Open access peer-reviewed chapter

Cancer Stem Cells — Perspectives and How to Target Them

Written By

Zhaopeng Tang, Qianfeng Wang, Sarah Shigdar, Wei Duan and Dongxi Xiang

Submitted: 02 April 2015 Reviewed: 28 October 2015 Published: 10 February 2016

DOI: 10.5772/61861

From the Edited Volume

Smart Drug Delivery System

Edited by Ali Demir Sezer

Chapter metrics overview

2,257 Chapter Downloads

View Full Metrics

Abstract

Cancer stem cell is a progressive concept moving forward to interpret the hard-to-cure nature of cancer and the relevant behavior in response to clinical therapies. Despite the remaining debates regarding the existence of cancer stem cells, the cancer stem cell model provides a potential approach for advanced innovative therapies targeting the “roots” of cancer, which has enhanced treatment outcomes. This chapter summarizes advanced perspectives in the field of cancer stem cell research, including experimental strategies for targeting these cells, highlights challenges of this theory, and explores feasible therapeutic strategies for overcoming the intrinsic resistance of cancer stem cells to clinical treatment.

Keywords

  • Cancer stem cell
  • stemness
  • chemoresistance
  • targeted therapy

1. Introduction

Cancer remains one of the leading causes of human death, and even though extraordinary efforts and budget have been spent, clinical trials in the eradication or control of cancer progression have generally created disappointing outcomes [1, 2]. Cancer develops originally from normal cells through the accumulation of multiple genetic alterations that ultimately convert to malignant phenotypes [3, 4]. Despite a better understanding of cancer biology and evolutionary genomic characteristics, translating these achievements into feasible and successful clinical outcomes continues to be a problem [5]. One attractive theory, the concept of cancer stem cells (CSCs), being explored recently in cancer research may hold the answer [6]. This chapter summarizes the major characteristics of CSCs and highlights several key approaches used for CSC study, where they could be of help for efficient CSC-targeted therapy.

Advertisement

2. CSC: An involving concept moving forward

2.1. Cancer genetic evolution

Cancer biology and genomics have increasingly validated cancer as a complex adaptive system. This landmark perspective was introduced by Peter Nowell in 1976, who viewed cancer development as an evolutionary process driven by stepwise mutations with sequential, subclonal selection at the nuclei acid level, a theory that is similar to Darwinian natural selection [7]. This theory includes several key aspects. (1) Genetic instability: cells possess a battery of mechanisms to preserve their DNA structural integrity. Cellular genetic structural varies (deletions, duplications, and rearrangements) or DNA point mutations can initiate cell biological changes that may lead to tumor formation. Thus, the perturbation of mechanisms controlling genomic stability is responsible for the oncogenic processes [8]. (2) Error-prone repair processes and a genotoxic exposure could result in particular mutational spectra of cancer cells, including cigarette carcinogens, ultraviolet light, and chemotherapeutics. (3) Recurrent, mutation specific traits in cancer can potentially affect clonal selection [9] (Figure 1).

Figure 1.

Genetic diversification and clonal dynamics of cancer development.

The microenvironment surrounding a tumor complexes with multiple cellular components that provide an adaptive landscape and necessary elements for natural selection of cancer growth [10]. The interaction between cancer cells and their surroundings is mutually beneficial [11]. Cancer cells can reset the extracellular environment to be specialized niches for further cancer growth and migration. The cancer niche in turn provides external signals for cancer-clone expansion and tumor cell survival and proliferation. The spatial heterogeneity is critical for evolving cancer cell to be a malignant phenotype and beneficial for cell migration and distant invasion [12]. During the process of cancer-clone expansion, migrating cells invade new habitats where they face new selective pressures, thereby increasing the rate of further cancer mutation [13].

The cancer niche is an unclosed system. In addition to the tissue positions and associated etiological environmental exposure of patients, the tissue microecosystem is manipulated by multiple systemic or external factors [14]. Genotoxic exposures, such as ultraviolet light, pathogenic infection, and long-term dietary habits, are able to modulate energy metabolism. They are speculated to be the primary etiological causes of tumor initiation and further evolution of cancer clones [14]. Cancer cell ecosystems can be altered following clinical treatment, in which most cancer cells together with healthy tissues are killed under intensive therapies. However, some specialized niches protect cells from cancer treatment, where variant or therapy-resistant cancer cells emerge [15]. On the way of cancer progression and tumor recurrence after therapy, the primary unit of selection is a specialized cell, known as CSC, which possesses extensive self-renewal potential (Figure 2).

Figure 2.

The cancer ecosystem.

2.2. Cancer stem cell model

Cancer research was previously dominated by the clonal evolution model, also known as a conventional stochastic model, a concept whereby all cells within a tumor have equal potential to propagate and maintain a tumor following stepwise genetic and/or epigenetic changes, but they are hard to identify the tumorigenic subset [7, 1619]. Recently, accumulated studies suggest cancers as hierarchical organizations, which is the basis of the hierarchical or CSC model [20]. In the CSC model, only a small subset of cancer cells possesses the ability to self-renew, differentiates, and reforms a tumor. CSCs, as “roots of cancer” operating in a hierarchical fashion, are defined by their abilities [2024] (1) to form new tumors with high efficiency that histologically resembles the original tumors when xenotransplanted into immunodeficient mice, (2) to generate descendant cells possessing unlimited self-renewal (regeneration) potential but uncommitted differentiation options, and (3) to generate large populations of differentiated offspring and progenitor cells that exit the stem cell state and lose the ability to self-renew, thus no longer possessing tumorigenic potential. Such differentiated daughter cells can undergo limited rounds of cell division because they lack the intrinsic clonogenic characteristics that are essential for tumor initiation and long-term progression of the malignancy. The increasingly widespread attraction of the CSC model is due to the fact that it can provide a plausible account for poorly understood clinical phenomena, such as therapy resistance, as CSCs can resume growth and contribute to a new relapse after therapy suspension. It is critical to appreciate that both the stochastic and CSC models share the same conviction that only a small population of cancer cells are capable of maintaining cancer. The main difference is that CSCs within the CSC model are characterized with the help of distinct cellular phenotypes [20].

Critically, the CSC model stands on the basis that CSCs are reliable and stable over time, and their unique traits could not be obtained by differentiated descendants. It is worth to note that the persuading of CSC model has been accompanying with intense debates amongst cancer researchers. As evident from a new concept, the dynamic CSC model suggests that the CSC phenotype is much more fluid than previously predicted and can be regulated by external signals [25, 26]. Thus, not only can CSCs self-renew to create new progeny and differentiate to non-CSCs but the de-differentiation of non-CSCs to CSCs can also occur and thus return to the malignancy growth cycle [2628]. This latest finding has significant implications for oncology in that future effective anticancer therapeutic strategies should aim at targeting both CSCs and non-CSCs (Figure 3).

Figure 3.

Stochastic cancer model versus hierarchical and dynamic CSC models of tumor heterogeneity.

Advertisement

3. Challenges and opportunities the CSC theory faces

Several challenges the CSC theory is now facing should be addressed before this concept can fetch benefits for the clinically relevant entity [29].

3.1. Frequency of CSCs

One challenge that is intensely debated is how many CSCs exist within a tumor. Some studies suggested that the frequency of CSCs is less than 1 in every 1000 cells in a tumor, which supports the hierarchical status between CSCs and non-CSCs [30, 31]. However, recent evidences reported that the number of cells that possess intrinsic tumorigenic potential is relatively higher [32, 33]. Hepatocytes possess the potential of self-renewal and unlimited proliferation under certain conditions [34, 35]. It is shown that various colorectal cancers do not follow the same hierarchy as their CSC frequencies vary [36]. More importantly, the prediction of the number of CSCs in a particular tumor type depends on multiple manual factors, including the experimental procedure designed, the choice of cell surface markers, and the types of immunodeficient mice into which the CSCs were injected. Chiba et al. used the technique of side population (SP) analysis to detect the subpopulations that act CSC functions and revealed that SP cells possess abilities of high proliferation and antiapoptosis in both Huh7 and PLC/PRF/5 cells with a percentage of 0.25% and 0.8%, respectively [37]. Kimura et al. determined the frequency of CSCs by analyzing cell surface markers and the percentage of CD133+ cells in Huh7 and PLC/PRF/5 were 2.7% and 23.5%, which is totally different with the number obtained on the base of EpCAM-positive cells, with a proportion of 89.1% and 40.7%, respectively [38]. However, Cao et al. found the CSC frequency of PLC/PRF/5 cells was 8.8% by using the sphere-forming assay [39]. Thus, considering a tumor as a hierarchical malignancy has intrinsic limitations, as a large population of cancer cells cooperate and contribute to tumor growth, which is consistent with the conventional nonhierarchical model of malignancies [40].

3.2. Therapy resistance of CSCs

The failure of cancer initial treatment is normally explained due to the presence of a subset of resistant cancer cells. CSCs are supposed to be one of such cells; they are logically resistant to traditional chemotherapies owing to their “stem-like” features, including enhanced abilities of DNA damage repair and cellular detoxification capacity via elevated aldehyde dehydrogenase (ALDH) activity, increased expression of enhancement of ATP-binding cassette (ABC) surface transporters, and their nature quiescence [4143]. CSCs with an up-regulation of antiapoptotic molecules or a high expression of drug efflux pumps respond quite differently when exposed to drugs; they can both survive after cancer treatment [44]. The rapid relapse of the malignancy suggests that the clonogenic core of the cancer was not effectively targeted, and this might be due to the de-differentiation of non-CSCs to CSCs [45]. Even though CSCs are believed to be the most promising candidates for overcoming chemoresistance and tumor recurrence, both CSCs and non-CSCs should be targeted eventually.

3.3. Isolation of CSCs

The isolation and subsequent assessment of CSCs are clearly a rapidly developing area in which diverse strategies were incorporated for obtaining higher CSC purities. One continual concern after isolation assays is that whether the proposed CSCs separated from the bulk of the tumor can still possess the intrinsic cellular properties. The selected CSCs require the distinct phenotypes that can generate a similar malignancy to the parental tumor when transplanted into immunodeficient mice [46]. The subsequent challenge is to optimize experimental procedures to allow xenotransplantation of viable single suspension cells into animals.

3.4. Targeting “confirmed” CSCs

Due to the relevance of cancer development, CSCs are ideal targets for molecular-directed therapies. The limitation is that CSCs are typically present at very low levels. In addition, CSC markers overlap with normal stem cells or between CSCs [47]. These drawbacks indicate that the identification of CSC markers for guiding molecular therapeutics is still in its infancy. Many CSC markers are not strictly CSC antigens since they present on normal cancer cells as well, but they are molecules that support CSCs in their niche.

The characterization of CSCs has led to some experimentally “confirmed” markers or markers involved in the process of targeting or interaction with CSCs. Epithelial cell adhesion molecule (EpCAM) is one of the most highly expressed tumor-associated markers, being found in a broad range of epithelial cancers. EpCAM has been “rediscovered” as a CSC antigen in breast, colon, prostate, and pancreatic cancers [4850]. In studies on colon cancer tumorigenicity, EpCAMhighCD44+ was considered as a robust marker of more “stem-like” subpopulations. Also, recruiting effector cells to tumors using an anti-EpCAM/anti-CD3 bispecific antibody has been shown to be a promising strategy in the treatment of cancer [51].

In addition to cell-surface markers that are frequently utilized to identify CSCs, the high activity of signal transduction routes can also contribute to CSCs features [52]. The self-renewal capacity of CSCs leads to a long-term clonogenicity mutation, which might be effectively therapeutically targeted. The activation of intracellular signaling pathways associated with the self-renew of CSCs, including Wnt pathway, Hedgehog (Hh) pathway, and Notch pathway, can stimulate a more immature tumor phenotype, facilitate tumor invasion, and promote therapy resistance [53]. The interference of such pathways that promote CSC function might provide an effective therapeutic window for drugs in the war against cancer. These stem cell-associated surface marker proteins and pathways are promising targets for anticancer drug development

Advertisement

4. Approaches used for studying CSCs

The primary challenge to study CSCs is the ability to identify and investigate CSCs in laboratories using both in vitro and in vivo assays. This section addresses the methodologies that have been widely used for isolation and characterization of CSCs.

4.1. In vivo xenotransplantation and limiting dilution assay: The gold standard

Limiting dilution assays (LDAs) have been used in a broad variety of biological fields. LDA is an experimental technique that attempts to quantify the frequency of biological particles that perform a particular function within a larger mixed population [54]. The aim of LDA is to obtain highly precise data at the macro level. To achieve this goal, several conditions must be considered [55, 56]. First, the cells should be Poisson distributed. Second, the assay should be designed with maximum information containing both negative and positive cultures. Moreover, the conditions must be performed at the state that the response of a single limiting particle can be detected. Thus, the culture conditions should be uniform in all paralleled groups and wells [56].

The ability to determine the number of CSCs is a critical step for the success of in vivo transplantation. However, there has been little or no attempt to standardize this approach [57]. In all conditions tested, the statistical behavior of the system (Es) and changes in condition over time and statistical noise were the two major contributors to overall uncertainty [57]. However, as expected from statistical principles, Es declines with increasing number of replicates analyzed. Also, increasing the number of replicates beyond 96 is unlikely to provide substantial decreases in error, and fewer wells can be used to obtain results with similar precision. Another requirement that arises in stem cell research is the need to accommodate small numbers of replicates in a statistically consistent and defensible manner. Operator error, mouse-to-mouse error, and other errors appear to play minor roles. These findings establish parameters that contribute to the variability of LDA and provide strategies for the optimization and interpretation of the LDA-based CSC estimates. To this end, extreme limiting dilution analysis (ELDA) has emerged as the preferred method that is based on sound statistical principle and methodologies [58]. ELDA works well when the number of replicates is small.

The CSC concept brings essential predictions of cancer research including susceptibility to chemotherapy, aggressiveness of the disease, and the pace of recurrence that may largely be influenced by the functional properties of CSCs [48, 49, 5961]. To functionally measure CSC potential, the operational assay of evaluating CSCs has been developed. The creation of an array of genetically modified immunodeficient mice enabling analogous xenograft experiments can be used to determine and quantify cells that obtain tumor-initiating activity in human tumors [48, 49, 5961]. These xenograft models are considered the “gold standard” in CSC research; they are able to guide the biology and therapeutic responses of human CSCs.

The enumeration of CSCs has been performed using phenotypic markers in in vitro limiting dilution transplantation assays. Some key aspects should be considered [32]: (1) inefficient engraftment in mice may result in underestimation of the actual CSC frequency; (2) phenotypic characterization of CSCs may lead to different rates depending on the antibody combinations used, and the use of specific antibodies may induce immune-mediated clearance of CSCs [48]; (3) the approach of tumor implantation (e.g., intravenously, intrafemorally) critically defines engraftment and affects the export of stem cell frequency [62]; and (4) the kind of mouse strain used as tumor recipient and the degree of mouse immunodeficiency are other factors need to be considered [32]. It is reported that the use of adjuvants/culture supplementary (e.g., Matrigel or growth factors) influences not only the measured frequency of CSCs but their phenotypes as well [30, 32].

4.2. The side population assay

In addition to the isolation and identification of CSCs through a set of cell surface markers (such as CD44+/CD24low/lin-/ALDH+, CD44+/CD24low, EpCAM+/CD44+/CD24–, or CD34+/CD38–). The SP discrimination assay is another flow cytometry method used to detect stem cells based on the properties of the Hoechst dye efflux via the ABC transporters [63]. The SP assay has been used to identify stem cell and progenitor populations in various tissues [64, 65]. However, several reports have stated that dye efflux is not a universal property of all stem cells although SPs are much abundant within stem cells [66]. SPs are not just restricted to the stem cell phenotypes as ABC transporters are also expressed by specialized cells in certain organs [67]. The ABC transporters are believed to play a key role in those tissues, protecting the cytotoxic effects of toxins [68]. However, the identification of CSCs has raised interests in the SP assay, and SP subpopulations isolated from some cancers possess capabilities of drug-resistant, self-renewal, and tumorigenicity when transplanted into immunocompromised mice [63]. The SP assay can play an extremely valuable role in the primary isolation and identification of potential stem/progenitor cells, when specific cell surface markers were absent.

4.3. Clonal sphere formation assay and proliferation (invasion assay) assay

The clonal sphere formation assay is another functional method used for the identification of CSCs and their purification from the rest of neighboring cells [69, 70]. This assay begins with reliable single cell suspensions originating from primary tumors or established cancer cell lines. Serial dilutions of the single cancer cell suspensions are seeded on ultra-low attachment substrata in the presence of serum-free media with growth factors [71]. CSCs can grow into 3D nonadherent structures called spheres, while non-CSCs cannot survive in such culture conditions. The results of the assay may become more reliable if the spheres are serially passaged. The self-renewal capacity of sphere-generating cells can be estimated by evaluating the sphere formation frequency through LDA and measuring sphere size [72]. The sphere formation assay has been used to identify adult stem cells or CSCs from a number of tissues, including mammary gland, brain, skin, and human melanoma [7376].

4.4. Enzymatic assay of cell-surface markers

CSCs can be isolated by the enzymatic activities (ALDH activity) [77, 78]. The measurement of ALDH activity by ALDEFLUOR staining is useful to screen tumor cells for resistance to alkylating agents and to identify heterogeneity within tumor cells [79]. The ALDH+ population is consistent with CSC characteristics, generating tumors that recapitulate the phenotypic heterogeneity of the initial tumor. The measurement of ALDH populations revealed that the ALDH+ cells were capable of self-renewing into both ALDH+ and ALDH− cells [80]. A recent study in melanoma described both ALDH+ and ALDH− cells derived from patient biopsies (100 or 2000 cells, respectively) were able to efficiently form tumors. It is also reported that ALDH cells alone may not be sufficient for CSC selection [32]. Despite this, studies in breast cancer combining other CSC surface markers with Aldefluor have improved tumorigenic enrichment and this combination may prove to be a better strategy for enrichment of CSCs [32].

4.5. Slow-cycling population assay

The slow-cycling population assay can be used to distinguish CSCs from progenitor cells, which determines the incorporation of labeled precursor of nucleotides for cellular DNA synthesis [81]. The slow-cycling cells, such as CSCs, maintaining sufficient labels allow their detection by the anti-BrdU antibody staining or radioactive label [82]. Some technical issues impede the accuracy of BrdU detection: (1) it is hard to demonstrate that cells have a similar cycling in vitro and in vivo because of the destructive nature of the BrdU detection procedure, (2) it is not expected that all the stem cells can be equally labeled because the BrdU incorporation occurs only during the S phase of cell cycle, and (3) the label retention depends on the length of the cell cycle, while the later status appears to alter as the organ matures [83]. Thus, quantification of stem cells through this assay requires further confirmation through other methods.

4.6. Lineage labeling assay

The lineage labeling assay was initially developed for understanding and tracing the biological developments of a cell, including the identification of cell origin, measuring lineage relationships, and determination of division patterns [84]. In this assay, cells are labeled (usually with a fluorescent dye or tag), followed by tracing in vitro and in vivo. There are several inherent limitations with this technique: (1) the experimental procedure can damage cells or some cells may have weak and/or transient expression of markers and unrestricted clonality [84] and (2) with whole-mount labeling, it is not possible to identify spatial relationships between stem cells and its transit amplifying progeny [85]. In this case, targeted cells are noninvasively labeled in their native environment, and the development of a progenitor cell and a composition of their lineages can be followed. It remains unclear whether the combination of these techniques can be extended to identify CSCs and metastatic initiating stem/progenitor cells.

While there has been a hot debate in recent years as to whether the CSC theory is correct, very recent lineage tracing studies have provided proof that a relatively small number of cells are capable of generating and maintaining a tumor. These studies used fluorescently labeled tumor cells and showed that the quiescent cells remained after further in vivo passages [86]. Furthermore, following treatment, these quiescent cells started to proliferate and generate proliferative progenitor cells that were capable of maintaining a tumor [8789]. By eliminating these quiescent cells, tumor growth was impaired. These results link back to the theory that leaving a small population of CSCs after the conventional treatment lead to a recurrence of the tumor. Therefore, it is necessary to eliminate both populations of cells within a tumor to produce effective anticancer therapeutic strategies [90].

4.7. CSC epigenome and next-generation sequencing

Whole genome sequencing has made tremendous contributions to cancer research, which enables researchers to discover and understand the rules of cancer development at the nucleic acid level [91, 92]. However, the genome instability and genetic variants limit to demonstrate cancer progression. It was speculated that there exist a senior layer of information, besides genome sequence for regulating differential gene expressions and thus determining cancer evolving. This concept was early prospected by Conrad Hal Waddington in 1942 and subsequently described as “epigenetic control system” by Nanney in 1958 [93, 94]. Epigenetics primarily refers to the study of chromosome variations that modulate gene transcription without alterations in the DNA sequences [95, 96]. Epigenome contains genetic information, not as stable as genome, representing the cellular epigenetic state varying with influence from external factors. Mechanisms produce such changes mainly include DNA methylation, histone modifications, nucleosome positioning, and chromatin remodeling [96]. These epigenetic regulation mechanisms are reported to be closely related with the gaining of stem cell-specific properties, whereby contributing to tumor inheterogeneous [97, 98]. Epigenetic alterations would offer survival benefits in CSC subpopulation which promotes the genetic expression to the self-renewal state, resulting in tumor initiation and further progression [99, 100]. The relevance of the DNA methylation and histone modifications in CSC regulation, subsequently with advancing tumor growth, were recently illustrated in various cancer models [97, 99, 101].

Next-generation sequencing (NGS), also known as high-throughput sequencing, has revolutionized the study of genomics and opened a new chapter of the epigenome research of cancer and stem cell [102, 103]. Four mainly NGS-based approaches have been developed to identify epigenome research, including methylated DNA immunoprecipitation sequencing (MeDIP-Seq), whole genome bisulfite sequencing (WGBS), reduced representation bisulfite sequencing (RRBS), and chromatin immunoprecipitation sequencing (ChIP-Seq). Compared with the previous approach, the innovative advantages of NGS have significantly accelerated the sequence of DNA and RNA and promoted the scientific sightings in CSC epigenome research.

Advertisement

5. Therapeutic strategies used for targeting CSCs

Current failure of cancer treatment is normally due to the existence and functionalities of CSCs, which possess more chemoresistance than their non-CSC counterparts, to conventional treatments. Recently, multiple strategies have been developed for targeting and eradicating CSCs while sparing healthy tissues, thereby minimizing the patients to face therapy risks.

5.1. Targeting CSCs via surface markers

Since CSCs are attractive targets for anticancer treatment, the CSC-correlated cell surface proteins have been monitored extensively for identification, probable isolation and monitoring the variation of leukemic and solid CSCs in preclinical and clinical settings [104]. Studies using certain ligands or antibodies against CSC surface maker proteins, including EpCAM, CD47, CD44, CD90, CD133, IL-3R, immunoglobulin mucin TIM-3, ALDH+, and others, have advanced the therapeutic efficacy [105108]. Limitations, such as the expression of such cell surface makers, may vary in different stages of cancer development; only a small subpopulation of CSCs standing in the summit among bunk of cancer cells, as well as overlapping of CSC-associated marker proteins with normal stem cells, impede the effectively targeting of CSCs [47]. For bench assays, it is recommended to employ functional studies such as in vivo LDA upon xenotransplantation to ascertain CSCs, rather than solely be dependent on the incidence of a single or a combination of multiple CSC markers. However, in case of in vivo CSC-based therapy, the CSC surface marker proteins present one of the very few feasible options for targeting CSCs in animals and patients, given the practical and ethical restrictions related with lineage tracing and xenotransplantation.

Accumulated attractions have been grasped in the development of aptamers (also known as chemical antibodies) and monoclonal antibodies for targeting CSCs via targeting surface marker proteins. Shigdar et al. have isolated two RNA aptamers using SELEX for targeting CSC surface markers, EpCAM, and CD133 [109, 110]. These CSC-targeting aptamers internalize into target cells through receptor-mediated endocytosis, which is capable of circumventing ATP-binding cassette transporters that function multiple drug resistance in CSCs [26]. In addition, this newly developed EpCAM RNA aptamer is more sensitive than counterpart antibodies for detecting the surface EpCAM proteins in formalin-fixed paraffin-embedded primary breast cancers, with no nonspecific staining or cross-reactivity with non-EpCAM-expression tissues [111]. This shows the potential of aptamers for specific targeting of cancers but with minimized side effects observation. To optimize promising approaches by targeting cell surface markers might be one of the answers for tracing CSCs and eventually eradiating CSCs, thereby preventing patients from suffering from cancer attacking.

5.2. Targeting ATP-driven efflux transporters

The failure of cancer chemotherapy mainly results from increased efflux of anticancer drugs from cancer cells, resulting in the impairment of drug cellular entrance and consequent reduction of chemotherapeutic sensitivity [112]. A considerable root of multidrug resistance (MDR) involves the augmented expression of the ABC transporter superfamily, many of these transmembrane proteins are responsible for effluxion of various xenobiotics (including anticancer agents) from cancer intracellular membranes [112, 113]. The well-characterized MDR transporters consist with ABCB1 (MDR1 or P-glycoprotein), ABCC1 (MRP1), and ABCG2 (BCRP or MXR) [112, 114116]. Although projects focusing on understanding the mechanisms of these ABC transporters behind chemotherapy failure have been broadly developed, clinical outcomes in these fields have been generally unsatisfactory [113, 117]. The recurrence of primary and metastatic cites can occur following the escaped chemotherapy, in which CSCs, possessing enhanced efflux of therapeutic agents through ABC transporters, have been proven to provide major contributions [114, 118, 119].

Due to the correlation of ABC transporters with CSC phenotypes, one approach circumventing CSC-based chemoresistance involves the use of specific molecular inhibitors against certain functions of individual MDR transporters [114]. Verapamil, an agent targeting general ABC transporters, has been moved to the clinical trials, but the further study was suspended due to the dose-limiting toxicity [120]. Thus, the more specific molecules targeting individual MDR transporters are required [121]. A considerable body of evidence has shown positive outcomes of overcoming CSC drug resistance by developing physical conjugation or chemical nanoparticle-functionalized drug delivery of anticancer agents. Chou et al. indicated that the conjugation of doxorubicin (DOX) into nanodiamonds reduced efflux of DOX in MDR1 overexpressing cancer cells and facilitated the anticancer efficacy in DOX-resistant cancer models [122]. While CSC-related chemoresistance and tumor relapse arise mainly due to the increased expression of ABC transporters, the therapeutic strategies developed for specifically disturbing actions of ABC transporter proteins would be an attractive potential for targeting and eradicating CSCs.

Advertisement

6. Conclusions

The CSC concept is still an evolving model moving forward. Recent lineage tracing strategy has proven the existence and functions of CSCs in progression of solid tumors. CSCs with intrinsic activities of chemoresistance are the culprit in tumor recurrence and the root of treatment failures. Challenges remain on how to efficiently identify and target CSCs in vivo and monitor CSCs posttreatment. Cancer biology has proofed that traditional clonal evolution, and CSC models are not exclusive but co-exist with each other in tumorigenesis. Therefore, future elucidation of molecular mechanisms underlying CSC biology should open a new window of efficacious novel therapy strategies that eliminate both CSCs and non-CSCs.

References

  1. 1. Jemal A, Siegel R, Ward E, et al. Cancer statistics, 2008. CA Cancer J Clin 2008; 58: 71–96. DOI: 10.3322/CA.2007.0010
  2. 2. Herter-Sprie GS, Greulich H, Wong K-K. Activating mutations in ERBB2 and their impact on diagnostics and treatment. Front Oncol 2013; 3: 86. DOI: 10.3389/fonc.2013.00086.
  3. 3. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011; 144: 646–674. DOI: 10.1016/j.cell.2011.02.013
  4. 4. Krogan NJ, Lippman S, Agard DA, Ashworth A, Ideker T. The cancer cell map initiative: defining the hallmark networks of cancer. Mol Cell 2015; 58: 690–698. DOI: 10.1016/j.molcel.2015.05.008
  5. 5. Stratton MR. Exploring the genomes of cancer cells: progress and promise. Science 2011; 331: 1553–1558. DOI: 10.1126/science.1204040
  6. 6. O’Connor ML, Xiang D, Shigdar S, et al. Cancer stem cells: a contentious hypothesis now moving forward. Cancer Lett 2014; 344: 180–187. DOI: doi: 10.1016/j.canlet.2013.11.012
  7. 7. Nowell PC. The clonal evolution of tumor cell populations. Science 1976; 194: 23–28.
  8. 8. Tsai AG, Lu H, Raghavan SC, Muschen M, Hsieh C-L, Lieber MR. Human chromosomal translocations at CpG sites and a theoretical basis for their lineage and stage specificity. Cell 2008; 135: 1130–1142. DOI: 10.1016/j.cell.2008.10.035
  9. 9. Bardelli A, Cahill DP, Lederer G, et al. Carcinogen-specific induction of genetic instability. Proc Natl Acad Sci U S A 2001; 98: 5770–5775. DOI: 10.1073/pnas.081082898
  10. 10. Gatenby RA, Gillies RJ. A microenvironmental model of carcinogenesis. Nat Rev Cancer 2008; 8: 56–61. DOI: 10.1038/nrc2255
  11. 11. Lathia JD, Heddleston JM, Venere M, Rich JN. Deadly teamwork: neural cancer stem cells and the tumor microenvironment. Cell Stem Cell 2011; 8: 482–485. DOI: 10.1016/j.stem.2011.04.013
  12. 12. Chen J, Sprouffske K, Huang Q, Maley CC. Solving the puzzle of metastasis: the evolution of cell migration in neoplasms. PloS One 2011; 6: e17933. DOI: 10.1371/journal.pone.0017933
  13. 13. Mazzone M, Dettori D, Leite de Oliveira R, et al. Heterozygous deficiency of PHD2 restores tumor oxygenation and inhibits metastasis via endothelial normalization. Cell 2009; 136: 839–851. DOI: 10.1016/j.cell.2009.01.020
  14. 14. Greaves M, Maley CC. Clonal evolution in cancer. Nature 2012; 481: 306–313. DOI: 10.1038/nature10762
  15. 15. Gilbert LA, Hemann MT. DNA damage-mediated induction of a chemoresistant niche. Cell 2010; 143: 355–366. DOI: 10.1016/j.cell.2010.09.043
  16. 16. Cabioglu N, Ozmen V, Kaya H, et al. Increased lymph node positivity in multifocal and multicentric breast cancer. J Am Coll Surg 2009; 208: 67–74. DOI: 10.1016/j.jamcollsurg.2008.09.001
  17. 17. Nguyen-Khac F, Lesty C, Eclache V, et al. Chromosomal abnormalities in transformed Ph-negative myeloproliferative neoplasms are associated to the transformation subtype and independent of JAK2 and the TET2 mutations. Genes Chromosomes Cancer 2010; 49: 919–927. DOI: 10.1002/gcc.20802
  18. 18. Visvader JE, Lindeman GJ. Cancer stem cells in solid tumours: accumulating evidence and unresolved questions. Nat Rev Cancer 2008; 8: 755–768. DOI: 10.1038/nrc2499
  19. 19. Nguyen-Khac F, Lesty C, Eclache V, et al. Chromosomal abnormalities in transformed Ph-negative myeloproliferative neoplasms are associated to the transformation subtype and independent of JAK2 and the TET2 mutations. Genes Chromosomes Cancer 2010; 49: 919–927. DOI: 10.1002/gcc.20802
  20. 20. Shackleton M, Quintana E, Fearon ER, Morrison SJ. Heterogeneity in cancer: cancer stem cells versus clonal evolution. Cell 2009; 138: 822–829. DOI: 10.1016/j.cell.2009.08.017
  21. 21. Lee HE, Kim JH, Kim YJ, et al. An increase in cancer stem cell population after primary systemic therapy is a poor prognostic factor in breast cancer. Br J Cancer 2011; 104: 1730–1738. DOI: 10.1038/bjc.2011.159
  22. 22. Beck B, Driessens G, Goossens S, et al. A vascular niche and a VEGF-Nrp1 loop regulate the initiation and stemness of skin tumours. Nature 2011; 478: 399–403. DOI: 10.1038/nature10525
  23. 23. Nguyen LV, Vanner R, Dirks P, Eaves CJ. Cancer stem cells: an evolving concept. Nat Rev Cancer 2012; 12: 133–143. DOI: 10.1038/nrc3184
  24. 24. Wenjing J, Jianhua P, Yue Z, Cho WCS, Kunlin J. The implications of cancer stem cells for cancer therapy. Int J Mol Sci 2012; 13: 16636–16657. DOI: 10.3390/ijms131216636
  25. 25. Vermeulen L, de Sousa e Melo F, Richel DJ, Medema JP. The developing cancer stem-cell model: clinical challenges and opportunities. Lancet Oncol 2012; 13: e83–e89. DOI: 10.1016/S1470-2045(11)70257-1
  26. 26. Xiang D, Shigdar S, Qiao G, et al. Nucleic acid aptamer-guided cancer therapeutics and diagnostics: the next generation of cancer medicine. Theranostics 2015; 5: 23–42. DOI: 10.7150/thno.10202
  27. 27. Chaffer CL, Brueckmann I, Scheel C, et al. Normal and neoplastic nonstem cells can spontaneously convert to a stem-like state. Proc Natl Acad Sci U S A 2011; 108: 7950–7955. DOI: 10.1073/pnas.1102454108
  28. 28. Scheel C, Eaton EN, Li SH-J, et al. Paracrine and autocrine signals induce and maintain mesenchymal and stem cell states in the breast. Cell 2011; 145: 926–940. DOI: 10.1016/j.cell.2011.04.029
  29. 29. O’Brien CA, Kreso A, Jamieson CHM. Cancer stem cells and self-renewal. Clin Cancer Res 2010; 16: 3113–3120. DOI: 10.1158/1078-0432.CCR-09-2824
  30. 30. Wei J, Wunderlich M, Fox C, et al. Microenvironment determines lineage fate in a human model of MLL-AF9 leukemia. Cancer Cell 2008; 13: 483–495. DOI: 10.1016/j.ccr.2008.04.020
  31. 31. Kim M, Moon HB, Spangrude GJ. Major age-related changes of mouse hematopoietic stem/progenitor cells. Ann N Y Acad Sci 2003; 996: 195–208. DOI: 10.1111/j.1749-6632.2003.tb03247.x
  32. 32. Quintana E, Shackleton M, Sabel MS, Fullen DR, Johnson TM, Morrison SJ. Efficient tumour formation by single human melanoma cells. Nature 2008; 456: 593–598. DOI: 10.1038/nature07567
  33. 33. Tan E, Ding XQ, Saadi A, Agarwal N, Naash MI, Al-Ubaidi MR. Expression of cone-photoreceptor-specific antigens in a cell line derived from retinal tumors in transgenic mice. Invest Ophthalmol Vis Sci 2004; 45: 764–768.
  34. 34. Limaye PB, Bowen WC, Orr AV, Luo J, Tseng GC, Michalopoulos GK. Mechanisms of hepatocyte growth factor-mediated and epidermal growth factor-mediated signaling in transdifferentiation of rat hepatocytes to biliary epithelium. Hepatology 2008; 47: 1702–1713. DOI: 10.1002/hep.22221
  35. 35. Michalopoulos GK, Barua L, Bowen WC. Transdifferentiation of rat hepatocytes into biliary cells after bile duct ligation and toxic biliary injury. Hepatology 2005; 41: 535–544. DOI: 10.1002/hep.20600
  36. 36. Vermeulen L, Snippert HJ. Stem cell dynamics in homeostasis and cancer of the intestine. Nat Rev Cancer 2014; 14: 468–480. DOI: 10.1038/nrc3744
  37. 37. Chiba T, Kita K, Zheng YW, et al. Side population purified from hepatocellular carcinoma cells harbors cancer stem cell-like properties. Hepatology 2006; 44: 240–251. DOI: 10.1002/hep.21227
  38. 38. Kimura O, Takahashi T, Ishii N, et al. Characterization of the epithelial cell adhesion molecule (EpCAM)+ cell population in hepatocellular carcinoma cell lines. Cancer Sci 2010; 101: 2145–2155. DOI: 10.1111/j.1349-7006.2010.01661.x
  39. 39. Cao L, Zhou Y, Zhai B, et al. Sphere-forming cell subpopulations with cancer stem cell properties in human hepatoma cell lines. BMC Gastroenterol 2011; 11: 71. DOI: 10.1186/1471-230X-11-71
  40. 40. Adams JM, Strasser A. Is tumor growth sustained by rare cancer stem cells or dominant clones? Cancer Res 2008; 68: 4018–4021. DOI: 10.1158/0008-5472
  41. 41. Resetkova E, Reis-Filho JS, Jain RK, et al. Prognostic impact of ALDH1 in breast cancer: a story of stem cells and tumor microenvironment. Breast Cancer Res Treat 2010; 123: 97–108. DOI: 10.1007/s10549-009-0619-3
  42. 42. Allikmets R, Schriml LM, Hutchinson A, Romano-Spica V, Dean M. A human placenta-specific ATP-binding cassette gene (ABCP) on chromosome 4q22 that is involved in multidrug resistance. Cancer Res 1998; 58: 5337–5339.
  43. 43. Abdullah LN, Chow EK. Mechanisms of chemoresistance in cancer stem cells. Clin Transl Med 2013; 2: 3. DOI: 10.1186/2001-1326-2-3
  44. 44. Todaro M, Francipane MG, Medema JP, Stassi G. Colon cancer stem cells: promise of targeted therapy. Gastroenterology 2010; 138: 2151–2162. DOI: 10.1053/j.gastro.2009.12.063
  45. 45. Sottoriva A, Verhoeff JJC, Borovski T, et al. Cancer stem cell tumor model reveals invasive morphology and increased phenotypical heterogeneity. Cancer Res 2010; 70: 46–56. DOI: 10.1158/0008-5472
  46. 46. Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med 1997; 3: 730–737.
  47. 47. Yang YM, Chang JW. Current status and issues in cancer stem cell study. Cancer Invest 2008; 26: 741–755. DOI: 10.1080/07357900801901856
  48. 48. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A 2003; 100: 3983–3988. DOI: 10.1073/pnas.0530291100
  49. 49. O’Brien CA, Pollett A, Gallinger S, Dick JE. A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature 2007; 445: 106–110. DOI: 10.1038/nature05372
  50. 50. Li C, Heidt DG, Dalerba P, et al. Identification of pancreatic cancer stem cells. Cancer Res 2007; 67: 1030–1037. DOI: 10.1158/0008-5472.CAN-06-2030
  51. 51. Shen J, Zhu Z. Catumaxomab, a rat/murine hybrid trifunctional bispecific monoclonal antibody for the treatment of cancer. Curr Opin Mol Ther 2008; 10: 273–284.
  52. 52. Medema JP, Vermeulen L. Microenvironmental regulation of stem cells in intestinal homeostasis and cancer. Nature 2011; 474: 318–326. DOI: 10.1038/nature10212
  53. 53. de Sousa EM, Vermeulen L, Richel D, Medema JP. Targeting Wnt signaling in colon cancer stem cells. Clin Cancer Res 2011; 17: 647–653. DOI: 10.1158/1078-0432.CCR-10-1204
  54. 54. Hu Y, Smyth GK. ELDA: extreme limiting dilution analysis for comparing depleted and enriched populations in stem cell and other assays. J Immunol Methods 2009; 347: 70–78. DOI: 10.1016/j.jim.2009.06.008
  55. 55. Parrish RS, Hazlett LJ. Design and estimation in the limiting dilution assay when concentrations vary widely among samples. J Biopharm Stat 2000; 10: 197–215. DOI: 10.1081/BIP-1001101022
  56. 56. Macken C. Design and analysis of serial limiting dilution assays with small sample sizes. J Immunol Methods 1999; 222: 13–29. DOI: 10.1016/S0022-1759(98)00133-1
  57. 57. Sieburg HB, Cho RH, Muller-Sieburg CE. Limiting dilution analysis for estimating the frequency of hematopoietic stem cells: uncertainty and significance. Exp Hematol 2002; 30: 1436–1443. DOI: 10.1016/S0301-472X(02)00963-3
  58. 58. Cheli Y, Giuliano S, Guiliano S, et al. Mitf is the key molecular switch between mouse or human melanoma initiating cells and their differentiated progeny. Oncogene 2011; 30: 2307–2318. DOI: 10.1038/onc.2010.598
  59. 59. Shultz LD, Sidman CL. Genetically determined murine models of immunodeficiency. Ann Rev Immunol 1987; 5: 367–403. DOI: 10.1146/annurev.iy.05.040187.002055
  60. 60. Ricci-Vitiani L, Lombardi DG, Pilozzi E, et al. Identification and expansion of human colon-cancer-initiating cells. Nature 2007; 445: 111–115. DOI: 10.1038/nature05384
  61. 61. Maitland NJ, Frame FM, Polson ES, Lewis JL, Collins AT. Prostate cancer stem cells: do they have a basal or luminal phenotype? Horm Cancer 2011; 2: 47–61. DOI: 10.1007/s12672-010-0058-y
  62. 62. Barabe F, Kennedy JA, Hope KJ, Dick JE. Modeling the initiation and progression of human acute leukemia in mice. Science 2007; 316: 600–604. DOI: 10.1126/science.1139851
  63. 63. Howarth KD, Blood KA, Ng BL, et al. Array painting reveals a high frequency of balanced translocations in breast cancer cell lines that break in cancer-relevant genes. Oncogene 2008; 27: 3345–3359. DOI: 10.1038/sj.onc.1210993
  64. 64. Summer R, Kotton DN, Sun X, Ma B, Fitzsimmons K, Fine A. Side population cells and Bcrp1 expression in lung. Am J Physiol Lung Cell Mol Physiol 2003; 285: L97–L104. DOI: 10.1152/ajplung.00009.2003
  65. 65. Kim M, Moon H-B, Spangrude GJ. Major age-related changes of mouse hematopoietic stem/progenitor cells. Ann N Y Acad Sci 2003; 996: 195–208. DOI: 10.1111/j.1749-6632.2003.tb03247.x
  66. 66. Triel C, Vestergaard ME, Bolund L, Jensen TG, Jensen UB. Side population cells in human and mouse epidermis lack stem cell characteristics. Exp Cell Res 2004; 295: 79–90. DOI: 10.1016/j.yexcr.2003.11.032
  67. 67. Choo CR, Danjoux C, Morton GC. Should pathological T3 and/or margin positive prostate cancer receive adjuvant therapy? A radiation oncologist’s view. Can J Urol 2000; 7: 1043–1050.
  68. 68. Fromm MF, Kauffmann HM, Fritz P, et al. The effect of rifampin treatment on intestinal expression of human MRP transporters. Am J Pathol 2000; 157: 1575–1580. DOI: 10.1016/S0002-9440(10)64794-3
  69. 69. Pece S, Tosoni D, Confalonieri S, et al. Biological and molecular heterogeneity of breast cancers correlates with their cancer stem cell content. Cell 2010; 140: 62–73. DOI: 10.1016/j.cell.2009.12.007
  70. 70. Kim S, Alexander CM. Tumorsphere assay provides more accurate prediction of in vivo responses to chemotherapeutics. Biotechnol Lett 2014; 36: 481–488. DOI: 10.1007/s10529-013-1393-1
  71. 71. Willan PM, Farnie G. Application of stem cell assays for the characterization of cancer stem cells. In: Cancer Stem Cells in Solid Tumors. Springer; 2011. p. 259–282. DOI: 10.1007/978-1-61779-246-5_15
  72. 72. Charrier C, Coronas V, Fombonne J, et al. Characterization of neural stem cells in the dorsal vagal complex of adult rat by in vivo proliferation labeling and in vitro neurosphere assay. Neuroscience 2006; 138: 5–16. DOI: 10.1016/j.neuroscience.2005.10.046
  73. 73. Ayyanan A, Civenni G, Ciarloni L, et al. Increased Wnt signaling triggers oncogenic conversion of human breast epithelial cells by a Notch-dependent mechanism. Proc Natl Acad Sci U S A 2006; 103: 3799–3804. DOI: 10.1073/pnas.0600065103
  74. 74. Singh SK, Hawkins C, Clarke ID, et al. Identification of human brain tumour initiating cells. Nature 2004; 432: 396–401. DOI: 10.1038/nature03128
  75. 75. Toma JG, McKenzie IA, Bagli D, Miller FD. Isolation and characterization of multipotent skin-derived precursors from human skin. Stem Cells 2005; 23: 727–737. DOI: 10.1634/stemcells.2004-0134
  76. 76. Fang D, Nguyen TK, Leishear K, et al. A tumorigenic subpopulation with stem cell properties in melanomas. Cancer Res 2005; 65: 9328–9337. DOI: 10.1158/0008-5472.CAN-05-1343
  77. 77. Croker AK, Goodale D, Chu J, et al. High aldehyde dehydrogenase and expression of cancer stem cell markers selects for breast cancer cells with enhanced malignant and metastatic ability. J Cell Mol Med 2009; 13: 2236–2252. DOI: 10.1111/j.1582-4934.2008.00455.x
  78. 78. Cariati M, Naderi A, Brown JP, et al. Alpha-6 integrin is necessary for the tumourigenicity of a stem cell-like subpopulation within the MCF7 breast cancer cell line. Int J Cancer 2008; 122: 298–304. DOI: 10.1002/ijc.23103
  79. 79. Chute JP, Muramoto GG, Whitesides J, et al. Inhibition of aldehyde dehydrogenase and retinoid signaling induces the expansion of human hematopoietic stem cells. Proc Natl Acad Sci U S A 2006; 103: 11707–11712. DOI: 10.1073/pnas.0603806103
  80. 80. Ginestier C, Hur MH, Charafe-Jauffret E, 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. DOI: 10.1016/j.stem.2007.08.014
  81. 81. Braun KM, Watt FM. Epidermal label-retaining cells: background and recent applications. J Investig Dermatol Symp Proc 2004; 9: 196–201. DOI: 10.1111/j.1087-0024.2004.09313.x
  82. 82. Kase S, Yoshida K, Ohgami K, Shiratori K, Ohno S, Nakayama KI. Phosphorylation of p27(KIP1) in the mitotic cells of the corneal epithelium. Curr Eye Res 2006; 31: 307–312. DOI: 10.1080/02713680600584687
  83. 83. Oliver JA, Maarouf O, Cheema FH, Martens TP, Al-Awqati Q. The renal papilla is a niche for adult kidney stem cells. J Clin Invest 2004; 114: 795–804. DOI: 10.1172/JCI200420921
  84. 84. Bossing T, Technau GM. The fate of the CNS midline progenitors in Drosophila as revealed by a new method for single cell labelling. Development 1994; 120: 1895–1906.
  85. 85. Jensen UB, Lowell S, Watt FM. The spatial relationship between stem cells and their progeny in the basal layer of human epidermis: a new view based on whole-mount labelling and lineage analysis. Development 1999; 126: 2409–2418.
  86. 86. Tao L, van Bragt MPA, Laudadio E, Li Z. Lineage tracing of mammary epithelial cells using cell-type-specific cre-expressing adenoviruses. Stem Cell Reports 2014; 2: 770–779. DOI: 10.1016/j.stemcr.2014.04.004
  87. 87. Chen J, Li Y, Yu T-S, et al. A restricted cell population propagates glioblastoma growth after chemotherapy. Nature 2012; 488: 522–526. DOI: 10.1038/nature11287
  88. 88. Driessens G, Beck B, Caauwe A, Simons BD, Blanpain C. Defining the mode of tumour growth by clonal analysis. Nature 2012; 488: 527–530. DOI: 10.1038/nature11344
  89. 89. Schepers AG, Snippert HJ, Stange DE, et al. Lineage tracing reveals Lgr5+ stem cell activity in mouse intestinal adenomas. Science 2012; 337: 730–735. DOI: 10.1126/science.1224676
  90. 90. Shigdar S, Lin J, Li Y, et al. Cancer stem cell targeting: the next generation of cancer therapy and molecular imaging. Ther Deliv 2012; 3: 227–244.
  91. 91. Nater A, Burri R, Kawakami T, Smeds L, Ellegren H. Resolving evolutionary relationships in closely related species with whole-genome sequencing data. Syst Biol 2015; DOI: 10.1093/sysbio/syv045
  92. 92. Hehir-Kwa JY, Pfundt R, Veltman JA. Exome sequencing and whole genome sequencing for the detection of copy number variation. Expert Rev Mol Diagn 2015; 15: 1023–1032. DOI: 10.1586/14737159.2015.1053467
  93. 93. Li J, Wang T, Zhang X, Yang X. The contribution of next generation sequencing technologies to epigenome research of stem cell and tumorigenesis. Hum Genet Embryol 2011; 01: 1–10. DOI: 10.4172/2161-0436.s2-001
  94. 94. Nanney DL. Epigenetic control systems. Proc Natl Acad Sci U S A 1958; 44: 712–717.
  95. 95. Morales La Madrid A, Hashizume R, Kieran MW. Future clinical trials in DIPG: bringing epigenetics to the clinic. Front Oncol 2015; 5: 148. DOI: 10.3389/fonc.2015.00148
  96. 96. Busch C, Burkard M, Leischner C, Lauer UM, Frank J, Venturelli S. Epigenetic activities of flavonoids in the prevention and treatment of cancer. Clin Epigenetics 2015; 7: 64. DOI: 10.1186/s13148-015-0095-z
  97. 97. Munoz P, Iliou MS, Esteller M. Epigenetic alterations involved in cancer stem cell reprogramming. Mol Oncol 2012; 6: 620–636. DOI: 10.1016/j.molonc.2012.10.006
  98. 98. Esteller M. Epigenetics in cancer. N Engl J Med 2008; 358: 1148–1159. DOI: 10.1056/NEJMra072067
  99. 99. Volkel P, Dupret B, Le Bourhis X, Angrand P-O. Diverse involvement of EZH2 in cancer epigenetics. Am J Transl Res 2015; 7: 175–193.
  100. 100. Jeter CR, Yang T, Wang J, Chao HP, Tang DG. Concise review: NANOG in cancer stem cells and tumor development: an update and outstanding questions. Stem Cells 2015; 33: 2381–2390. DOI: 10.1002/stem.2007
  101. 101. Shukla S, Meeran SM. Epigenetics of cancer stem cells: pathways and therapeutics. Biochim Biophys Acta 2014; 1840: 3494–3502. DOI: 10.1016/j.bbagen.2014.09.017
  102. 102. Mardis ER. The impact of next-generation sequencing technology on genetics. Trends Genet 2008; 24: 133–141. DOI: 10.1016/j.tig.2007.12.007
  103. 103. Morozova O, Marra MA. Applications of next-generation sequencing technologies in functional genomics. Genomics 2008; 92: 255–264. DOI: 10.1016/j.ygeno.2008.07.001
  104. 104. Visvader JE, Lindeman GJ. Cancer stem cells in solid tumours: accumulating evidence and unresolved questions. Nat Rev Cancer 2008; 8: 755–768. DOI: 10.1038/nrc2499
  105. 105. Chen K, Huang Y-h, Chen J-l. Understanding and targeting cancer stem cells: therapeutic implications and challenges. Acta Pharmacol Sin 2013; 34: 732–740. DOI: 10.1038/aps.2013.27
  106. 106. Pollyea DA, Gutman JA, Gore L, Smith CA, Jordan CT. Targeting acute myeloid leukemia stem cells: a review and principles for the development of clinical trials. Haematologica 2014; 99: 1277–1284. DOI: 10.3324/haematol.2013.085209
  107. 107. Hoang VT, Buss EC, Wang W, et al. The rarity of ALDH(+) cells is the key to separation of normal versus leukemia stem cells by ALDH activity in AML patients. Int J Cancer 2015; 137: 525–536. DOI: 10.1002/ijc.29410
  108. 108. Fan D, Li Z, Zhang X, et al. AntiCD3Fv fused to human interleukin-3 deletion variant redirected T cells against human acute myeloid leukemic stem cells. J Hematol Oncol 2015; 8: 18. DOI: 10.1186/s13045-015-0109-5
  109. 109. Shigdar S, Qiao L, Zhou S-F, et al. RNA aptamers targeting cancer stem cell marker CD133. Cancer Lett 2013; 330: 84–95. DOI: 10.1016/j.canlet.2012
  110. 110. Shigdar S, Lin J, Yu Y, Pastuovic M, Wei M, Duan W. RNA aptamer against a cancer stem cell marker epithelial cell adhesion molecule. Cancer Sci 2011; 102: 991–998. DOI: 10.1111/j.1349-7006.2011.01897.x
  111. 111. Shigdar S, Qian C, Lv L, et al. The use of sensitive chemical antibodies for diagnosis: detection of low levels of EpCAM in breast cancer. PLoS One 2013; 8: e57613. DOI: 10.1371/journal.pone.0057613
  112. 112. Fletcher JI, Haber M, Henderson MJ, Norris MD. ABC transporters in cancer: more than just drug efflux pumps. Nat Rev Cancer 2010; 10: 147–156. DOI: 10.1038/nrc2789
  113. 113. Gottesman MM, Fojo T, Bates SE. Multidrug resistance in cancer: role of ATP-dependent transporters. Nat Rev Cancer 2002; 2: 48–58. DOI: 10.1038/nrc706
  114. 114. Abdullah LN, Chow EK-H. Mechanisms of chemoresistance in cancer stem cells. Clin Transl Med 2013; 2: 3. DOI: 10.1186/2001-1326-2-3
  115. 115. Jonker JW, Merino G, Musters S, et al. The breast cancer resistance protein BCRP (ABCG2) concentrates drugs and carcinogenic xenotoxins into milk. Nat Med 2005; 11: 127–129. DOI: 10.1038/nm1186
  116. 116. Zhou S, Schuetz JD, Bunting KD, et al. The ABC transporter Bcrp1/ABCG2 is expressed in a wide variety of stem cells and is a molecular determinant of the side-population phenotype. Nat Med 2001; 7: 1028–1034. DOI: 10.1038/nm0901-1028
  117. 117. Szakacs G, Paterson JK, Ludwig JA, Booth-Genthe C, Gottesman MM. Targeting multidrug resistance in cancer. Nat Rev Drug Discov 2006; 5: 219–234. DOI: 10.1038/nrd1984
  118. 118. Scharenberg CW, Harkey MA, Torok-Storb B. The ABCG2 transporter is an efficient Hoechst 33342 efflux pump and is preferentially expressed by immature human hematopoietic progenitors. Blood 2002; 99: 507–512. DOI: 10.1182/blood.V99.2.507
  119. 119. Chow EK-H, Fan L-l, Chen X, Bishop JM. Oncogene-specific formation of chemoresistant murine hepatic cancer stem cells. Hepatology 2012; 56: 1331–1341. DOI: 10.1002/hep.25776
  120. 120. Dalton WS, Crowley JJ, Salmon SS, et al. A phase III randomized study of oral verapamil as a chemosensitizer to reverse drug resistance in patients with refractory myeloma. A Southwest Oncology Group study. Cancer 1995; 75: 815–820.
  121. 121. Lancet JE, Baer MR, Duran GE, et al. A phase I trial of continuous infusion of the multidrug resistance inhibitor zosuquidar with daunorubicin and cytarabine in acute myeloid leukemia. Leuk Res 2009; 33: 1055–1061. DOI: 10.1016/j.leukres.2008.09.015
  122. 122. Chow EK, Zhang XQ, Chen M, et al. Nanodiamond therapeutic delivery agents mediate enhanced chemoresistant tumor treatment. Sci Transl Med 2011; 3: 73ra21. DOI: 10.1126/scitranslmed.3001713

Written By

Zhaopeng Tang, Qianfeng Wang, Sarah Shigdar, Wei Duan and Dongxi Xiang

Submitted: 02 April 2015 Reviewed: 28 October 2015 Published: 10 February 2016