [Google Scholar] [CrossRef] [Green Version]
  58. Hope, K.J.; Jin, L.; Dick, J.E. Acute myeloid leukemia originates from a hierarchy of leukemic stem cell classes that differ in self-renewal capacity. Nat. Immunol. 2004, 5, 738–743. [Google Scholar] [CrossRef]
  59. Inoue, A.; Kobayashi, C.I.; Shinohara, H.; Miyamoto, K.; Yamauchi, N.; Yuda, J.; Akao, Y.; Minami, Y. Chronic myeloid leukemia stem cells and molecular target therapies for overcoming resistance and disease persistence. Int. J. Hematol. 2018, 108, 365–370. [Google Scholar] [CrossRef] [Green Version]
  60. Corbin, A.S.; Agarwal, A.; Loriaux, M.; Cortes, J.; Deininger, M.W.; Druker, B.J. Human chronic myeloid leukemia stem cells are insensitive to imatinib despite inhibition of BCR-ABL activity. J. Clin. Investig. 2011, 121, 396–409. [Google Scholar] [CrossRef] [Green Version]
  61. Foster, R.; Buckanovich, R.J.; Rueda, B.R. Ovarian cancer stem cells: Working towards the root of stemness. Cancer Lett. 2013, 338, 147–157. [Google Scholar] [CrossRef] [PubMed]
  62. Burgos-Ojeda, D.; Rueda, B.R.; Buckanovich, R.J. Ovarian cancer stem cell markers: Prognostic and therapeutic implications. Cancer Lett. 2012, 322, 1–7. [Google Scholar] [CrossRef] [Green Version]
  63. Collins, A.T.; Maitland, N.J. Prostate cancer stem cells. Eur. J. Cancer 2006, 42, 1213–1218. [Google Scholar] [CrossRef]
  64. Chen, X.; Li, Q.; Liu, X.; Liu, C.; Liu, R.; Rycaj, K.; Zhang, D.; Liu, B.; Jeter, C.; Calhoun-Davis, T.; et al. Defining a Population of Stem-like Human Prostate Cancer Cells That Can Generate and Propagate Castration-Resistant Prostate Cancer. Clin. Cancer Res. 2016, 22, 4505–4516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Liu, X.; Chen, X.; Rycaj, K.; Chao, H.-P.; Deng, Q.; Jeter, C.R.; Liu, C.; Honorio, S.; Li, H.; Davis, T.; et al. Systematic dissection of phenotypic, functional, and tumorigenic heterogeneity of human prostate cancer cells. Oncotarget 2015, 6, 23959–23986. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Hermann, P.C.; Huber, S.L.; Herrler, T.; Aicher, A.; Ellwart, J.W.; Guba, M.; Bruns, C.J.; Heeschen, C. Distinct Populations of Cancer Stem Cells Determine Tumor Growth and Metastatic Activity in Human Pancreatic Cancer. Cell Stem Cell 2007, 1, 313–323. [Google Scholar] [CrossRef] [Green Version]
  67. Lee, C.J.; Dosch, J.; Simeone, D.M. Pancreatic Cancer Stem Cells. J. Clin. Oncol. 2008, 26, 2806–2812. [Google Scholar] [CrossRef]
  68. Li, C.; Lee, C.J.; Simeone, D.M. Identification of Human Pancreatic Cancer Stem Cells. In Advanced Structural Safety Studies; Springer Science and Business Media LLC: Berlin/Heidelberg, Germany, 2009; Volume 568, pp. 161–173. [Google Scholar]
  69. La Porta, C.A.M. Cancer Stem Cells: Lessons from Melanoma. Stem Cell Rev. Rep. 2008, 5, 61–65. [Google Scholar] [CrossRef]
  70. Kumar, D.; Gorain, M.; Kundu, G.; Kundu, G.C. Therapeutic implications of cellular and molecular biology of cancer stem cells in melanoma. Mol. Cancer 2017, 16, 7. [Google Scholar] [CrossRef] [Green Version]
  71. Chinn, S.B.; Darr, O.A.; Owen, J.H.; Bellile, E.; McHugh, J.B.; Spector, M.E.; Papagerakis, S.M.; Chepeha, U.B.; Bradford, C.R.; Carey, T.E.; et al. Cancer stem cells: Mediators of tumorigenesis and metastasis in head and neck squamous cell carcinoma. Head Neck 2014, 37, 317–326. [Google Scholar] [CrossRef] [Green Version]
  72. Kim, J.; Shin, J.H.; Chen, C.-H.; Cruz, L.; Farnebo, L.; Yang, J.; Borges, P.; Kang, G.; Mochly-Rosen, D.; Sunwoo, J.B. Targeting aldehyde dehydrogenase activity in head and neck squamous cell carcinoma with a novel small molecule inhibitor. Oncotarget 2017, 8, 52345–52356. [Google Scholar] [CrossRef] [Green Version]
  73. Lan, J.; Huang, B.; Liu, R.; Ju, X.; Zhou, Y.; Jiang, J.; Liang, W.; Shen, Y.; Li, F.; Pang, L. Expression of cancer stem cell markers and their correlation with pathogenesis in vascular tumors. Int. J. Clin. Exp. Pathol. 2015, 8, 12621–12633. [Google Scholar]
  74. Veselska, R.; Skoda, J.; Neradil, J. Detection of cancer stem cell markers in sarcomas. Klin. Onkol. 2012, 25, 16–20. [Google Scholar]
  75. Visvader, J.E.; Lindeman, G.J. Cancer Stem Cells: Current Status and Evolving Complexities. Cell Stem Cell 2012, 10, 717–728. [Google Scholar] [CrossRef] [Green Version]
  76. Miyoshi, N.; Mizushima, T.; Doki, Y.; Mori, M. Cancer stem cells in relation to treatment. Jpn. J. Clin. Oncol. 2018, 49, 232–237. [Google Scholar] [CrossRef]
  77. Toh, T.B.; Lim, J.J.; Chow, E.K.-H. Epigenetics in cancer stem cells. Mol. Cancer 2017, 16, 29. [Google Scholar] [CrossRef] [Green Version]
  78. Dawood, S.; Austin, L.; Cristofanilli, M. Cancer stem cells: Implications for cancer therapy. Oncology (Williston Park. N. Y.) 2014, 28, 1101–1107. [Google Scholar]
  79. Steinbichler, T.B.; Dudas, J.; Skvortsov, S.; Ganswindt, U.; Riechelmann, H.; Skvortsova, I.-I. Therapy resistance mediated by cancer stem cells. Semin. Cancer Boil. 2018, 53, 156–167. [Google Scholar] [CrossRef]
  80. Dymock, B.W. The rise of epigenetic drug discovery. Futur. Med. Chem. 2016, 8, 1523–1524. [Google Scholar] [CrossRef]
  81. Hsu, L.C.; Chang, W.C.; Hoffmann, I.; Duester, G. Molecular analysis of two closely related mouse aldehyde dehydrogenase genes: Identification of a role for Aldh1, but not Aldh-pb, in the biosynthesis of retinoic acid. Biochem. J. 1999, 339, 387–395. [Google Scholar] [CrossRef]
  82. Yoshida, A.; Rzhetsky, A.; Hsu, L.C.; Chang, C. Human aldehyde dehydrogenase gene family. JBIC J. Boil. Inorg. Chem. 1998, 251, 549–557. [Google Scholar] [CrossRef]
  83. Eirew, P.; Kannan, N.; Knapp, D.J.H.F.; Vailllant, F.; Emerman, J.T.; Lindeman, G.J.; Visvader, J.E.; Eaves, C.J. Aldehyde Dehydrogenase Activity Is a Biomarker of Primitive Normal Human Mammary Luminal Cells. Stem Cells 2012, 30, 344–348. [Google Scholar] [CrossRef]
  84. Ioannou, M.; Serafimidis, I.; Arnes, L.; Sussel, L.; Singh, S.; Vasiliou, V.; Gavalas, A. ALDH1B1 is a potential stem/progenitor marker for multiple pancreas progenitor pools. Dev. Boil. 2012, 374, 153–163. [Google Scholar] [CrossRef] [Green Version]
  85. Singh, S.; Brocker, C.; Koppaka, V.; Chen, Y.; Jackson, B.C.; Matsumoto, A.; Thompson, D.C.; Vasiliou, V. Aldehyde Dehydrogenases in Cellular Responses to Oxidative/electrophilic Stress. Free Radic. Boil. Med. 2012, 56, 89–101. [Google Scholar] [CrossRef] [Green Version]
  86. Vassalli, G. Aldehyde Dehydrogenases: Not Just Markers, but Functional Regulators of Stem Cells. Stem Cells Int. 2019, 2019, 1–15. [Google Scholar] [CrossRef] [Green Version]
  87. Ciccone, V.; Terzuoli, E.; Donnini, S.; Giachetti, A.; Morbidelli, L.; Ziche, M. Stemness marker ALDH1A1 promotes tumor angiogenesis via retinoic acid/HIF-1α/VEGF signalling in MCF-7 breast cancer cells. J. Exp. Clin. Cancer Res. 2018, 37, 311. [Google Scholar] [CrossRef]
  88. Vogler, T.; Kriegl, L.; Horst, D.; Engel, J.; Sagebiel, S.; Schäffauer, A.J.; Kirchner, T.; Jung, A. The expression pattern of aldehyde dehydrogenase 1 (ALDH1) is an independent prognostic marker for low survival in colorectal tumors. Exp. Mol. Pathol. 2012, 92, 111–117. [Google Scholar] [CrossRef]
  89. Hoogen, C.V.D.; Van Der Horst, G.; Cheung, H.; Buijs, J.T.; Lippitt, J.M.; Hamdy, F.C.; Eaton, C.L.; Thalmann, G.N.; Cecchini, M.G.; Pelger, R.C.; et al. High Aldehyde Dehydrogenase Activity Identifies Tumor-Initiating and Metastasis-Initiating Cells in Human Prostate Cancer. Cancer Res. 2010, 70, 5163–5173. [Google Scholar] [CrossRef] [Green Version]
  90. Ueda, K.; Ogasawara, S.; Akiba, J.; Nakayama, M.; Todoroki, K.; Ueda, K.; Sanada, S.; Suekane, S.; Noguchi, M.; Matsuoka, K.; et al. Aldehyde Dehydrogenase 1 Identifies Cells with Cancer Stem Cell-Like Properties in a Human Renal Cell Carcinoma Cell Line. PLoS ONE 2013, 8, e75463. [Google Scholar] [CrossRef] [Green Version]
  91. Ginestier, C.; Hur, M.H.; Charafe-Jauffret, E.; Monville, F.; Dutcher, J.; Brown, M.; Jacquemier, J.; Viens, P.; Kleer, C.G.; Liu, S.; et al. ALDH1 Is a Marker of Normal and Malignant Human Mammary Stem Cells and a Predictor of Poor Clinical Outcome. Cell Stem Cell 2007, 1, 555–567. [Google Scholar] [CrossRef] [Green Version]
  92. Ma, S.K.Y.; Lee, T.K.; Zheng, B.-J.; Chan, K.W.; Guan, X.-Y.; Lee, T.K.W. CD133+ HCC cancer stem cells confer chemoresistance by preferential expression of the Akt/PKB survival pathway. Oncogene 2007, 27, 1749–1758. [Google Scholar] [CrossRef] [Green Version]
  93. Jin, X.; Zhao, Y.; Qian, J.; Tang, J.; Zhan, X.-D. Aldehyde dehydrogenase 1 can be used as a new marker of cancer stem cells in laryngeal cancer cells in vitro. Zhonghua Zhong Liu Za Zhi (Chin. J. Oncol.) 2011, 33, 900–904. [Google Scholar]
  94. Afify, S.M.M.; Seno, M. Conversion of Stem Cells to Cancer Stem Cells: Undercurrent of Cancer Initiation. Cancers 2019, 11, 345. [Google Scholar] [CrossRef] [Green Version]
  95. Tanei, T.; Morimoto, K.; Shimazu, K.; Kim, S.J.; Tanji, Y.; Taguchi, T.; Tamaki, Y.; Noguchi, S. Association of Breast Cancer Stem Cells Identified by Aldehyde Dehydrogenase 1 Expression with Resistance to Sequential Paclitaxel and Epirubicin-Based Chemotherapy for Breast Cancers. Clin. Cancer Res. 2009, 15, 4234–4241. [Google Scholar] [CrossRef] [Green Version]
  96. Morimoto, K.; Kim, S.J.; Tanei, T.; Shimazu, K.; Tanji, Y.; Taguchi, T.; Tamaki, Y.; Terada, N.; Noguchi, S. Stem cell marker aldehyde dehydrogenase 1-positive breast cancers are characterized by negative estrogen receptor, positive human epidermal growth factor receptor type 2, and high Ki67 expression. Cancer Sci. 2009, 100, 1062–1068. [Google Scholar] [CrossRef]
  97. Rausch, V.; Liu, L.; Kallifatidis, G.; Baumann, B.; Mattern, J.; Gladkich, J.; Wirth, T.; Schemmer, P.; Buchler, 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]
  98. Yip, N.C.; Fombon, I.S.; Liu, P.; Brown, S.; Kannappan, V.; Armesilla, A.L.; Xu, B.; Cassidy, J.; Darling, J.L.; Wang, W. Disulfiram modulated ROS–MAPK and NFκB pathways and targeted breast cancer cells with cancer stem cell-like properties. Br. J. Cancer 2011, 104, 1564–1574. [Google Scholar] [CrossRef] [Green Version]
  99. Locher, K.P. Mechanistic diversity in ATP-binding cassette (ABC) transporters. Nat. Struct. Mol. Boil. 2016, 23, 487–493. [Google Scholar] [CrossRef] [Green Version]
  100. Begicevic, R.-R.; Falasca, M. ABC Transporters in Cancer Stem Cells: Beyond Chemoresistance. Int. J. Mol. Sci. 2017, 18, 2362. [Google Scholar] [CrossRef] [Green Version]
  101. Dean, M.; Fojo, T.; Bates, S. Tumour stem cells and drug resistance. Nat. Rev. Cancer 2005, 5, 275–284. [Google Scholar] [CrossRef]
  102. Lario, A.P.; García, C.B.; Elizondo, M.E.; Lobo, C. Expression of proteins associated with multidrug resistance and resistance to chemotherapy in lung cancer. Arch. Bronconeumol. (Engl. Ed.) 2007, 43, 479–484. [Google Scholar] [CrossRef]
  103. Leonessa, F.; Clarke, R. ATP binding cassette transporters and drug resistance in breast cancer. Endocr. Relat. Cancer 2003, 10, 43–73. [Google Scholar] [CrossRef] [Green Version]
  104. Boesch, M.; Zeimet, A.G.; Rumpold, H.; Gastl, G.; Sopper, S.; Wolf, D. Drug Transporter-Mediated Protection of Cancer Stem Cells from Ionophore Antibiotics. Stem Cells Transl. Med. 2015, 4, 1028–1032. [Google Scholar] [CrossRef]
  105. Xiong, B.; Ma, L.; Hu, X.; Zhang, C.; Cheng, Y. Characterization of side population cells isolated from the colon cancer cell line SW480. Int. J. Oncol. 2014, 45, 1175–1183. [Google Scholar] [CrossRef] [Green Version]
  106. Wright, M.H.; Calcagno, A.M.; Salcido, C.D.; Carlson, M.D.; Ambudkar, S.V.; Varticovski, L. Brca1 breast tumors contain distinct CD44+/CD24- and CD133+ cells with cancer stem cell characteristics. Breast Cancer Res. 2008, 10, R10. [Google Scholar] [CrossRef] [Green Version]
  107. Frank, M.H.; Margaryan, A.; Sadée, W.; Huang, Y.; Schatton, T.; Waaga-Gasser, A.M.; Waaga-Gasser, A.M.; Sayegh, M.H. ABCB5-Mediated Doxorubicin Transport and Chemoresistance in Human Malignant Melanoma. Cancer Res. 2005, 65, 4320–4333. [Google Scholar] [CrossRef] [Green Version]
  108. Shi, G.-M.; Xu, Y.; Fan, J.; Zhou, J.; Yang, X.-R.; Qiu, S.-J.; Liao, Y.; Wu, W.-Z.; Ji, Y.; Ke, A.-W.; et al. Identification of side population cells in human hepatocellular carcinoma cell lines with stepwise metastatic potentials. J. Cancer Res. Clin. Oncol. 2008, 134, 1155–1163. [Google Scholar] [CrossRef]
  109. Moitra, K. Overcoming Multidrug Resistance in Cancer Stem Cells. BioMed Res. Int. 2015, 2015, 1–8. [Google Scholar] [CrossRef] [Green Version]
  110. Lou, H.; Dean, M. Targeted therapy for cancer stem cells: The patched pathway and ABC transporters. Oncogene 2007, 26, 1357–1360. [Google Scholar] [CrossRef] [Green Version]
  111. Marcelletti, J.F.; Multani, P.S.; Lancet, J.E.; Baer, M.R.; Sikic, B.I. Leukemic blast and natural killer cell P-glycoprotein function and inhibition in a clinical trial of zosuquidar infusion in acute myeloid leukemia. Leuk. Res. 2009, 33, 769–774. [Google Scholar] [CrossRef]
  112. Tang, R.; Faussat, A.-M.; Perrot, J.-Y.; Marjanovic, Z.; Cohen, S.; Storme, T.; Morjani, H.; Legrand, O.; Marie, J.-P. Zosuquidar restores drug sensitivity in P-glycoprotein expressing acute myeloid leukemia (AML). BMC Cancer 2008, 8, 51. [Google Scholar] [CrossRef] [Green Version]
  113. Hanahan, D.; Weinberg, R.A. The Hallmarks of Cancer. Cell 2000, 100, 57–70. [Google Scholar] [CrossRef] [Green Version]
  114. Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The Next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
  115. Sun, H.-R.; Wang, S.; Yan, S.-C.; Zhang, Y.; Nelson, P.J.; Jia, H.-L.; Qin, L.-X.; Dong, Q.-Z. Therapeutic Strategies Targeting Cancer Stem Cells and Their Microenvironment. Front. Oncol. 2019, 9, 9. [Google Scholar] [CrossRef]
  116. Strasser, A.; Harris, A.W.; Bath, M.L.; Cory, S. Novel primitive lymphoid tumours induced in transgenic mice by cooperation between myc and bcl-2. Nature 1990, 348, 331–333. [Google Scholar] [CrossRef]
  117. Todaro, M.; Alea, M.P.; Di Stefano, A.B.; Cammareri, P.; Vermeulen, L.; Iovino, F.; Tripodo, C.; Russo, A.; Gulotta, G.; Medema, J.P.; et al. Colon Cancer Stem Cells Dictate Tumor Growth and Resist Cell Death by Production of Interleukin-4. Cell Stem Cell 2007, 1, 389–402. [Google Scholar] [CrossRef] [Green Version]
  118. Sun, Q.; Wang, Y.; Desgrosellier, J.S. Combined Bcl-2/Src inhibition synergize to deplete stem-like breast cancer cells. Cancer let. 2019, 457, 40–46. [Google Scholar] [CrossRef] [PubMed]
  119. Viale, A.; De Franco, F.; Orleth, A.; Cambiaghi, V.; Giuliani, V.; Bossi, D.; Ronchini, C.; Ronzoni, S.; Muradore, I.; Monestiroli, S.; et al. Cell-cycle restriction limits DNA damage and maintains self-renewal of leukaemia stem cells. Nature 2009, 457, 51–56. [Google Scholar] [CrossRef] [PubMed]
  120. Gascoigne, K.E.; Taylor, S.S.; Sridharan, D.; Brown, M.; Lambert, W.C.; McMahon, L.W. How do anti-mitotic drugs kill cancer cells? J. Cell Sci. 2009, 122, 2579–2585. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Mccord, A.M.; Jamal, M.; Williams, E.S.; Camphausen, K.; Tofilon, P.J. CD133+ glioblastoma stem-like cells are radiosensitive with a defective DNA damage response compared with established cell lines. Clin. Cancer Res. 2009, 15, 5145–5153. [Google Scholar] [CrossRef] [Green Version]
  122. Gallmeier, E.; Hermann, P.C.; Mueller, M.-T.; Machado, J.G.; Ziesch, A.; De Toni, E.N.; Palagyi, A.; Eisen, C.; Ellwart, J.W.; Rivera, J.; et al. Inhibition of Ataxia Telangiectasia- and Rad3 -Related Function Abrogates the In Vitro and In Vivo Tumorigenicity of Human Colon Cancer Cells Through Depletion of the CD133+ Tumor-Initiating Cell Fraction. Stem Cells 2011, 29, 418–429. [Google Scholar] [CrossRef] [PubMed]
  123. Bao, S.; Wu, Q.; McLendon, R.E.; Hao, Y.; Shi, Q.; Hjelmeland, A.B.; Dewhirst, M.W.; Bigner, D.D.; Rich, J.N. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature 2006, 444, 756–760. [Google Scholar] [CrossRef] [PubMed]
  124. Cohen, A.A.; Geva-Zatorsky, N.; Eden, E.; Morgenstern, M.F.; Issaeva, I.; Sigal, A.; Milo, R.; Cohen-Saidon, C.; Liron, Y.; Kam, Z.; et al. Dynamic Proteomics of Individual Cancer Cells in Response to a Drug. Science 2008, 322, 1511–1516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Raha, D.; Wilson, T.R.; Peng, J.; Peterson, D.; Yue, P.; Evangelista, M.; Wilson, C.; Merchant, M.; Settleman, J. The Cancer Stem Cell Marker Aldehyde Dehydrogenase Is Required to Maintain a Drug-Tolerant Tumor Cell Subpopulation. Cancer Res. 2014, 74, 3579–3590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Rizzo, S.; Hersey, J.M.; Mellor, P.; Dai, W.; Santos-Silva, A.; Liber, D.; Luk, L.; Titley, I.; Carden, C.P.; Box, G.; et al. Ovarian cancer stem cell like side populations are enriched following chemotherapy and overexpress EZH2. Mol. Cancer Ther. 2011, 10, 325–335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Levina, V.; Marrangoni, A.M.; Demarco, R.; Gorelik, E.; Lokshin, A.E. Drug-Selected Human Lung Cancer Stem Cells: Cytokine Network, Tumorigenic and Metastatic Properties. PLoS ONE 2008, 3, e3077. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Chen, J.; Li, Y.; Yu, T.-S.; McKay, R.M.; Burns, D.K.; Kernie, S.G.; Parada, L.F. A restricted cell population propagates glioblastoma growth after chemotherapy. Nature 2012, 488, 522–526. [Google Scholar] [CrossRef] [Green Version]
  129. Qiu, Z.-K.; Shen, N.; Chen, Y.-S.; Yang, Q.-Y.; Guo, C.-C.; Feng, B.-H.; Chen, Z.-P. Enhanced MGMT expression contributes to temozolomide resistance in glioma stem-like cells. Chin. J. Cancer 2014, 33, 115–122. [Google Scholar] [CrossRef]
  130. Kurtova, A.V.; Xiao, J.; Mo, Q.; Pazhanisamy, S.; Krasnow, R.E.; Lerner, S.P.; Chen, F.; Roh, T.; Lay, E.; Ho, P.L.; et al. Blocking PGE2-induced tumour repopulation abrogates bladder cancer chemoresistance. Nature 2014, 517, 209–213. [Google Scholar] [CrossRef]
  131. Saito, Y.; Uchida, N.; Tanaka, S.; Suzuki, N.; Tomizawa-Murasawa, M.; Sone, A.; Najima, Y.; Takagi, S.; Aoki, Y.; Wake, A.; et al. Induction of cell cycle entry eliminates human leukemia stem cells in a mouse model of AML. Nat. Biotechnol. 2010, 28, 275–280. [Google Scholar] [CrossRef] [Green Version]
  132. Lombardo, Y.; Scopelliti, A.; Cammareri, P.; Todaro, M.; Iovino, F.; Vitiani, L.R.; Gulotta, G.; Dieli, F.; De Maria, R.; Stassi, G. Bone Morphogenetic Protein 4 Induces Differentiation of Colorectal Cancer Stem Cells and Increases Their Response to Chemotherapy in Mice. Gastroenterology 2011, 140, 297–309.e6. [Google Scholar] [CrossRef] [PubMed]
  133. Wang, J.-Y.; Chang, C.-C.; Chiang, C.-C.; Chen, W.-M.; Hung, S.-C. Silibinin suppresses the maintenance of colorectal cancer stem-like cells by inhibiting PP2A/AKT/mTOR pathways. J. Cell. Biochem. 2011, 113, 1733–1743. [Google Scholar] [CrossRef] [PubMed]
  134. Nowak, D.; Stewart, D.; Koeffler, H.P. Differentiation therapy of leukemia: 3 decades of development. Blood 2009, 113, 3655–3665. [Google Scholar] [CrossRef] [PubMed]
  135. Zhou, G.; Zhang, J.; Wang, Z.-Y.; Chen, S.-J.; Chen, Z. Treatment of acute promyelocytic leukaemia with all-trans retinoic acid and arsenic trioxide: A paradigm of synergistic molecular targeting therapy. Philos. Trans. R Soc. B Boil. Sci. 2007, 362, 959–971. [Google Scholar] [CrossRef] [Green Version]
  136. Hanahan, D.; Folkman, J. Patterns and Emerging Mechanisms of the Angiogenic Switch during Tumorigenesis. Cell 1996, 86, 353–364. [Google Scholar] [CrossRef] [Green Version]
  137. Pour, L.; Hájek, R.; Buchler, T.; Maisnar, V.; Smolej, L. Angiogenesis and antiangiogenic cancer therapy. Vnitrni Lek. 2004, 50, 930–938. [Google Scholar]
  138. Folkman, J. Role of angiogenesis in tumor growth and metastasis. Semin. Oncol. 2002, 29, 15–18. [Google Scholar] [CrossRef]
  139. Pandya, N.; Dhalla, N.S.; Santani, D.D. Angiogenesis—A new target for future therapy. Vasc. Pharmacol. 2006, 44, 265–274. [Google Scholar] [CrossRef]
  140. Furuya, M. Complexity of tumor vasculature and molecular targeting therapies. Front. Biosci. 2011, 3, 549–561. [Google Scholar] [CrossRef] [Green Version]
  141. Hida, K.; Maishi, N.; Torii, C.; Hida, Y. Tumor angiogenesis—characteristics of tumor endothelial cells. Int. J. Clin. Oncol. 2016, 21, 206–212. [Google Scholar] [CrossRef]
  142. Pugh, C.W.; Ratcliffe, P.J. Regulation of angiogenesis by hypoxia: Role of the HIF system. Nat. Med. 2003, 9, 677–684. [Google Scholar] [CrossRef]
  143. Gilbertson, R.J.; Rich, J.N. Making a tumour’s bed: Glioblastoma stem cells and the vascular niche. Nat. Rev. Cancer 2007, 7, 733–736. [Google Scholar] [CrossRef]
  144. Soeda, A.; Park, M.; Lee, D.; Mintz, A.; Androutsellis-Theotokis, A.; McKay, R.D.; Engh, J.; Iwama, T.; Kunisada, T.; Kassam, A.B.; et al. Hypoxia promotes expansion of the CD133-positive glioma stem cells through activation of HIF-1α. Oncogene 2009, 28, 3949–3959. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Phi, L.T.H.; Sari, I.N.; Yang, Y.-G.; Lee, S.-H.; Jun, N.; Kim, K.S.; Lee, Y.K.; Kwon, H.Y. Cancer Stem Cells (CSCs) in Drug Resistance and their Therapeutic Implications in Cancer Treatment. Stem Cells Int. 2018, 2018, 1–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Heddleston, J.M.; Li, Z.; McLendon, R.E.; Hjelmeland, A.B.; Rich, J.N. The hypoxic microenvironment maintains glioblastoma stem cells and promotes reprogramming towards a cancer stem cell phenotype. Cell Cycle 2009, 8, 3274–3284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Heddleston, J.M.; Li, Z.; Lathia, J.D.; Bao, S.; Hjelmeland, A.B.; Rich, J.N. Hypoxia inducible factors in cancer stem cells. Br. J. Cancer 2010, 102, 789–795. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Ricciuti, B.; Foglietta, J.; Bianconi, V.; Sahebkar, A.; Pirro, M. Enzymes involved in tumor-driven angiogenesis: A valuable target for anticancer therapy. Semin. Cancer Boil. 2019, 56, 87–99. [Google Scholar] [CrossRef] [PubMed]
  149. Jeong, H.; Kim, S.; Hong, B.-J.; Lee, C.-J.; Kim, Y.-E.; Bok, S.; Oh, J.-M.; Gwak, S.-H.; Yoo, M.Y.; Lee, M.S.; et al. Tumor-associated macrophages enhance tumor hypoxia and aerobic glycolysis. Cancer Res. 2019. [Google Scholar] [CrossRef] [Green Version]
  150. Tanriover, G.; Aytac, G. Mutualistic Effects of the Myeloid-Derived Suppressor Cells and Cancer Stem Cells in the Tumor Microenvironment. Crit. Rev. Oncog. 2019, 24, 61–67. [Google Scholar] [CrossRef]
  151. Guo, Y.; Feng, K.; Wang, Y.; Han, W. Targeting cancer stem cells by using chimeric antigen receptor-modified T cells: A potential and curable approach for cancer treatment. Protein Cell 2017, 9, 516–526. [Google Scholar] [CrossRef] [Green Version]
  152. Miele, L.; Espinoza, I.; Pochampally, R.R.; Watabe, K.; Xing, F. Notch signaling: Targeting cancer stem cells and epithelial-to-mesenchymal transition. OncoTargets Ther. 2013, 6, 1249–1259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. He, S.; Liu, Z.; Oh, D.-Y.; Thiele, C.J. MYCN and the epigenome. Front. Oncol. 2013, 3, 1. [Google Scholar] [CrossRef] [Green Version]
  154. Guo, Z.; Jiang, J.-H.; Zhang, J.; Yang, H.-J.; Yang, F.-Q.; Qi, Y.-P.; Zhong, Y.-P.; Su, J.; Yang, R.-R.; Li, L.-Q.; et al. COX-2 Promotes Migration and Invasion by the Side Population of Cancer Stem Cell-Like Hepatocellular Carcinoma Cells. Medicine 2015, 94, e1806. [Google Scholar] [CrossRef] [PubMed]
  155. D’Angelo, R.C.; Ouzounova, M.; Davis, A.; Choi, D.; Tchuenkam, S.M.; Kim, G.; Luther, T.; Quraishi, A.A.; Şenbabaoğlu, Y.; Conley, S.J.; et al. Notch reporter activity in breast cancer cell lines identifies a subset of cells with stem cell activity. Mol. Cancer Ther. 2015, 14, 779–787. [Google Scholar] [CrossRef] [Green Version]
  156. Lee, S.H.; Do, S.I.; Lee, H.J.; Kang, H.J.; Koo, B.S.; Lim, Y.C. Notch1 signaling contributes to stemness in head and neck squamous cell carcinoma. Lab. Investig. 2016, 96, 508–516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Hawley, T.S.; Riz, I.; Yang, W.; Wakabayashi, Y.; DePalma, L.; Chang, Y.-T.; Peng, W.; Zhu, J.; Hawley, R. Identification of an ABCB1 (P-glycoprotein)-positive carfilzomib-resistant myeloma subpopulation by the pluripotent stem cell fluorescent dye CDy1. Am. J. Hematol. 2013, 88, 265–272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Fender, A.W.; Nutter, J.M.; Fitzgerald, T.L.; Bertrand, F.E.; Sigounas, G. Notch-1 Promotes Stemness and Epithelial to Mesenchymal Transition in Colorectal Cancer. J. Cell. Biochem. 2015, 116, 2517–2527. [Google Scholar] [CrossRef]
  159. Benedito, R.; Roca, C.; Sörensen, I.; Adams, S.; Gossler, A.; Fruttiger, M.; Adams, R.H. The Notch Ligands Dll4 and Jagged1 Have Opposing Effects on Angiogenesis. Cell 2009, 137, 1124–1135. [Google Scholar] [CrossRef] [Green Version]
  160. Bhowmick, N.A.; Neilson, E.G.; Moses, H.L. Stromal fibroblasts in cancer initiation and progression. Nature 2004, 432, 332–337. [Google Scholar] [CrossRef]
  161. Owen, J.L.; Zadeh, M. Macrophages and chemokines as mediators of angiogenesis. Front. Physiol. 2013, 4, 159. [Google Scholar] [CrossRef] [Green Version]
  162. Meier, K.; Lehr, C.-M.; Daum, N. Differentiation potential of human pancreatic stem cells for epithelial- and endothelial-like cell types. Ann. Anat. Anat. Anz. 2009, 191, 70–82. [Google Scholar] [CrossRef] [PubMed]
  163. Wang, R.; Chadalavada, K.; Wilshire, J.; Kowalik, U.; Hovinga, K.E.; Geber, A.; Fligelman, B.; Leversha, M.; Brennan, C.W.; Tabar, V. Glioblastoma stem-like cells give rise to tumour endothelium. Nature 2010, 468, 829–833. [Google Scholar] [CrossRef] [PubMed]
  164. Grillet, F.; Bayet, E.; Villeronce, O.; Zappia, L.; Lagerqvist, E.L.; Lunke, S.; Charafe-Jauffret, E.; Pham, K.; Molck, C.; Rolland, N.; et al. Circulating tumour cells from patients with colorectal cancer have cancer stem cell hallmarks in ex vivo culture. Gut 2017, 66, 1802–1810. [Google Scholar] [CrossRef] [Green Version]
  165. Burgess, D.J. Breast cancer: Circulating and dynamic EMT. Nat. Rev. Cancer 2013, 13, 148–149. [Google Scholar] [CrossRef] [PubMed]
  166. Tam, W.L.; Weinberg, R.A. The epigenetics of epithelial-mesenchymal plasticity in cancer. Nat. Med. 2013, 19, 1438–1449. [Google Scholar] [CrossRef] [Green Version]
  167. Zhao, Y.; Bao, Q.; Renner, A.; Camaj, P.; Eichhorn, M.; Ischenko, I.; Angele, M.; Kleespies, A.; Jauch, K.W.; Bruns, C.J. Cancer stem cells and angiogenesis. Int. J. Dev. Boil. 2011, 55, 477–482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Bao, S. Stem Cell-like Glioma Cells Promote Tumor Angiogenesis through Vascular Endothelial Growth Factor. Cancer Res. 2006, 66, 7843–7848. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Monzani, E.; Facchetti, F.; Galmozzi, E.; Corsini, E.; Benetti, A.; Cavazzin, C.; Gritti, A.; Piccinini, A.; Porro, D.; Santinami, M.; et al. Melanoma contains CD133 and ABCG2 positive cells with enhanced tumourigenic potential. Eur. J. Cancer 2007, 43, 935–946. [Google Scholar] [CrossRef]
  170. Maeda, S.; Shinchi, H.; Kurahara, H.; Mataki, Y.; Maemura, K.; Sato, M.; Natsugoe, S.; Aikou, T.; Takao, S. CD133 expression is correlated with lymph node metastasis and vascular endothelial growth factor-C expression in pancreatic cancer. Br. J. Cancer 2008, 98, 1389–1397. [Google Scholar] [CrossRef] [Green Version]
  171. Gao, H.; Chakraborty, G.; Lee-Lim, A.P.; Mo, Q.; Decker, M.; Vonica, A.; Shen, R.; Brogi, E.; Brivanlou, A.H.; Giancotti, F.G. The BMP inhibitor Coco reactivates breast cancer cells at lung metastatic sites. Cell 2012, 150, 764–779. [Google Scholar] [CrossRef] [Green Version]
  172. Giancotti, F.G. Mechanisms Governing Metastatic Dormancy and Reactivation. Cell 2013, 155, 750–764. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Kemper, K.; Grandela, C.; Medema, J.P. Molecular identification and targeting of colorectal cancer stem cells. Oncotarget 2010, 1, 387–395. [Google Scholar] [CrossRef] [Green Version]
  174. Zhang, J.; Yuan, B.; Zhang, H.; Li, H. Human epithelial ovarian cancer cells expressing CD105, CD44 and CD106 surface markers exhibit increased invasive capacity and drug resistance. Oncol. Lett. 2019, 17, 5351–5360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Liu, J.; Chen, L.; Deng, H.; Xu, B.; Li, M.; Zheng, X.; Wu, C.; Jiang, J.-T. Epithelial-to-mesenchymal transition in human esophageal cancer associates with tumor progression and patient’s survival. Int. J. Clin. Exp. Pathol. 2014, 7, 6943–6949. [Google Scholar] [PubMed]
  176. Calon, A.; Lonardo, E.; Berenguer, A.; Espinet, E.; Hernando-Momblona, X.; Iglesias, M.; Sevillano, M.; Palomo-Ponce, S.; Tauriello, D.V.F.; Byrom, D.; et al. Stromal gene expression defines poor-prognosis subtypes in colorectal cancer. Nat. Genet. 2015, 47, 320–329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Chang, J.C. Cancer stem cells: Role in tumor growth, recurrence, metastasis, and treatment resistance. Medicine (Baltimore) 2016, 95, S20–S25. [Google Scholar] [CrossRef]
  178. Hermann, P.C.; Sainz, B. Pancreatic cancer stem cells: A state or an entity? Semin. Cancer Boil. 2018, 53, 223–231. [Google Scholar] [CrossRef] [Green Version]
  179. Todaro, M.; Gaggianesi, M.; Catalano, V.; Benfante, A.; Iovino, F.; Biffoni, M.; Apuzzo, T.; Sperduti, I.; Volpe, S.; Cocorullo, G.; et al. CD44v6 Is a Marker of Constitutive and Reprogrammed Cancer Stem Cells Driving Colon Cancer Metastasis. Cell Stem Cell 2014, 14, 342–356. [Google Scholar] [CrossRef] [Green Version]
  180. Yang, Z.; Ni, W.; Cui, C.; Qi, W.; Piao, L.; Xuan, Y. Identification of LETM1 as a marker of cancer stem-like cells and predictor of poor prognosis in esophageal squamous cell carcinoma. Hum. Pathol. 2018, 81, 148–156. [Google Scholar] [CrossRef]
  181. Gupta, P.B.; Önder, T.; Jiang, G.; Tao, K.; Kuperwasser, C.; Weinberg, R.A.; Lander, E.S. Identification of selective inhibitors of cancer stem cells by high-throughput screening. Cell 2009, 138, 645–659. [Google Scholar] [CrossRef] [Green Version]
  182. Bure, I.V.; Nemtsova, M.V.; Zaletaev, D.V. Roles of E-cadherin and Noncoding RNAs in the Epithelial–mesenchymal Transition and Progression in Gastric Cancer. Int. J. Mol. Sci. 2019, 20, 2870. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Zhou, Z.; Zhang, H.-S.; Liu, Y.; Zhang, Z.-G.; Du, G.-Y.; Li, H.; Yu, X.-Y.; Huang, Y. Loss of TET1 facilitates DLD1 colon cancer cell migration via H3K27me3-mediated down-regulation of E-cadherin. J. Cell. Physiol. 2017, 233, 1359–1369. [Google Scholar] [CrossRef] [PubMed]
  184. Wakefield, L.M.; Hill, C.S. Beyond TGFβ: Roles of other TGFβ superfamily members in cancer. Nat. Rev. Cancer 2013, 13, 328–341. [Google Scholar] [CrossRef] [PubMed]
  185. Parbin, S.; Kar, S.; Shilpi, A.; Sengupta, D.; Deb, M.; Rath, S.K.; Patra, S.K. Histone deacetylases: A saga of perturbed acetylation homeostasis in cancer. J. Histochem. Cytochem. 2014, 62, 11–33. [Google Scholar] [CrossRef] [Green Version]
  186. Ganai, S.A. Histone deacetylase inhibitors modulating non-epigenetic players: The novel molecular targets for therapeutic intervention. Curr. Drug Targets 2018, 17, 1. [Google Scholar] [CrossRef]
  187. Cao, Q.; Yu, J.; Han, S.; Kim, J.H.; Mani, R.-S.; Tomlins, S.A.; Mehra, R.; Laxman, B.; Cao, X.; Kleer, C.G.; et al. Repression of E-cadherin by the polycomb group protein EZH2 in cancer. Oncogene 2008, 27, 7274–7284. [Google Scholar] [CrossRef] [Green Version]
  188. Massague, J. TGFbeta signalling in context. Nat. Rev. Mol. Cell Biol. 2012, 13, 616–630. [Google Scholar] [CrossRef]
  189. Calon, A.; Tauriello, D.; Batlle, E. TGF-beta in CAF-mediated tumor growth and metastasis. Semin. Cancer Boil. 2014, 25, 15–22. [Google Scholar] [CrossRef]
  190. Park, S.-M.; Gaur, A.B.; Lengyel, E.; Peter, M.E. The miR-200 family determines the epithelial phenotype of cancer cells by targeting the E-cadherin repressors ZEB1 and ZEB2. Genes Dev. 2008, 22, 894–907. [Google Scholar] [CrossRef] [Green Version]
  191. Guan, T.; Dominguez, C.X.; Amezquita, R.A.; Laidlaw, B.J.; Cheng, J.; Henao-Mejia, J.; Williams, A.; Flavell, R.A.; Lu, J.; Kaech, S.M. ZEB1, ZEB2, and the miR-200 family form a counterregulatory network to regulate CD8+ T cell fates. J. Exp. Med. 2018, 215, 1153–1168. [Google Scholar] [CrossRef]
  192. Tellez, C.S.; Juri, D.E.; Do, K.; Bernauer, A.M.; Thomas, C.L.; Damiani, L.A.; Tessema, M.; Leng, S.; Belinsky, S.A. EMT and stem cell-like properties associated with miR-205 and miR-200 epigenetic silencing are early manifestations during carcinogen-induced transformation of human lung epithelial cells. Cancer Res. 2011, 71, 3087–3097. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Menéndez, J.; Alarcón, T. Metabostemness: A New Cancer Hallmark. Front. Oncol. 2014, 4, 262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Lee, K.-M.; Giltnane, J.M.; Balko, J.M.; Schwarz, L.J.; Guerrero-Zotano, A.L.; Hutchinson, K.E.; Nixon, M.J.; Estrada, M.V.; Sánchez, V.; Sanders, M.E.; et al. MYC and MCL1 Cooperatively Promote Chemotherapy-Resistant Breast Cancer Stem Cells via Regulation of Mitochondrial Oxidative Phosphorylation. Cell Metab. 2017, 26, 633–647.e7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Chiche, J.; Brahimi-Horn, M.C.; Pouysségur, J. Tumour hypoxia induces a metabolic shift causing acidosis: A common feature in cancer. J. Cell. Mol. Med. 2010, 14, 771–794. [Google Scholar] [CrossRef] [Green Version]
  196. Chiche, J.; Ilc, K.; Laferrière, J.; Trottier, E.; Dayan, F.; Mazure, N.M.; Brahimi-Horn, M.C.; Pouyssegur, J. Hypoxia-Inducible Carbonic Anhydrase IX and XII Promote Tumor Cell Growth by Counteracting Acidosis through the Regulation of the Intracellular pH. Cancer Res. 2009, 69, 358–368. [Google Scholar] [CrossRef] [Green Version]
  197. Pollyea, D.A.; Stevens, B.M.; Jones, C.L.; Winters, A.; Pei, S.; Minhajuddin, M.; D’Alessandro, A.; Culp-Hill, R.; Riemondy, K.A.; Gillen, A.E.; et al. Venetoclax with azacitidine disrupts energy metabolism and targets leukemia stem cells in patients with acute myeloid leukemia. Nat. Med. 2018, 24, 1859–1866. [Google Scholar] [CrossRef]
  198. Nakada, D. Venetolax with Azacitidine Drains Fuel from AML Stem Cells. Cell Stem Cell 2019, 24, 7–8. [Google Scholar] [CrossRef] [Green Version]
  199. Fiorillo, M.; Lamb, R.; Tanowitz, H.B.; Mutti, L.; Krstic-Demonacos, M.; Cappello, A.R.; Martinez-Outschoorn, U.E.; Sotgia, F.; Lisanti, M.P. Repurposing atovaquone: Targeting mitochondrial complex III and OXPHOS to eradicate cancer stem cells. Oncotarget 2016, 7, 34084–34099. [Google Scholar] [CrossRef] [Green Version]
  200. Jones, C.L.; Stevens, B.M.; D’Alessandro, A.; Reisz, J.A.; Culp-Hill, R.; Nemkov, T.; Pei, S.; Khan, N.; Adane, B.; Ye, H.; et al. Inhibition of Amino Acid Metabolism Selectively Targets Human Leukemia Stem Cells. Cancer Cell 2018, 34, 724–740.e4. [Google Scholar] [CrossRef] [Green Version]
  201. Ye, H.; Adane, B.; Khan, N.; Sullivan, T.; Minhajuddin, M.; Gasparetto, M.; Stevens, B.; Pei, S.; Balys, M.; Ashton, J.; et al. Leukemic Stem Cells Evade Chemotherapy by Metabolic Adaptation to an Adipose Tissue Niche. Cell Stem Cell 2016, 19, 23–37. [Google Scholar] [CrossRef] [Green Version]
  202. Amsalem, Z.; Arif, T.; Shteinfer-Kuzmine, A.; Chalifa-Caspi, V.; Shoshan-Barmatz, V. The Mitochondrial Protein VDAC1 at the Crossroads of Cancer Cell Metabolism: The Epigenetic Link. Cancers 2020, 12, 1031. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Skoda, J.; Borankova, K.; Jansson, P.; Huang, M.L.-H.; Veselska, R.; Richardson, D.R. Pharmacological targeting of mitochondria in cancer stem cells: An ancient organelle at the crossroad of novel anti-cancer therapies. Pharmacol. Res. 2019, 139, 298–313. [Google Scholar] [CrossRef] [PubMed]
  204. Sancho, P.; Barneda, D.; Heeschen, C. Hallmarks of cancer stem cell metabolism. Br. J. Cancer 2016, 114, 1305–1312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Song, I.-S.; Jeong, Y.J.; Jeong, S.H.; Heo, H.J.; Kim, H.K.; Bae, K.B.; Park, Y.-H.; Kim, S.-U.; Kim, J.-M.; Kim, N.; et al. FOXM1-Induced PRX3 Regulates Stemness and Survival of Colon Cancer Cells via Maintenance of Mitochondrial Function. Gastroenterology 2015, 149, 1006–1016.e9. [Google Scholar] [CrossRef] [PubMed]
  206. Isayev, O.; Rausch, V.; Bauer, N.; Liu, L.; Fan, P.; Zhang, Y.; Gladkich, J.; Nwaeburu, C.C.; Mattern, J.; Mollenhauer, M.; et al. Inhibition of glucose turnover by 3-bromopyruvate counteracts pancreatic cancer stem cell features and sensitizes cells to gemcitabine. Oncotarget 2014, 5, 5177–5189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. Arif, T.; Paul, A.; Krelin, Y.; Shteinfer-Kuzmine, A.; Shoshan-Barmatz, V. Mitochondrial VDAC1 Silencing Leads to Metabolic Rewiring and the Reprogramming of Tumour Cells into Advanced Differentiated States. Cancers 2018, 10, 499. [Google Scholar] [CrossRef] [Green Version]
  208. Arif, T.; Krelin, Y.; Nakdimon, I.; Benharroch, D.; Paul, A.; Dadon-Klein, D.; Shoshan-Barmatz, V. VDAC1 is a molecular target in glioblastoma, with its depletion leading to reprogrammed metabolism and reversed oncogenic properties. Neuro Oncol. 2017, 19, 951–964. [Google Scholar] [CrossRef]
  209. Dawson, M.A. The cancer epigenome: Concepts, challenges, and therapeutic opportunities. Science 2017, 355, 1147–1152. [Google Scholar] [CrossRef]
  210. Chatterjee, A.; Rodger, E.J.; Eccles, M. Epigenetic drivers of tumourigenesis and cancer metastasis. Semin. Cancer Boil. 2018, 51, 149–159. [Google Scholar] [CrossRef]
  211. Ahuja, N.; Sharma, A.R.; Baylin, S.B. Epigenetic Therapeutics: A New Weapon in the War Against Cancer. Annu. Rev. Med. 2016, 67, 73–89. [Google Scholar] [CrossRef] [Green Version]
  212. Borley, J.; Brown, R. Epigenetic mechanisms and therapeutic targets of chemotherapy resistance in epithelial ovarian cancer. Ann. Med. 2015, 47, 359–369. [Google Scholar] [CrossRef]
  213. Arrowsmith, C.; Bountra, C.; Fish, P.V.; Lee, K.; Schapira, M. Epigenetic protein families: A new frontier for drug discovery. Nat. Rev. Drug Discov. 2012, 11, 384–400. [Google Scholar] [CrossRef] [Green Version]
  214. Gates, L.A.; Shi, J.; Rohira, A.D.; Feng, Q.; Zhu, B.; Bedford, M.T.; Sagum, C.A.; Jung, S.Y.; Qin, J.; Tsai, M.-J.; et al. Acetylation on histone H3 lysine 9 mediates a switch from transcription initiation to elongation. J. Boil. Chem. 2017, 292, 14456–14472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Wolny, E.; Braszewska-Zalewska, A.; Kroczek, D.; Hasterok, R. Histone H3 and H4 acetylation patterns are more dynamic than those of DNA methylation in Brachypodium distachyon embryos during seed maturation and germination. Protoplasma 2017, 254, 2045–2052. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Cruz, C.; Della Rosa, M.; Krueger, C.; Gao, Q.; Horkai, D.; King, M.; Field, L.; Houseley, J. Tri-methylation of histone H3 lysine 4 facilitates gene expression in ageing cells. eLife 2018, 7, 34081. [Google Scholar] [CrossRef] [PubMed]
  217. Huang, H.; Weng, H.; Zhou, K.; Wu, T.; Zhao, B.S.; Sun, M.; Chen, Z.; Deng, X.; Xiao, G.; Auer, F.; et al. Histone H3 trimethylation at lysine 36 guides m6A RNA modification co-transcriptionally. Nature 2019, 567, 414–419. [Google Scholar] [CrossRef] [PubMed]
  218. Li, F.; Wu, R.; Cui, X.; Zha, L.; Yu, L.; Shi, H.; Xue, B. Histone Deacetylase 1 (HDAC1) Negatively Regulates Thermogenic Program in Brown Adipocytes via Coordinated Regulation of Histone H3 Lysine 27 (H3K27) Deacetylation and Methylation. J. Boil. Chem. 2016, 291, 4523–4536. [Google Scholar] [CrossRef] [Green Version]
  219. Möller, M.; Schotanus, K.; Soyer, J.L.; Haueisen, J.; Happ, K.; Stralucke, M.; Happel, P.; Smith, K.M.; Connolly, L.R.; Freitag, M.; et al. Destabilization of chromosome structure by histone H3 lysine 27 methylation. PLoS Genet. 2019, 15, e1008093. [Google Scholar] [CrossRef] [Green Version]
  220. Bird, A. CpG-rich islands and the function of DNA methylation. Nature 1986, 321, 209–213. [Google Scholar] [CrossRef]
  221. Strahl, B.D.; Allis, C.D. The language of covalent histone modifications. Nature 2000, 403, 41–45. [Google Scholar] [CrossRef]
  222. Cosgrove, M.S.; Wolberger, C. How does the histone code work? Biochem. Cell. Biol. 2005, 83, 468–476. [Google Scholar] [CrossRef] [PubMed]
  223. Cloney, R. Gene regulation: Optical control of epigenetics. Nat. Rev. Genet. 2016, 17, 254. [Google Scholar] [PubMed]
  224. Harvey, Z.; Chen, Y.; Jarosz, D.F. Protein-Based Inheritance: Epigenetics beyond the Chromosome. Mol. Cell 2017, 69, 195–202. [Google Scholar] [CrossRef] [PubMed]
  225. Kim, M.Y.; Lee, J.E.; Kim, L.K.; Kim, T. Epigenetic memory in gene regulation and immune response. BMB Rep. 2019, 52, 127–132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Goldman, M. CpG Islands. In Encyclopedia of Genetics; Elsevier BV: Amsterdam, The Netherlands, 2001; p. 477. [Google Scholar]
  227. Deaton, A.M.; Bird, A. CpG islands and the regulation of transcription. Genes Dev. 2011, 25, 1010–1022. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  228. Ohtani-Fujita, N.; Fujita, T.; Aoike, A.; Osifchin, N.E.; Robbins, P.D.; Sakai, T. CpG methylation inactivates the promoter activity of the human retinoblastoma tumor-suppressor gene. Oncogene 1993, 8, 8. [Google Scholar]
  229. Di Croce, L.; Helin, K. Transcriptional regulation by Polycomb group proteins. Nat. Struct. Mol. Boil. 2013, 20, 1147–1155. [Google Scholar] [CrossRef]
  230. Morey, L.; Helin, K. Polycomb group protein-mediated repression of transcription. Trends Biochem. Sci. 2010, 35, 323–332. [Google Scholar] [CrossRef]
  231. Corso-Díaz, X.; Jaeger, C.; Chaitankar, V.; Swaroop, A. Epigenetic control of gene regulation during development and disease: A view from the retina. Prog. Retin. Eye Res. 2018, 65, 1–27. [Google Scholar] [CrossRef]
  232. Collinson, A.; Collier, A.; Morgan, N.P.; Sienerth, A.; Chandra, T.; Andrews, S.; Rugg-Gunn, P.J. Deletion of the Polycomb-Group Protein EZH2 Leads to Compromised Self-Renewal and Differentiation Defects in Human Embryonic Stem Cells. Cell Rep. 2016, 17, 2700–2714. [Google Scholar] [CrossRef]
  233. Wainwright, E.N.; Scaffidi, P. Epigenetics and Cancer Stem Cells: Unleashing, Hijacking, and Restricting Cellular Plasticity. Trends Cancer 2017, 3, 372–386. [Google Scholar] [CrossRef] [Green Version]
  234. Donaldson, M.C.; Sungalee, S.; Zufferey, M.; Tavernari, D.; Katanayeva, N.; Battistello, E.; Mina, M.; Douglass, K.M.; Rey, T.; Raynaud, F.; et al. EZH2 oncogenic mutations drive epigenetic, transcriptional, and structural changes within chromatin domains. Nat. Genet. 2019, 51, 517–528. [Google Scholar] [CrossRef]
  235. Woods, B.A.; Levine, R.L. The role of mutations in epigenetic regulators in myeloid malignancies. Immunol. Rev. 2014, 263, 22–35. [Google Scholar] [CrossRef]
  236. Yi, J.M.; Tsai, H.-C.; Glöckner, S.C.; Lin, S.; Ohm, J.E.; Easwaran, H.; James, C.D.; Costello, J.F.; Riggins, G.; Eberhart, C.G.; et al. Abnormal DNA methylation of CD133 in colorectal and glioblastoma tumors. Cancer Res. 2008, 68, 8094–8103. [Google Scholar] [CrossRef] [Green Version]
  237. Tabu, K.; Sasai, K.; Kimura, T.; Wang, L.; Aoyanagi, E.; Kohsaka, S.; Tanino, M.; Nishihara, H.; Tanaka, K. Promoter hypomethylation regulates CD133 expression in human gliomas. Cell Res. 2008, 18, 1037–1046. [Google Scholar] [CrossRef] [Green Version]
  238. Gorodetska, I.V.; Lukiyanchuk, V.; Peitzsch, C.; Kozeretska, I.; Dubrovska, A. BRCA1 and EZH2 cooperate in regulation of prostate cancer stem cell phenotype. Int. J. Cancer 2019, 145, 2974–2985. [Google Scholar] [CrossRef]
  239. Mani, S.A.; Guo, W.; Liao, M.-J.; Eaton, E.N.; Ayyanan, A.; Zhou, A.Y.; Brooks, M.; Reinhard, F.; Zhang, C.C.; Shipitsin, M.; et al. The Epithelial-Mesenchymal Transition Generates Cells with Properties of Stem Cells. Cell 2008, 133, 704–715. [Google Scholar] [CrossRef] [Green Version]
  240. Cai, C.; Zhu, X. The Wnt/beta-catenin pathway regulates self-renewal of cancer stem-like cells in human gastric cancer. Mol. Med. Rep. 2012, 5, 1191–1196. [Google Scholar]
  241. Matsui, W.H. Cancer stem cell signaling pathways. Medicine 2016, 95, S8–S19. [Google Scholar] [CrossRef]
  242. Zhu, C.; Pan, Y.; Ma, S.; Cao, K.; Zhou, S.; Zhao, A.; Li, M.; Qian, F. Therapeutic approaches targeting cancer stem cells. J. Cancer Res. Ther. 2018, 14, 1469. [Google Scholar] [CrossRef]
  243. Lai, E. Notch signaling: Control of cell communication and cell fate. Development 2004, 131, 965–973. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  244. Dessaud, E.; McMahon, A.P.; Briscoe, J. Pattern formation in the vertebrate neural tube: A sonic hedgehog morphogen-regulated transcriptional network. Development 2008, 135, 2489–2503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  245. Takebe, N.; Miele, L.; Harris, P.J.; Jeong, W.; Bando, H.; Kahn, M.; Yang, S.X.; Ivy, S.P. Targeting Notch, Hedgehog, and Wnt pathways in cancer stem cells: Clinical update. Nat. Rev. Clin. Oncol. 2015, 12, 445–464. [Google Scholar] [CrossRef]
  246. Zhang, J.; Shao, X.; Sun, H.; Liu, K.; Ding, Z.; Chen, J.; Fang, L.; Su, W.; Hong, Y.; Li, H.; et al. NUMB negatively regulates the epithelial-mesenchymal transition of triple-negative breast cancer by antagonizing Notch signaling. Oncotarget 2016, 7, 61036–61053. [Google Scholar] [CrossRef] [Green Version]
  247. Polakis, P. The many ways of Wnt in cancer. Curr. Opin. Genet. Dev. 2007, 17, 45–51. [Google Scholar] [CrossRef]
  248. Gujral, T.S.; Chan, M.; Peshkin, L.; Sorger, P.K.; Kirschner, M.W.; MacBeath, G. A noncanonical Frizzled2 pathway regulates epithelial-mesenchymal transition and metastasis. Cell 2014, 159, 844–856. [Google Scholar] [CrossRef] [Green Version]
  249. Yang, L.; Xie, G.; Fan, Q.; Xie, J. Activation of the hedgehog-signaling pathway in human cancer and the clinical implications. Oncogene 2009, 29, 469–481. [Google Scholar] [CrossRef] [Green Version]
  250. Clement, V.; Sanchez, P.; De Tribolet, N.; Radovanovic, I.; i Altaba, A.R. HEDGEHOG-GLI1 signaling regulates human glioma growth, cancer stem cell self-renewal, and tumorigenicity. Curr. Biol. 2007, 17, 165–172. [Google Scholar] [CrossRef]
  251. Steinway, S.N.; Zañudo, J.G.; Ding, W.; Rountree, C.B.; Feith, D.J.; Loughran, T.P.; Albert, R. Network Modeling of TGF Signaling in Hepatocellular Carcinoma Epithelial-to-Mesenchymal Transition Reveals Joint Sonic Hedgehog and Wnt Pathway Activation. Cancer Res. 2014, 74, 5963–5977. [Google Scholar] [CrossRef] [Green Version]
  252. Fan, H.-X.; Wang, S.; Zhao, H.; Liu, N.; Chen, D.; Sun, M.; Zheng, J. Sonic hedgehog signaling may promote invasion and metastasis of oral squamous cell carcinoma by activating MMP-9 and E-cadherin expression. Med. Oncol. 2014, 31, 1–8. [Google Scholar] [CrossRef]
  253. Suzuki, H.; Watkins, D.N.; Jair, K.-W.; Schuebel, K.E.; Markowitz, S.D.; Chen, W.D.; Pretlow, T.P.; Yang, B.; Akiyama, Y.; Van Engeland, M.; et al. Epigenetic inactivation of SFRP genes allows constitutive WNT signaling in colorectal cancer. Nat. Genet. 2004, 36, 417–422. [Google Scholar] [CrossRef] [PubMed]
  254. Krishnamurthy, N.; Kurzrock, R. Targeting the Wnt/beta-catenin pathway in cancer: Update on effectors and inhibitors. Cancer Treat. Rev. 2018, 62, 50–60. [Google Scholar] [CrossRef] [PubMed]
  255. Hussain, M.; Rao, M.; Humphries, A.E.; Hong, J.A.; Liu, F.; Yang, M.; Caragacianu, D.; Schrump, D.S. Tobacco Smoke Induces Polycomb-Mediated Repression of Dickkopf-1 in Lung Cancer Cells. Cancer Res. 2009, 69, 3570–3578. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  256. Ghoshal, P.; Nganga, A.J.; Moran-Giuati, J.; Szafranek, A.; Johnson, T.R.; Bigelow, A.J.; Houde, C.M.; Avet-Loiseau, H.; Smiraglia, D.J.; Ersing, N.; et al. Loss of the SMRT/NCoR2 Corepressor Correlates with JAG2 Overexpression in Multiple Myeloma. Cancer Res. 2009, 69, 4380–4387. [Google Scholar] [CrossRef] [Green Version]
  257. Wu, Y.; Gong, L.; Xu, J.; Mou, Y.; Xu, X.; Qian, Z. The clinicopathological significance of HES1 promoter hypomethylation in patients with colorectal cancer. OncoTargets Ther. 2017, 10, 5827–5834. [Google Scholar] [CrossRef] [Green Version]
  258. Benoit, Y.D.; Laursen, K.B.; Witherspoon, M.S.; Lipkin, S.M.; Gudas, L.J. Inhibition of PRC2 histone methyltransferase activity increases TRAIL-mediated apoptosis sensitivity in human colon cancer cells. J. Cell. Physiol. 2013, 228, 764–772. [Google Scholar] [CrossRef] [Green Version]
  259. Han, X.; Ranganathan, P.; Tzimas, C.; Weaver, K.L.; Jin, K.; Astudillo, L.; Zhou, W.; Zhu, X.; Li, B.; Robbins, D.J.; et al. Notch Represses Transcription by PRC2 Recruitment to the Ternary Complex. Mol. Cancer Res. 2017, 15, 1173–1183. [Google Scholar] [CrossRef] [Green Version]
  260. Jagani, Z.; Mora-Blanco, E.L.; Sansam, C.G.; McKenna, E.S.; Wilson, B.; Chen, D.; Klekota, J.; Tamayo, P.; Nguyen, P.T.L.; Tolstorukov, M.; et al. Loss of the tumor suppressor Snf5 leads to aberrant activation of the Hedgehog-Gli pathway. Nat. Med. 2010, 16, 1429–1433. [Google Scholar] [CrossRef] [Green Version]
  261. McKenna, E.S.; Tamayo, P.; Cho, Y.-J.; Tillman, E.J.; Mora-Blanco, E.L.; Sansam, C.G.; Koellhoffer, E.C.; Pomeroy, S.L.; Roberts, C.W.M. Epigenetic inactivation of the tumor suppressor BIN1 drives proliferation of SNF5-deficient tumors. Cell Cycle 2012, 11, 1956–1965. [Google Scholar] [CrossRef] [Green Version]
  262. Coni, S.; Mancuso, A.B.; Di Magno, L.; Sdruscia, G.; Manni, S.; Serrao, S.M.; Rotili, D.; Spiombi, E.; Bufalieri, F.; Petroni, M.; et al. Selective targeting of HDAC1/2 elicits anticancer effects through Gli1 acetylation in preclinical models of SHH Medulloblastoma. Sci. Rep. 2017, 7, 44079. [Google Scholar] [CrossRef]
  263. Canettieri, G.; Di Marcotullio, L.; Greco, A.; Coni, S.; Antonucci, L.; Infante, P.; Pietrosanti, L.; De Smaele, E.; Ferretti, E.; Miele, E.; et al. Histone deacetylase and Cullin3-REN(KCTD11) ubiquitin ligase interplay regulates Hedgehog signalling through Gli acetylation. Nat. Cell. Biol. 2010, 12, 132–142. [Google Scholar]
  264. Niewiadomski, P.; Niedziółka, S.M.; Markiewicz, Ł.; Uśpieński, T.; Baran, B.; Chojnowska, K. Gli Proteins: Regulation in Development and Cancer. Cells 2019, 8, 147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  265. Michalak, E.M.; Visvader, J.E. Dysregulation of histone methyltransferases in breast cancer – Opportunities for new targeted therapies? Molecular Oncol. 2016, 10, 1497–1515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  266. Rao, R.C.; Dou, Y. Hijacked in cancer: The KMT2 (MLL) family of methyltransferases. Nat. Rev. Cancer 2015, 15, 334–346. [Google Scholar] [CrossRef] [Green Version]
  267. Cozzio, A.; Passegué, E.; Ayton, P.M.; Karsunky, H.; Cleary, M.L.; Weissman, I.L. Similar MLL-associated leukemias arising from self-renewing stem cells and short-lived myeloid progenitors. Genes Dev. 2003, 17, 3029–3035. [Google Scholar] [CrossRef] [Green Version]
  268. Ono, R.; Masuya, M.; Nakajima, H.; Enomoto, Y.; Miyata, E.; Nakamura, A.; Ishii, S.; Suzuki, K.; Shibata-Minoshima, F.; Katayama, N.; et al. Plzf drives MLL-fusion–mediated leukemogenesis specifically in long-term hematopoietic stem cells. Blood 2013, 122, 1271–1283. [Google Scholar] [CrossRef] [Green Version]
  269. Krivtsov, A.V.; Twomey, D.; Feng, Z.; Stubbs, M.C.; Wang, Y.; Faber, J.; Levine, J.E.; Wang, J.; Hahn, W.C.; Gilliland, D.G.; et al. Transformation from committed progenitor to leukaemia stem cell initiated by MLL–AF9. Nature 2006, 442, 818–822. [Google Scholar] [CrossRef]
  270. Somervaille, T.C.; Matheny, C.J.; Spencer, G.J.; Iwasaki, M.; Rinn, J.; Witten, D.M.; Chang, H.Y.; Shurtleff, S.A.; Downing, J.R.; Cleary, M.L. Hierarchical Maintenance of MLL Myeloid Leukemia Stem Cells Employs a Transcriptional Program Shared with Embryonic Rather Than Adult Stem Cells. Cell Stem Cell 2009, 4, 129–140. [Google Scholar] [CrossRef] [Green Version]
  271. Lewis, P.W.; Müller, M.M.; Koletsky, M.S.; Cordero, F.; Lin, S.; Banaszynski, L.A.; Garcia, B.A.; Muir, T.W.; Becher, O.J.; Allis, C.D. Inhibition of PRC2 Activity by a Gain-of-Function H3 Mutation Found in Pediatric Glioblastoma. Science 2013, 340, 857–861. [Google Scholar] [CrossRef] [Green Version]
  272. Benetatos, L.; Vartholomatos, G. On the potential role of DNMT1 in acute myeloid leukemia and myelodysplastic syndromes: Not another mutated epigenetic driver. Ann. Hematol. 2016, 95, 1571–1582. [Google Scholar] [CrossRef]
  273. Brunetti, L.; Gundry, M.C.; A Goodell, M. DNMT3A in Leukemia. Cold Spring Harb. Perspect. Med. 2016, 7, a030320. [Google Scholar] [CrossRef] [PubMed]
  274. Sun, Y.; Chen, B.-R.; Deshpande, A. Epigenetic Regulators in the Development, Maintenance, and Therapeutic Targeting of Acute Myeloid Leukemia. Front. Oncol. 2018, 8, 41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  275. Munro, M.; Wickremesekera, S.K.; Peng, L.; Tan, S.T.; Itinteang, T. Cancer stem cells in colorectal cancer: A review. J. Clin. Pathol. 2017, 71, 110–116. [Google Scholar] [CrossRef]
  276. Todaro, M.; Francipane, M.G.; Medema, J.P.; Stassi, G. Colon Cancer Stem Cells: Promise of Targeted Therapy. Gastroenterology 2010, 138, 2151–2162. [Google Scholar] [CrossRef]
  277. Hammoud, S.S.; Cairns, B.R.; Jones, D.A. Epigenetic regulation of colon cancer and intestinal stem cells. Curr. Opin. Cell Boil. 2013, 25, 177–183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  278. Campbell, P.M.; Szyf, M. Human DNA methyltransferase gene DNMT1 is regulated by the APC pathway. Carcinog. 2003, 24, 17–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  279. Liang, T.-J.; Wang, H.-X.; Zheng, Y.-Y.; Cao, Y.-Q.; Wu, X.; Zhou, X.; Dong, S.-X. APC hypermethylation for early diagnosis of colorectal cancer: A meta-analysis and literature review. Oncotarget 2017, 8, 46468–46479. [Google Scholar] [CrossRef] [Green Version]
  280. Paschall, A.V.; Liu, K. Epigenetic and Immune Regulation of Colorectal Cancer Stem Cells. Curr. Color. Cancer Rep. 2015, 11, 414–421. [Google Scholar] [CrossRef]
  281. Pathania, R.; Ramachandran, S.; Elangovan, S.; Padia, R.; Yang, P.; Cinghu, S.; Veeranan-Karmegam, R.; Arjunan, P.; Gnana-Prakasam, J.P.; Sadanand, F.; et al. DNMT1 is essential for mammary and cancer stem cell maintenance and tumorigenesis. Nat. Commun. 2015, 6, 1–11. [Google Scholar] [CrossRef]
  282. Xia, H.; Cao, J.; Li, Q.; Lv, Y.; Jia, W.; Ren, W.; Cheng, Q.; Song, X.; Xu, G. Hepatocellular Carcinoma-propagating Cells are Detectable by Side Population Analysis and Possess an Expression Profile Reflective of a Primitive Origin. Sci. Rep. 2016, 6, 34856. [Google Scholar] [CrossRef] [Green Version]
  283. Lawson, D.A.; Bhakta, N.R.; Kessenbrock, K.; Prummel, K.D.; Yu, Y.; Takai, K.; Zhou, A.; Eyob, H.; Balakrishnan, S.; Wang, C.-Y.; et al. Single-cell analysis reveals a stem-cell program in human metastatic breast cancer cells. Nature 2015, 526, 131–135. [Google Scholar] [CrossRef] [PubMed]
  284. Boesch, M.; Zeimet, A.G.; Reimer, D.; Schmidt, S.; Gastl, G.; Parson, W.; Spoeck, F.; Hatina, J.; Wolf, D.; Sopper, S. The side population of ovarian cancer cells defines a heterogeneous compartment exhibiting stem cell characteristics. Oncotarget 2014, 5, 7027–7039. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  285. Visvader, J.E.; Lindeman, G.J. Cancer stem cells in solid tumours: Accumulating evidence and unresolved questions. Nat. Rev. Cancer 2008, 8, 755–768. [Google Scholar] [CrossRef] [PubMed]
  286. Hatina, J.; Boesch, M.; Sopper, S.; Kripnerova, M.; Wolf, D.; Reimer, D.; Marth, C.; Zeimet, A.G. Ovarian Cancer Stem Cell Heterogeneity. Single Mol. Single Cell Seq. 2019, 1139, 201–221. [Google Scholar] [CrossRef]
  287. Smith, L.M.; Nesterova, A.; Ryan, M.C.; Duniho, S.; Jonas, M.; Anderson, M.; Zabinski, R.F.; Sutherland, M.K.; Gerber, H.-P.; Van Orden, K. CD133/prominin-1 is a potential therapeutic target for antibody-drug conjugates in hepatocellular and gastric cancers. Br. J. Cancer 2008, 99, 100–109. [Google Scholar] [CrossRef]
  288. Swaminathan, S.K.; Roger, E.; Toti, U.; Niu, L.; Ohlfest, J.R.; Panyam, J. CD133-targeted paclitaxel delivery inhibits local tumor recurrence in a mouse model of breast cancer. J. Control. Release 2013, 171, 280–287. [Google Scholar] [CrossRef] [Green Version]
  289. Krampitz, G.W.; George, B.M.; Willingham, S.B.; Volkmer, J.-P.; Weiskopf, K.; Jahchan, N.; Newman, A.M.; Sahoo, D.; Zemek, A.J.; Yanovsky, R.L.; et al. Identification of tumorigenic cells and therapeutic targets in pancreatic neuroendocrine tumors. Proc. Nat. Acad. Sci. USA 2016, 113, 4464–4469. [Google Scholar] [CrossRef] [Green Version]
  290. Piccione, E.C.; Juarez, S.; Tseng, S.; Liu, J.; Stafford, M.; Narayanan, C.; Wang, L.; Weiskopf, K.; Majeti, R. SIRPα-Antibody Fusion Proteins Selectively Bind and Eliminate Dual Antigen-Expressing Tumor Cells. Clin. Cancer Res. 2016, 22, 5109–5119. [Google Scholar] [CrossRef] [Green Version]
  291. Boesch, M.; Onder, L.; Cheng, H.-W.; Novkovic, M.; Mörbe, U.; Sopper, S.; Gastl, G.; Jochum, W.; Ruhstaller, T.; Knauer, M.; et al. Interleukin 7-expressing fibroblasts promote breast cancer growth through sustenance of tumor cell stemness. OncoImmunology 2018, 7, e1414129. [Google Scholar] [CrossRef] [Green Version]
  292. Advani, R.; Flinn, I.; Popplewell, L.; Forero, A.; Bartlett, N.L.; Ghosh, N.; Kline, J.; Roschewski, M.; LaCasce, A.; Collins, G.; et al. CD47 Blockade by Hu5F9-G4 and Rituximab in Non-Hodgkin’s Lymphoma. N. Engl. J. Med. 2018, 379, 1711–1721. [Google Scholar] [CrossRef]
  293. Sallman, D.A.; Donnellan, W.B.; Asch, A.S.; Lee, D.J.; Al Malki, M.; Marcucci, G.; Pollyea, D.A.; Kambhampati, S.; Komrokji, R.S.; Van Elk, J.; et al. The first-in-class anti-CD47 antibody Hu5F9-G4 is active and well tolerated alone or with azacitidine in AML and MDS patients: Initial phase 1b results. J. Clin. Oncol. 2019, 37, 7009. [Google Scholar] [CrossRef]
  294. Ajani, J.A.; Song, S.; Hochster, H.S.; Steinberg, I.B. Cancer Stem Cells: The Promise and the Potential. Semin. Oncol. 2015, 42, S3–S17. [Google Scholar] [CrossRef] [PubMed]
  295. Kanwar, S.S.; Yu, Y.; Nautiyal, J.; Patel, B.B.; Majumdar, A.P. The Wnt/beta-catenin pathway regulates growth and maintenance of colonospheres. Mol. Cancer 2010, 9, 212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  296. Lenz, H.-J.; Kahn, M. Safely targeting cancer stem cells via selective catenin coactivator antagonism. Cancer Sci. 2014, 105, 1087–1092. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  297. El-Khoueiry, A.B.; Ning, Y.; Yang, D.; Cole, S.; Kahn, M.; Zoghbi, M.; Berg, J.; Fujimori, M.; Inada, T.; Kouji, H.; et al. A phase I first-in-human study of PRI-724 in patients (pts) with advanced solid tumors. J. Clin. Oncol. 2013, 31, 2501. [Google Scholar] [CrossRef]
  298. Jang, G.-B.; Hong, I.-S.; Kim, R.-J.; Lee, S.-Y.; Park, S.-J.; Lee, E.-S.; Park, J.H.; Yun, C.-H.; Chung, J.-U.; Lee, K.-J. Wnt/β-catenin small-molecule inhibitor CWP232228 preferentially inhibits the growth of breast cancer stem-like cells. Cancer Res. 2015, 75, 1691–1702. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  299. Kim, J.; Lee, H.; Park, K.; Choi, Y.-K.; Nam, J.; Hong, I. CWP232228 targets liver cancer stem cells through Wnt/β-catenin signaling: A novel therapeutic approach for liver cancer treatment. Oncotarget 2016, 7, 20395–20409. [Google Scholar] [CrossRef] [Green Version]
  300. Xu, R.; Shimizu, F.; Hovinga, K.; Beal, K.; Karimi, S.; Droms, L.; Peck, K.K.; Gutin, P.; Iorgulescu, J.B.; Kaley, T.J.; et al. Molecular and Clinical Effects of Notch Inhibition in Glioma Patients: A Phase 0/I Trial. Clin. Cancer Res. 2016, 22, 4786–4796. [Google Scholar] [CrossRef] [Green Version]
  301. Zhao, Z.-L.; Zhang, L.; Huang, C.-F.; Ma, S.-R.; Bu, L.-L.; Liu, J.-F.; Yu, G.-T.; Liu, B.; Gutkind, J.S.; Kulkarni, A.B.; et al. NOTCH1 inhibition enhances the efficacy of conventional chemotherapeutic agents by targeting head neck cancer stem cell. Sci. Rep. 2016, 6, 24704. [Google Scholar] [CrossRef] [Green Version]
  302. Sun, M.; Zhang, N.; Wang, X.; Li, Y.; Qi, W.; Zhang, H.; Li, Z.; Yang, Q. Hedgehog pathway is involved in nitidine chloride induced inhibition of epithelial-mesenchymal transition and cancer stem cells-like properties in breast cancer cells. Cell Biosci. 2016, 6, 44. [Google Scholar] [CrossRef] [Green Version]
  303. Sharma, N.; Nanta, R.; Sharma, J.; Gunewardena, S.; Singh, K.P.; Shankar, S.; Srivastava, R.K. PI3K/AKT/mTOR and sonic hedgehog pathways cooperate together to inhibit human pancreatic cancer stem cell characteristics and tumor growth. Oncotarget 2015, 6, 32039–32060. [Google Scholar] [CrossRef] [PubMed]
  304. Miyazaki, Y.; Matsubara, S.; Ding, Q.; Tsukasa, K.; Yoshimitsu, M.; Kosai, K.-I.; Takao, S. Efficient elimination of pancreatic cancer stem cells by hedgehog/GLI inhibitor GANT61 in combination with mTOR inhibition. Mol. Cancer 2016, 15, 49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  305. Kim, E.J.; Sahai, V.; Abel, E.V.; Griffith, K.A.; Greenson, J.K.; Takebe, N.; Khan, G.N.; Blau, J.; Craig, R.; Balis, U.G.; et al. Pilot clinical trial of hedgehog pathway inhibitor GDC-0449 (vismodegib) in combination with gemcitabine in patients with metastatic pancreatic adenocarcinoma. Clin. Cancer Res. 2014, 20, 5937–5945. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  306. Catenacci, D.V.; Junttila, M.R.; Karrison, T.; Bahary, N.; Horiba, M.N.; Nattam, S.R.; Marsh, R.; Wallace, J.; Kozloff, M.; Rajdev, L.; et al. Randomized Phase Ib/II Study of Gemcitabine Plus Placebo or Vismodegib, a Hedgehog Pathway Inhibitor, in Patients With Metastatic Pancreatic Cancer. J. Clin. Oncol. 2015, 33, 4284–4292. [Google Scholar] [CrossRef] [PubMed]
  307. Li, Y.; Rogoff, H.A.; Keates, S.; Gao, Y.; Murikipudi, S.; Mikule, K.; Leggett, D.; Li, W.; Pardee, A.B.; Li, C.J. Suppression of cancer relapse and metastasis by inhibiting cancer stemness. Proc. Nat. Acad. Sci. USA 2015, 112, 1839–1844. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  308. El-Rayes, B.F.; Shahda, S.; Starodub, A.; O’Neil, B.H.; Hanna, W.T.; Shaib, W.L.; Oh, C.; Li, W.; Li, Y.; Borodyansky, L.; et al. A phase Ib extension study of cancer stemness inhibitor BB608 (napabucasin) in combination with gemcitabine and nab-paclitaxel (nab-PTX) in patients (pts) with metastatic pancreatic cancer. J. Clin. Oncol. 2016, 34, 4128. [Google Scholar] [CrossRef]
  309. Langleben, A.; Supko, J.G.; Hotte, S.J.; Batist, G.; Hirte, H.W.; Rogoff, H.; Li, Y.; Li, W.; Kerstein, D.; Leggett, D.; et al. A dose-escalation phase I study of a first-in-class cancer stemness inhibitor in patients with advanced malignancies. J. Clin. Oncol. 2013, 31, 2542. [Google Scholar] [CrossRef]
  310. Jonker, D.J.; Nott, L.; Yoshino, T.; Gill, S.; Shapiro, J.; Ohtsu, A.; Zalcberg, J.; Vickers, M.M.; Wei, A.C.; Gao, Y.; et al. Napabucasin versus placebo in refractory advanced colorectal cancer: A randomised phase 3 trial. Lancet Gastroenterol. Hepatol. 2018, 3, 263–270. [Google Scholar] [CrossRef]
  311. Hubbard, J.M.; Grothey, A. Napabucasin: An Update on the First-in-Class Cancer Stemness Inhibitor. Drugs 2017, 77, 1091–1103. [Google Scholar] [CrossRef]
  312. Okkenhaug, K.; Graupera, M.; Vanhaesebroeck, B. Targeting PI3K in Cancer: Impact on Tumor Cells, Their Protective Stroma, Angiogenesis, and Immunotherapy. Cancer Discov. 2016, 6, 1090–1105. [Google Scholar] [CrossRef] [Green Version]
  313. Kolev, V.N.; Wright, Q.; Vidal, C.M.; Ring, J.E.; Shapiro, I.M.; Ricono, J.; Weaver, D.T.; Padval, M.V.; Pachter, J.A.; Xu, Q. PI3K/mTOR Dual Inhibitor VS-5584 Preferentially Targets Cancer Stem Cells. Cancer Res. 2014, 75, 446–455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  314. Naujokat, C.; Steinhart, R. Salinomycin as a Drug for Targeting Human Cancer Stem Cells. J. Biomed. Biotechnol. 2012, 2012, 1–17. [Google Scholar] [CrossRef] [PubMed]
  315. Zhao, P.; Xia, G.; Dong, S.; Jiang, Z.-X.; Chen, M. An iTEP-salinomycin nanoparticle that specifically and effectively inhibits metastases of 4T1 orthotopic breast tumors. Biomaterials 2016, 93, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  316. Ferrara, N.; Hillan, K.J.; Gerber, H.-P.; Novotny, W. Discovery and development of bevacizumab, an anti-VEGF antibody for treating cancer. Nat. Rev. Drug Discov. 2004, 3, 391–400. [Google Scholar] [CrossRef]
  317. Hurwitz, H.; Fehrenbacher, L.; Novotny, W.; Cartwright, T.; Hainsworth, J.; Heim, W.; Berlin, J.; Baron, A.; Griffing, S.; Holmgren, E.; et al. Bevacizumab plus Irinotecan, Fluorouracil, and Leucovorin for Metastatic Colorectal Cancer. N. Engl. J. Med. 2004, 350, 2335–2342. [Google Scholar] [CrossRef] [Green Version]
  318. Penson, R.T.; Huang, H.Q.; Wenzel, L.B.; Monk, B.J.; Stockman, S.; Long, H.J.; Ramondetta, L.M.; Landrum, L.M.; Oaknin, A.; Reid, T.J.; et al. Bevacizumab for advanced cervical cancer: Patient-reported outcomes of a randomised, phase 3 trial (NRG Oncology-Gynecologic Oncology Group protocol 240). Lancet Oncol. 2015, 16, 301–311. [Google Scholar] [CrossRef] [Green Version]
  319. Coleman, R.L.; Brady, M.F.; Herzog, T.J.; Sabbatini, P.; Armstrong, D.K.; Walker, J.L.; Kim, B.-G.; Fujiwara, K.; Tewari, K.S.; O’Malley, D.M.; et al. Bevacizumab and paclitaxel–carboplatin chemotherapy and secondary cytoreduction in recurrent, platinum-sensitive ovarian cancer (NRG Oncology/Gynecologic Oncology Group study GOG-0213): A multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 2017, 18, 779–791. [Google Scholar] [CrossRef] [Green Version]
  320. Aalders, K.C.; Tryfonidis, K.; Senkus-Konefka, E.; Cardoso, F. Anti-angiogenic treatment in breast cancer: Facts, successes, failures and future perspectives. Cancer Treat. Rev. 2017, 53, 98–110. [Google Scholar] [CrossRef]
  321. Miller, K.; Wang, M.; Gralow, J.; Dickler, M.; Cobleigh, M.; Perez, E.A.; Shenkier, T.; Cella, D.; Davidson, N.E. Paclitaxel plus Bevacizumab versus Paclitaxel Alone for Metastatic Breast Cancer. N. Engl. J. Med. 2007, 357, 2666–2676. [Google Scholar] [CrossRef] [Green Version]
  322. Escudier, B.; Eisen, T.; Stadler, W.M.; Szczylik, C.; Oudard, S.; Siebels, M.; Négrier, S.; Chevreau, C.; Solska, E.; Desai, A.A.; et al. Sorafenib in Advanced Clear-Cell Renal-Cell Carcinoma. N. Engl. J. Med. 2007, 356, 125–134. [Google Scholar] [CrossRef]
  323. Wilhelm, S.; Carter, C.; Lynch, M.; Lowinger, T.; Dumas, J.; Smith, R.A.; Schwartz, B.; Simantov, R.; Kelley, S. Discovery and development of sorafenib: A multikinase inhibitor for treating cancer. Nat. Rev. Drug Discov. 2006, 5, 835–844. [Google Scholar] [CrossRef] [PubMed]
  324. Gounder, M.; Mahoney, M.R.; Van Tine, B.A.; Ravi, V.; Attia, S.; Deshpande, H.A.; Gupta, A.A.; Milhem, M.; Conry, R.M.; Movva, S.; et al. Sorafenib for Advanced and Refractory Desmoid Tumors. N. Engl. J. Med. 2018, 379, 2417–2428. [Google Scholar] [CrossRef] [PubMed]
  325. Kim, S.Y.; Kim, S.-M.; Chang, H.-J.; Kim, B.-W.; Lee, Y.S.; Park, C.-S.; Park, K.-C.; Chang, H.-S. SoLAT (Sorafenib Lenvatinib alternating treatment): A new treatment protocol with alternating Sorafenib and Lenvatinib for refractory thyroid Cancer. BMC Cancer 2018, 18, 956. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  326. Liu, C.; Cao, F.; Xing, W.; Si, T.; Yu, H.; Yang, X.; Guo, Z. Efficacy of cryoablation combined with sorafenib for the treatment of advanced renal cell carcinoma. Int. J. Hyperth. 2019, 36, 219–227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  327. Elgebaly, A.S.; Menshawy, A.; El Ashal, G.; Osama, O.; Ghanem, E.; Omar, A.; Negida, A. Sunitinib alone or in combination with chemotherapy for the treatment of advanced breast cancer: A systematic review and meta-analysis. Breast Dis. 2016, 36, 91–101. [Google Scholar] [CrossRef]
  328. Ferrari, S.M.; Centanni, M.; Virili, C.; Miccoli, M.; Ferrari, P.; Ruffilli, I.; Ragusa, F.; Antonelli, A.; Fallahi, P.; Martinaa, C.M.F.S. Sunitinib in the Treatment of Thyroid Cancer. Curr. Med. Chem. 2019, 26, 963–972. [Google Scholar] [CrossRef]
  329. McDermott, D.F.; Huseni, M.A.; Atkins, M.B.; Motzer, R.J.; Rini, B.I.; Escudier, B.; Fong, L.; Joseph, R.W.; Pal, S.K.; Reeves, J.A.; et al. Clinical activity and molecular correlates of response to atezolizumab alone or in combination with bevacizumab versus sunitinib in renal cell carcinoma. Nat. Med. 2018, 24, 749–757. [Google Scholar] [CrossRef]
  330. Pons, F.; Bellmunt, J. Sunitinib malate in the treatment of urothelial cancer. Expert Opin. Investig. Drugs 2013, 23, 115–124. [Google Scholar] [CrossRef]
  331. Lu, K.V.; Chang, J.P.; Parachoniak, C.A.; Pandika, M.M.; Aghi, M.K.; Meyronet, D.; Isachenko, N.; Fouse, S.D.; Phillips, J.J.; Cheresh, D.A.; et al. VEGF inhibits tumor cell invasion and mesenchymal transition through a MET/VEGFR2 complex. Cancer Cell 2012, 22, 21–35. [Google Scholar] [CrossRef] [Green Version]
  332. Jain, R.K. Normalization of Tumor Vasculature: An Emerging Concept in Antiangiogenic Therapy. Science 2005, 307, 58–62. [Google Scholar] [CrossRef]
  333. Kessenbrock, K.; Plaks, V.; Werb, Z. Matrix metalloproteinases: Regulators of the tumor microenvironment. Cell 2010, 141, 52–67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  334. Ramnath, N.; Creaven, P.J. Matrix metalloproteinase inhibitors. Curr. Oncol. Rep. 2004, 6, 96–102. [Google Scholar] [CrossRef] [PubMed]
  335. Hua, H.; Li, M.; Luo, T.; Yin, Y.; Jiang, Y. Matrix metalloproteinases in tumorigenesis: An evolving paradigm. Cell. Mol. Life Sci. 2011, 68, 3853–3868. [Google Scholar] [CrossRef] [PubMed]
  336. Coussens, L.M.; Fingleton, B.; Matrisian, L.M. Matrix Metalloproteinase Inhibitors and Cancer--Trials and Tribulations. Science 2002, 295, 2387–2392. [Google Scholar] [CrossRef]
  337. Winer, A.; Adams, S.; Mignatti, P. Matrix Metalloproteinase Inhibitors in Cancer Therapy: Turning Past Failures into Future Successes. Mol. Cancer Ther. 2018, 17, 1147–1155. [Google Scholar] [CrossRef] [Green Version]
  338. Tlatli, R.; El Ayeb, M. MMP inhibitors and cancer treatment trials, limitations and hopes for the future. Arch. l’Institut Pasteur Tunis 2013, 90, 3–21. [Google Scholar]
  339. Díaz, N.; Suárez, D. Molecular Dynamics Studies of Matrix Metalloproteases. Breast Cancer 2017, 1579, 111–134. [Google Scholar]
  340. Ray, J.M.; Stetler-Stevenson, W.G. The role of matrix metalloproteases and their inhibitors in tumour invasion, metastasis and angiogenesis. Eur Respir J. 1994, 7, 2062–2072. [Google Scholar]
  341. Vartak, D.G.; Gemeinhart, R.A. Matrix metalloproteases: Underutilized targets for drug delivery. J. Drug Target. 2007, 15, 1–20. [Google Scholar] [CrossRef] [Green Version]
  342. Levin, M.; Udi, Y.; Solomonov, I.; Sagi, I. Next generation matrix metalloproteinase inhibitors — Novel strategies bring new prospects. Biochim. Biophys. Acta (BBA) Bioenerg. 2017, 1864, 1927–1939. [Google Scholar] [CrossRef]
  343. Meisel, J.E.; Chang, M. Selective small-molecule inhibitors as chemical tools to define the roles of matrix metalloproteinases in disease. Biochim. Biophys. Acta (BBA) Bioenerg. 2017, 1864, 2001–2014. [Google Scholar] [CrossRef] [PubMed]
  344. Song, J.; Tang, J.; Guo, F. Identification of Inhibitors of MMPS Enzymes via a Novel Computational Approach. Int. J. Boil. Sci. 2018, 14, 863–871. [Google Scholar] [CrossRef] [PubMed]
  345. Zhong, Y.; Lu, Y.-T.; Sun, Y.; Shi, Z.-H.; Li, N.-G.; Tang, Y.-P.; Duan, J.-A. Recent opportunities in matrix metalloproteinase inhibitor drug design for cancer. Expert Opin. Drug Discov. 2017, 13, 75–87. [Google Scholar] [CrossRef] [PubMed]
  346. Pan, E.; Supko, J.G.; Kaley, T.J.; Butowski, N.A.; Cloughesy, T.; Jung, J.; Desideri, S.; Grossman, S.; Ye, X.; Park, D.M. Phase I study of RO4929097 with bevacizumab in patients with recurrent malignant glioma. J. Neuro Oncol. 2016, 130, 571–579. [Google Scholar] [CrossRef] [Green Version]
  347. Scala, S. Molecular Pathways: Targeting the CXCR4-CXCL12 Axis-Untapped Potential in the Tumor Microenvironment. Clin. Cancer Res. 2015, 21, 4278–4285. [Google Scholar] [CrossRef] [Green Version]
  348. Gaggianesi, M.; Turdo, A.; Chinnici, A.; Lipari, E.; Apuzzo, T.; Benfante, A.; Sperduti, I.; Di Franco, S.; Meraviglia, S.; Presti, E.L.; et al. IL4 Primes the Dynamics of Breast Cancer Progression via DUSP4 Inhibition. Cancer Res. 2017, 77, 3268–3279. [Google Scholar] [CrossRef] [Green Version]
  349. Ginestier, C.; Liu, S.; Diebel, M.E.; Korkaya, H.; Luo, M.; Brown, M.; Wicinski, J.; Cabaud, O.; Charafe-Jauffret, E.; Birnbaum, D.; et al. CXCR1 blockade selectively targets human breast cancer stem cells in vitro and in xenografts. J. Clin. Investig. 2010, 120, 485–497. [Google Scholar] [CrossRef]
  350. Goldstein, L.J.; Perez, R.P.; Yardley, D.; Han, L.K.; Reuben, J.M.; Gao, H.; McCanna, S.; Butler, B.; Ruffini, P.A.; Liu, Y. A window-of-opportunity trial of the CXCR1/2 inhibitor reparixin in operable HER-2-negative breast cancer. Breast Cancer Res. 2020, 22, 1–9. [Google Scholar] [CrossRef]
  351. Marcucci, F.; Rumio, C.; Lefoulon, F. Anti-Cancer Stem-like Cell Compounds in Clinical Development—An Overview and Critical Appraisal. Front. Oncol. 2016, 6, 115. [Google Scholar] [CrossRef] [Green Version]
  352. Singh, J.K.; Farnie, G.; Bundred, N.J.; Simoes, B.M.; Shergill, A.; Landberg, G.; Howell, S.J.; Clarke, R.B. Targeting CXCR1/2 significantly reduces breast cancer stem cell activity and increases the efficacy of inhibiting HER2 via HER2-dependent and -independent mechanisms. Clin. Cancer Res. 2013, 19, 643–656. [Google Scholar] [CrossRef] [Green Version]
  353. Gassenmaier, M.; Chen, D.; Buchner, A.; Henkel, L.; Schiemann, M.; Mack, B.; Schendel, D.J.; Zimmermann, W.; Pohla, H. CXC Chemokine Receptor 4 is Essential for Maintenance of Renal cell Carcinoma-Initiating Cells and Predicts Metastasis. Stem Cells 2013, 31, 1467–1476. [Google Scholar] [CrossRef] [PubMed]
  354. Trautmann, F.; Cojoc, M.; Kurth, I.; Melin, N.; Bouchez, L.C.; Dubrovska, A.; Peitzsch, C. CXCR4 as biomarker for radioresistant cancer stem cells. Int. J. Radiat. Boil. 2014, 90, 687–699. [Google Scholar] [CrossRef] [PubMed]
  355. Ventola, C.L. Cancer Immunotherapy, Part 2: Efficacy, Safety, and Other Clinical Considerations. Pharm. Ther. 2017, 42, 452. [Google Scholar]
  356. Sambi, M.; Bagheri, L.; Szewczuk, M.R. Current Challenges in Cancer Immunotherapy: Multimodal Approaches to Improve Efficacy and Patient Response Rates. J. Oncol. 2019, 2019, 4508794. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  357. Abril-Rodriguez, G.; Ribas, A. SnapShot: Immune Checkpoint Inhibitors. Cancer Cell 2017, 31, 848–848.e1. [Google Scholar] [CrossRef] [PubMed]
  358. Darvin, P.; Toor, S.M.; Nair, V.S.; Elkord, E. Immune checkpoint inhibitors: Recent progress and potential biomarkers. Exp. Mol. Med. 2018, 50, 1–11. [Google Scholar] [CrossRef] [Green Version]
  359. Li, B.; Chan, H.L.; Chen, P. Immune Checkpoint Inhibitors: Basics and Challenges. Curr. Med. Chem. 2019, 26, 3009–3025. [Google Scholar] [CrossRef]
  360. Mittica, G.; Genta, S.; Aglietta, M.; Valabrega, G. Immune Checkpoint Inhibitors: A New Opportunity in the Treatment of Ovarian Cancer? Int. J. Mol. Sci. 2016, 17, 1169. [Google Scholar] [CrossRef] [Green Version]
  361. Hwang, S.J.E.; Carlos, G.; Wakade, D.; Byth, K.; Kong, B.Y.; Chou, S.; Carlino, M.S.; Kefford, R.; Peñas, P.F. Cutaneous adverse events (AEs) of anti-programmed cell death (PD)-1 therapy in patients with metastatic melanoma: A single-institution cohort. J. Am. Acad. Dermatol. 2016, 74, 455–461.e1. [Google Scholar] [CrossRef] [Green Version]
  362. Xu, D.-L.; Li, Z.-Q.; Zhang, G. Research Advances on Programmed Cell Death Receptor-1 Antibody in the Treatment of Lymphoma—Review. Zhongguo Shi Yan Xue Ye Xue Za Zhi 2019, 27, 2019–2023. [Google Scholar]
  363. Zimmerman, M.P.; Mehr, S.R. Targeted programmed cell death in lung cancer treatment. Am. J. Manag. Care 2014, 20, E3. [Google Scholar] [PubMed]
  364. Okazaki, T.; Chikuma, S.; Iwai, Y.; Fagarasan, S.; Honjo, T. A rheostat for immune responses: The unique properties of PD-1 and their advantages for clinical application. Nat. Immunol. 2013, 14, 1212–1218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  365. Du, X.; Liu, M.; Su, J.; Zhang, P.; Tang, F.; Ye, P.; Devenport, M.; Wang, X.; Zhang, Y.; Liu, Y.; et al. Uncoupling therapeutic from immunotherapy-related adverse effects for safer and effective anti-CTLA-4 antibodies in CTLA4 humanized mice. Cell Res. 2018, 28, 433–447. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  366. Liu, Y.; Zheng, P. How Does an Anti-CTLA-4 Antibody Promote Cancer Immunity? Trends Immunol. 2018, 39, 953–956. [Google Scholar] [CrossRef]
  367. Rowshanravan, B.; Halliday, N.; Sansom, D. CTLA-4: A moving target in immunotherapy. Blood 2018, 131, 58–67. [Google Scholar] [CrossRef]
  368. Ribas, A.; Wolchok, J.D. Cancer immunotherapy using checkpoint blockade. Science 2018, 359, 1350–1355. [Google Scholar] [CrossRef] [Green Version]
  369. Weber, J.S.; Mandalà, M.; Del Vecchio, M.; Gogas, H.; Arance, A.; Cowey, C.L.; Dalle, S.; Schenker, M.; Sileni, V.C.; Marquez-Rodas, I.; et al. Adjuvant Nivolumab versus Ipilimumab in Resected Stage III or IV Melanoma. N. Engl. J. Med. 2017, 377, 1824–1835. [Google Scholar] [CrossRef]
  370. Gao, X.; McDermott, D.F. Ipilimumab in combination with nivolumab for the treatment of renal cell carcinoma. Expert Opin. Boil. Ther. 2018, 18, 947–957. [Google Scholar] [CrossRef]
  371. Overman, M.J.; Lonardi, S.; Wong, K.Y.M.; Lenz, H.-J.; Gelsomino, F.; Aglietta, M.; Morse, M.A.; Van Cutsem, E.; McDermott, R.; Hill, A.; et al. Durable Clinical Benefit with Nivolumab Plus Ipilimumab in DNA Mismatch Repair–Deficient/Microsatellite Instability–High Metastatic Colorectal Cancer. J. Clin. Oncol. 2018, 36, 773–779. [Google Scholar] [CrossRef]
  372. Schachter, J.; Ribas, A.; Long, G.V.; Arance, A.; Grob, J.-J.; Mortier, L.; Daud, A.N.A.; Carlino, M.S.; McNeil, C.; Lotem, M.; et al. Pembrolizumab versus ipilimumab for advanced melanoma: Final overall survival results of a multicentre, randomised, open-label phase 3 study (KEYNOTE-006). Lancet 2017, 390, 1853–1862. [Google Scholar] [CrossRef]
  373. Lee, H.T.; Lee, S.H.; Heo, Y.-S. Molecular Interactions of Antibody Drugs Targeting PD-1, PD-L1, and CTLA-4 in Immuno-Oncology. Molecules 2019, 24, 1190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  374. Shen, X.; Zhao, B. Efficacy of PD-1 or PD-L1 inhibitors and PD-L1 expression status in cancer: Meta-analysis. BMJ 2018, 362, k3529. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  375. Zhang, J.; Medeiros, L.J.; Young, K.H. Cancer Immunotherapy in Diffuse Large B-Cell Lymphoma. Front. Oncol. 2018, 8, 351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  376. Amraee, A.; Evazi, M.R.; Shakeri, M.; Roozbeh, N.; Ghazanfarpour, M.; Ghorbani, M.; Ansari, J.; Darvish, L. Efficacy of nivolumab as checkpoint inhibitor drug on survival rate of patients with relapsed/refractory classical Hodgkin lymphoma: A meta-analysis of prospective clinical study. Clin. Transl. Oncol. 2019, 21, 1093–1103. [Google Scholar] [CrossRef] [PubMed]
  377. Bair, S.M.; Strelec, L.E.; Feldman, T.A.; Ahmed, G.; Armand, P.; Shah, N.N.; Singavi, A.N.; Reddy, N.; Khan, N.; Andreadis, C.; et al. Outcomes and Toxicities of Programmed Death-1 (PD-1) Inhibitors in Hodgkin Lymphoma Patients in the United States: A Real-World, Multicenter Retrospective Analysis. Oncologist 2019, 24, 955–962. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  378. Falchi, L.; Sawas, A.; Deng, C.; Amengual, J.E.; Colbourn, D.S.; Lichtenstein, E.A.; Khan, K.A.; Schwartz, L.H.; Connor, O.J.O. High rate of complete responses to immune checkpoint inhibitors in patients with relapsed or refractory Hodgkin lymphoma previously exposed to epigenetic therapy. J. Hematol. Oncol. 2016, 9, 132. [Google Scholar] [CrossRef] [Green Version]
  379. Khunger, M.; Rakshit, S.; Pasupuleti, V.; Hernandez, A.V.; Mazzone, P.; Stevenson, J.; Pennell, N.A.; Velcheti, V. Incidence of Pneumonitis with Use of Programmed Death 1 and Programmed Death-Ligand 1 Inhibitors in Non-Small Cell Lung Cancer. Chest 2017, 152, 271–281. [Google Scholar] [CrossRef] [Green Version]
  380. Wang, D.Y.; Salem, J.-E.; Cohen, J.V.; Chandra, S.; Menzer, C.; Ye, F.; Zhao, S.; Das, S.; Beckermann, K.E.; Ha, L.; et al. Fatal Toxic Effects Associated with Immune Checkpoint Inhibitors: A Systematic Review and Meta-analysis. JAMA Oncol. 2018, 4, 1721–1728. [Google Scholar] [CrossRef] [Green Version]
  381. Su, S.; Zhao, J.; Xing, Y.; Zhang, X.; Liu, J.; Ouyang, Q.; Chen, J.; Su, F.; Liu, Q.; Song, E. Immune Checkpoint Inhibition Overcomes ADCP-Induced Immunosuppression by Macrophages. Cell 2018, 175, 442–457.e23. [Google Scholar] [CrossRef] [Green Version]
  382. Li, X.; Xu, P.; Wang, C.; Xu, N.; Xu, A.; Xu, Y.; Sadahira, T.; Araki, M.; Wada, K.; Matsuura, E.; et al. Synergistic effects of the immune checkpoint inhibitor CTLA-4 combined with the growth inhibitor lycorine in a mouse model of renal cell carcinoma. Oncotarget 2017, 8, 21177–21186. [Google Scholar] [CrossRef] [Green Version]
  383. Tsukamoto, H.; Fujieda, K.; Miyashita, A.; Fukushima, S.; Ikeda, T.; Kubo, Y.; Senju, S.; Ihn, H.; Nishimura, Y.; Oshiumi, H. Combined Blockade of IL6 and PD-1/PD-L1 Signaling Abrogates Mutual Regulation of Their Immunosuppressive Effects in the Tumor Microenvironment. Cancer Res. 2018, 78, 5011–5022. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  384. Yi, M.; Jiao, D.; Qin, S.; Chu, Q.; Wu, K.; Li, A. Synergistic effect of immune checkpoint blockade and anti-angiogenesis in cancer treatment. Mol. Cancer 2019, 18, 60. [Google Scholar] [CrossRef] [PubMed]
  385. Spranger, S.; Bao, R.; Gajewski, T.F. Melanoma-intrinsic beta-catenin signalling prevents anti-tumour immunity. Nature 2015, 523, 231–235. [Google Scholar] [CrossRef] [PubMed]
  386. Hugo, W.; Zaretsky, J.M.; Sun, L.; Song, C.; Moreno, B.H.; Hu-Lieskovan, S.; Berent-Maoz, B.; Pang, J.; Chmielowski, B.; Cherry, G.; et al. Genomic and Transcriptomic Features of Response to Anti-PD-1 Therapy in Metastatic Melanoma. Cell 2016, 165, 35–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  387. Hsu, J.-M.; Xia, W.; Hsu, Y.-H.; Chan, L.-C.; Yu, W.-H.; Cha, J.-H.; Chen, C.-T.; Liao, H.-W.; Kuo, C.-W.; Khoo, K.H.; et al. STT3-dependent PD-L1 accumulation on cancer stem cells promotes immune evasion. Nat. Commun. 2018, 9, 1908. [Google Scholar] [CrossRef]
  388. Lee, Y.; Shin, J.H.; Longmire, M.; Wang, H.; Kohrt, H.E.; Chang, H.Y.; Sunwoo, J.B. CD44+ Cells in Head and Neck Squamous Cell Carcinoma Suppress T-Cell-Mediated Immunity by Selective Constitutive and Inducible Expression of PD-L1. Clin. Cancer Res. 2016, 22, 3571–3581. [Google Scholar] [CrossRef] [Green Version]
  389. Wu, Y.; Chen, C.; Xu, Z.P.; Gu, W. Increased PD-L1 Expression in Breast and Colon Cancer Stem Cells. Clin. Exp. Pharmacol. Physiol. 2017, 44, 602–604. [Google Scholar] [CrossRef]
  390. Bruttel, V.S.; Wischhusen, J.; Wischhusen, J. Cancer Stem Cell Immunology: Key to Understanding Tumorigenesis and Tumor Immune Escape? Front. Immunol. 2014, 5, 360. [Google Scholar] [CrossRef] [Green Version]
  391. Sorrentino, C.; Ciummo, S.L.; Cipollone, G.; Caputo, S.; Bellone, M.; Di Carlo, E. Interleukin-30/IL27p28 Shapes Prostate Cancer Stem-like Cell Behavior and Is Critical for Tumor Onset and Metastasization. Cancer Res 2018, 78, 2654–2668. [Google Scholar] [CrossRef] [Green Version]
  392. >Szaryńska, M.; Olejniczak, A.; Kobiela, J.; Łaski, D.; Śledziński, Z.; Kmieć, Z. Cancer stem cells as targets for DC-based immunotherapy of colorectal cancer. Sci. Rep. 2018, 8, 12042. [Google Scholar] [CrossRef]
  393. Aliru, M.L.; Schoenhals, E.J.; Venkatesulu, B.P.; Anderson, C.C.; Barsoumian, H.B.; Younes, A.I.; Mahadevan, L.S.K.; Soeung, M.; Aziz, K.E.; Welsh, J.W.; et al. Radiation therapy and immunotherapy: What is the optimal timing or sequencing? Immunotherapy 2018, 10, 299–316. [Google Scholar] [CrossRef] [PubMed]
  394. Gotwals, P.; Cameron, S.; Cipolletta, D.; Cremasco, V.; Crystal, A.; Hewes, B.; Mueller, B.; Quaratino, S.; Sabatos-Peyton, C.; Petruzzelli, L.; et al. Prospects for combining targeted and conventional cancer therapy with immunotherapy. Nat. Rev. Cancer 2017, 17, 286–301. [Google Scholar] [CrossRef] [PubMed]
  395. Platten, M. How to integrate immunotherapy into standard of care in glioblastoma. Neuro Oncol. 2019, 21, 699–700. [Google Scholar] [CrossRef] [PubMed]
  396. Grupp, S.A.; Kalos, M.; Barrett, D.; Aplenc, R.; Porter, D.L.; Rheingold, S.R.; Teachey, D.T.; Chew, A.; Hauck, B.; Wright, J.F.; et al. Chimeric Antigen Receptor–Modified T Cells for Acute Lymphoid Leukemia. N. Engl. J. Med. 2013, 368, 1509–1518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  397. Mount, C.W.; Majzner, R.G.; Sundaresh, S.; Arnold, E.P.; Kadapakkam, M.; Haile, S.; Labanieh, L.; Hulleman, E.; Woo, P.J.; Rietberg, S.P.; et al. Potent antitumor efficacy of anti-GD2 CAR T cells in H3-K27M+ diffuse midline gliomas. Nat. Med. 2018, 24, 572–579. [Google Scholar] [CrossRef] [PubMed]
  398. Feng, K.-C.; Guo, Y.-L.; Liu, Y.; Dai, H.-R.; Wang, Y.; Lv, H.-Y.; Huang, J.-H.; Yang, Q.-M.; Han, W. Cocktail treatment with EGFR-specific and CD133-specific chimeric antigen receptor-modified T cells in a patient with advanced cholangiocarcinoma. J. Hematol. Oncol. 2017, 10, 4. [Google Scholar] [CrossRef] [Green Version]
  399. Guo, Y.-L.; Feng, K.-C.; Liu, Y.; Wu, Z.; Dai, H.; Yang, Q.-M.; Wang, Y.; Jia, H.; Han, W. Phase I Study of Chimeric Antigen Receptor–Modified T Cells in Patients with EGFR-Positive Advanced Biliary Tract Cancers. Clin. Cancer Res. 2017, 24, 1277–1286. [Google Scholar] [CrossRef] [Green Version]
  400. Cai, B.; Guo, M.; Wang, Y.; Zhang, Y.; Yang, J.; Guo, Y.; Dai, H.; Yu, C.; Sun, Q.; Qiao, J.; et al. Co-infusion of haplo-identical CD19-chimeric antigen receptor T cells and stem cells achieved full donor engraftment in refractory acute lymphoblastic leukemia. J. Hematol. Oncol. 2016, 9, 1–6. [Google Scholar] [CrossRef] [Green Version]
  401. Noh, K.H.; Kim, B.W.; Song, K.-H.; Cho, H.; Lee, Y.-H.; Kim, J.H.; Chung, J.-Y.; Kim, J.-H.; Hewitt, S.M.; Seong, S.-Y.; et al. Nanog signaling in cancer promotes stem-like phenotype and immune evasion. J. Clin. Investig. 2012, 122, 4077–4093. [Google Scholar] [CrossRef] [Green Version]
  402. Bhadury, J.; Nilsson, L.M.; Muralidharan, S.V.; Green, L.C.; Li, Z.; Gesner, E.M.; Hansen, H.C.; Keller, U.B.; McLure, K.G.; Nilsson, J.A. BET and HDAC inhibitors induce similar genes and biological effects and synergize to kill in Myc-induced murine lymphoma. Proc. Natl. Acad. Sci. USA 2014, 111, E2721–E2730. [Google Scholar] [CrossRef] [Green Version]
  403. Negmeldin, A.T.; Knoff, J.R.; Pflum, M.K.H. The structural requirements of histone deacetylase inhibitors: C4-modified SAHA analogs display dual HDAC6/HDAC8 selectivity. Eur. J. Med. Chem. 2018, 143, 1790–1806. [Google Scholar] [CrossRef] [PubMed]
  404. Nieto, Y.; Valdez, B.C.; Thall, P.F.; Jones, R.B.; Wei, W.; Myers, A.; Hosing, C.; Ahmed, S.; Popat, U.; Shpall, E.J.; et al. Double epigenetic modulation of high-dose chemotherapy with azacitidine and vorinostat for patients with refractory or poor-risk relapsed lymphoma. Cancer 2016, 122, 2680–2688. [Google Scholar] [CrossRef] [PubMed]
  405. Amiri-Kordestani, L.; Luchenko, V.; Peer, C.J.; Ghafourian, K.; Reynolds, J.; Draper, D.; Frye, R.; Woo, S.; Venzon, D.; Wright, J.; et al. Phase I trial of a new schedule of romidepsin in patients with advanced cancers. Clin. Cancer Res. 2013, 19, 4499–4507. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  406. Fouladi, M.; Furman, W.L.; Chin, T.; Freeman, B.B.; Dudkin, L.; Stewart, C.F.; Krailo, M.D.; Speights, R.; Ingle, A.M.; Houghton, P.J.; et al. Phase I Study of Depsipeptide in Pediatric Patients With Refractory Solid Tumors: A Children’s Oncology Group Report. J. Clin. Oncol. 2006, 24, 3678–3685. [Google Scholar] [CrossRef] [PubMed]
  407. Dhalluin, C.; Carlson, J.E.; Zeng, L.; He, C.; Aggarwal, A.K.; Zhou, M.-M. Structure and ligand of a histone acetyltransferase bromodomain. Nature 1999, 399, 491–496. [Google Scholar] [CrossRef]
  408. Dey, A.; Chitsaz, F.; Abbasi, A.; Misteli, T.; Ozato, K. The double bromodomain protein Brd4 binds to acetylated chromatin during interphase and mitosis. Proc. Natl. Acad. Sci. USA 2003, 100, 8758–8763. [Google Scholar] [CrossRef] [Green Version]
  409. Jang, J.E.; Eom, J.I.; Jeung, H.-K.; Cheong, J.-W.; Lee, J.Y.; Kim, J.S.; Min, Y.H. AMPK–ULK1-Mediated Autophagy Confers Resistance to BET Inhibitor JQ1 in Acute Myeloid Leukemia Stem Cells. Clin. Cancer Res. 2016, 23, 2781–2794. [Google Scholar] [CrossRef] [Green Version]
  410. Wen, N.; Guo, B.; Zheng, H.; Xu, L.; Liang, H.; Wang, Q.; Wang, D.; Chen, X.; Zhang, S.; Li, Y.; et al. Bromodomain inhibitor jq1 induces cell cycle arrest and apoptosis of glioma stem cells through the VEGF/PI3K/AKT signaling pathway. Int. J. Oncol. 2019, 55, 879–895. [Google Scholar] [CrossRef]
  411. Filippakopoulos, P.; Knapp, S. Targeting bromodomains: Epigenetic readers of lysine acetylation. Nat. Rev. Drug Discov. 2014, 13, 337–356. [Google Scholar] [CrossRef]
  412. Alqahtani, A.; Choucair, K.; Ashraf, M.; Hammouda, D.M.; Alloghbi, A.; Khan, T.; Senzer, N.; Nemunaitis, J. Bromodomain and extra-terminal motif inhibitors: A review of preclinical and clinical advances in cancer therapy. Futur. Sci. OA 2019, 5, FSO372. [Google Scholar] [CrossRef] [Green Version]
  413. Fu, L.-L.; Tian, M.; Li, X.; Li, J.-J.; Huang, J.; Ouyang, L.; Zhang, Y.; Liu, B. Inhibition of BET bromodomains as a therapeutic strategy for cancer drug discovery. Oncotarget 2015, 6, 5501–5516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  414. Chen, L.; Alexe, G.; Dharia, N.V.; Ross, L.; Iniguez, A.B.; Conway, A.S.; Wang, E.J.; Veschi, V.; Lam, N.; Qi, J.; et al. CRISPR-Cas9 screen reveals a MYCN-amplified neuroblastoma dependency on EZH2. J. Clin. Investig. 2017, 128, 446–462. [Google Scholar] [CrossRef] [PubMed]
  415. Shahbazi, J.; Liu, T.; Atmadibrata, B.; Bradner, E.J.; Marshall, G.M.; Lock, R.B. The bromodomain inhibitor JQ1 and the histone deacetylase inhibitor panobinostat synergistically reduce N-Myc expression and induce anticancer effects. Clin. Cancer Res. 2016, 22, 2534–2544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  416. Li, J.; Hao, D.; Wang, L.; Wang, H.; Wang, Y.; Zhao, Z.; Li, P.; Deng, C.; Di, L.-J. Epigenetic targeting drugs potentiate chemotherapeutic effects in solid tumor therapy. Sci. Rep. 2017, 7, 4035. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  417. Raynal, N.J.-M.; Da Costa, E.M.; Lee, J.T.; Gharibyan, V.; Ahmed, S.; Zhang, H.; Sato, T.; Malouf, G.G.; Issa, J.-P.J. Repositioning FDA-Approved Drugs in Combination with Epigenetic Drugs to Reprogram Colon Cancer Epigenome. Mol. Cancer Ther. 2016, 16, 397–407. [Google Scholar] [CrossRef] [Green Version]
  418. Klaus, C.R.; Iwanowicz, D.; Johnston, D.; Campbell, C.A.; Smith, J.J.; Moyer, M.P.; Copeland, R.A.; Olhava, E.J.; Scott, M.P.; Pollock, R.M.; et al. DOT1L Inhibitor EPZ-5676 Displays Synergistic Antiproliferative Activity in Combination with Standard of Care Drugs and Hypomethylating Agents in MLL-Rearranged Leukemia Cells. J. Pharmacol. Exp. Ther. 2014, 350, 646–656. [Google Scholar] [CrossRef] [Green Version]
  419. Ramadoss, M.; Mahadevan, V. Targeting the cancer epigenome: Synergistic therapy with bromodomain inhibitors. Drug Discov. Today 2018, 23, 76–89. [Google Scholar] [CrossRef]
  420. Abbaszadegan, M.R.; Bagheri, V.; Razavi, M.S.; Momtazi-Borojeni, A.A.; Sahebkar, A.; Gholamin, M. Isolation, identification, and characterization of cancer stem cells: A review. J. Cell. Physiol. 2017, 232, 2008–2018. [Google Scholar] [CrossRef]
  421. Qin, W.; Zheng, Y.; Qian, B.; Zhao, M. Prostate Cancer Stem Cells and Nanotechnology: A Focus on Wnt Signaling. Front. Pharmacol. 2017, 8, 153. [Google Scholar] [CrossRef] [Green Version]
  422. Xu, Y.; Chenna, V.; Hu, C.; Sun, H.-X.; Khan, M.; Bai, H.; Yang, X.-R.; Zhu, Q.-F.; Sun, Y.-F.; Maitra, A.; et al. Polymeric Nanoparticle Encapsulated Hedgehog Pathway Inhibitor HPI-1 (“NanoHHI”) Inhibits Systemic Metastases in an Orthotopic Model of Human Hepatocellular Carcinoma. Clin. Cancer Res. 2011, 18, 1291–1302. [Google Scholar] [CrossRef] [Green Version]
  423. Burke, A.R.; Singh, R.; Carroll, D.L.; Wood, J.C.S.; D’Agostino, R.B.; Ajayan, P.M.; Torti, F.M.; Torti, S.V. The resistance of breast cancer stem cells to conventional hyperthermia and their sensitivity to nanoparticle-mediated photothermal therapy. Biomaterials 2012, 33, 2961–2970. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  424. Yao, H.-J.; Zhang, Y.; Sun, L.; Liu, Y. The effect of hyaluronic acid functionalized carbon nanotubes loaded with salinomycin on gastric cancer stem cells. Biomaterials 2014, 35, 9208–9223. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Cancer stem cells are able to resist conventional therapies and form new tumors, unless targeted by cancer stem cell (CSC)-specific therapy. Adapted from Dzobo et al. [30].
Figure 1. Cancer stem cells are able to resist conventional therapies and form new tumors, unless targeted by cancer stem cell (CSC)-specific therapy. Adapted from Dzobo et al. [30].
Cells 09 01896 g001
Figure 2. Cancer stem cell markers expressed in some human cancers are shown in the figure. Figure adapted from Dzobo et al. [34]. See Table 1 for references. The list of CSC markers is not exhaustive. The CSC markers continue to be refined based on new data.
Figure 2. Cancer stem cell markers expressed in some human cancers are shown in the figure. Figure adapted from Dzobo et al. [34]. See Table 1 for references. The list of CSC markers is not exhaustive. The CSC markers continue to be refined based on new data.
Cells 09 01896 g002
Figure 3. Hallmarks of cancer stem cells include increased expression of ATP-binding cassette (ABC) membrane transporters, enhanced survival signaling, increased drug in activation as well as increased DNA repair processes compared to cancer cells. This allows CSCs to survive conventional therapy and thus contribute to chemoresistance for example. Adapted from Senthebane et al. [3].
Figure 3. Hallmarks of cancer stem cells include increased expression of ATP-binding cassette (ABC) membrane transporters, enhanced survival signaling, increased drug in activation as well as increased DNA repair processes compared to cancer cells. This allows CSCs to survive conventional therapy and thus contribute to chemoresistance for example. Adapted from Senthebane et al. [3].
Cells 09 01896 g003
Figure 4. Cancer stem cells are able to reside deep within the tumor in hypoxic regions that are normally toxic to normal cells, whilst CSCs are able to release factors such as hypoxia-inducible factor 1 which induces the release of proangiogenic factors, this position means CSCs are inaccessible to drugs or are exposed to reduced drug doses. Adapted from Senthebane et al. [3].
Figure 4. Cancer stem cells are able to reside deep within the tumor in hypoxic regions that are normally toxic to normal cells, whilst CSCs are able to release factors such as hypoxia-inducible factor 1 which induces the release of proangiogenic factors, this position means CSCs are inaccessible to drugs or are exposed to reduced drug doses. Adapted from Senthebane et al. [3].
Cells 09 01896 g004
Table 1. CSC markers expressed in different human cancers *.
Table 1. CSC markers expressed in different human cancers *.
CancerCSC MarkersReferences
CervicalCD133+, CD49f+, CK-17+[35,36,37]
EsophagealCD44+, ALDH1+, Integrin α7+[38,39]
KidneyCD24-, CD44+, CD105+, CD133+[40,41,42]
Lung cancerCD44+, CD90+, CD133+, ABCG2+, ALDH+ [24,33]
Colon cancerCD24+, CD44+, CD133+, EpCAM+, ALDH+[43,44,45,46]
Liver cancerCD24+, CD44+, CD90+, CD133+, ALDH+, ABCG2+ [47,48]
Breast cancerCD24-, CD44+, CD133+, ALDH-1+[5,49,50]
GastricCD44+, CD133+[51,52,53]
Glioma CD44+, CD133+, A2B5+, BCRP1+, SSEA-1+[54,55]
Leukemia (AML)CD34+, CD38−, CD123+[56,57,58]
Leukemia (CML)CD25+, CD26+, CD44+, CD93+, IL1RAP+[59,60]
Ovarian CD44+, CD117+, CD133+, ALDH1+[61,62]
Prostate cancerCD44+, CD133+, α2β1+, ALDH+[63,64,65]
Pancreatic cancerCD44+, CD133+, ABCG2+, ALDH+, EpCAM+[66,67,68]
MelanomaABCB5+, CD20+[69,70]
Head and neck cancerCD44+, CD133+[71,72]
SarcomaCD29+, CD117+, CD133+, Nestin+, Stro-1+[73,74]
* The list of CSC markers is not exhaustive. The CSC markers continue to be refined based on new data.
Table 2. Drugs currently under trial in combination with chemotherapy and radiotherapy for the treatment of different cancers.
Table 2. Drugs currently under trial in combination with chemotherapy and radiotherapy for the treatment of different cancers.
Cancer TypeChemotherapy/Radiotherapy/ImmunotherapyClinical Trial Identifier
Breast Ruxolitinib + ChemotherapyNCT02876302
Lapatinib + RadiotherapyNCT01868503
Paclitaxel + ReparixinNCT02370238
Paclitaxel + ReparixinNCT02001974
Vorinostat + LapatinibNCT01118975
MK-0752 + Docetaxel + PegfilgrastimNCT00645333
ColorectalOMP-305B83 + FOLFIRI + FOLFOXNCT03035253
Napabucasin + Fluorouracil + Leucovorin + Irinotecan + BevacizumabNCT02753127
OMP-21M18NCT01189942
EsophagealDietary Supplement: FursultiamineNCT02423811
GastrointestinalPhase 1: BBI608
Phase 2: Fluorouracil + Oxaliplatin + Leucovorin + Irinotecan + Bevacizumab + Capecitabine + Regorafenib
NCT02024607
Glioma3-Dimensional Conformal Radiation Therapy + Gamma-Secretase Inhibitor RO4929097 + Intensity-Modulated Radiation Therapy + Temozolomide NCT01119599
ChemoID assay + ChemotherapyNCT03632135
Stem Cell Radiotherapy (ScRT) + TemozolomideNCT02039778
Head and NeckIPI-926 + CetuximabNCT01255800
HematologicAzacitidine + SL-401 + VenetoclaxNCT03113643
Lenalidomide + Dexamethasone + MEDI-551NCT01861340
ZileutonNCT01130688
HepatocellularBBI608 + BBI503 + SorafenibNCT02279719
MetforminNCT01442870
OvarianChemotherapyNCT03632798
Carboplatin + Paclitaxel + Ruxolitinib + Ruxolitinib PhosphateNCT02713386
MetforminNCT01579812
PancreaticGamma-secretase/Notch signaling pathway inhibitor RO4929097NCT01192763
Demcizumab + Abraxane® + GemcitabineNCT01189929
Cyberknife radiation + gemcitabineNCT01051284

Share and Cite

MDPI and ACS Style

Dzobo, K.; Senthebane, D.A.; Ganz, C.; Thomford, N.E.; Wonkam, A.; Dandara, C. Advances in Therapeutic Targeting of Cancer Stem Cells within the Tumor Microenvironment: An Updated Review. Cells 2020, 9, 1896. https://doi.org/10.3390/cells9081896

AMA Style

Dzobo K, Senthebane DA, Ganz C, Thomford NE, Wonkam A, Dandara C. Advances in Therapeutic Targeting of Cancer Stem Cells within the Tumor Microenvironment: An Updated Review. Cells. 2020; 9(8):1896. https://doi.org/10.3390/cells9081896

Chicago/Turabian Style

Dzobo, Kevin, Dimakatso Alice Senthebane, Chelene Ganz, Nicholas Ekow Thomford, Ambroise Wonkam, and Collet Dandara. 2020. "Advances in Therapeutic Targeting of Cancer Stem Cells within the Tumor Microenvironment: An Updated Review" Cells 9, no. 8: 1896. https://doi.org/10.3390/cells9081896

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