You are on page 1of 10

Cancer Letters 372 (2016) 147–156

Contents lists available at ScienceDirect

Cancer Letters
j o u r n a l h o m e p a g e : w w w. e l s e v i e r. c o m / l o c a t e / c a n l e t

Mini-review

Lung cancer stem cells: The root of resistance


Lauren MacDonagh a, Steven G. Gray a, Eamon Breen b, Sinead Cuffe a, Stephen P. Finn a,c,
Kenneth J. O’Byrne d, Martin P. Barr a,*
a Thoracic Oncology Research Group, School of Clinical Medicine, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St. James’s Hospital and

Trinity College Dublin, Ireland


b Flow Cytometry Core Facility, Institute of Molecular Medicine, Trinity Centre for Health Sciences, St. James’s Hospital and Trinity College Dublin, Ireland
c Department of Histopathology, St. James’s Hospital and Trinity College Dublin, Ireland
d Cancer & Ageing Research Program, Queensland University of Technology, Brisbane, Australia

A R T I C L E I N F O A B S T R A C T

Article history: In the absence of specific treatable mutations, platinum-based chemotherapy remains the gold stan-
Received 10 December 2015 dard of treatment for lung cancer patients. However, 5-year survival rates remain poor due to the
Received in revised form 6 January 2016 development of resistance and eventual relapse. Resistance to conventional cytotoxic therapies pres-
Accepted 7 January 2016
ents a significant clinical challenge in the treatment of this disease. The cancer stem cell (CSC) hypothesis
suggests that tumors are arranged in a hierarchical structure, with the presence of a small subset of stem-
Keywords:
like cells that are responsible for tumor initiation and growth. This CSC population has a number of key
Lung cancer
properties such as the ability to asymmetrically divide, differentiate and self-renew, in addition to having
Cancer stem cells
Chemotherapy increased intrinsic resistance to therapy. While cytotoxic chemotherapy kills the bulk of tumor cells, CSCs
Resistance are spared and have the ability to recapitulate the heterogenic tumor mass. The identification of lung
Markers CSCs and their role in tumor biology and treatment resistance may lead to innovative targeted thera-
pies that may ultimately improve clinical outcomes in lung cancer patients. This review will focus on
lung CSC markers, their role in resistance and their relevance as targets for future therapies.
© 2016 Elsevier Ireland Ltd. All rights reserved.

Introduction and mortality [4,5]. Despite intense efforts to overcome such re-
sistance in lung cancer and other cancer types using novel agents,
Lung cancer and resistance alone and in combination with chemo- and radiotherapy, the un-
derlying mechanisms conferring this resistant phenotype in lung
Lung cancer is the leading cause of cancer-related death world- cancer remain largely unknown [6]. It is now well established that
wide and is classified into two main subtypes; non-small cell lung CSCs constitute a unique subset of cells which are distinct from the
cancer (NSCLC) which accounts for approximately 85% of all lung bulk of tumor cells by their exclusive ability to perpetuate the growth
cancers and small-cell lung cancer (SCLC) which is diagnosed in 15% of a malignant population of cells, indefinitely. This may explain the
of cases [1,2]. Histologically, NSCLC is further divided into three sub- ineffectiveness of many conventional therapies and patient relapse
types; adenocarcinoma, squamous-cell carcinoma and large cell [7]. These important clinical observations have stimulated intense
carcinoma [3]. Smoking is a major risk factor for lung cancer and interest in experimental approaches for further investigation of CSCs
is associated with all histological subtypes, in particular, squa- and their role in the treatment of drug resistant lung cancer.
mous cell carcinomas. Adenocarcinoma on the other hand, is the
predominant lung cancer subtype commonly seen in never smokers. The cancer stem cell hypothesis
Despite improved advances in the treatment of lung cancer, the
5-year survival rate remains low, largely due to the emergence of The CSC hypothesis is now a well-accepted and widely studied
resistance prior to and during the course of treatment with che- field within oncology. Cellular heterogeneity is a histological hall-
motherapy and radiation therapy. This resistance to therapy mark of many solid tumors and the CSC hypothesis suggests that
represents a significant clinical challenge in the treatment of lung this heterogeneity within a tumor is due to a hierarchical cell
cancer and contributes largely to disease progression, recurrence dynamic and the presence of a small subpopulation of cells that
display the properties of normal somatic stem cells [8]. These CSCs
are multipotent, and have two key characteristics of stem cells; the
* Corresponding author. Tel.: +353 1 8963620; fax: +353 1 8963503. abilities to self-renew and to differentiate, properties necessary for
E-mail address: mbarr@stjames.ie; barrma@tcd.ie (M.P. Barr). tumor initiation and progression (Fig. 1A). CSCs can asymmetri-

http://dx.doi.org/10.1016/j.canlet.2016.01.012
0304-3835/© 2016 Elsevier Ireland Ltd. All rights reserved.
148 L. MacDonagh et al./Cancer Letters 372 (2016) 147–156

Lung cancer stem cells

Conventional anti-cancer therapies kill the bulk of the tumor,


however, CSCs exhibit robust intrinsic resistance and survive therapy
due to increased telomere length, activation of anti-apoptotic path-
ways, increased membrane transporter activity and their ability to
migrate and metastasize [23].
Telomeres are comprised of a repetitive G-rich sequence and an
abundance of associated proteins that form an effective cap that pro-
tects chromosome ends. Telomerase, also known as telomere
terminal transferase, is a ribonucleoprotein complex that adds the
telomere DNA sequence repeats to lengthen the telomere. It confers
limitless proliferative capacity to cells through its ability to elon-
gate telomeres [24,25]. Telomerase activity is readily detected in most
cancers but not in somatic tissues and is essential for stem cell in-
tegrity and longevity [26]. Increased telomere length has previously
been shown to confer resistance to a number of anti-cancer thera-
pies; inversely, shortened telomere length is associated with drug
sensitivity [27]. Telomerase inhibition enhanced the pro-apoptotic
response to anti-cancer therapies resulting in delayed relapse fol-
lowing chemotherapeutic treatment [27–29]. Lung cancer stem cells
display longer telomeres than their non-CSC counterparts. Treat-
ment with the specific telomerase inhibitor, MST312, has a strong
and preferential anti-proliferative effect on the lung CSC popula-
tion in vitro and in vivo [14]. Inhibition of telomerase using Imetelstat,
either alone or in combination with Trastuzumab, decreases breast
Fig. 1. Division strategies of cancer stem cells. (A) Cancer stem cells (purple) must CSCs and inhibits their capacity to self-renew in HER2 positive breast
accomplish the dual task of self-renewal and generation of differentiated progeny cancer cells [30].
(blue). (B–D) Strategies that maintain a balance of stem cells and differentiated Several mechanisms have been postulated to play a role in re-
progeny. (B) Symmetric cell division. Each cancer stem cell can give rise to two cancer
sistance, including impaired apoptotic machinery, increased DNA-
stem cells or to two differentiated progeny. (C) Asymmetric cell division, each cancer
stem cell gives rise to one cancer stem cell and one differentiated cell. (D) Cancer repair mechanisms, up-regulation of multidrug resistance proteins
stem cell maintenance may be a combination of symmetric and asymmetric divi- and membrane efflux transporters [31]. Such mechanisms may be
sion strategies. A population strategy provides dynamic control over the balance responsible, at least in part, for driving CSCs into a phenotype of
between stem cells and differentiated progeny, a capacity necessary for the main- reduced apoptotic cell death, which in turn forms the basis of tumor
tenance or expansion of cancer stem cells.
progression. ATP-binding cassette (ABC) transporters, such as
p-glycoprotein and multidrug resistant associated protein (MRP1),
are membrane transporters that can pump structurally unrelated
small molecules, such as cytotoxic chemotherapeutic drugs out of
cally divide (Fig. 1C), which enables them to simultaneously self- the cell. Normal stem cells and lung CSCs express high levels of ABC-
perpetuate and to generate differentiated progeny and give rise to transporters resulting in low intracellular drug concentrations [32,33].
heterogenic tumor, with a consistently maintained CSC subpopu- For example, the ABCG2 gene is highly expressed in hematopoi-
lation [9,10]. Although asymmetric division allows for both self- etic and lung cancer stem cells but is switched off in most terminally
renewal and differentiated progeny, it does not allow the CSC differentiated progeny [34].
subpopulation to expand. However, the stem cell niche has been Cancer stem cells are chemoresistant and metastatic, two fea-
shown to expand during development. Similarly, the CSC popula- tures that correlate with poor prognosis and tumor recurrence [35].
tion expands during periods of stress, suggesting that they can also Lung adenocarcinoma CSCs have increased expression of galectin-
symmetrically divide (Fig. 1B) [11–13]. CSCs can rely on either asym- 1, a multifunctional protein which promotes invasiveness and
metric or symmetric division or a combination of the two (Fig. 1D). metastasis and is associated with poor overall survival and lymph
Previous studies have shown that a pure lung CSC population is node metastasis [36]. Epithelial to mesenchymal transition (EMT)
capable of giving rise to a heterogenic progeny of CSCs and non- is an evolutionary conserved developmental process. Studies indi-
CSC cells through asymmetric cell division, whereas non-CSC lung cate that metastatic cancer cells, cells which have undergone EMT,
cancer cells undergo only symmetric division resulting in a pure non- exhibit a CSC phenotype [37,38]. Numerous pathways, including the
CSC cell population [14]. Wnt/β-catenin [39–42], Notch [43,44], Hedgehog [45–47] and NFκB
Cellular stress, such as chemotherapy treatment, can induce sym- [48] regulate CSC maintenance as well as EMT and may prove to
metric division of rare CSC populations and apoptosis of non-CSC be key targets in eradicating the CSC population and therefore the
cells, therefore enriching the population [15]. Symmetric division root of resistance and metastasis [37]. Induction of EMT confers re-
of the CSC population during initial cycles of chemotherapy may sistance of NSCLC cells to EGFR-tyrosine kinase inhibitors, however,
trigger relapse in the form of a chemoresistant tumor [16]. Despite enforced inhibition of the Hedgehog pathway re-sensitizes the NSCLC
an initial response to anti-cancer treatments, chemotherapeutic treat- cells to EGFR-tyrosine kinase inhibitors through mediation of the
ment can lead to the symmetric propagation of a small subpopulation EMT process [47].
of drug-tolerant cells with stem cell features [17–20]. Cisplatin treat- Previously identified lung CSC subsets have been shown to confer
ment causes the lung CSC population to expand in patient ex- resistance to conventional chemotherapeutics, biological mol-
plants and in H460-xenografted nude mice. Similar studies have also ecules, targeted therapies and radiotherapy used in the current
shown side-population CSC expansion following treatment with cy- management of lung cancer. The elimination of lung CSCs is of
totoxic drugs such as 5’fluorouracil, an anti-metabolite commonly utmost importance at the time of therapeutic intervention in order
used in the treatment of several tumor types [21,22]. to prevent CSC expansion and subsequent tumor recurrence, relapse
L. MacDonagh et al./Cancer Letters 372 (2016) 147–156 149

Fig. 2. Anti-cancer therapies induce CSC expansion. Treatment with conventional chemotherapeutic agents or radiotherapy kills the bulk of tumor cells (blue). However
the intrinsically resistant CSCs (purple) survive. Cellular stress caused by chemotherapeutic treatment can induce expansion of these CSCs. The survival and expansion of
CSCs promotes tumor relapse and metastasis.

and metastasis (Fig. 2). Identifying a lung CSC population and elu- The Notch pathway is a highly conserved embryogenesis sig-
cidating its mechanisms of intrinsic resistance may highlight a key naling pathway involved in the determination of cell fate [60].
molecule or pathway targeting strategy that may prevent tumor re- This pathway has been identified as a key pathway associated
sistance and recurrence at its source, leading to more effective lung with the CSC maintenance of ALDH1-positive lung CSCs. Suppres-
cancer treatments. While little is currently known regarding lung sion of Notch signaling with a γ-secretase inhibitor results in a
cancer stem cell biology, a number of CSC markers have be iden- much reduced ALDH1 expressing population and decreased pro-
tified and studied. These include ALDH1, CD133, side population liferation and clonogenic capacity of tumor cells [59]. In a study
(Hoechst-negative), CD44, CD87 and CD117, all of which have been by Barr et al., an isogenic panel of cisplatin resistant NSCLC cell
linked to chemoresistance in a number of first line anti-cancer lines was generated by chronic exposure to cisplatin. Increased
therapies. ALDH1 activity correlated with significantly increased expression
of CSC and EMT markers, Nanog, Oct-4, Sox-2, c-Met and β-catenin
Aldehyde dehydrogenase and resistance in the cisplatin resistant cell lines relative to their sensitive
counterparts [61].
The aldehyde dehydrogenase (ALDH) family members are cy- Treatment with paclitaxel has also been shown to induce a greater
tosolic isoenzymes responsible for oxidizing intracellular aldehydes, ALDH1 CSC population in H460 and H1299 lung cancer cell lines,
including the oxidation of retinol (vitamin A) to retinoic acid, an whereas treatment with the selective CSC inhibitor, salinomycin, de-
important modulator of gene expression and cell differentiation. creased the presence of the ALDH1 population and reduced the
ALDH is highly expressed in hematopoietic stem cells [49,50]. In tumorsphere-forming potential of these cells, with no effect on the
recent years ALDH1 has emerged as a prominent marker for CSCs ALDH1-negative population [62]. While paclitaxel treatment in an
in a number of solid tumors such as breast, colon, head and neck, in vivo model decreased tumor volume, it increased the number of
prostate [51–55], and lung cancer [56]. ALDH1 is expressed in a metastatic nodules, most likely due to the expansion of an ALDH1-
number of NSCLC cell lines, where ALDH1-positive cell subpopu- positive tumor initiating CSC population. Salinomycin alone did not
lations show distinct stem-like characteristics and gene expression reduce the size of the primary tumor but decreased metastasis [62].
profiles. Low numbers of ALDH1-positive cells, when injected into In addition to chemotherapy, radiotherapy has also been shown to
nude mice have the ability to initiate tumor growth and generate induce and expand the ALDH1-positive CSC population in lung ad-
heterogenic tumors. On the other hand, their ALDH1-negative coun- enocarcinoma A549 cells. Repeated 4 Gy radiation treatment was
terparts do not demonstrate this ability, indicating that the ALDH1- used to generate an A549 radioresistant cell line, which showed
positive fraction is capable of self-renewal and can give rise to a greater proliferative and clonogenic capacity when challenged with
differentiated cell population. ALDH1-positive cells are highly re- radiation, compared to their parental counterparts. Expansion of the
sistant to chemotherapeutic agents commonly used as first-line ALDH1-positive subpopulation correlated with the development of
therapy in the clinical setting, such as cisplatin, gemcitabine, doxo- radioresistance and increased doses of radiation [63].
rubicin, vinorelbine and docetaxel, whereas ALDH1-negative cells Resistance to epidermal growth factor receptor (EGFR) tyro-
are sensitive to the cytotoxic activity of these drugs [56]. ALDH1 sine kinase inhibitors is a major problem in the treatment of lung
overexpression is associated with poor prognosis in NSCLC pa- cancer. A model of acquired resistance to gefitinib was estab-
tients, where high ALDH1 expression is significantly associated with lished in a number of EGFR mutated cell lines, EGFR-mutant HCC827
a more aggressive and advanced pathological grade and stage [56,57]. (exon19del E746–A750), PC-9 (exon19del E746–A750), HCC4006
Furthermore, increased ALDH1 expression has been associated with (exon19del L747–E749), and HCC4011 (L858R) cells [64]. Cell lines
increased metastasis in multiple cancers [58,59]. were treated using a stepwise dose escalation or high-dose gefitinib
150 L. MacDonagh et al./Cancer Letters 372 (2016) 147–156

Table 1 conditions select undifferentiated CD133-positive cells and promote


Potential strategies targeting CSC markers. their enrichment and expansion and induce sphere formation [93,94].
CSC Marker Drug Reference Overexpression of the embryonic stem cell transcription factors
ALDH1 Salinomycin [62] Oct-4 and Nanog confirmed the undifferentiated immature phe-
Silibinin [67] notype of CD133-positive cells [93]. In the presence of serum, tumor-
DEAB [68,69] initiating spheres adhered to the plastic and acquired the
Disulfiram [70–73] morphological features of differentiated cells. CD133 expression
CD133 Pantoprazole [21]
Neuropeptide antagonists [21]
was lost during differentiation. Subcutaneous injection of lung
Side population Verapamil [74] sphere-forming cells or pure CD133-positive populations into im-
EGFR Inhibitors [75,76] munodeficient mice resulted in the growth of tumors with
Obatoclax [77] morphological features similar to those seen in the human tumor
GDC-0449 [78]
from which they were derived, confirming the tumorigenic poten-
Mithramycin A [79]
Nicardipine [80] tial of these CD133 expressing sphere-forming cells [93]. Injection
Pravastatin [80] of differentiated cells or CD133-negative subpopulations using the
Secalonic Acid D [81] same animal model resulted in decreased tumorigenic potential
Low Weight Molecular [82] and loss of key stemness characteristics. Lung tumor CSC-spheres
Heparin
Axitinib [83]
and CD133-positive cells showed resistance to cisplatin, gemcitabine,
CD117 Imatinib [84] paclitaxel, doxorubicin and etoposide when treated with peak plasma
Axitinib [83] concentrations. Resistance was directly correlated with increased
A number of cancer stem cell (CSC) markers identified to date may offer potential expression of the ABC-transporter, ABCG2 [93,95]. Knock-down of
as specific targets for therapy, particularly in chemoresistant lung cancer. Combi- the embryonic cell marker Oct-4 specifically blocked the ability of
nation treatments using novel agents or drugs targeting stem-like cells with CD133-positive cells to form spheres and further promoted their
conventional anti-cancer treatments may further enhance the efficacy of chemo- differentiation into CD133-negative cells, thereby reducing the
therapeutic drugs. As such, co-treatment strategies may re-sensitize drug resistant
lung cancer cells to therapy, while at the same time inhibiting expansion of the CSC
CD133-positive population. Oct-4 inhibition decreased the inva-
population within the tumor. sive and tumorigenic potential of CD133-positive cells in vivo and
increased apoptosis of the CD133-positive cells in response to ra-
diotherapy in vitro [95].
to generate resistant sublines. Treatment with high-dose gefitinib Platinum-based chemotherapy, such as cisplatin and carboplatin,
induced ALDH1 overexpression, increased EMT-associated markers is an integral first-line therapy for patients with NSCLC. However,
and self-renewal capabilities. These results suggest that acquired tumor recurrence and resistance is commonly seen during the course
resistance is based, at least in part, on the expansion of a stem cell- of treatment with these platinum agents. Cisplatin treatment, either
like population [64,65]. Gefitinib resistant cells exhibit a higher acute or chronic, resulted in an 8-fold enrichment of CD133-
tumor-initiating capability than their parental controls when in- positive fractions. Analysis of CD133 in patient tumor resections
jected subcutaneously into NOD/SCID mice. Gefitinib resistant identified a CD133-positive subtype as a biomarker for predicting
sublines displayed resistance to the commonly used chemothera- shorter progression-free survival [96]. This enrichment was thought
peutic agents, docetaxel and paclitaxel, an effect that may be due to be due to drug resistance and recurrence. Low-dose cisplatin treat-
to the expansion of the ALDH1 CSC population [64]. Isolated ALDH1 ment, sufficient to cause DNA-damage but not cell death, expanded
cells from gefitinib resistant PC-9 cells showed increased resis- the CD133-positive population and up-regulated ABCG2 and ABCB1
tance to cisplatin, etoposide and fluorouracil compared to ALDH1- drug efflux-associated proteins in H460 and H661 cell lines [21].
negative cells [66]. A similar induction of an ALDH1-positive CD133 expansion and stem-cell associated gene expression was de-
subpopulation was observed during the development of NSCLC re- pendent on cisplatin concentration and exposure time. Cisplatin-
sistance to the EGFR tyrosine kinase inhibitor, erlotinib. ALDH1- primed cells show increased tumor-sphere formation in soft agar
positive cells showed a refractory, inherent resistance to this EGFR colony-forming assays. Cells pre-treated with cisplatin showed in-
TKI. Treatment with a natural polyphenol, silibinin, specifically targets creased cross-resistance to doxorubicin and paclitaxel. Treatment
erlotinib-refractory ALDH1-positive cells, reducing population size of side population cells with a specific ABCG2 inhibitor
and circumventing erlotinib resistance in vivo, minimizing the ability (pantoprazole) or a pan-inhibitor of ABC transporters (Verapamil)
of the CSCs to evade death and therefore reducing the possibility completely abrogated cisplatin-induced resistance [21].
of recurrence [67]. Targeting ALDH1, in addition to one or more CSC Tumor-hypoxia has long been considered a therapeutic problem
surface markers, may offer potential as novel strategies for over- in many solid tumors. Recent studies have shown that hypoxic con-
coming the development of treatment resistance in lung cancer ditions significantly increase CD133-positive and Oct-4 expressing
(Table 1). cells, which correlate with increased gefitinib resistance. Hypoxic
expansion of gefitinib resistant cells was mediated through the up-
CD133 and resistance regulation of HIF-1α and insulin-like growth factor-1 expression and
the subsequent activation of insulin-like growth factor-1 receptor
The CD133 antigen, known as prominin 1, is a 120 kDa pentaspan [97]. Inhibition of HIF-1α or insulin-like growth factor-1 receptor
transmembrane protein, encoded by the prom1 gene. It is highly significantly decreases CD133 and Oct-4 positive populations and
expressed on undifferentiated cells, such as hematopoietic stem overcomes gefitinib resistance [97]. Elevated CD133 expression is
cells, endothelial progenitor cells and neural stem cells [85–87]. associated with intrinsic chemotherapy and radiotherapy resis-
CD133 is a well-documented CSC marker that represents a tumor- tance due to reduced apoptotic response, diminished cell cycle arrest
initiating cell subset in breast, colon, prostate, liver and ovarian and enhanced DNA repair capacity [98]. SCLC chemoresistant CD133-
solid tumors, as well as SCLC and NSCLC [88–93]. Immunohisto- positive cells show increased expression of the mitogenic
chemical analysis of patient-derived tumors showed variation in neuropeptide receptor for gastrin-releasing peptide and arginine va-
CD133 positivity among patients, but was not detectable in normal sopressin. These CD133-positive cells show increased sensitivity to
lung tissues [93]. SCLC and NSCLC tumors were dissociated and the growth inhibitory and pro-apoptotic effects of broad spec-
cultured at a low density in serum-free media supplemented with trum neuropeptide antagonists, highlighting a potential role for
epidermal growth factor and basic fibroblast growth factor. Such neuropeptide antagonists as CSC targeting agents in SCLC [99].
L. MacDonagh et al./Cancer Letters 372 (2016) 147–156 151

A meta-analysis of 11 studies, consisting of 1004 NSCLC patient population occurrence has been shown to re-sensitize adenocar-
tissues, further confirmed that high CD133 expression correlates with cinoma (A549) and malignant pleural mesothelioma (ZL55 and
significantly worse 5-year overall survival compared to those with SDM103T2) cell lines to doxorubicin and mitoxantrane, respective-
low CD133 expression. CD133 expression was associated with a ly [109,110]. In a similar study, treatment of A549 and Calu-6 cell
number of clinical parameters such as tumor stage, grade, differ- lines with mithramycin A (a selective inhibitor of the binding of tran-
entiation status and lymph node metastasis [100,101]. scription factor Sp1 to the ABCG2 promoter region) repressed smoke-
induced expression of ABCG2 and side population frequency [79,111].
Side population and resistance Occurrence of the side population has been associated with re-
The flow cytometry based side population strategy was initially sistance to many chemotherapeutic agents and targeted therapies
conceived to enrich or isolate hematopoietic stem cells. It is based used today due to its characteristic excessive efflux capacity. Tar-
on the ability of stem cells to efficiently efflux Hoechst 33342, and geting side population cells may prove to be an important mechanism
therefore present as a Hoechst-negative population on a dual wave- to overcome resistance to a number of therapies by reducing drug
length flow cytometry plot. This technique has been used to isolate efflux from cells, thereby increasing drug retention within the cells.
putative cancer stem cells from a number of solid tumor types, in- In a recent study by Wu et al., NSCLC patients exhibiting chemo-
cluding prostate, breast, malignant pleural mesothelioma and lung resistance to platinum-based chemotherapy had elevated serum
[74,102–105]. Tumor side population cells pump out fluorescent levels of cholesterol and increased expression of ABCG2 [80]. In an
Hoechst 33342 dye (Hoechst-negative), whereas the main bulk pop- in vitro model of NSCLC using A549 cells, cholesterol increased che-
ulation, retain the dye (Hoechst-positive). The efflux capacity of the moresistance while inhibition of ABCG2 with Nicardipine reversed
side population phenotype is determined by ATP-binding cassette cholesterol-induced platinum-resistance. Similarly, treatment with
membrane transporter G2 (ABCG2), also known as breast cancer re- the statin Pravastatin, re-sensitized A549 cells to cisplatin, carboplatin
sistance protein 1 (BCRP1) and is associated with tumor growth, and oxaliplatin. This study suggests that lung cancer patients with
progression and metastasis [106]. ABCG2 actively effluxes a wide high cholesterol may respond to platinum-based therapy when com-
variety of chemically and structurally unrelated compounds from bined with a statin [80]. Secalonic acid D, a marine fungal-derived
cells; ABCG2 overexpression in tumor cells confers multidrug re- metabolite, has a potent anti-cancer effect through its inhibition of
sistance to a multitude of anti-cancer drugs [107]. Hoechst staining ABCB1, ABCC1 and most significantly ABCG2, in multidrug resis-
of NSCLC cell lines confirmed the presence of a Hoechst-negative tant cells. Treatment with Secalonic acid D significantly increased
side population. However, treatment of cells with verapamil, a ABCG2 degradation via the activation of caplain 1, decreased the
calcium-channel blocker and inhibitor of ABCG2, completely elimi- percentage of side population cells and inhibited the sphere-
nated the presence of the side population, therefore confirming the forming abilities of the side population CSC cells [81]. Low weight
transporter ABCG2 as an integral efflux pump in CSCs. NSCLC side molecular heparin has been suggested to affect the biological prop-
population cells show increased proliferative capacity, tumorige- erties of side population cells. Treatment of side population cells
nicity and overexpression of ABCG2 compared to non-side population with low weight molecular heparin decreased the sphere-forming
cells [33,74]. abilities of the CSC population through the degradation of ABCG2.
NSCLC side population cells are enriched in the cell cycle G0/G1 While treatment of the cisplatin resistant side population cells with
phase, have higher ALDH1 activity, higher clonogenic survivability low weight molecular heparin alone did not affect their prolifera-
and increased self-renewal and differentiation capacities compared tive capacity, the side population cells were re-sensitized to cisplatin-
to the main cell population. Treatment of NSCLC cell lines (A549, H1650 induced apoptosis when used in combination with cisplatin [82].
and H1975) with EGFR tyrosine kinase inhibitors resulted in de-
creased side population frequency and ABCG2 expression. Downstream
regulation of the EGFR-Src-Akt signaling axis resulted in significant CD44 and resistance
inhibition of Sox-2. This inhibition resulted in the abrogation of the CD44 is an integral membrane glycoprotein that functions as a
self-renewal and metastatic abilities of the side population cells [75,76]. receptor for the extracellular matrix glycan, hyaluronan. CD44 is ex-
β-arrestin is involved in the progression of NSCLC, where its inhibi- pressed on the surface of specific glandular, fibroblastoid and
tion has been shown to significantly decrease the presence of the side hematopoietic cells [112]. CD44 has been identified as an impor-
population and inhibit the growth and expansion of A549 cells. The tant marker of CSCs in breast, prostate, pancreatic, squamous head
Bcl-2 family member, MCL-1, is overexpressed in the side popula- and neck, and in more recent years, lung cancer [113–116]. NSCLC
tion and is associated with the self-renewal of CSCs. MCL-1 specific cells expressing CD44 are enriched for stem-like properties. CD44-
inhibition, with Obatoclax, prevented self-renewal of EGFR–resistant positive cells showed increased spheroid body formation, enhanced
NSCLC cells [77]. The Hedgehog pathway has long been implicated self-renewal abilities and increased tumor initiating capacity in vivo
in the maintenance of CSCs. Treatment of adenocarcinoma cells compared to their negative counterparts, and the heterogenic CD44
(HCC) and SCLC cells (H1339) with the Hedgehog inhibitor GDC- population from which they were derived [117]. CD44-positive cells
0449, inhibited cell growth and significantly decreased side population showed increased expression of pluripotency markers (Nanog, Oct-
frequency. Hedgehog inhibition in combination with cisplatin in- 4, Sox-2) and EMT markers (SNAI1, CDH2 and VIM) and increased
creased the cytotoxic effects of cisplatin when compared to cisplatin- resistance to cisplatin [117]. NSCLC cells surviving 5 Gy radiation
only treatment [78]. Small molecule inhibition or gene silencing of therapy were enriched for CD44 expressing cells, and showed in-
CK2α resulted in the down-regulation of the Hedgehog pathway and creased tumor initiating potential, indicating an inherent
subsequently induced a reduction of the side population percent- radioresistance within the CD44-positive subpopulation [118]. Both
age [108]. lung cancer and malignant pleural mesothelioma cells co-expressing
Cigarette smoking at diagnosis or during lung cancer treat- CD44 with ALDH1 showed increased drug resistance, sphere-
ment is associated with poor prognosis and cigarette smoke has now forming capacity and tumorigenicity [15,68].
been shown to promote drug resistance via the expansion of the Immunohistochemical staining of 159 resected NSCLC tumors
CSC side population [109]. Treatment of A549 adenocarcinoma cells showed that CD44 expression is more frequently detected in squa-
with cigarette smoke condensate resulted in a significant increase mous cell carcinoma compared to adenocarcinoma histology. High
in side population frequency, ABCG2 expression and doxorubicin CD44 expression was also significantly correlated with advanced
efflux. This effect was mediated by PI3K/Akt signaling. PI3K inhi- regional lymph node metastasis and is a significant predictor of a
bition and subsequent ABCG2 down-regulation and decreased side more advanced tumor status in squamous cell carcinomas [119].
152 L. MacDonagh et al./Cancer Letters 372 (2016) 147–156

CD87 and resistance exhibit increased expression of CD133, CD117, Oct-4, TRA-1-81, in-
Numerous solid tumors, including lung cancer, show increased creased nuclear expression of β-catenin and resistance to
expression of urokinase plasminogen activator (uPA) and its recep- chemotherapy [84]. CSC-spheres also showed high expression of SCF,
tor, urokinase plasminogen activator receptor (uPAR), otherwise whereas SCF was not detectable in the bulk cell population. Exog-
known as CD87. CD87 is highly involved in migration and in the reg- enous SCF expression increased CSC-sphere size and stimulated their
ulation of cell adhesion. High expression of CD87 is strongly proliferation. Treatment of the CD117-expressing CSC-spheres with
correlated with an unfavorable clinical outcome and a signifi- Imatinib (Gleevec), an FDA-approved tyrosine kinase CD117 inhib-
cantly shorter overall survival in SCLC [120,121]. In an in vivo model itor commonly used in the treatment of AML and GIST, dramatically
of NSCLC, simultaneous inhibition of CD87 and MMP-9 inhibited reduced CSC-sphere size and number. Imatinib given in combina-
tumor growth, invasion, metastasis and angiogenesis [122]. Reduc- tion with cisplatin or doxorubicin led to a potent elimination of both
tion of CD87 signaling increased cisplatin-induced apoptosis and CSC-sphere forming cells and the bulk cell population [84].
anoikis, a method of programmed cell death induced by anchorage-
dependent cells detaching from the extracellular matrix [123]. Gutova Targeting CSCs to overcome therapeutic resistance in lung cancer
et al. identified a rare population of CD87-positive cells in a panel
of human SCLC cell lines (H1688, H1417, H69AR, H211, H1882 and Specific targeting of cancer stem cells in combination with first-
H250). Isolation of these CD87-positive cells confirmed the expres- line chemotherapeutic agents holds great promise as a strategy to
sion of stemness and CSC markers, such as CD44 and MDR1, overcome chemoresistance, tumor relapse and metastasis (Fig. 3).
respectively. The CD87-positive population demonstrated high However, such specific targeting of CSCs has not been extensively
sphere-forming capabilities, increased tumor initiating potential, as explored as a therapeutic option in the treatment of lung cancer.
well as significant resistance to numerous traditional chemothera- Inhibition of CSC markers, such as ABCG2, has been shown to
peutic agents, including cisplatin, etoposide and paclitaxel enhance the efficacy of chemotherapeutic agents in CSC cells [83].
[120,124,125]. Axitinib is an oral, potent, small molecule ATP competitive multi-
target tyrosine kinase inhibitor. It inhibits CSC markers CD117 and
CD117 and resistance ABCG2. Axitinib has also been shown to reverse multidrug resis-
CD117 (KIT, c-KIT) is a type III receptor tyrosine kinase that is tance via ABCG2 inhibition both in vitro and in vivo. Axitinib–
phosphorylated upon binding of its ligand, stem cell factor (SCF), doxorubicin combination treatment promoted intracellular
leading to a signaling cascade that culminates in the alteration of accumulation of doxorubicin within side population CSC cells, and
a number of biological processes, such as apoptosis, cell differen- significantly enhanced the cytotoxic effects of doxorubicin [83].
tiation, cell adhesion and proliferation [126]. SCF and CD117 are An increasing body of evidence is beginning to emerge in rela-
overexpressed in a number of human malignancies, including gas- tion to the inhibition of ALDH1 in stem-like tumor cells that are
trointestinal stromal tumors (GIST) [127], breast cancer [128], acute resistant to therapy [132]. Conventional chemotherapy has had little
myeloid leukaemia (AML) [129] and lung cancer [130]. High SCF ex- effect in the treatment of malignant pleural mesothelioma. However,
pression in lung adenocarcinoma is strongly associated with smoking combination treatment of ALDH-high, CD44-positive subpopula-
status and poor prognosis [131]. Overexpression of CD117 within tions isolated from three mesothelioma cell lines (H28, H2052 and
lung tumors is also associated with poor prognosis, lower survival Meso4) with the ALDH1 inhibitor, diethylaminobenzaldehyde (DEAB),
rates and chemoresistance [84]. NSCLC cell lines, H460 and A549, increased cisplatin sensitivity and significantly reduced cell viabil-
when grown in CSC selective media, gave rise to tumorspheres that ity [68]. Knockdown (siRNA) or chemical inhibition (using DEAB or

Fig. 3. Future implications for CSC-targeted therapies. (A) Current treatment strategies focus on debulking the tumor (blue) but not the intrinsically resistant CSC subset
(purple). Chemotherapy kills the majority of cells within the main tumor cell population. However the CSC subset survives and gives rise to new, therapeutically resistant
heterogenic tumors and metastases. (B) Combining current cytotoxic therapies with novel CSC-targeted therapies may eradicate this CSC subset and therefore inhibit the
root of chemoresistance, tumor recurrence and metastasis, thereby improving clinical outcomes.
L. MacDonagh et al./Cancer Letters 372 (2016) 147–156 153

disulfiram) of ALDH1A1 in A549 and H522 lung cancer cells de- many have been shown to be well tolerated and show enhanced
creased the cells proliferative and migratory capabilities [69]. Of efficacy when used in combination with conventional cytotoxic
interest is the recent re-purposing of the FDA-approved pan- chemotherapies.
ALDH1 inhibitor, disulfiram (Antabuse), originally used in the
treatment of chronic alcoholism [133]. In more recent years, a Acknowledgments
number of studies have demonstrated the potential use of disulfi-
ram in targeting ALDH1 expressing CSCs in breast, cervical, prostate, This work was funded by Molecular Medicine Ireland (MMI), as
melanoma and lung tumors [134–138]. As such, disulfiram was used part of the Clinical & Translational Research Scholars Programme
to examine its effects in re-sensitizing cells to current therapies or (CTRSP).
enhancing the cytotoxic effects of platinum-based chemotherapy
and radiation therapy [70,71]. When disulfiram is administered in
combination with copper chloride in vitro, it acts as an effective Conflict of interest
proteasome inhibitor and as a copper ionophore. Disulfiram–
copper complexes are formed and induce copper-dependent The authors have no conflict of interest to declare.
oxidative stress, exerting its potent apoptotic and anti-cancer effects
[72]. In a small phase IIb trial, disulfiram in combination with References
cisplatin and vinorelbine was well-tolerated and significantly pro-
longed overall patient survival in metastatic NSCLC patients [73]. [1] A. Jemal, F. Bray, M.M. Center, J. Ferlay, E. Ward, D. Forman, Global cancer
Interestingly, of the forty patients included in the trial, there were statistics, CA Cancer J. Clin. 61 (2011) 69–90.
[2] J. Ferlay, I. Soerjomataram, R. Dikshit, S. Eser, C. Mathers, M. Rebelo, et al.,
two long-term survivors, both of whom were in the disulfiram com- Cancer incidence and mortality worldwide: sources, methods and major
bination treatment group. patterns in GLOBOCAN 2012, Int. J. Cancer 136 (2015) E359–E386.
The presence of hyper-malignant or CSCs in lung cancer and other [3] R.S. Herbst, J.V. Heymach, S.M. Lippman, Lung cancer, N. Engl. J. Med. 359
(2008) 1367–1380.
tumor types are largely responsible for the failure of current anti- [4] F.H. Sarkar, Y. Li, Z. Wang, D. Kong, S. Ali, Implication of microRNAs in drug
cancer treatments with metastasis and relapse accounting for a high resistance for designing novel cancer therapy, Drug Resist. Updat. 13 (2010)
percentage of patients. In a recent study by Li et al., the small mol- 57–66.
[5] K.E. Allen, G.J. Weiss, Resistance may not be futile: microRNA biomarkers for
ecule inhibitor BBI608 inhibited gene transcription driven by the chemoresistance and potential therapeutics, Mol. Cancer Ther. 9 (2010)
signaling transducer and activator of transcription 3 (STAT3) [139]. 3126–3136.
BBI608 abrogated cancer stem cell properties including tumorsphere [6] G. Szakacs, J.K. Paterson, J.A. Ludwig, C. Booth-Genthe, M.M. Gottesman,
Targeting multidrug resistance in cancer, Nat. Rev. Drug Discov. 5 (2006)
formation and self-renewal capabilities through the depletion of
219–234.
cancer stem cells. BBI608 treatment effectively blocks cancer relapse [7] L.V. Nguyen, R. Vanner, P. Dirks, C.J. Eaves, Cancer stem cells: an evolving
and metastasis in mice and holds great promise as a future anti- concept, Nat. Rev. Cancer 12 (2012) 133–143.
stemness therapy [139]. [8] T. Reya, S.J. Morrison, M.F. Clarke, I.L. Weissman, Stem cells, cancer, and cancer
stem cells, Nature 414 (2001) 105–111.
[9] S.J. Morrison, J. Kimble, Asymmetric and symmetric stem-cell divisions in
Conclusion development and cancer, Nature 441 (2006) 1068–1074.
[10] C. Cocola, S. Sanzone, S. Astigiano, P. Pelucchi, E. Piscitelli, L. Vilardo, et al., A
rat mammary gland cancer cell with stem cell properties of self-renewal and
Despite the development of targeted therapies, there has been multi-lineage differentiation, Cytotechnology 58 (2008) 25–32.
little improvement in the prognosis of lung cancer patients. The [11] M. Diehn, R.W. Cho, N.A. Lobo, T. Kalisky, M.J. Dorie, A.N. Kulp, et al., Association
of reactive oxygen species levels and radioresistance in cancer stem cells,
cancer stem cell hypothesis has provided a new avenue of focus in
Nature 458 (2009) 780–783.
overcoming resistance to therapy, preventing tumor relapse and im- [12] A.B. Hjelmeland, Q. Wu, J.M. Heddleston, G.S. Choudhary, J. MacSwords, J.D.
proving progression-free and overall survival rates in lung cancer Lathia, et al., Acidic stress promotes a glioma stem cell phenotype, Cell Death
Differ. 18 (2011) 829–840.
patients. Signaling pathways associated with stem cell properties,
[13] J. Tower, Stress and stem cells, Wiley Interdiscip. Rev. Dev. Biol. 1 (2012)
such as differentiation and self-renewal capacities in addition to the 789–802.
identification of CSC markers, offer potential targets for novel anti- [14] D. Serrano, A.M. Bleau, I. Fernandez-Garcia, T. Fernandez-Marcelo, P. Iniesta,
cancer strategies. While a number of pre-clinical and clinical studies C. Ortiz-de-Solorzano, et al., Inhibition of telomerase activity preferentially
targets aldehyde dehydrogenase-positive cancer stem-like cells in lung cancer,
(Table 2) examining CSC-targeted agents are still in their infancy, Mol. Cancer 10 (2011) 96.
[15] J. Liu, Z. Xiao, S.K. Wong, V.P. Tin, K.Y. Ho, J. Wang, et al., Lung cancer
tumorigenicity and drug resistance are maintained through ALDH(hi)CD44(hi)
tumor initiating cells, Oncotarget 4 (2013) 1698–1711.
Table 2 [16] G. Hamilton, U. Olszewski, Chemotherapy-induced enrichment of cancer stem
Clinical trials targeting CSCs. cells in lung cancer, J. Bioanal. Biomed. S9 (2013) 003, doi:10.4172/1948-
593X.S9-003.
Target Agent Phase Clinical Trials
[17] S.V. Sharma, D.Y. Lee, B. Li, M.P. Quinlan, F. Takahashi, S. Maheswaran, et al.,
KRAS Defactinib II NCT01951690 A chromatin-mediated reversible drug-tolerant state in cancer cell
CD117 Imetelstat II NCT01197968 subpopulations, Cell 141 (2010) 69–80.
DLL4 Demcizumab I/II NCT01189968 [18] M. Wintzell, L. Lofstedt, J. Johansson, A.B. Pedersen, J. Fuxe, M. Shoshan,
Repeated cisplatin treatment can lead to a multiresistant tumor cell population
NCT02259582
with stem cell features and sensitivity to 3-bromopyruvate, Cancer Biol. Ther.
STAT3 Methformin II NCT01717482
13 (2012) 1454–1462.
STAT3 BBI608 I/II NCT02347917
[19] V. Levina, A.M. Marrangoni, R. DeMarco, E. Gorelik, A.E. Lokshin, Drug-selected
NCT02432326 human lung cancer stem cells: cytokine network, tumorigenic and metastatic
NCT02467361 properties, PLoS ONE 3 (2008) e3077.
NCT01775423 [20] A.M. Calcagno, C.D. Salcido, J.P. Gillet, C.P. Wu, J.M. Fostel, M.D. Mumau, et al.,
Notch RO49290971 I/II NCT01193868 Prolonged drug selection of breast cancer cells and enrichment of cancer stem
NCT01193881 cell characteristics, J. Natl Cancer Inst. 102 (2010) 1637–1652.
CSC Immunotherapy Vaccine I/II NCT02115958 [21] Y.P. Liu, C.J. Yang, M.S. Huang, C.T. Yeh, A.T. Wu, Y.C. Lee, et al., Cisplatin selects
NCT02084823 for multidrug-resistant CD133+ cells in lung adenocarcinoma by activating
Sp1 Mithramycin A II NCT01624090 Notch signaling, Cancer Res. 73 (2013) 406–416.
ALDH1 Disulfiram II/III NCT00312819 [22] M.M. Shi, Y.L. Xiong, X.S. Jia, X. Li, L. Zhang, X.L. Li, et al., Fluorouracil selectively
enriches stem-like cells in the lung adenocarcinoma cell line SPC, Tumour Biol.
Therapeutics specifically targeting CSC-associated markers are currently being as- 34 (2013) 1503–1510.
sessed as individual therapies and in combination with conventional chemotherapies [23] J. Sleeman, P.S. Steeg, Cancer metastasis as a therapeutic target, Eur. J. Cancer
in lung cancer. 46 (2010) 1177–1180.
154 L. MacDonagh et al./Cancer Letters 372 (2016) 147–156

[24] S.M. Bailey, J.P. Murnane, Telomeres, chromosome instability and cancer, [52] A. Shenoy, E. Butterworth, E.H. Huang, ALDH as a marker for enriching
Nucleic Acids Res. 34 (2006) 2408–2417. tumorigenic human colonic stem cells, Methods Mol. Biol. 916 (2012) 373–
[25] A. Tomas-Loba, I. Flores, P.J. Fernandez-Marcos, M.L. Cayuela, A. Maraver, A. 385.
Tejera, et al., Telomerase reverse transcriptase delays aging in cancer-resistant [53] M.R. Clay, M. Tabor, J.H. Owen, T.E. Carey, C.R. Bradford, G.T. Wolf, et al.,
mice, Cell 135 (2008) 609–622. Single-marker identification of head and neck squamous cell carcinoma cancer
[26] M. Armanios, C.W. Greider, Telomerase and cancer stem cells, Cold Spring Harb. stem cells with aldehyde dehydrogenase, Head Neck 32 (2010) 1195–1201.
Symp. Quant. Biol. 70 (2005) 205–208. [54] C. van den Hoogen, G. van der Horst, H. Cheung, J.T. Buijs, J.M. Lippitt, N.
[27] R.J. Ward, C. Autexier, Pharmacological telomerase inhibition can sensitize Guzman-Ramirez, et al., High aldehyde dehydrogenase activity identifies
drug-resistant and drug-sensitive cells to chemotherapeutic treatment, Mol. tumor-initiating and metastasis-initiating cells in human prostate cancer,
Pharmacol. 68 (2005) 779–786. Cancer Res. 70 (2010) 5163–5173.
[28] M.A. Cerone, J.A. Londono-Vallejo, C. Autexier, Telomerase inhibition enhances [55] T. Li, Y. Su, Y. Mei, Q. Leng, B. Leng, Z. Liu, et al., ALDH1A1 is a marker for
the response to anticancer drug treatment in human breast cancer cells, Mol. malignant prostate stem cells and predictor of prostate cancer patients’
Cancer Ther. 5 (2006) 1669–1675. outcome, Lab. Invest. 90 (2010) 234–244.
[29] C. Bruedigam, F.O. Bagger, F.H. Heidel, C. Paine Kuhn, S. Guignes, A. Song, et al., [56] F. Jiang, Q. Qiu, A. Khanna, N.W. Todd, J. Deepak, L. Xing, et al., Aldehyde
Telomerase inhibition effectively targets mouse and human AML stem cells dehydrogenase 1 is a tumor stem cell-associated marker in lung cancer, Mol.
and delays relapse following chemotherapy, Cell Stem. Cell 15 (2014) 775–790. Cancer Res. 7 (2009) 330–338.
[30] J.E. Koziel, B.S. Herbert, The telomerase inhibitor imetelstat alone, and in [57] K. Okudela, T. Woo, H. Mitsui, T. Suzuki, M. Tajiri, Y. Sakuma, et al.,
combination with trastuzumab, decreases the cancer stem cell population and Downregulation of ALDH1A1 expression in non-small cell lung carcinomas–its
self-renewal of HER2(+) breast cancer cells, Breast Cancer Res. Treat. 149 (2015) clinicopathologic and biological significance, Int. J. Clin. Exp. Pathol. 6 (2013)
607–618. 1–12.
[31] M. Signore, L. Ricci-Vitiani, R. De Maria, Targeting apoptosis pathways in cancer [58] E. Charafe-Jauffret, C. Ginestier, F. Iovino, C. Tarpin, M. Diebel, B. Esterni, et al.,
stem cells, Cancer Lett. 332 (2013) 374–382. Aldehyde dehydrogenase 1-positive cancer stem cells mediate metastasis and
[32] K. Chen, Y.H. Huang, J.L. Chen, Understanding and targeting cancer stem cells: poor clinical outcome in inflammatory breast cancer, Clin. Cancer Res. 16
therapeutic implications and challenges, Acta Pharmacol. Sin. 34 (2013) (2010) 45–55.
732–740. [59] J.P. Sullivan, M. Spinola, M. Dodge, M.G. Raso, C. Behrens, B. Gao, et al., Aldehyde
[33] M.M. Ho, A.V. Ng, S. Lam, J.Y. Hung, Side population in human lung cancer dehydrogenase activity selects for lung adenocarcinoma stem cells dependent
cell lines and tumors is enriched with stem-like cancer cells, Cancer Res. 67 on notch signaling, Cancer Res. 70 (2010) 9937–9948.
(2007) 4827–4833. [60] J. Wang, Z.H. Li, J. White, L.B. Zhang, Lung cancer stem cells and implications
[34] M. Dean, T. Fojo, S. Bates, Tumour stem cells and drug resistance, Nat. Rev. for future therapeutics, Cell Biochem. Biophys. 69 (2014) 389–398.
Cancer 5 (2005) 275–284. [61] M.P. Barr, S.G. Gray, A.C. Hoffmann, R.A. Hilger, J. Thomale, J.D. O’Flaherty, et al.,
[35] R. Perona, B.D. Lopez-Ayllon, J. de Castro Carpeno, C. Belda-Iniesta, A role for Generation and characterisation of Cisplatin-resistant non-small cell lung
cancer stem cells in drug resistance and metastasis in non-small-cell lung cancer cell lines displaying a stem-like signature, PLoS ONE 8 (2013) e54193.
cancer, Clin. Transl. Oncol. 13 (2011) 289–293. [62] L. Larzabal, N. El-Nikhely, M. Redrado, W. Seeger, R. Savai, A. Calvo, Differential
[36] X. Zhou, D. Li, X. Wang, B. Zhang, H. Zhu, J. Zhao, Galectin-1 is overexpressed effects of drugs targeting cancer stem cell (CSC) and non-CSC populations on
in CD133+ human lung adenocarcinoma cells and promotes their growth and lung primary tumors and metastasis, PLoS ONE 8 (2013) e79798.
invasiveness, Oncotarget 6 (2015) 3111–3122. [63] J. Mihatsch, M. Toulany, P.M. Bareiss, S. Grimm, C. Lengerke, R. Kehlbach, et al.,
[37] A. Singh, J. Settleman, EMT, cancer stem cells and drug resistance: an emerging Selection of radioresistant tumor cells and presence of ALDH1 activity in vitro,
axis of evil in the war on cancer, Oncogene 29 (2010) 4741–4751. Radiother. Oncol. 99 (2011) 300–306.
[38] J.J. Wamsley, M. Kumar, D.F. Allison, S.H. Clift, C.M. Holzknecht, S.J. Szymura, [64] K. Shien, S. Toyooka, H. Yamamoto, J. Soh, M. Jida, K.L. Thu, et al., Acquired
et al., Activin upregulation by NF-kappaB is required to maintain mesenchymal resistance to EGFR inhibitors is associated with a manifestation of stem cell-like
features of cancer stem-like cells in non-small cell lung cancer, Cancer Res. properties in cancer cells, Cancer Res. 73 (2013) 3051–3061.
75 (2015) 426–435. [65] R.R. Arasada, J.M. Amann, M.A. Rahman, S.S. Huppert, D.P. Carbone, EGFR
[39] I. Malanchi, H. Peinado, D. Kassen, T. Hussenet, D. Metzger, P. Chambon, et al., blockade enriches for lung cancer stem-like cells through Notch3-dependent
Cutaneous cancer stem cell maintenance is dependent on beta-catenin signaling, Cancer Res. 74 (2014) 5572–5584.
signalling, Nature 452 (2008) 650–653. [66] C.P. Huang, M.F. Tsai, T.H. Chang, W.C. Tang, S.Y. Chen, H.H. Lai, et al., ALDH-
[40] I.H. Oh, Microenvironmental targeting of Wnt/beta-catenin signals for positive lung cancer stem cells confer resistance to epidermal growth factor
hematopoietic stem cell regulation, Expert Opin. Biol. Ther. 10 (2010) receptor tyrosine kinase inhibitors, Cancer Lett. 328 (2013) 144–151.
1315–1329. [67] B. Corominas-Faja, C. Oliveras-Ferraros, E. Cuyas, A. Segura-Carretero, J. Joven,
[41] H. Wang, G. Zhang, H. Zhang, F. Zhang, B. Zhou, F. Ning, et al., Acquisition of B. Martin-Castillo, et al., Stem cell-like ALDH(bright) cellular states in
epithelial-mesenchymal transition phenotype and cancer stem cell-like EGFR-mutant non-small cell lung cancer: a novel mechanism of acquired
properties in cisplatin-resistant lung cancer cells through AKT/beta-catenin/ resistance to erlotinib targetable with the natural polyphenol silibinin, Cell
Snail signaling pathway, Eur. J. Pharmacol. 723 (2014) 156–166. Cycle 12 (2013) 3390–3404.
[42] Y. Teng, X. Wang, Y. Wang, D. Ma, Wnt/beta-catenin signaling regulates cancer [68] L. Cortes-Dericks, L. Froment, R. Boesch, R.A. Schmid, G. Karoubi, Cisplatin-
stem cells in lung cancer A549 cells, Biochem. Biophys. Res. Commun. 392 resistant cells in malignant pleural mesothelioma cell lines show
(2010) 373–379. ALDH(high)CD44(+) phenotype and sphere-forming capacity, BMC Cancer 14
[43] A.W. Duncan, F.M. Rattis, L.N. DiMascio, K.L. Congdon, G. Pazianos, C. Zhao, (2014) 304.
et al., Integration of Notch and Wnt signaling in hematopoietic stem cell [69] J.S. Moreb, H.V. Baker, L.J. Chang, M. Amaya, M.C. Lopez, B. Ostmark, et al., ALDH
maintenance, Nat. Immunol. 6 (2005) 314–322. isozymes downregulation affects cell growth, cell motility and gene expression
[44] K.A. Hassan, L. Wang, H. Korkaya, G. Chen, I. Maillard, D.G. Beer, et al., Notch in lung cancer cells, Mol. Cancer 7 (2008) 87.
pathway activity identifies cells with cancer stem cell-like properties and [70] A. O’Brien, J.E. Barber, S. Reid, N. Niknejad, J. Dimitroulakos, Enhancement of
correlates with worse survival in lung adenocarcinoma, Clin. Cancer Res. 19 cisplatin cytotoxicity by disulfiram involves activating transcription factor 3,
(2013) 1972–1980. Anticancer Res. 32 (2012) 2679–2688.
[45] V. Justilien, A.P. Fields, Molecular pathways: novel approaches for improved [71] Y. Wang, W. Li, S.S. Patel, J. Cong, N. Zhang, F. Sabbatino, et al., Blocking the
therapeutic targeting of Hedgehog signaling in cancer stem cells, Clin. Cancer formation of radiation-induced breast cancer stem cells, Oncotarget 5 (2014)
Res. 21 (2015) 505–513. 3743–3755.
[46] M. Zuo, A. Rashid, C. Churi, J.N. Vauthey, P. Chang, Y. Li, et al., Novel therapeutic [72] J.L. Allensworth, M.K. Evans, F. Bertucci, A.J. Aldrich, R.A. Festa, P. Finetti, et al.,
strategy targeting the Hedgehog signalling and mTOR pathways in biliary tract Disulfiram (DSF) acts as a copper ionophore to induce copper-dependent
cancer, Br. J. Cancer 112 (2015) 1042–1051. oxidative stress and mediate anti-tumor efficacy in inflammatory breast cancer,
[47] A. Ahmad, M.Y. Maitah, K.R. Ginnebaugh, Y. Li, B. Bao, S.M. Gadgeel, et al., Mol. Oncol. 9 (2015) 1155–1168.
Inhibition of Hedgehog signaling sensitizes NSCLC cells to standard therapies [73] H. Nechushtan, Y. Hamamreh, S. Nidal, M. Gotfried, A. Baron, Y.I. Shalev, et al.,
through modulation of EMT-regulating miRNAs, J. Hematol. Oncol. 6 (2013) A phase IIb trial assessing the addition of disulfiram to chemotherapy for the
77. treatment of metastatic non-small cell lung cancer, Oncologist 20 (2015)
[48] M. Kumar, D.F. Allison, N.N. Baranova, J.J. Wamsley, A.J. Katz, S. Bekiranov, et al., 366–367.
NF-kappaB regulates mesenchymal transition for the induction of non-small [74] Y. Shi, X. Fu, Y. Hua, Y. Han, Y. Lu, J. Wang, The side population in human lung
cell lung cancer initiating cells, PLoS ONE 8 (2013) e68597. cancer cell line NCI-H460 is enriched in stem-like cancer cells, PLoS ONE 7
[49] M.B. Kastan, E. Schlaffer, J.E. Russo, O.M. Colvin, C.I. Civin, J. Hilton, Direct (2012) e33358.
demonstration of elevated aldehyde dehydrogenase in human hematopoietic [75] S. Singh, J. Trevino, N. Bora-Singhal, D. Coppola, E. Haura, S. Altiok, et al.,
progenitor cells, Blood 75 (1990) 1947–1950. EGFR/Src/Akt signaling modulates Sox2 expression and self-renewal of
[50] L.C. Hsu, W.C. Chang, I. Hoffmann, G. Duester, Molecular analysis of two closely stem-like side-population cells in non-small cell lung cancer, Mol. Cancer 11
related mouse aldehyde dehydrogenase genes: identification of a role for Aldh1, (2012) 73.
but not Aldh-pb, in the biosynthesis of retinoic acid, Biochem. J. 339 (Pt 2) [76] R. Xiang, D. Liao, T. Cheng, H. Zhou, Q. Shi, T.S. Chuang, et al., Downregulation
(1999) 387–395. of transcription factor SOX2 in cancer stem cells suppresses growth and
[51] C. Ginestier, M.H. Hur, E. Charafe-Jauffret, F. Monville, J. Dutcher, M. Brown, metastasis of lung cancer, Br. J. Cancer 104 (2011) 1410–1417.
et al., ALDH1 is a marker of normal and malignant human mammary stem [77] S. Singh, N. Bora-Singhal, J. Kroeger, H. Laklai, S.P. Chellappan, betaArrestin-1
cells and a predictor of poor clinical outcome, Cell Stem. Cell 1 (2007) 555– and Mcl-1 modulate self-renewal growth of cancer stem-like side-population
567. cells in non-small cell lung cancer, PLoS ONE 8 (2013) e55982.
L. MacDonagh et al./Cancer Letters 372 (2016) 147–156 155

[78] F. Tian, J. Mysliwietz, J. Ellwart, F. Gamarra, R.M. Huber, A. Bergner, Effects of [104] M.A. Goodell, K. Brose, G. Paradis, A.S. Conner, R.C. Mulligan, Isolation and
the Hedgehog pathway inhibitor GDC-0449 on lung cancer cell lines are functional properties of murine hematopoietic stem cells that are replicating
mediated by side populations, Clin. Exp. Med. 12 (2012) 25–30. in vivo, J. Exp. Med. 183 (1996) 1797–1806.
[79] M. Zhang, A. Mathur, Y. Zhang, S. Xi, S. Atay, J.A. Hong, et al., Mithramycin [105] K. Kai, S. D’Costa, B.I. Yoon, A.R. Brody, R.C. Sills, Y. Kim, Characterization of
represses basal and cigarette smoke-induced expression of ABCG2 and inhibits side population cells in human malignant mesothelioma cell lines, Lung Cancer
stem cell signaling in lung and esophageal cancer cells, Cancer Res. 72 (2012) 70 (2010) 146–151.
4178–4192. [106] M. Christgen, M. Ballmaier, U. Lehmann, H. Kreipe, Detection of putative cancer
[80] Y. Wu, R. Si, H. Tang, Z. He, H. Zhu, L. Wang, et al., Cholesterol reduces the stem cells of the side population phenotype in human tumor cell cultures,
sensitivity to platinum-based chemotherapy via upregulating ABCG2 in lung Methods Mol. Biol. 878 (2012) 201–215.
adenocarcinoma, Biochem. Biophys. Res. Commun. 457 (2015) 614–620. [107] B. Sarkadi, C. Ozvegy-Laczka, K. Nemet, A. Varadi, ABCG2—a transporter for
[81] Y.P. Hu, L.Y. Tao, F. Wang, J.Y. Zhang, Y.J. Liang, L.W. Fu, Secalonic acid D reduced all seasons, FEBS Lett. 567 (2004) 116–120.
the percentage of side populations by down-regulating the expression of [108] S. Zhang, Y. Wang, J.H. Mao, D. Hsieh, I.J. Kim, L.M. Hu, et al., Inhibition of
ABCG2, Biochem. Pharmacol. 85 (2013) 1619–1625. CK2alpha down-regulates Hedgehog/Gli signaling leading to a reduction of
[82] Q. Niu, W. Wang, Y. Li, D.M. Ruden, F. Wang, J. Song, et al., Low molecular a stem-like side population in human lung cancer cells, PLoS ONE 7 (2012)
weight heparin ablates lung cancer cisplatin-resistance by inducing e38996.
proteasome-mediated ABCG2 protein degradation, PLoS ONE 7 (2012) e41035. [109] Y. An, A. Kiang, J.P. Lopez, S.Z. Kuo, M.A. Yu, E.L. Abhold, et al., Cigarette smoke
[83] F. Wang, Y.J. Mi, X.G. Chen, X.P. Wu, Z. Liu, S.P. Chen, et al., Axitinib targeted promotes drug resistance and expansion of cancer stem cell-like side
cancer stemlike cells to enhance efficacy of chemotherapeutic drugs via population, PLoS ONE 7 (2012) e47919.
inhibiting the drug transport function of ABCG2, Mol. Med. 18 (2012) 887– [110] B. Fischer, C. Frei, U. Moura, R. Stahel, E. Felley-Bosco, Inhibition of
898. phosphoinositide-3 kinase pathway down regulates ABCG2 function and
[84] V. Levina, A. Marrangoni, T. Wang, S. Parikh, Y. Su, R. Herberman, et al., sensitizes malignant pleural mesothelioma to chemotherapy, Lung Cancer 78
Elimination of human lung cancer stem cells through targeting of the stem (2012) 23–29.
cell factor-c-kit autocrine signaling loop, Cancer Res. 70 (2010) 338–346. [111] W.J. Yang, M.J. Song, E.Y. Park, J.J. Lee, J.H. Park, K. Park, et al., Transcription
[85] P.A. Horn, H. Tesch, P. Staib, D. Kube, V. Diehl, D. Voliotis, Expression of AC133, factors Sp1 and Sp3 regulate expression of human ABCG2 gene and
a novel hematopoietic precursor antigen, on acute myeloid leukemia cells, chemoresistance phenotype, Mol. Cells 36 (2013) 368–375.
Blood 93 (1999) 1435–1437. [112] M.B. Penno, J.T. August, S.B. Baylin, M. Mabry, R.I. Linnoila, V.S. Lee, et al.,
[86] N. Sanai, A. Alvarez-Buylla, M.S. Berger, Neural stem cells and the origin of Expression of CD44 in human lung tumors, Cancer Res. 54 (1994) 1381–
gliomas, N. Engl. J. Med. 353 (2005) 811–822. 1387.
[87] A. Pircher, C.M. Kahler, S. Skvortsov, M. Dlaska, G. Kawaguchi, T. Schmid, et al., [113] T.M. Phillips, W.H. McBride, F. Pajonk, The response of CD24(-/low)/CD44+
Increased numbers of endothelial progenitor cells in peripheral blood and breast cancer-initiating cells to radiation, J. Natl Cancer Inst. 98 (2006)
tumor specimens in non-small cell lung cancer: a methodological challenge 1777–1785.
and an ongoing debate on the clinical relevance, Oncol. Rep. 19 (2008) [114] E.M. Hurt, B.T. Kawasaki, G.J. Klarmann, S.B. Thomas, W.L. Farrar, CD44+ CD24(-)
345–352. prostate cells are early cancer progenitor/stem cells that provide a model for
[88] M.H. Wright, A.M. Calcagno, C.D. Salcido, M.D. Carlson, S.V. Ambudkar, L. patients with poor prognosis, Br. J. Cancer 98 (2008) 756–765.
Varticovski, Brca1 breast tumors contain distinct CD44+/CD24- and CD133+ [115] C. Li, D.G. Heidt, P. Dalerba, C.F. Burant, L. Zhang, V. Adsay, et al., Identification
cells with cancer stem cell characteristics, Breast Cancer Res. 10 (2008) R10. of pancreatic cancer stem cells, Cancer Res. 67 (2007) 1030–1037.
[89] C.A. O’Brien, A. Pollett, S. Gallinger, J.E. Dick, A human colon cancer cell capable [116] V. Orian-Rousseau, CD44, a therapeutic target for metastasising tumours, Eur.
of initiating tumour growth in immunodeficient mice, Nature 445 (2007) J. Cancer 46 (2010) 1271–1277.
106–110. [117] E.L. Leung, R.R. Fiscus, J.W. Tung, V.P. Tin, L.C. Cheng, A.D. Sihoe, et al., Non-small
[90] J. Miki, B. Furusato, H. Li, Y. Gu, H. Takahashi, S. Egawa, et al., Identification cell lung cancer cells expressing CD44 are enriched for stem cell-like properties,
of putative stem cell markers, CD133 and CXCR4, in hTERT-immortalized PLoS ONE 5 (2010) e14062.
primary nonmalignant and malignant tumor-derived human prostate epithelial [118] R. Gomez-Casal, C. Bhattacharya, N. Ganesh, L. Bailey, P. Basse, M. Gibson, et al.,
cell lines and in prostate cancer specimens, Cancer Res. 67 (2007) 3153–3161. Non-small cell lung cancer cells survived ionizing radiation treatment display
[91] S. Yin, J. Li, C. Hu, X. Chen, M. Yao, M. Yan, et al., CD133 positive hepatocellular cancer stem cell and epithelial-mesenchymal transition phenotypes, Mol.
carcinoma cells possess high capacity for tumorigenicity, Int. J. Cancer 120 Cancer 12 (2013) 94.
(2007) 1444–1450. [119] Y.H. Ko, H.S. Won, E.K. Jeon, S.H. Hong, S.Y. Roh, Y.S. Hong, et al., Prognostic
[92] M.D. Curley, V.A. Therrien, C.L. Cummings, P.A. Sergent, C.R. Koulouris, A.M. significance of CD44s expression in resected non-small cell lung cancer, BMC
Friel, et al., CD133 expression defines a tumor initiating cell population in Cancer 11 (2011) 340.
primary human ovarian cancer, Stem Cells 27 (2009) 2875–2883. [120] M. Gutova, J. Najbauer, A. Gevorgyan, M.Z. Metz, Y. Weng, C.C. Shih, et al.,
[93] A. Eramo, F. Lotti, G. Sette, E. Pilozzi, M. Biffoni, A. Di Virgilio, et al., Identification Identification of uPAR-positive chemoresistant cells in small cell lung cancer,
and expansion of the tumorigenic lung cancer stem cell population, Cell Death PLoS ONE 2 (2007) e243.
Differ. 15 (2008) 504–514. [121] C.E. Almasi, L. Drivsholm, H. Pappot, G. Hoyer-Hansen, I.J. Christensen, The
[94] V. Tirino, R. Camerlingo, R. Franco, D. Malanga, A. La Rocca, G. Viglietto, et al., liberated domain I of urokinase plasminogen activator receptor—a new tumour
The role of CD133 in the identification and characterisation of tumour- marker in small cell lung cancer, APMIS 121 (2013) 189–196.
initiating cells in non-small-cell lung cancer, Eur. J. Cardiothorac Surg. 36 (2009) [122] J.S. Rao, C. Gondi, C. Chetty, S. Chittivelu, P.A. Joseph, S.S. Lakka, Inhibition of
446–453. invasion, angiogenesis, tumor growth, and metastasis by adenovirus-mediated
[95] Y.C. Chen, H.S. Hsu, Y.W. Chen, T.H. Tsai, C.K. How, C.Y. Wang, et al., Oct-4 transfer of antisense uPAR and MMP-9 in non-small cell lung cancer cells, Mol.
expression maintained cancer stem-like properties in lung cancer-derived Cancer Ther. 4 (2005) 1399–1408.
CD133-positive cells, PLoS ONE 3 (2008) e2637. [123] D. Alfano, I. Iaccarino, M.P. Stoppelli, Urokinase signaling through its receptor
[96] G. Bertolini, L. Roz, P. Perego, M. Tortoreto, E. Fontanella, L. Gatti, et al., Highly protects against anoikis by increasing BCL-xL expression levels, J. Biol. Chem.
tumorigenic lung cancer CD133+ cells display stem-like features and are spared 281 (2006) 17758–17767.
by cisplatin treatment, Proc. Natl. Acad. Sci. U.S.A. 106 (2009) 16281–16286. [124] T. Kubo, N. Takigawa, M. Osawa, D. Harada, T. Ninomiya, N. Ochi, et al.,
[97] A. Murakami, F. Takahashi, F. Nurwidya, I. Kobayashi, K. Minakata, M. Subpopulation of small-cell lung cancer cells expressing CD133 and CD87 show
Hashimoto, et al., Hypoxia increases gefitinib-resistant lung cancer stem cells resistance to chemotherapy, Cancer Sci. 104 (2013) 78–84.
through the activation of insulin-like growth factor 1 receptor, PLoS ONE 9 [125] X. Qiu, Z. Wang, Y. Li, Y. Miao, Y. Ren, Y. Luan, Characterization of sphere-
(2014) e86459. forming cells with stem-like properties from the small cell lung cancer cell
[98] L. Lundholm, P. Haag, D. Zong, T. Juntti, B. Mork, R. Lewensohn, et al., Resistance line H446, Cancer Lett. 323 (2012) 161–170.
to DNA-damaging treatment in non-small cell lung cancer tumor-initiating [126] M. Miettinen, J. Lasota, KIT (CD117): a review on expression in normal and
cells involves reduced DNA-PK/ATM activation and diminished cell cycle arrest, neoplastic tissues, and mutations and their clinicopathologic correlation, Appl.
Cell Death Dis. 4 (2013) e478. Immunohistochem. Mol. Morphol. 13 (2005) 205–220.
[99] S. Sarvi, A.C. Mackinnon, N. Avlonitis, M. Bradley, R.C. Rintoul, D.M. Rassl, et al., [127] H. Joensuu, Treatment of inoperable gastrointestinal stromal tumor (GIST) with
CD133+ cancer stem-like cells in small cell lung cancer are highly tumorigenic Imatinib (Glivec, Gleevec), Med. Klin. (Munich) 97 (Suppl. 1) (2002) 28–30.
and chemoresistant but sensitive to a novel neuropeptide antagonist, Cancer [128] J. Liu, X. Liu, X. Feng, S. Lv, W. Zhang, Y. Niu, C-kit overexpression correlates
Res. 74 (2014) 1554–1565. with KIT gene copy numbers increases in phyllodes tumors of the breast, Breast
[100] H. Qu, R. Li, Z. Liu, J. Zhang, R. Luo, Prognostic value of cancer stem cell marker Cancer Res. Treat. 149 (2015) 395–401.
CD133 expression in non-small cell lung cancer: a systematic review, Int. J. [129] S. Ferrari, A. Grande, P. Zucchini, R. Manfredini, E. Tagliafico, E. Rossi, et al.,
Clin. Exp. Pathol. 6 (2013) 2644–2650. Overexpression of c-kit in a leukemic cell population carrying a trisomy 4 and
[101] W. Wang, Y. Chen, J. Deng, J. Zhou, Y. Zhou, S. Wang, The prognostic value of its relationship with the proliferative capacity, Leuk. Lymphoma 9 (1993)
CD133 expression in non-small cell lung cancer: a meta-analysis, Tumour Biol. 495–501.
35 (2014) 9769–9775. [130] A.D. Donnenberg, L. Zimmerlin, R.J. Landreneau, J.D. Luketich, V.S. Donnenberg,
[102] L.E. Pascal, A.J. Oudes, T.W. Petersen, Y.A. Goo, L.S. Walashek, L.D. True, et al., KIT (CD117) expression in a subset of non-small cell lung carcinoma (NSCLC)
Molecular and cellular characterization of ABCG2 in the prostate, BMC Urol. patients, PLoS ONE 7 (2012) e52885.
7 (2007) 6. [131] D. Perumal, S. Pillai, J. Nguyen, C. Schaal, D. Coppola, S.P. Chellappan, Nicotinic
[103] K.M. Britton, J.A. Kirby, T.W. Lennard, A.P. Meeson, Cancer stem cells and side acetylcholine receptors induce c-Kit ligand/Stem Cell Factor and promote
population cells in breast cancer and metastasis, Cancers (Basel) 3 (2011) stemness in an ARRB1/ beta-arrestin-1 dependent manner in NSCLC,
2106–2130. Oncotarget 5 (2014) 10486–10502.
156 L. MacDonagh et al./Cancer Letters 372 (2016) 147–156

[132] R. Januchowski, K. Wojtowicz, M. Zabel, The role of aldehyde dehydrogenase [136] C. Thoma, Prostate cancer: copper unlocks therapeutic potential of disulfiram,
(ALDH) in cancer drug resistance, Biomed. Pharmacother. 67 (2013) 669–680. Nat. Rev. Urol. 11 (2014) 664.
[133] R.G. Bell, H.W. Smith, Preliminary report on clinical trials of antabuse, Can. [137] B.W. Morrison, N.A. Doudican, K.R. Patel, S.J. Orlow, Disulfiram induces
Med. Assoc. J. 60 (1949) 286–288. copper-dependent stimulation of reactive oxygen species and activation of
[134] D. Chen, Q.C. Cui, H. Yang, Q.P. Dou, Disulfiram, a clinically used anti-alcoholism the extrinsic apoptotic pathway in melanoma, Melanoma Res. 20 (2010) 11–20.
drug and copper-binding agent, induces apoptotic cell death in breast cancer [138] L. Duan, H. Shen, G. Zhao, R. Yang, X. Cai, L. Zhang, et al., Inhibitory effect of
cultures and xenografts via inhibition of the proteasome activity, Cancer Res. Disulfiram/copper complex on non-small cell lung cancer cells, Biochem.
66 (2006) 10425–10433. Biophys. Res. Commun. 446 (2014) 1010–1016.
[135] P. Boyd, I. Major, W. Wang, C. McConville, Development of disulfiram-loaded [139] Y. Li, H.A. Rogoff, S. Keates, Y. Gao, S. Murikipudi, K. Mikule, et al., Suppression
vaginal rings for the localised treatment of cervical cancer, Eur. J. Pharm. of cancer relapse and metastasis by inhibiting cancer stemness, Proc. Natl.
Biopharm. 88 (2014) 945–953. Acad. Sci. U.S.A. 112 (2015) 1839–1844.

You might also like