Open access

Multidrug Resistance Transporters – Roles in maintaining Cancer Stem-Like Cells

Written By

To, Kenneth K.W. and Fu, L.W.

Submitted: 30 November 2010 Published: 23 August 2011

DOI: 10.5772/23867

From the Edited Volume

Stem Cells in Clinic and Research

Edited by Ali Gholamrezanezhad

Chapter metrics overview

2,950 Chapter Downloads

View Full Metrics

1. Introduction

Cancer has become the leading cause of death worldwide in the year 2010, according to a new edition of the World Cancer Report from the International Agency for Research on Cancer (World Health Organization, 2008). Despite the advance in the development of novel chemotherapeutic drugs, the dismal prognosis facing most cancer patients may result from the ability of cancer to withstand drug treatment, recur and metastasize after initial therapies. There is accumulating evidence in support of a central role for cancer stem cells (CSCs) in the initiation, propagation and recurrence of human cancers. Therefore, targeting CSCs has become an attractive research topic for the improvement of treatment outcome and prolongation of patient survival. However, CSCs are endowed with the ability to survive against chemo- and radiation therapy. A better understanding of the mechanisms underlying CSC resistance is badly needed. This chapter provides a review about evidence supporting a fundamental role for CSCs in cancer progression, and summarizes potential mechanisms of CSC resistance to treatment, with emphasis on the involvement of multidrug resistance transporters and their regulation in CSCs.

Advertisement

2. The Cancer Stem Cell (CSC) hypothesis

For many years, tumors have been described as the proliferation of cell clones in which multiple genetic alterations had occurred over time (Nowell, 1976). This “clonal evolution” model is a non-hierarchical model that proposes all cells within a tumor would have an equal chance of acquiring genetic mutations necessary for driving the tumor growth. Subsequently, under selective pressures, the more aggressive cells would drive the tumor progression and lead to therapy resistance. Distinct from this notion, an emerging “cancer stem cell model” is a hierarchical model, which proposes that only a subset of cells called “cancer stem cells” (CSCs) or “tumor-initiating cells” can initiate and propagate a tumor. The CSCs can self-renew, propagate the tumor, and differentiate into the diverse types of cells that are found in the original tumor, thereby mimicking stem cells.

The emergence of the CSC model can be dated back to the mid-19th century when a German pathologist Rudolf Virchow proposed that cancers arise from the activation of dormant, embryonic-like cells present in mature tissue (Virchow, 1855). His speculation was based on the histological similarities between the developing fetus and certain types of cancer such as teratocarcinomas. Later, the term “cancer stem cells” was probably first coined by Hamburger and Salmon who postulated that a subpopulation of cells in a tumor capable of growing in soft agar are the cell renewal source of a neoplasm and also serve as the seeds of metastatic spread of cancer (Hamburger & Salmon, 1977). More recently, the first conclusive evidence for CSCs was reported in 1994 when John Dick and colleagues isolated the cancerous stem cells from acute myelogenous leukemia and documented their self-renewing capacity (Lapidot et al., 1994).

Since then, the CSC hypothesis has shifted the paradigm in our understanding of cancer tumorigenesis and has important implications in cancer chemotherapy. With respect to tumor development and progression, it could explain the well-known heterogeneous nature of cells in a tumor (Park et al., 1971). While CSCs represent the only cells with self-renewal capability driving the tumor growth, the remaining actively proliferating cells making up the bulk of the tumor could still differentiate and are therefore destined to die. Therefore, the goal of cancer chemotherapy should be to target these CSCs for complete eradication of the tumor.

It should be noted that the CSC hypothesis indeed does not specify the origin of the cancer initiating cells. They could originate from a stem, progenitor or differentiated cell. Therefore, the term “tumor-initiating cell” is often used instead of cancer stem cells to avoid the confusion. The prevailing thought is that CSCs are derived through an activation process involving one of three possible pathways (Figure 1): (1) from normal stem cells losing growth regulation; (2) from progenitor (Jamieson et al., 2004; Krivtsov et al., 2006) or differentiated cells acquiring the self-renewal capacity; or (3) by the fusion of normal stem cells with cancer cells (Pawelek & Chakraborty, 2008; Dittmar et al., 2009).

2.1. Detection and identification of CSCs

The general consensus nowadays is that CSCs can only be ultimately defined experimentally by their ability to recapitulate the generation of a continuously growing tumor (Clarke et al., 2006). However, due to technical difficulty of tumor repopulation in vivo, three other popular molecular or phenotypic characteristics of CSCs are being exploited for their identification and prospective isolation from tumor specimens and cancer cell lines. These include (1) the “side population (SP)” phenotype manifested by the exclusion of Hoechst 33342 dye in flow cytometric assays; (2) cell surface markers; and (3) anchorage-independent sphere formation ability. The putative CSC population thus identified will usually be further validated by their ability to initiate a tumor and subsequently recapitulate the heterogeneity of the primary tumor.

2.1.1. The “side population (SP)” phenotype

CSCs and the normal stem cells alike express high levels of the ATP-binding cassette (ABC) transporters, which help protect them from cytotoxic insult throughout their long lifespan. By using the energy of ATP hydrolysis, ABC transporters actively efflux drugs out of the cells, thereby protecting them from toxic xenobiotics (Gottesman et al., 2002). Importantly, this drug-efflux capability conferred by ABC transporters (including ABCG2 and P-gp/MDR1) has been used as a marker in the isolation and analysis of haematopoietic stem cells (HSCs). Goodell and colleagues were the first to report that when mouse bone marrow-derived cells are incubated with the dye Hoechst 33342 and then analyzed by dual-wavelength flow cytometry, a small population of cells does not accumulate an appreciable amount of the fluorescent dye and is thus identified as a Hoechst-dim side population (SP) (Goodell et al., 1997). Remarkably, the side population is highly enriched in HSCs (Goodell et al., 1996). When isolated from mice and transplanted into irradiated mice, small numbers of these SP cells can reconstitute the bone marrow, demonstrating that these cells are pluripotent. Later, it was demonstrated that the transporter Abcg2, but not P-gp/Mdr1, was responsible for the SP in mouse bone marrow (Zhou et al., 2001). Human ABCG2 was subsequently also found to be responsible for the SP phenotype in human bone marrow (Scharenberg et al., 2002).

Figure 1.

Origin of CSCs (tumor-initiating cells). A CSC may arise from one of the following molecular pathways: (i) a stem cell undergoing a mutation; (iia & iib) a progenitor/differentiated cell undergoing several mutations, thus allowing them to acquire the self-renewal ability; (iii) fusion of a cancer cell with a normal stem cell, thereby equipping the former with self-renewal capability (not shown in the figure). Like normal stem cells, CSCs are capable of long-term self-renewal and dividing asymmetrically to recapitulate the generation of a continuously growing tumor (pluripotency). In all scenarios, the resulting CSC has lost normal growth regulation and progress into malignancy.

Since its initial application in bone marrow HSCs, the side population technique based on Hoechst 33342 efflux has been adapted to identify putative stem cells and progenitors in many normal tissues (Zhou et al., 2001; Asakura & Rudnucki, 2002; Leckner et al., 2002; Alvi et al., 2003; Summer et al., 2003; Budak et al., 2005; Du et al., 2005, Hussain, et al., 2005). SP cells have also been found in a number of established cancer cell lines as well as tumor samples and have been shown to have stem cell-like properties, overexpress ABCG2, and possess inherent drug-resistance (Kondo et al., 2004; Hirschmann-Jax et al., 2004; Haraguchi et al., 2005; Seigel et al., 2005, Chiba et al., 2006). Figure 2 shows the existence of such a SP in a ABCG2-overexpressing mitoxantrone-selected resistant pancreatic cell line. The nearly complete elimination of all SP cells after treatment with the specific ABCG2 inhibitor, fumitremorgin C (FTC), suggests that ABCG2 is a major molecular determinant for the SP phenotype.

Figure 2.

Identification and isolation of SP cells for the study of putative CSCs. FACS analysis was performed for a mitoxantrone-selected drug-resistant and ABCG2-overexpressing pancreatic cancer cell line after incubation with Hoechst 33342 dye for 1 h. The gated R4 region represents a Hoechst staining-resistant cell population (i.e. SP cells); their abundance are indicated by the number in the figure.

Despite the initial excitment about using SP to identify CSCs, the ABCG2-highly expressing SP and ABCG2-negative non-SP tumor cells have been reported to be similarly tumorigenic (Patrawala et al., 2005). It is believed that the SP fraction obtained is not a pure stem-cell population, which is greatly affected by the isolation method (Montanaro et al., 2004). There were also report demonstrating that SP cells do not identify stem cell (Triel et al., 2004). Moreover, ABCG2, the molecular determinant for Hoechst exclusion, is not an absolute requirement for stem cells. Abcg2-deficient mice are viable and demonstrate no defect in steady state hematopoiesis, though the bone marrow of Abcg2-deficient mice does lack a SP (Zhou et al., 2002). Nevertheless, since CSCs lack distinct molecular markers, Hoechst 33342-dependent cell sorting remains the most widely employed technique for the identification and purification of putative CSCs.

It is also noteworthy that expression of drug transporters (especially MDR1/Pgp) can be part of the differentiated phenotype of cells in normal tissue (Triel et al., 2004). Histopathological and molecular biological studies have reported increased expression of ABCB1 in more differentiated tumors (Mizoguchi et al., 1990; Nishiyama et al., 1993; Bates et al., 1989; Mickley et al., 1989). We have also reported the cell type-specific upregulation of ABCG2 by romidepsin, a differentiating agent and anticancer drug, in cancer cell lines (To et al, 2008, 2011). Furthermore, P-gp and/or ABCG2 are usually overexpressed with the onset of multidrug resistance in cancer cell populations. In these situations, the SP phenotype will not definitively identify CSCs, but because the overexpression of these transporters allows the cells to effectively exclude the Hoechst dye.

2.1.2. CSC-specific cell surface markers

Another common way to identify putative CSCs from patient-derived tissues or cancer cell lines is by labeling the isolated cells with antibodies against various cell surface markers already known in normal stem cells. Cells bearing these cell-surface markers can be isolated by fluorescence activated cell sorting (FACS) (Woodward et al., 2005) or magnetic bead columns (Dou et al., 2007). These enriched cell populations are then tested for their ability to initiate tumorigenesis in immune deficient mice.

Several cell surface markers have been used to detect CSCs (Table 1). Among them, the cell surface protein CD133 (Prominin 1, a transmembrane glycoprotein) is probably the most extensively used marker, which was also known to define stem and progenitor cells in varuous tissues (Shmelkov et al., 2005). A cautionary technical note is worth mentioning. CD133 expression is in fact found to be indifferent to the differentiation status of most cells. On the other hand, its posttranslational glycoslyated form was found to be downregulated upon cell differentiation (Florek et al., 2005). Therefore, upon dedifferentation of the committed cells to generate CSCs as observed in oncogenesis (Figure 1), the glycosylation of CD133 (AC133, the glycosylated epitope of CD133) is expected to increase and therefore serve as a marker for the tumorigenic potential of putative CSCs. In other words, antibody against AC133, but not CD133, should be used for the prospective identification of putative CSCs. It is also noteworthy that, since tumor initiating CSCs are heterogeneous, a specific marker or set of markers has not been found to unequivocally identify CSCs in solid tumors (Welte et al., 2010). CSCs identified from solid tumors may also express other organ-specific markers.

Tumor typePutative CSC cell surface markersReference
BreastCD44+ CD24-/lowAl-Hajj et al. (2003); Wang et al. (2010)
ColonCD133+Fang et al. (2010)
ColonCD44+ EpCamhigh CD166+Kanwar et al. (2010)
CNSCD133+Pallini et al. (2011)
HNSCCCD44+ ALDH+Chen et al. (2010)
LiverCD13+Haraquchi et al. (2010)
MelanomaCD20+Schmidt et al. (2011)
Multiple myelomaCD138-Singh et al. (2004)
NSCLCCD133+Salnikov et al. (2010)
PancreasCD44+ CD24+ ESA+Hong et al. (2009)
ProstateCD44+ α2β1+ CD133+Collins et al. (2005)

Table 1.

Commonly employed CSC cell surface markers in various tissues. (HNSCC = head and neck squamous cell carcinoma; NSCLC = non small cell lung cancer)

2.1.3. Anchorage-independent sphere formation assay

Putative CSCs have also been identified based on their ability to form colonies in vitro. Typically, putative CSCs fractions are seeded onto culture dishes coated with serum-free media containing epidermal growth factor and basic fibroblast growth factor. The growth of spherical colonies after a few weeks is considered indicative of self-renewal ability, and would be consistent with a CSC phenotype. Sphere-forming ability as a measure of stem cells was first developed for central nervous system (CNS) cells, where it has been shown that a subset of cells isolated from human fetal brain, and subsequently from human CNS tumors, can form spheres when cultured under the appropriate conditions (Tamaki et al., 2002). These spheres can self renew in vitro, and differentiate into all of the neuronal lineages, both in vitro and in vivo. More importantly, it was subsequently demonstrated for brain tumors that the neurosphere-forming cells could be prospectively isolated from fresh tissue using the cell surface marker CD133. These CD133+ cells did indeed initiate brain tumors in vivo, without any in vitro manipulation, indicating that they do in fact represent CSCs (Singh et al., 2003).

2.2. CSCs in hematopoietic malignancies

The hematopoietic system is the best characterized somatic tissue with respect to stem cell biology. Many of the physical, biologic, and developmental features of normal hematopoietic stem cells have been defined and useful methods for studying stem cells have been established. It is therefore not surprising that CSCs were first identified in human acute myelogenous leukemia (AML), an aggressive malignancy of immature hematopoietic cells in the bone marrow. The leukemia-initiating activity of primary human AML cells in immunodeficient mice was first demonstrated by John Dick & colleagues, where they found that the “leukemic stem cells (LSCs)” were exclusively found in the CD34+CD38- subpopulation (Bonnet & Dick, 1997). As normal hematopoietic stem cells (HSC) share the CD34+CD38- immunophenotype, it was proposed that AML stem cells arise from HSC.

2.3. CSCs in solid tumors and cancer cell lines

CSCs have been more difficult to identify in non-haematopoietic cancers because fewer well-developed phenotypic markers and definitive assay systems are available. Al-Hajj et al. were the first to identify and prospectively isolate a minority subpopulation of cells from a human solid breast cancer cell line, based on the expression of surface markers and their potential to form tumor after injection into the mammary fat pad of NOD/SCID mice (Al-Hajj et al., 2003). Cells with the phenotype of epithelial-specific antigen (ESA)+ Lineage marker (Lin)-CD24-/lowCD44+ were found to generate tumor that were histologically similar to those of primary breast tumors in mice when as few as 100 cells were transplanted. Similar findings were also published for human brain tumors (GBMs and medulloblastomas) (Singh et al., 2003; Hemmati et al., 2003). These CSCs can differentiate into cells that have characteristics of both neurons and glial cels, self-renew in vitro at higher levels than normal neuronal stem cells, and grow and differentiate in neonatal rat brains. Interestingly, the putative CSCs isolated from these brain tumors overexpressing CD133 were found to regenerate identical brain tumors in NOD/SCID mice. Furthermore, these tumors could also be serially transplanted (Singh et al., 2004). It is likely that, as suitable markers and assay systems become available, more solid tumor CSCs will be described.

2.4. Signaling pathways supporting the self-renewal of CSCs

There are several signaling pathways including Notch (McGovern et al., 2009), Wnt/β-catenin (Reya & Clevers, 2005), Hedgehog (Medina et al., 2009), and PI3K/Akt (Hu et al., 2005), which have known roles in maintenance and/or control of normal and cancer stem cell compartments, as well as being implicated in cancer. Since they are playing an important functional role in CSC self-renewal and survival, they also represent attractive novel therapeutic targets for complete eradication of tumor. A short list of candidate chemotherapeutic drugs designed to target these signaling pathways, currently under preclinical development or in clinical trials, is compiled in Table 2 and Table 3, respectively. Selected signaling cascades are also discussed in more detail as follows.

TargetNovel agent/combinationCSCs from specific tumor type testedReference
Bcl-2TRAIL + ABT-737 (Abbott)BrainTagscherer et al., 2008
CD44CD44 antibodiesAMLJin et al., 2006
Death receptorsTRAIL + chemotherapyAML progenitorsPlasilova et al., 2002
Fatty acid synthase (F A S)ResveratrolBreastPandey et al., 2010
HedgehogCyclopamineMedulloblastomaBerman et al., 2002
IL-4IL-4 blocking antibodies + chemotherapyColonFrancipane et al., 2008
Notchγ-secretase inhibitor (GSI-18)MedulloblastomaFan et al., 2006
PI3K/AktA-443654 (Abbott)BrainGallia et al., 2009
SHH/mTORCyclopamine + rapamycin + chemotherapyPancreasMueller et al., 2009
TGF-βTGF-β + imatinibCMLNaka et al., 2010
Wnt/β-catenin/lef-1CGP049090, PKF115-584 (Novartis)CLLGandhirajan et al., 2010
Wnt (Canonical)ZTM000990, PKF118-310, anti-WNT1 & anti-WNT2 antibodies--Barker & Clevers, 2006
XIAPSmall molecular XIAP inhibitors (Pfizer) + γ-irradiationBrainVellanki et al., 2009

Table 2.

Preclinical studies of novel drug candidates targeting various signaling pathways associated with CSCs.

TargetDrugCancer type (Phase)SponsorClinical trial identifier
NotchMK0752Breast (I)MerckNCT00106145
Pancreatic (I, II)Cancer Research, UKNCT01098344
PF-03084014Leukemia (I)PfizerNCT00878189
RO4929097Renal cell (II)U Health Network, TorontoNCT01141569
Sonic HedgehogBMS-833923Basal cell (I)Bristol-Myers SquibbNCT00670189
GDC-0449Solid tumors (I)GenentechNCT00968981
Colorectal (II)GenentechNCT00636610
LDE225Medulloblastoma (I)NovartisNCT00880308
PF-04449913Hematologic (I)PfizerNCT00953758
WntResveratrolColon (I, II)U California, IrvineNCT00256334

Table 3.

Clinical trials on new drugs specifically targeting CSC signaling pathways.

2.4.1. Notch

The Notch/γ-secretase/Jagged signaling pathway is involved in cellular response to intrinsic or extrinsic developmental cues to execute specific developmental programs (Artavanis-Tsakonas et al., 1999). It is an important regulator of differentiation and helps control cell fate. It is also involved in vasculogenesis and angiogenesis. Extensive crosstalk has been shown to exist between Notch and other developmental signaling pathways (Hedgehog and Wnt, see below). Notch signaling is activated by ligand binding. The Notch ligands (Jagged 1 & 2, and Delta 1-3) induce the release of the Notch intracellular (Notch-IC) domain via enzymatic cleavage by α- and γ-secretases. The released Notch-IC will then translocate to the nucleus where it turns on the transcription of Notch responsive genes (Artavanis-Tsakonas et al., 1999; Lehar et al., 2005). Notch signaling pathways are activated in both breast CSCs (Phillips et al., 2006) and in endothelial cells (Scharpfenecker et al., 2009) in response to radiation. γ-secretase inhibitors have been developed to inhibit Notch signaling to block CSC self-renewal and were shown to inhibit the engraftment of medulloblastoma in animal model (Fan et al., 2006).

2.4.2. Wnt/β-catenin

The Wnt signaling pathway is an ancient system that has been highly conserved during evolution. It has been implicated in a wide range of biological processes from maintaining stem cells in their pluripotent state to the induction of specific tissues and organs during development. Canonical Wnt signals are transduced through Frizzled family receptors and LRP5/LRP6 coreceptor to the β-catenin signaling cascade (comprehensively reviewed by Wend et al., 2010). This Wnt/β-catenin signaling pathway is important for self-renewal in stem cells and has been found to be dysregulated in solid and haematopoietic cancers (Zhao et al., 2007; Katoh & Katoh, 2007). The pathway has also been shown to promote genomic instability, thereby enhancing the DNA damage tolerance in CSCs (Eyler & Rich, 2008). Conditional knockout of the key Wnt mediator protein β-catenin in hematopoietic progenitor cells have been shown to delay the development of CML in a bone marrow transplantation model in mice (Zhao et al., 2007).

2.4.3. Sonic hedgehog (SHH)

The SHH pathway is regulated by the binding of Shh (ligand) on the transmembrane receptor patched (Ptch). In the absence of Shh, Ptch inhibits the activity of another transmembrane protein, smoothened (Smo), resulting in inactivation of the SHH pathway (Pasca di Magliano et al., 2003). Binding of Shh to Ptch abrogates the inhibitory effect of Ptch. Smo is derepressed and the transcription factor Gli (Gli1-3) is activated. Gli1 is a potent activator of a number of downstream target genes, including HNF-3β, cyclins D1, IGFBP-6, Snail, CXCR4, and Bcl-2, to regulate neural development, cell proliferation, oncogenesis, survival, epithelial-mesenchymal transition, migration, invasion and metastasis, respectively (Katoh & Katoh, 2007). As positive and negative feedbacks, GLI1 protein respectively activates its own expression and that of PTCH1 (Agren et al., 2004). Therefore, Gli1 is considered a marker of abnormal activation of SHH pathway. While both SHH and Wnt pathways are commonly hyperactivated in tumors to sustain tumor growth, crosstalk between the two pathways has been reported (He et al., 2006), which adds to the complexity of regulation of CSCs. With the development of specific SHH inhibitors such as cyclopamine, the SHH signaling pathway has been proposed to be a druggable target in CSCs (Medina et al., 2009).

2.4.4. PI3K/PTEN/Akt

The PI3K/PTEN/Akt pathway is one of the most extensively studied signal transduction axes that control cell growth, survival, and proliferation (Sarker et al., 2009). The loss of PTEN and the consequent enhancement of Akt pathway activity has been found to constitute the major molecular events accompanying the increased stem cell character and chemoresistance of gliomas (Hu et al., 2005). Activate Akt pathway is also associated with the occurrence of a population of radiation resistant cancer stem-like cells in medulloblastomas, where Akt inhibition appears to sensitize the cells for radiation-induced apoptosis (Hambardzumyan et al., 2008).

With a better appreciation of the CSC-specific signaling pathways, it becomes logical in an attempt to eradicate the tumor by combining these CSC-targeted therapies with standard chemotherapies. Since the aforementioned pathways also govern normal stem cell development and maintenance, it will be critical to establish a dose and schedule where the tumor is suppressed or eliminated without undue toxicity of normal stem cells. Recent data on mouse leukemia models suggest that the PTEN-dependence of CSCs may be exploited for their specific targeting, while sparing the normal haematopoietic stem cells (Yilmaz et al., 2006). Rapamycin was found to selectively kill the leukemia initiating cells in mice harboring a conditional deletion of PTEN, illustrating that novel therapies may be devised specifically for CSCs.

Advertisement

3. Multidrug resistance and cancer stem cells

3.1. Working models of cancer drug resistance

Clinical drug resistance to anticancer therapy is well-known to be multifactorial, involving alteration in drug targets, inactivation of drug, decreased drug uptake, increased drug efflux, and dysregulation of apoptosis pathways (Gottesman, 2002). Usually, cancers that recur after an initial response to chemotherapeutic drugs become resistant to multiple drugs, giving rise to the phenomenon of multidrug resistance (MDR). The traditional belief is that a few cells in the tumor have acquired genetic alteration(s) to confer drug resistance (i.e. “clonal evoluation” model, Figure 3A). These resistant clones have a selective advantage that enables them to outgrow the rest of the tumor following chemotherapy.

Figure 3.

Models of cancer drug resistance

With better appreciation of the role of CSCs in cancer biology, MDR is generally considered to be ultimately caused by CSCs. As discussed above, CSCs share many properties of the normal stem cells, which help protect them from cytotoxic insult throughout their long lifespan. These properties include quiescence, resistance to xenobiotics through the expression of several ATP-binding cassette (ABC) transporters, active DNA repair capacity, and resistance to apoptosis, which collectively make CSCs naturally resistant to chemotherapy. Therefore, after exposure to conventional chemotherapeutic drugs, CSCs survive and are able to repopulate the tumor both with another CSC and with differentiated cells originated from the CSCs (Figure 3B).

This working model where the intrinsic protective mechanisms of CSCs alone provide the basis for drug resistance might be considered too simplistic. A modified “acquired resistance” stem-cell model was thus proposed to more closely resemble the real clinical situation (Figure 3C). This can be exemplified in the recent studies of imatinib resistance in leukemia patients. Imatinib, a tyrosine kinase inhibitor, is a promising molecularly targeted chemotherapeutic agent. It has been shown to be both a substrate and inhibitor of ABCG2, thus allowing its efflux by a stem cell that express this ABC transporter (Houghton et al., 2004; Burger et al., 2004). In-depth mechanistic studies in imatinib-resistant leukemia cells revealed several “acquired” mutations in the kinase domain of ABL in patient with CML or with AML associated with t(9;22)(q34;q11). These findings therefore suggest that CSCs expressing the drug transporter could facilitate, but not be solely responsible for, the acquisition of acquired mechanisms of drug resistance. As for imatinib, the acquired mutation in ABL, the ultimate drug target, could confer higher levels of drug resistance.

Different tumor types may respond differently to chemotherapy. Cancers that respond to initial chemotherapy may appear to acquire drug resistance during the course of treatment. Other cancers may appear to be intrinsically resistant. In either case, the CSC model of drug resistance applies. It is the quiescent CSCs with the innate drug resistance that survive the chemotherapy; and more importantly, they are capable of repopulating the tumor following chemotherapy. On the other hand, acquired drug resistance in more differentiated cancer cells, through gene mutation, amplification or rearrangement, may contribute to an aggressive phenotype, but it is not the primary reason for cancer recurrence or spread after therapy. Therapeutic strategies that specifically target the CSCs should eradicate tumors more effectively than current treatments and reduce the risk of relapse and metastasis.

3.2. ABC Transporters and normal stem cells/CSCs

Among the several protective mechanisms for CSCs, the overexpression of the ATP-binding cassette (ABC) efflux transporters is probably the most important. The ABC transporters belong to the largest superfamily of transport proteins (Gottesman & Ambudkar, 2001). A total of 49 ABC transporter genes have been identified in the human genome and they were grouped into seven subfamilies (designated A to G) according to their structural and sequence homologues (Vasiliou et al., 2009). By using the energy of ATP hydrolysis, these transporters actively efflux drugs from cells, serving to protect them from cytotoxic substances. The two ABC transporter-encoding genes that have been studied most extensively in stem cells are ABCB1 (MDR1), which encodes P-glycoprotein, and ABCG2. Together with ABCC1 (MRP1), they represent the three major multidrug resistance genes that have been identified in cancer cells. Table 4 summarizes the different ABC transporters that have been found to contribute to cancer drug resistance.

GeneProtein / aliasLocation on chromosomeChemotherapeutic drugs effluxedOther important substrates
ABCA2ABCA29q34.3estramustine-
ABCA3ABCA316p13.3daunorubicinsurfactant?
ABCB1P-gp/
MDR1
7q21.12colchicine, doxorubicin, etoposide, vinblastine, paclitaxeldigoxin, saquinivir, rhodamine
ABCB4MDR37q21.12paclitaxel, vinblastinebile salt
ABCB5ABC197p15.3doxorubicin-
ABCB11BSEP/SPGP2q21.3paclitaxel, vinblastinebile salt, pravastatin
ABCC1MRP116p13.12doxorubicin, daunorubicin, vincristine, etoposide, colchicine, camptothecins, methotrexaterhodamine
ABCC2MRP210q24.2vinblastine, cisplatin, doxorubicin, methotrexatesulfinpyrazone, bilirubin
ABCC3MRP317q21.33methotrexate, etoposide-
ABCC4MRP413q32.16-mercaptopurine, 6-thioguanine, methotrexatecAMP, cGMP
ABCC5MRP53q27.16-mercaptopurine, 6-thioguanine,cAMP, cGMP
ABCC6MRP616p13.12etoposide-
ABCC10MRP76p21.1paciltaxelE217βG
ABCC11MRP816q12.15-fluorouracilcAMP, cGMP
ABCG2ABCG2/BCRP4q22mitoxantrone, topotecan, doxorubicin, daunorubicin, irinotecan, imatinib, methotrexatepheophorbide A, Hoechst 33342, rhodamine

Table 4.

ABC transporters involved in drug resistance

3.2.1. ABCB1

ABCB1, also commonly known as P-glycoprotein (P-gp), is the most extensively studied multidrug resistance transporter, which was discovered more than 30 years ago (Jiliano & Ling, 1976). It has been found to be expressed in > 50% of all drug-resistant tumors. Human ABCB1 is the product of the MDR1 gene and acts as an ATP-dependent pump for a multitude of structurally unrelated hydrophobic compounds, including numerous anticancer and antimicrobial drugs (Gottesman & Ambudkar, 2001).

In Hoechst dye exclusion assay using human cancer cell lines, the expression of ABCB1 (usually together with ABCG2) has been found to be higher in the isolated SP cells. As described above, the SP population has an enhanced capacity for the efflux of Hoechst dye, presumably due to ABCB1 and/or ABCG2 expression. Although Zhou et al. reported that ABCB1 may not contribute to the SP phenotype because bone marrow cells from Mdr1a/1b-/- mice are completely lacking in the SP population (Zhou et al., 2001), ABCB1 is still generally considered to be important in protecting CSCs from toxic insult. Result by Zhou et al. may just represent a tumor type-specific observation. Moreover, SP indeed does not define CSCs. The SP fraction is composed of both stem and non-stem cells, and some stem cells are not located in the SP compartment (Zhou et al., 2002).

3.2.2. ABCG2

ABCG2 is a more recently discovered ABC transporter responsible for cancer drug resistance. It was discovered almost simultaneously by three research groups, giving it three different names (BCRP/ABCP/MXR) in the 1990s (Doyle et al., 1998, Allikmets et al., 1998; Miyake et al., 1999). Subsequently, the sequences for these genes turned out to be nearly identical, thereafter the gene was assigned an official name ABCG2 by the Human Gene Nomenclature Committee, which falls into the “G” subfamily of ABC transporters comprising only of half-transporters.

The list of ABCG2 substrates has been expanding rapidly, which highlights the important role of this transporter in drug disposition and treatment outcomes (Polgar et al., 2008). Numerous cancer chemotherapeutic drugs have been identified as ABCG2 substrates, such as mitoxantrone, flavopiridol, topotecan, and some of the newly developed tyrosine kinase inhibitors. There is considerable overlap in substrate drug specificity of ABCG2 and other multidrug resistance transporters, including ABCB1, ABCC1, ABCC2, and some solute carrier transporters. Besides anticancer drugs, several other therapeutic classes have also been described as ABCG2 substrates, including antibiotics, antivirals, flavonoids, and antihyperlipidemic drugs.

Numerous studies have indicated that ABCG2 overexpression plays a possible role in cancer drug resistance, particularly in leukemia (Ross et al., 2010). For example, higher expression of ABCG2 was found to be associated with AML cases (Ross et al., 2000) or with a poor response to remission induction therapy in AML cases (Steinbach et al., 2002). Of note, ABCG2 is often found to be expressed together with P-gp in AML cases, resulting in poor prognosis (Galimberti et al., 2004; van den Heuvel-Eibrink et al., 2007). Interestingly, ABCG2 and ABCB1 mRNA level was found to be higher in non-responding AML cases, only when the primitive subset of CD34+/CD38- leukaemia stem cells were analyzed (Ho et al., 2008). Although the self renewal capability was not evaluated for the CD34+/CD38- cell population in these studies, it appears that they are the tumor-initiating CSCs protected by the increased expression of the transporters.

3.2.3. Physiological role of ABCB1 and ABCG2 in CSCs

Although high expression of ABCB1 and ABCG2 is generally believed to be a marker for normal and/or cancer stem cells, their physiological role in still not clear. Mice deficient in either Abcb1, Abcc1, or Abcg2 are viable, fertile and have normal stem cell compartments (Schinkel et al., 1994; Zhou et al., 2002; Jonker al., 2002). This indicates that none of these transporter genes are necessary for stem cell growth or maintenance. On the other hand, these knockout mice are more sensitive to the effects of drugs such as vinblastine, ivermectin, topotecan and mitoxantrone, consistent with a role for these ABC transporters in protecting cells from toxins.

As discussed above, both ABCB1 and ABCG2 have been proposed to be survival factors for normal stem cells or CSCs by excluding various xenotoxins out of the cells. However and interestingly, ABCB1/P-gp expression generally tracks with the cell differentiation status, where more differentiated tumors tend to have higher expression of the transporter (Mizoguchi et al., 1990; Nishiyama et al, 1993). Moreover, ABCB1/P-gp expression is almost universally found to be upregulated, accompanied by increased expression of markers of maturation, in cancer cell lines treated with differentiating agents (Bates et al., 1989; Mickley et al., 1989). Given that CSCs need to maintain their pluripotent state for self-renewal and repopulating the rest of the tumor, they should be minimally differentiated. It follows that ABCB1/P-gp may not be an important CSC survival factor per se. In contrast, high level functional expression of ABCG2 has been reported in undifferentiated human embryonic stem cells (hESCs) (Apati et al., 2008). The therapeutic implication of these observations is that the undifferentiated and ABCG2-overexpressing cancer cells within a tumor may represent the most chemoresistant putative CSCs that need to be targeted for complete eradication of the tumor.

3.2.4. Overcoming drug resistance by transporter inhibition

3.2.4.1. Early generations of transporter inhibitors

An obvious strategy to restore drug sensitivity in MDR cancer cells caused by ABC drug transporters is to block transporter-mediated drug efflux. Over the past decade, tremendous efforts have been made to discover and synthesize such inhibitors/modulators. Of note, efforts to combat drug resistance caused by the MDR transporters have focused mostly on the use of functional modulators or reversal agents, rather than modulation of the transporter gene regulation. The most well-known inhibitors that have been tested for targeting ABCB1/P-gp (verapamil, cyclosporine A, and valspodar (PSC833)) and ABCG2 (fumitremorgin C (FTC) and Ko143) are also useful research tools for studying modulation of these transporters. A few of these inhibitors, including tariquidar (XR9576) (Kuhnle et al., 2009), can interact with both ABCB1/P-gp and ABCG2. They represent promising lead compounds for further development because drug-resistant tumors usually have overexpression of more than one MDR transporters.

Numerous clinical trials have been performed to evaluate the combination of P-gp modulators with standard chemotherapy regimens in enhancing anticancer efficacy (Sandor et al., 1998). However, they were mostly disappointing and failed to prove the MDR reversal hypothesis, partly because of the lack of specific and potent inhibitors against the MDR transporters. On the other hand, unpredictable pharmacokinetic drug interactions, simultaneous involvement of several drug transporters in tumor tissues, as well as the variability in drug transporter expression levels among individuals, remain obstacles to using modulators to restore drug sensitivity in the clinic.

3.2.4.2. Novel transporter inhibitors may hold promise to target CSCs

The abrupt termination of a phase III clinical trial of a second generation ABCB1/P-gp inhibitor, valspodar (also known as PSC833), due to unexpected toxicity to the patients probably have an enormous negative impact in the field. It was just until recently when the discovery of potent and specific inhibition of P-gp and/or ABCG2 by tyrosine kinase inhibitors (TKIs) has renewed the research interest in developing drug transporter inhibitors for the circumvention of MDR. TKIs are an important new class of molecularly targeted chemotherapeutic agents that specifically inhibit several oncogenic tyrosine kinases, thereby regulating cancer proliferation, invasion, metastasis and angiogenesis. The first TKI that was approved for CML, imatinib, has been shown to reverse MDR by inhibiting ABCB1(P-gp) (Hegedus et al., 2002) and ABCG2 (Houghton et al., 2004). A few other TKIs were also demonstrated to reverse drug resistance mediated by MDR transporters in various in vitro and in vivo models (reviewed in Wang & Fu, 2010). However, it is still controversial as to whether TKIs are substrates or inhibitors of ABCB1 (P-gp) and/or ABCG2, which may depend on the concentration used. Moreover, since these TKIs are acting on oncogenic tyrosine kinases, which may have interactions/crosstalk with the other CSC-specific signaling pathways described before, the novel TKIs may prove to be good drug candidates targeting CSCs.

Given the central role played by ABCB1 and/or ABCG2 in protecting CSCs, specific transporter inhibitors theoretically could be employed as “cancer stem cell sensitizing agents” that allow the most crucial and drug resistant cells in a tumor to be destroyed. These therapies would be predicted to have toxic effects on the patients’ normal stem cells. Since both ABCG2 and ABCB1 are also known to constitute the blood-brain barrier, this approach has to be carefully titrated to avoid excessive toxicity.

Advertisement

4. Regulation of MDR transporters and its relevance to CSCs

As mentioned above, the MDR transporter ABCG2 may be the bona fide CSC survival factor. Therefore, our discussion in this section will focus on ABCG2. Recently, an increasing number of studies have focused on unravelling the molecular regulation of ABCG2 because ABCG2 expression is highly sensitive to various developmental and environmental stimuli.

4.1. Transcriptional regulation of ABCG2 at the promoter level

Early studies examining the regulation of ABCG2 have focused at the transcriptional level. A few functional cis-elements have been identified at the ABCG2 promoter, including hypoxia (Krishnamurthy et al., 2004), estrogen (Ee et al., 2004), progesterone (Wang et al., 2008), and the xenobiotic (aryl hydrocarbon receptor) response elements (Tan et al., 2010; To et al., 2011), which tightly control ABCG2 expression and serve as cellular defense mechanisms against various stimuli. A PPAR-γ response element upstream of the ABCG2 gene has also been shown to facilitate the upregulation of ABCG2 for protecting dendritic cells (Szatmari et al., 2006). Cytokines and growth factors have also been reported to affect ABCG2 levels, though the exact mechanism is not clear.

Other studies on ABCG2 regulation are mostly related to its overexpression in drug-resistant cancer cell lines. The overexpression of ABCG2 has been found to correlate with increased binding of a set of permissive histone modification marks, RNA polymerase II and a chromatin remodelling factor Brg-1, but decreased association of a repressive histone mark, HDAC-1 and Sp1 with the proximal ABCG2 promoter (To et al., 2008a). It has been demonstrated that chromatin dynamics and structure contribute significantly to the maintenance of pluripotency and regulation of differentiation in embryonic stem cells (Shafa et al., 2010). To this end, prolonged drug selection has been found to enrich the resulting subline with CSC characteristics (Calcagno et al., 2010). Therefore, we speculate that the chromatin remodelling observed at the ABCG2 promoter may coincide with the enrichment of the pluripotent CSCs in the drug-selected resistant cells (Figure 4).

Figure 4.

Chromatin remodelling at the ABCG2 promoter allows overexpression of the transporter in drug-selected resistant cells (To et al., 2008a).

A closer look at the ABCG2 promoter also reveal that there are a few putative binding sites for the stem cell transcription factors Oct4 and Nanog (Figure 5), which are known to promote self-renewal and pluripotency (Boyer et al., 2005). To this end, ABCG2 and Oct4/POU5F1 were found to be highly coexpressed in the resistant subline selected from the parental K562 leukemia cells (Marques et al., 2010). These observations are therefore consistent with the notion that ABCG2 is a survival factor for the pluripotent CSCs.

Figure 5.

Putative binding sites for the stem cell transcription factors Oct4 and Nanog at the ABCG2 promoter.

4.2. MicroRNA-mediated regulation of ABCG2

MicroRNAs (miRNAs) are small noncoding RNAs that repress gene expression in a variety of eukaryotic organisms. They play important roles in several cellular processes, such as proliferation, differentiation, apoptosis, and development, by simultaneously controlling the expression levels of hundred of genes. In human cancer, recent studies have shown that miRNA expression profiles differ between normal tissues and derived cancers and between cancer types (Lu et al., 2005). MiRNAs can also act as oncogenes or tumor suppressors, exerting a key function in tumorigenesis (Esquela-Kerscher et al., 2006; Hammond, 2007). Gene regulation by miRNAs is mediated by the formation of imperfect hybrids with the 3’untranslated region (3’UTR) sequences of the target mRNAs, leading to mRNA degradation and/or translational inhibition.

Evidence pointing to the role of miRNAs in determining drug sensitivity and MDR is emerging. First, miRNA expression is largely dysregulated in drug-resistant cancer cells (Zhu et al., 2008; Pan et al., 2009). Second, the miRNA expression patterns in the NCI-60 drug screen cell lines are significantly correlated to the sensitivity patterns of the cancer cells for a large set of anticancer agents (Blower et al., 2008). Third, numerous miRNAs have been found to regulate drug resistance genes such as DHFR (Mishra et al., 2007) and BCL2 (Xia et al., 2008).

We and others have independently identified three miRNAs (miR-519c, -520h, and -328) regulating ABCG2 expression and determining the sensitivity of cancer cells (To et al., 2008b & 2009; Liao et al., 2008; Wang et al., 2010; Pan et al., 2009, respectively). We reported that ABCG2 mRNA is more stable in drug-selected and ABCG2-overexpressing resistant cell lines than in their parental counterparts (To et al., 2008b & 2009). This increase in mRNA stability was tied to a missing miR-519c binding site (also miR-328) in the truncated 3’UTR of ABCG2 mRNA in drug resistant cells (Figure 6). Intriguingly, the truncation of the ABCG2 3’UTR has also been reported in an undifferentiated human embryonic stem (HuES) cell line where its high ABCG2 expression was associated with the short 3’UTR variant forms (Apati et al., 2008). In contrast, another differentiated HuES cell line with lower ABCG2 levels possesses a longer 3’UTR variant (Apati et al., 2008). Sandberg et al. also found that rapidly proliferating cells express ABCG2 mRNA with shorter 3’UTRs, presumably to escape miRNA regulation (Sandberg et al., 2008). Therefore, in the resistant cells, miR-328 and miR-519c (though a proximal miR-519c binding site present also in the truncated 3’UTR has been recently discovered (Li et al., 2011)) cannot bind to ABCG2 mRNA because of the shorter 3’UTR, and thus miRNA-mediated mRNA degradation and/or protein translation block are relieved, contributing to ABCG2 overexpression (Figure 6). In a human retinoblastoma cell line model, it has been further demonstrated that low expressions of all three miRNAs (miR-328, -519c, & -520h) correlate very well with high ABCG2 expression, with concomitant expression of other stem cell markers including CD133 and ALDH1A1 (Li et al., 2011). On the other hand, hsa-miR-520h has been reported to promote differentiation of hematopoietic stem cells by inhibiting ABCG2 expression (Liao et al., 2008). These findings collectively support an important role played by miRNAs in maintaining high ABCG2 level in CSCs. It will be interesting to verify if the same phenomenon is also observed in patient tumor samples.

The regulation of the other two major multidrug resistance transporters, P-glycoprotein/MDR1 and MRP1, by miR-451, -27a and -326, respectively, have also been reported (Kovalchuk et al., 2008; Li et al., 2010; Liang et al., 2010). More importantly, modulation of miRNA expression or function can alter sensitivity of cancer cells to anticancer drugs (Zhu et al., 2008; Pan et al., 2009; Blower et al., 2008). This could be achieved by inhibiting the function of up-regulated miRNAs or restoring the expression of down-regulated miRNAs. Together, miRNAs may represent important players in intrinsic or acquired MDR in cancer cells.

With the general appreciation of the importance of miRNAs in gene regulation, an emerging role of miRNAs in regulating stem cell self-renewal and differentiation has been revealed (Kashyap et al., 2009), which are important for proper stem cell function and maintenance. Recently, the coordinated regulation of miRNAs and various stem cell transcription factors including OCT4, SOX2 and Nanog have emerged as the master regulatory mechanism for stem cells pluripotency and differentiation. Given that ABCG2 could be downstream target of these stem cell transcription factors, it remains to be seen if the miRNA/stem cell transcription factors network could intercept with the regulation of the MDR transporters in contributing to the pluripotent state and chemoresistance of the CSCs.

Figure 6.

A proposed model for ABCG2 upregulation in drug-resistant cells by escaping miRNA repression (To et al., 2009; Li et al., 2011)

Advertisement

5. Conclusion

The CSC model of drug resistance offers an appealing explanation as to why cancers that show an apparent complete clinical response to chemotherapy can relapse months or even years later. Numerous novel strategies to circumvent multidrug resistance have been designed to target the putative CSCs by exploiting pathways involved in MDR transporters-mediated drug resistance, or forcing these cells to proliferate and differentiate thus converting them into a target of conventional therapies. Given the complicated microRNA/pluripotency transcription factor/MDR transporters/CSCs network described above, a better understanding of the various molecular mechanisms regulating pluripotency is pivotal to realizing the therapeutic potential of the novel treatment modalities.

Advertisement

Acknowledgments

We acknowledge the researchers who have contributed to the advancements in cancer stem cells and ABC transporter research and whose works have not been cited here because of space limitations. The work described in this chapter was supported in part by a grant from the NSFC/RGC Joint Research Scheme sponsored by the Research Grants Council of Hong Kong and the National Natural Science Foundation of China (Project No. N_CUHK443/10) and the Seed Research Funding provided by the School of Pharmacy (CUHK) to Kenneth To.

References

  1. 1. AgrenM.KogermanP.LlemanM. I.WesslingM.ToftgardR.2004Expression of the PTCH1 tumor suppressor gene is regulated by alternative promoters and a single functional Gli-binding site. Gene, 330April 2004), 101114
  2. 2. Al-HajjM.WichaM. S.Benito-HernandezA.MorrisonS. J.ClarkeM. F.2003Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci USA, 1007April 2003), 39833988
  3. 3. AllikmetsR.SchrimlL. M.HutchinsonA.Romano-SpicaV.DeanM.1998A human placenta-specific ATP-binding cassette gene (ABCP) on chromosome 4q22 that is involved in multidrug resistance. Cancer Res, 5823December 1998), 53375339
  4. 4. AlviA. J.ClaytonH.JoshiC.EnverT.AshworthA.VivancoM. M.DaleT. C.SmalleyM. J.2003Functional and molecular characterisation of mammary side population cells. Breast Cancer Res, 51October 2002), R1R8
  5. 5. ApatiA.OrbanT. I.VargaN.NemethA.SchambergerA.KrizsikV.Erdelyi-BelleB.HomolyaL.VaradyG.PadanyiR.KarasziE.KemnaE.NemetK.SarkadiB.2008High level functional expression of the ABCG2 multidrug transporter in undifferentiated human embryonic stem cells. Biochim Biophy Acta, 177812December 2008), 27002709
  6. 6. Artavanis-TsakonasS.RandM. D.LakeR. J.1999Notch signaling: cell fate control and signal integration in development. Science, 2845415April 1999), 770776
  7. 7. AsakuraA.RudnickiM. A.2002Side population cells from diverse adult tissues are capable of in vitro hematopoietic differentiation. Exp Hematol, 3011November 2002), 13391345
  8. 8. BatesS. E.MickleyL. A.ChenY. N.RichertN.RudickJ.BiedlerJ. L.FojoA. T.1989Expression of a drug resistance gene in human neuroblastoma cell lines: modulation by retinoic acid-induced differentiation. Mol Cell Biol, 910October 1989), 43374344
  9. 9. BermanD. M.KarhadkarS. S.HallahanA. R.PritchardJ. I.EberhartC. G.WatkinsD. N.ChenJ. K.CooperM. K.TaipaleJ.OlsonJ. M.BeachyP. A.2002Medulloblastoma growth inhibition by hedgehog pathway blockade. Science, 2975586August 2002), 15591561
  10. 10. BlowerP. E.ChungJ. H.VerducciJ. S.LinS.ParkJ. K.DaiZ.LiuC. G.SchmmittgenT. D.ReinholdW. C.CroceC. M.WeinsteinJ. N.SadeeW.2008MicroRNAs modulate the chemosensitivity of tumor cells. Mol Cancer Ther, 71January 2008), 19
  11. 11. BonnetD.DickJ. E.1997Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med, 37July 1997), 730737
  12. 12. BoyerL. A.LeeT. I.ColeM. F.JohnstoneS. E.LevineS. S.ZuckerJ. P.GuentherM. G.KumarR. M.MurrayH. L.JennerR. G.GiffordD.K.MeltonD. A.JaenischR.YoungR. A.2005Core transcriptional regulatory circuitry in human embryonic stem cells. Cell, 1226September 2005), 947956
  13. 13. BudakM. T.AlpdoganO. S.ZhouM.LavkerR. M.MAAkinciWolosin. J. M.2005Ocular surface epithelia contain ABCG2-dependent side population cells exhibiting features associated with stem cells. J Cell Sci, 118No.Pt 8, (April 2005), 17151724
  14. 14. BurgerH.van TolH.BoersmaA. W.BrokM.WiemerE. A.StoterG.NooterK.Imatinibmesylate. . S. T.I57isa.substratefor.thebreast.cancerresistance.protein. B. C. R. P.drugA. B. C. G.pumpBlood, 1049November 200429402942
  15. 15. CalcagnoA. M.SalcidoC. D.GilletJ. P.WuC. P.FostelJ. M.MumauM. D.GottesmanM. M.VarticovskiL.AmbudkarS. V.2010Prolonged drug selection of breast cancer cells and enrichment of cancer stem cell characteristics. J Natl Cancer Inst, 10221November 2010), 16371652
  16. 16. ChenY. W.ChenK. H.HuangP. I.ChenY. C.ChiouG. Y.LoW. L.TsengL.HsuH. S.ChangK. W.ChiouS. H.2010Cucurbitacin I suppressed stem-like property and enhanced radiation-induced apoptosis in head and neck squamous carcinoma-derived CD44(+) ALDH(+) cells. Mol Cancer Ther, 911Novermber 2010), 28792892
  17. 17. ChibaT.KitaK.ZhengY. W.YokosukaO.SaishoH.IwamaA.NakauchiH.TaniguchiH.2006Side population purified from hepatocellular carcinoma cells harbors cancer stem cell-like properties. Hepatology, 441July 2006), 240251
  18. 18. ClarkeM. F.DickJ. F.DirksP. B.EavesC. J.JamiesonC. H.JonesD. L.VisvaderJ.WeissmanI. L.WahlG. M.2006Cancer stem cells- perspectives on current status and future directions: AACR Workshop on cancer stem cells. Cancer Res, 6619October 2006), 93399344
  19. 19. CollinsA. T.BerryP. A.HydeC.StowerM. J.MaitlandN. J.2005Prospective identification of tumorigenic prostate cancer stem cells. Cancer Res, 6523December 2005), 1094610951
  20. 20. DittmarT.NaglerC.SchwitallaS.ReithG.NiggemannB.ZankerK. S.2009Recurrence cancer stem cells- Made by cell fusion? Med Hypotheses, 734October 2009), 542547
  21. 21. DouJ.PanM.WenP.LiY.TangQ.ChuL.ZhaoF.JiangC.HuW.HuK.GuN.2007Isolation and identification of cancer stem-like cells from murine melanoma cell lines. Cell Mol Immunol, 46December 2007), 467472
  22. 22. DoyleL. A.YangW.AbruzzoL. V.KrogmannT.GaoY.RishiA. K.RossD. D.1998A multidrug resistance transporter from human MCF-7 breast cancer cells. Proc Natl Acad Sci USA, 9526December 1998), 1566515670
  23. 23. DuY.FunderburghM. L.MannM. M.SundarRaji. N.FunderburghJ. L.2005Multipotent stem cells in human corneal stroma. Stem Cells, 239October 2005), 12661275
  24. 24. Esquela-KerscherA.SlackF. J.2006OncomiRs-microRNAs with a role in cancer. Nat Rev Cancer, 64April 2006), 259269
  25. 25. EylerC. E.RichJ. N.2008Survival of the fittest: cancer stem cells in therapeutic resistance and angiogenesis. J Clin Oncol, 2617June 2008), 28392845
  26. 26. FanX.MatsuiW.KhakiL.StearnsD.ChunJ.LiY. M.EberhartC. G.2006Notch pathway inhibition depletes stem-like cells and blocks engraftment in embryonal brain tumors. Cancer Res, 6615August 2006), 74457452
  27. 27. FangD. D.KimY. J.LeeC. N.AggarwalS.Mc KinnonK.MesmerD.NortonJ.BirseC. E.HeT.RubenS. M.MooreP. A.2010Expansion of CD133(+) colon cancer cultures retaining stem cell properties to enable cancer stem cell target discovery. Br J Cancer, 1028April 2010), 12651275
  28. 28. FlorekM.HaaseM.MarzescoA. M.FreundD.EhningerG.HuttnerW. B.CorbeilD.2005Prominin-1/CD133, a neural and hematopoietic stem cell marker, is expressed in adult human differentiated cells and certain typrs of kidney cancer. Cell Tissue Res, 3191January 2005), 1526
  29. 29. FrancipaneM. G.AleaM. P.LombardoY.TodaroM.MedemaJ. P.StassiG.2008Crucial role of interleukin-4 in the survival of colon cancer stem cells. Cancer Res, 6811Jun 2008), 40224025
  30. 30. GalliaG. L.TylerB. M.HannC. L.SiuI. M.GirandaV. L.VescoviA. L.BremH.RigginsG. J.2009Inhibition of Akt inhibits growth of glioblastoma and glioblastoma stem-like cells. Mol Cancer Ther, 82February 2009), 386393
  31. 31. GalimbertiS.GuerrinoF.PalumboG. A.ConsoliU.FazziR.MorabitoF.SantiniV.PetriniM.2004Evaluation of BCRP and MDR-1 co-expression by quantitative molecular assessment in AML patients. Leuk Res, 284April 2004), 367372
  32. 32. GandhirajanR. K.StaibP. A.MinkeK.GehrkeI.PlickertG.SchlosserA.SchmittE. K.HallekM.KreuzerK. A.2010Small molecule inhibitors of Wnt/beta-catenin/lef-1 signaling induces apoptosis in chronic lymphocytic leukaemia cells in vitro and in vivo. Neoplasia, 124April 2010), 326335
  33. 33. GoodellM. A.BroseK.ParadisG.ConnerA. S.MulliganR. C.1996Isolation and functional properties of murine hematopoietic stem cells that are replicating in vivo. J Exp Med, 1834April 1996), 17971806
  34. 34. GoodellM. A.RosenzweigM.KimH.MarksD. F.De MariaM.ParadisG.GruppS. A.SieffC. A.MulliganR. C.JohnsonR. P.1997Dye efflux studies suggest that hematopoietic stem cells expressing low or undetectable levels of CD34 antigen exist in multiple species. Nat Med, 312December 1997), 13371345
  35. 35. GottesmanM. M.2002Mechanisms of cancer drug resistance. Annu Rev Med, 53615627
  36. 36. GottesmanM. M.AmbudkarS. V.2001Overview: ABC transporters and human disease. J Bioenerg Biomembr, 336December 2001), 453458
  37. 37. GottesmanM. M.FojoT.BatesS. E.2002Multidrug resistance in cancer: role of ATP-dependent transporters. Nat Rev Cancer, 21January 2002), 4858
  38. 38. HambardzumyanD.BecherO. J.RosenblumM. K.PandolfiP. P.Manova-TodorovaK.HollandE. C.2008PI3K pathway regulates survival of cancer stem cells residing in the pervascular niche following radiation in medulloblastoma in vivo. Genes Dev, 224February), 436448
  39. 39. HamburgerA. W.SalmonS. E.1977Primary bioassay of human tumor stem cells. Science, 1974302July), 461463
  40. 40. HammondS. M.2007MicroRNAs as tumor suppressors. Nat Genet, 395May), 582583
  41. 41. HaraquchiN.IshiiH.MimoriK.TanakaF.OhkumaM.KimH. M.AkitaH.TakiuchiD.HatanoH.NaqanoH.BarnardG. F.DokiY.MoriM.2010CD13 is a therapeutic target in human liver cancer stem cells. J Clin Invest, 1209September), 33263339
  42. 42. HeJ.ShengT.StelterA. A.LiC.ZhangX.SinhaM.LuxonB. A.XieJ.2006Suppressing Wnt signalling by the hedgehog pathway through sFRP-1. J Biol Chem, 28147November), 3559835602
  43. 43. HegedusT.OrfiL.SeprodiA.VaradiA.SarkadiB.KeriG.2002Interaction of tyrosine kinase inhibitors with the human multidrug transporter proteins, MDR1 and MRP1. Biochim Biophys Acta, 15872-3July), 318325
  44. 44. HemmatiH. D.NakanoI.LazareffJ. A.Masterman-SmithM.GeschwindD. H.Bronner-FraserM.KornblumH. I.2003Cancerous stem cells can arise from pediatric brain tumors. Proc Natl Acad Sci USA, 10025December), 1517815183
  45. 45. Hirchmann-JaxC.FosterA. E.WulfG. G.NuchternJ. G.JaxT. W.GobelU.GoodellM. A.BrennerM. K.2004A distinct “side population” of cells with high drug efflux capacity in human tumor cells. Proc Natl Acad Sci USA, 10139September), 1422814233
  46. 46. HoM. M.HoggeD. E.LingV.2008MDR1 and BCRP1 expression in leukemic progenitors correlates with chemotherapy response in acute myeloid leukemia. Exp Hematol, 364April), 433442
  47. 47. HongS. P.WenJ.BangS.ParkS.SongS. Y.2009CD44-positive cells are responsible for gemcitabine resistance in pancreatic cancer cells. Int J Cancer, 12510November), 23232331
  48. 48. HoughtonP. J.GermainG. S.HarwoodF. C.SchuetzJ. D.StewartC. F.BuchdungerE.TraxlerP.2004Imatinib mesylate is a potent inhibitor of the ABCG2 (BCRP) transporter and reverses resistance to topotecan and SN-38 in vitro. Cancer Res, 647April), 23332337
  49. 49. HuX.PandolfiP.LiY.KoutcherJ. A.RosenblumM.HollandE. C.2005mTOR promotes survival and astrocytic characteristics induced by Pten/AKT signalling in glioblastoma. Neoplasia, 74April), 356368
  50. 50. HussainS. Z.StromS. C.KirbyM. R.BurnsS.LangemeigerS.UedaT.HsiehM.TisdaleJ. F.2005Side population cells derived from adult human liver generate hepatocyte-like cells in vitro. Dig Dis Sci, 5010October), 17551763
  51. 51. JamiesonC. H.AillesL. E.DyllaS. J.MuijtjensM.JonesC.ZehnderJ. L.GotlibJ.LiK.ManzM. G.KeatingA.SawyersC. L.WeissmanI. L.2004Granulocyte-macrophage progenitors as candidate leukemia stem cells in blast-crisis CML. N Engl J Med, 3517August), 657667
  52. 52. JonkerJ. W.BuitelaarM.WagenaarE.van der ValkM. A.SchefferG. L.ScheperR. J.PloschT.KuipersF.ElferinkR. P.RosingH.BeijnenJ. H.SchinkelA. H.2002The breast cancer resistance protein protects against a major chlorophyll-derived dietary phototoxin and protoporphyria. Proc Natl Acad Sci USA, 9924November), 1564915654
  53. 53. JulianoR. L.LingV.1976A surface glycoprotein modulating drug permeability in Chinese hamster ovary cell mutants. Biochim Biophys Acta, 4551November), 152162
  54. 54. KashyapV.RezendeN. C.ScotlandK. B.ShafferS. M.PerssonJ. L.GudasL. J.MonganN. P.2009Regulation of stem cell pluriopotency and differentiation involves a mutual regulatory circuit of the NANOG, OCT4, and SOX2 pluriopotency transcription factors with polycomb repressive complexes and stem cell microRNAs. Stem Cells Dev, 187September), 10931108
  55. 55. KatohM.KatohM.2007Wnt signaling pathway and stem cell signaling network. Clin Cancer Res, 1314July), 40424045
  56. 56. KatohY.KatohM.2009Hedgehog target genes: mechanisms of carcinogenesis induced by aberrant hedgehog signalling activation. Curr Mol Med, 97September), 873886
  57. 57. KondoT.SetoguchiT.TagaT.2004Persistence of a small population of cancer stem-like cells in the C6 glioma cell line. Proc Natl Acad Sci USA, 1013January), 781786
  58. 58. KovalchukO.FilkowskiJ.MeservyJ.IlnytskyyY.TryndyakV. P.ChekhunV. F.PogribnyI. P.2008Involvement of microRNA-451 in resistance of the MCF-7 breast cancer cells to chemotherapeutic drug doxorubicin. Mol Cancer Ther, 77July), 21522159
  59. 59. KrivtsovA. V.TwomeyD.FengZ.et al.2006Transformation from committed progenitor to leukemia stem cell initiated by MLL-AF9. Nature, 4427104August), 818822
  60. 60. KuhnleM.EggerM.MullerC.MahringerA.BernhardtG.FrickerG.KonigB.BuschauerA.2009Potent and selective inhibitors of breast cancer resistance protein (ABCG2) derived from the p-glycoprotein (ABCB1) modulator tariquidar. J Med Chem, 524February), 11901197
  61. 61. LapidotT.SirardC.VormoorJ.MurdochB.HoangT.Caceres-CortesJ.MindenM.PatersonB.CaligiuriM. A.DickJ. E.1994A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature, 3676464Februaru), 645648
  62. 62. LechnerA.LeechC. A.AbrahamE. J.NolanA. L.HabenerJ. F.2002Nestin-positive progenitor cells derived from adult human pancreatic islets of Langerhans contain side population (SP) cells defined by expression of the ABCG2 (BCRP1) ATP-binding cassette transporter. Biochem Biophys Res Comm, 2932May), 670674
  63. 63. LeharS. M.DooleyJ.FarrA. G.BevanM. J.2005Notch ligands delta1 and jagged1 transmit distinct signals to T-cell precursors. Blood, 1054February), 14401447
  64. 64. LiZ.HuS.WangJ.CaiJ.XiaoL.YuL.WangZ.2010MiR-27a modulates MDR1/P-glycoprotein expression by targeting HIPK2 in human ovarian cancer cells. Gynecol Oncol, 1191October), 125130
  65. 65. LiangZ.WuH.XiaJ.LiY.ZhangY.HuangK.WagarN.YoonY.ChoH. T.ScalaS.ShimH.2010Involvement of miR-326 in chemotherapy resistance of breast cancer through modulating expression of multidrug resistance-associated protein 1. Biochem Pharmacol, 796March), 817824
  66. 66. LiaoR.SunJ.ZhangL.LouG.ChenM.ZhouD.ChenZ.ZhangS.2008MicroRNAs play a role in the development of human hematopoietic stem cells. J Cell Biochem, 1043June), 805817
  67. 67. LiX.PanY. Z.SeigelG. M.HuZ. H.HuangM.YuA. M.2011Breast cancer resistance protein BCRP/ABCG2 regulatory microRNAs (hsa-miR-328,-519c,-520h) and their differential expression in stem-like ABCG2+ cancer cells. Biochem Pharmacol, 816March), 783792
  68. 68. LuJ.GetzG.MiskaE. A.Alvarez-SaavedraE.LambJ.PeckD.Sweet-CorderoA.EbertB. L.MakR. H.FerradnoA. A.DowningJ. R.JacksT.HorvitzH. R.GolubT. R.2005MicroRNA expression profiles classify human cancers. Nature, 4357043June), 834838
  69. 69. MarquesD. S.SandriniJ. Z.BoyleR. T.MarinsL. F.TrindadeG. S.2010Relationships between multidrug resistance (MDR) and stem cell markers in human chronic myeloid leukemia cell lines. Leuk Res, 346June), 757762
  70. 70. MickleyL. A.BatesS. E.RichertN. D.CurrierS.TanakaS.FossF.RosenN.FojoA. T.1989Modulation of the expression of a multidrug resistance gene (mdr-1/P-glycoprotein) by differentiating agents. J Biol Chem, 26430October), 1803118040
  71. 71. Mc GovernM.VoutevR.MaciejowskiJ.CorsiA. K.HubbardE. J.2009A “latent niche” mechanism for tumor initiation. Proc Natl Acad Sci USA, 10628July), 1161711622
  72. 72. MedinaV.CalvoM. B.Diaz-PradoS.EspadaJ.2009Hedgehog signalling as a target in cancer stem cells. Clin Transl Oncol, 114April), 199207
  73. 73. MishraP. J.HumeniukR.MishraP. J.2007A miR-24 microRNA binding site polymorphism in dihydrofolate reductase gene leads to methotrexate resistance. Proc Natl Acad Sci USA, 10433August), 1351313518
  74. 74. MiyakeK.MickleyL.LitmanT.ZhanZ.RobeyR.CristensenB.BrangiM.GreenbergerK.DeanM.FojoT.BatesS. E.1999Molecular cloning of cDNAs which are highly overexpressed in mitoxantrone-resistant cells: demonstration of homology to ABC transport genes. Cancer Res, 591January), 813
  75. 75. MizoguchiT.YamadaK.FurukawaT.HidakaK.HisatsuguT.ShimazuH.TsuruoT.SumizawaT.AkiyamaS.1990Expression of the MDR1 gene in human gastric and colorectal carcinomas. J Natl Cancer Inst, 8221November), 16791683
  76. 76. MontanaroF.LiadakiK.SchiendaJ.FlintA.GussoniE.KunkelL. M.2004Demystifying SP cell purification: Viability, yield, and phenotype are defined by isolation parameters. Exp Cell Res, 2981August), 144154
  77. 77. MuellerM. T.HermannP. C.WitthauerJ.Rubio-ViqueiraB.LeichtS. F.HuberS.EllwartJ. W.MustafaM.BartensteinP.D’HaeseJ. G.SchoenbergM. H.BergerF.JauchK. W.HidalgoM.HeeschenC.2009Combined targeted treatment to eliminate tumorigenic cancer stem cells in human pancreatic cancer. Gastroenterology, 1373September), 11021113
  78. 78. NakaK.HoshiiT.MuraguchiT.TadokoroY.OoshioT.KondoY.NakaoS.MotoyamaN.HiraoA.2010TGF-beta-FOXO signalling maintains leukaemia-initiating cells in chronic myeloid leukaemia. Nature, 4637281February), 676680
  79. 79. NishiyamaK.ShirahamaT.YoshimuraA.SumizawaT.FurukawaT.Ichikawa-HaraguchiM.AkiyamaS.OhiY.1993Expression of the multidrug transporter, P-glycoprotein, in renal and transitional cell carcinomas. Cancer, 7111June), 36113619
  80. 80. NowellP. C.1976The clonal evolution of tumor cell populations. Science, 19442602328
  81. 81. PalliniR.Ricci-VitianiL.MontanoN.MollinariC.BiffoniM.CenciT.PiercontiF.MartiniM.De MariaR.LaroccaL. M.2011Expression of the stem cell marker CD133 in recurrent glioblastoma and its value for prognosis. Cancer, 1171January), 162174
  82. 82. PanY. Z.MorrisM. E.YuZ. M.2009MicroRNA-328 negatively regulates the expression of breast cancer resistance protein (BCRP/ABCG2) in human cancer cells. Mol Pharmacol, 756June), 13741379
  83. 83. ParkC. H.BergsagelD. E.Mc CullochE. A.1971Mouse myeloma tumor stem cells: a primary cell culture assay. J Natl Cancer Inst, 462February), 411422
  84. 84. PandeyP. R.OkudaH.WatabeM.PaiS. K.LiuW.KobayashiA.XingF.FukudaK.HirotaS.SugaiT.WakabayashiG.KoedaK.KashiwabaM.SuzukiK.ChibaT.EndoM.FujiokaT.TanjiS.MoY. Y.CaoD.WilberA. C.WatabeK.2010Resveratrol suppresses growth of cancer stem-like cells by inhibiting fatty acid synthase. Breast Cancer Res Treat, (December), DOIs10549-010-1300-6
  85. 85. Pasca diMagliano. M.HebrokM.2003Hedgehog signalling in cancer formation and maintenance. Nat Rev Cancer, 312December), 903911
  86. 86. PawelekJ. M.ChakrabortyA. K.2008Fusion of tumor cells with bone marrow-derived cells: a unifying explanation for metastasis. Nat Rev Cancer, 85May), 377386
  87. 87. PatrawalaL.CalhounT.Schneider-BroussardR.ZhouJ.ClaypoolK.TangD. G.2005Side population is enriched in tumorigenic, stem-like cancer cells, whereas ABCG2+ and ABCG2- cancer cells are similarly tumorigenic. Cancer Res, 6514July), 62076219
  88. 88. PhillipsT. M.Mc BrideW. H.PajonkF.2006The response of CD24-/low/CD44+ breast cancer-initiaiting cells to radiation. J Natl Cancer Inst, 9824December), 17771785
  89. 89. PlasilovaM.ZivnyJ.JelinelJ.NeuwirtovaR.CermakJ.EncasE.AnderaL.StopkaT.2002TRAIL (Apo2L) suppresses growth of primary human leukaemia and myelodysplasia progenitors. Leukemia, 161January), 6773
  90. 90. PolgarO.RobeyR. W.BatesS. E.2008ABCG2: structure, function and role in drug response. Expert Opin Drug Metab Toxicol, 41January), 115
  91. 91. ReyaT.CleversH.2005Wnt signalling in stem cells and cancer. Nature, 4347035April), 843850
  92. 92. RossD. D.HakanishiT.2010Impact of breast cancer resistance protein on cancer treatment outcomes, In: Multidrug resistance in cancer, Humana Press.
  93. 93. SalnikovA. V.GladkichJ.MoldenhauerG.VolmM.MatternJ.HerrI.2010CD133 is indicative for a resistance phenotype but does not represent a prognostic marker for survival of non-small cell lung cancer patients. Int J Cancer, 1264February), 950958
  94. 94. SandorV.FojoT.BatesS. E.1998Future perspectives for the development of P-glycoprotein modulators. Drug Resist Updat, 13190200
  95. 95. SarkerD.ReidA. H.YapT. A.de BonoJ. S.2009Targeting the PI3K/AKT pathway for the treatment of prostate cancer. Clin Cancer Res, 1515August), 47994805
  96. 96. ScharenbergC. W.HarkeyM. A.Torok-StorbB.2002The ABCG2 transporter is an efficient Hoechst 33342 efflux pump and is preferentially expressed by immature human hematopoietic progenitors. Blood, 992January), 507512
  97. 97. ScharpfeneckerM.KruseJ. J.SprongD.RussellN. S.tenDijke. P.StewartF. A.2009Ionizing radiation shifts the PAI-1/ID-1 balance and activates notch signaling in endothelial cells. Int J Rad Oncol Biol Phys, 732February), 506513
  98. 98. SchinkelA. H.SmitJ. J.van TellingenO.BeijnenJ. H.WagenaarE.van DeemterL.MolC. A.van der ValkM. A.Robanus-MaandagE. C.teRiele. H. P.1994Disruption of the mouse mdr1a P-glycoprotein gene leads to a deficiency in the blood-brain barrier and to increased sensitivity to drugs. Cell, 774May), 491502
  99. 99. SchmidtP.KopeckyC.HombachA.ZigrinoP.MauchC.AbkenH.2011Eradication of melanomas by targeted elimination of a minor subset of tumor cells. Proc Natl Acad Sci USA, 1086February), 24742479
  100. 100. SeigelG. M.CampbellL. M.NarayanM.Gonzalez-FernandezF.2005Cancer stem cell characteristics in retinoblastoma. Mol Vis, 11September), 729737
  101. 101. ShafaM.KrawetzR.RancourtD. E.2010Returning to the stem state: epigenetics of recapitulating pre-differentiation chromatin structure. Bioessays, 329September), 791799
  102. 102. ShmelkovS. V.StClair. R.LydenD.RafiiS.2005AC133/CD133/prominin-1. Int J Biochem Cel Biol, 374April), 715719
  103. 103. SinghS. K.ClarkeI. D.TerasakiM.BonnV. E.HawkinsC.SquireJ.DirksP. B.2003Identification of a cancer stem cell in human brain tumors. Cancer Res, 6318September), 58215828
  104. 104. SinghS. K.HawkinsC.ClarkeI. D.SquireJ. A.BayaniJ.HideT.HenkelmanR. M.CusimanoM. D.DirksP. B.2004Identification of human brain tumor initiating cells. Nature, 4327015November), 396401
  105. 105. StadlerB. M.Ruohola-BakerH.2008Small RNAs: keeping stem cells in line. Cell, 1324February), 563566
  106. 106. SteinbachD.SellW.VoigtA.HermannJ.ZintlF.SauerbreyA.2002BCRP gene expression is associated with a poor response to remission induction therapy in childhood acute myeloid leukemia. Leukemia, 168August), 14431447
  107. 107. SummerR.KottonD. N.SunX.MaB.FitzsimmonsK.FineA.2003Side population cells and Bcrp1 expression in lung. Am J Physiol Lung Cell Mol Physiol, 2851July), L97L104
  108. 108. TagschererK. E.FasslA.CamposB.FarhadiM.KraemerA.BockB. C.Macher-GoeppingerS.RadlwimmerB.WiestlerO. D.Herold-MendeC.RothW.2008Apoptosis-based treatment of glioblastomas with ABT-737, a novel small molecule inhibitor of Bcl-2 family proteins. Oncogene, 2752November), 66466656
  109. 109. TamakiS.EckertK.HeD.SuttonR.DosheM.JainG.TushinskiR.ReitsmaM.HarrisB.TsukamotoA.GageF.WeissmanI.UchidaN.2002Engraftment of sorted/expanded human central nervous system stem cells from fetal brain. J Neurosci Res, 696September), 976986
  110. 110. ToK. K.PolgarO.HuffL. M.MorisakiK.BatesS. E.2008aHistone modifications at the ABCG2 promoter following treatment with histone deacetylase inhibitor mirror those in multidrug-resistant cells. Mol Cancer Res, 61January), 151164
  111. 111. ToK. K.ZhanZ.LitmanT.BatesS. E.2008bRegulation of ABCG2 expression at the 3’untranslated region of its mRNA through modulation of transcript stability and protein translation by a putative microRNA in the S1 colon cancer cell line. Mol Cell Biol, 2817September), 51475161
  112. 112. ToK. K.RobeyR. W.KnutsenT.ZhanZ.RiedT.BatesS. E.2009Escape from hsa-miR-519c enables drug-resistant cells to maintain high expression of ABCG2. Mol Cancer Ther, 810October), 29592968
  113. 113. ToK. K.RobeyR. W.ZhanZ.BangioloL.BatesS. E.2011Upregulation of ABCG2 by romidepsin via the aryl hydrocarbon receptor pathway. Mol Cancer Res, (Feb) [Epub ahead of print].
  114. 114. TrielC.VestergaardM. E.BolundL.JensenT. G.JensenU. B.2004Side population cells in human can mouse epidermis lack stem cell characteristic. Exp Cell Res, 2951April), 7990
  115. 115. Van den-EibrinkHeuvel.M. M.van der HoltB.BurnettA. K.KnaufW. U.FeyM. F.VerhoefG. E.VellengaE.OssenkoppeleG. J.LowenbergB.SonneveldP.2007CD34-related coexpression of MDR1 and BCRP indicates a clinically resistant phenotype in patients with acute myeloid leukemia (AML) of older age. Ann Hematol, 865329337
  116. 116. VasiliouV.VasiliouK.KebertD. W.2009Human ATP-binding cassette (ABC) transporter family. Hum Genomics, 33April), 281290
  117. 117. VirchowR.1855Editorial. Virchows Arch Pathol Anat Physiol Klin Med, 323
  118. 118. WangF.XueX.WeiJ.AnY.YaoJ.CaiH.WuJ.DaiC.QianZ.XuZ.MaioY.2010hsa-miR-520h downregulates ABCG2 in pancreatic cancer cells to inhibit migration, invasion, and side populations. Br J Cancer, 1034August), 567574
  119. 119. WangK. H.KaoA. P.ChangC. C.LeeJ. N.HouM. F.LongC. Y.ChenH. S.TsaiE. M.2010Increasing CD44+/CD24(-) tumor stem cells, and upregulation of COX-2 and HDAC6, as major functions of HER2 in breast tumorigenesis. Mol Cancer, 9November), 288
  120. 120. WangX. K.FuL. W.2010Interaction of tyrosine kinase inhibitors with the MDR-related ABC transporter proteins. Curr Drug Metab, 117September), 618628
  121. 121. WelteY.AdjayeJ.LehrachH. R.RegenbrechtC. R.2010Cancer stem cells in solid tumors: elusive or illusive? Cell Commun Signal, 81May), 6
  122. 122. WendP.HollandJ. D.ZieboldU.BirchmeierW.2010Wnt signaling in stem and cancer stem cells. Semin Cell Dev Biol, 218October), 855863
  123. 123. WoodwardW. A.ChenM. S.BehbodF.RosenJ. M.2005On mammary stem cells. J Cell Sci, 118Pt16August), 35853594
  124. 124. World Health Organization2008World Cancer Report 2008. 978-9-28320-423-7WHO, Geneva.
  125. 125. XiaL.ZhangD.DuR.PanY.ZhaoL.SunS.HongL.LiuJ.DanD.2008miR-15b and miR-16 modulate multidrug resistance by targeting BCL2 in human gastric cancer cells. Int J Cancer, 1232July), 372379
  126. 126. YilmazO. H.ValdezR.TheisenB. K.GuoW.FergusonD. O.WuH.MorrisonS. J.2006Pten dependence distinguishes haematopoietic stem cells from leukemia-initiating cells. Nature, 4417092May), 475482
  127. 127. ZhaoC.BlumJ.ChenA.KwonH. Y.JungS. H.CookJ. M.LagooA.ReyT.2007Loss of β-catenin impairs the renewal of normal and CML stem cells in vivo. Cancer Cell, 126December), 528541
  128. 128. ZhouS.SchuetzJ. D.BuntingK. D.ColapietroA. M.SampathJ.MorrisJ. J.LagutinaI.GrosveldG. C.OsawaM.NakauchiH.SorrentinoB. P.2001The 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, 79September), 10281034
  129. 129. ZhouS.MorrisJ. J.BarnesY.LanL.SchuetzJ. D.SorrentinoB. P.2002Bcrp1 gene expression is required for normal numbers of side population stem cells in mice, and confers relative protection to mitoxantrone in hematopoietic cells in vivo. Proc Natl Acad Sci USA, 9919September), 1233911344
  130. 130. ZhuH.WuH.LiuX.EvansB. R.MedinaD. J.LiuC. G.YangJ. M.2008Role of microRNA miR-27a and miR-451 in the regulation of MDR1/P-glycoprotein expression in human cancer cells. Biochem Pharmacol, 765September), 582588

Written By

To, Kenneth K.W. and Fu, L.W.

Submitted: 30 November 2010 Published: 23 August 2011