Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Apr 18.
Published in final edited form as: Respirology. 2013 Jul;18(5):757–764. doi: 10.1111/resp.12094

Cancer stem cells in lung cancer: Evidence and controversies

Muhammad ALAMGEER 1,2, Craig D PEACOCK 3, William MATSUI 3, Vinod GANJU 1,2, D Neil WATKINS 2
PMCID: PMC3991120  NIHMSID: NIHMS561110  PMID: 23586700

Abstract

The cancer stem cell (CSC) model is based on a myriad of experimental and clinical observations suggesting that the malignant phenotype is sustained by a subset of cells characterized by the capacity for self-renewal, differentiation and innate resistance to chemotherapy and radiation. CSC may be responsible for disease recurrence after definitive therapy and may therefore be functionally synonymous with minimal residual disease. Similar to other solid tumours, several putative surface markers for lung CSC have been identified, including CD133 and CD44. In addition, expression and/or activity of the cytoplasmic enzyme aldehyde dehydrogenase ALDH and capacity of cells to exclude membrane permeable dyes (known as the ‘side population’) correlate with stem-like function in vitro and in vivo. Embryonic stem cell pathways such as Hedgehog, Notch and WNT may also be active in lung cancers stem cells and therefore may be therapeutically targetable for maintenance therapy in patients achieving a complete response to surgery, radiotherapy or chemo-therapy. This paper will review the evidence regarding the existence and function of lung CSC in the context of the experimental and clinical evidence and discuss some ongoing controversies regarding this model.

Keywords: aldehyde dehydrogenase, cancer stem cell, lung cancer, side population

INTRODUCTION

Lung cancer is the most common cause of cancer-related deaths worldwide.1 It is a heterogeneous disease with two distinct pathological classes: non-small cell lung cancer (NSCLC, which makes up 80% of cases) and small cell lung cancer (SCLC, making up 20% of all lung cancers). The most common forms of NSCLC are adenocarcinoma (30–50% of NSCLC) and squamous cell carcinoma (30% of NSCLC). Future treatment is geared towards adapting management specific to the tumour gene profile. However, recent next generation sequencing studies show a marked degree of genomic heterogeneity within each histo-logical subtype, thus presenting ever greater challenges for personalized medicine based on targeted therapies.2

Although approximately 20% of NSCLC are operable at presentation, recurrence rates remain high at 30–50%.3 Locally advanced NSCLC can be treated with radical chemo-radiotherapy with intent to cure; however, overall 5-year survival rates remain low at around 7–20%.4 For most cases of advanced NSCLC, systemic chemotherapy response rates are below 20–50%, and the 5-year mortality is in the order of 95%.5 In a subset of patients with advanced adenocarcinoma, dramatic responses to tyrosine kinase inhibitors can be seen in tumours with activating mutations in epithelial growth factor receptor or analplastic lymphoma kinase. However, almost all patients succumb to drug-resistant recurrences within 2–3 years.6,7 Although similar targeted agents have not been identified for use in SCLC, most cases are highly responsive to platinum-based chemotherapy, with or without radiotherapy in disease limited to the chest.8 Once again, despite complete responses in some, drug-resistant relapses result in a 5-year survival of only 5%.9

The remarkable ability of lung cancer to recur despite definitive local and/or systemic therapy suggests that minimal residual disease contains a population with enormous capacity for self-renewal and regeneration, a biological function generally limited to normal somatic stem cells. Termed ‘cancer stem cells’ (CSC), these cells are believed to be a phenotypically distinct population that possesses tumourigenic potential. Initially reported in haematological malignancies, and subsequently in several solid tumours, CSC represent potential therapeutic targets, and several strategies are already in clinical development. If the CSC hypothesis is correct, then their therapeutic targeting has the potential to delay or prevent disease recurrence. For the purposes of this review, we will not extend the definition of CSC to the question of cancer cell-of-origin, but rather focus on the question of stem-like cells in established lung cancers.

CANCER STEM CELLS

All complex multicellular organisms develop and regenerate from a somatic stem cell population that gives rise to a hierarchy of committed progenitors, leading ultimately to terminally differentiated cells in mature organs. The CSC hypothesis is based on the simple concept that cancers, like all other organs, contain a similar hierarchy with respect to self-renewal, differentiation and innate drug resistance.10 Thus, CSC give rise to highly proliferative progenitor cells and differentiated cells comprising the bulk of tumours and ultimately define the histological type of the cancer. Importantly, this hypothesis also predicts that, although these proliferating cells are responsive to treatment, their partially differentiated state prevents them from renewing the entire tumour from a minimal residual population. Numerous reports in haematological malignancies,11 breast cancer,12 brain tumours13 and colorectal cancer14 support this notion, and show that in each of these models, only a small, phenotypically distinct subpopulation of cells could recapitulate the tumour phenotype in vitro.

Isolation of CSC

The capacity of CSC to undergo self-renewal and differentiation has been demonstrated experimentally in various assays. Purified cancer cells isolated from freshly dissected tissues are able to grow as spheres in non-adherent cultures, form colonies more efficiently than their differentiated progeny and can be continually repassaged at clonal density.15 However, the current gold standard assay for CSC tumourigenic potential is their ability to grow in vivo as serially transplantable tumours in immunodeficient hosts. This strategy has successfully identified CSC in several tumour types, such as brain, breast, haemato-logical malignancies12,13,16 as well as in lung cancer (Table 1).

Table 1.

Experimental studies performed to study putative cancer stem cell markers in lung cancer

Study Sample Marker to isolate stem cells Main results
Ho et al.11 Cell lines SP SP cells showed high invasiveness and resistance to chemotherapy and high telomerase activity. In vivo high tumourigenicity compared with non-SP cells
Levina et al.18 Cell lines SP Enrichment of cells with SP and CD133 after chemotherapy treatment.
Drug surviving cells were enriched in self-renewal, differentiation and tumourigenic capacity.
Chen et al.19 Cell lines/tumour specimens CD133 CD133-positive cells formed spheres, expressed ABCG2 and Oct-4
CD133+ cells possessed self renewal capacity and were resistant to chemotherapy and radiotherapy
Eramo et al.20 Tumour specimens CD133 CD133-positive cells showed sphere formation, differentiation and chemoresistance
CD133-positive cells expressed Oct-4, NANOG, EPCAM and NCAM
Bertolini et al.21 Cell lines/tumour specimens CD133 CD133-positive cells expressed Oct-4, NANOG, α-6 integrin and CXCR4
CD133-positive cells were resistance to cisplatin and in patient's samples predicted poor response to treatment.
Jiang et al.22 Lung cancer/cell lines ALDH ALDH-positive cells had increased growth and formed large colonies in culture.
ALDH-positive cells showed differentiation and invasion potential.
ALDH-positive patients had poor survival 60% patient samples were also positive for CD133
Shi et al.23 Cell lines SP SP cell formed spheres and expressed ABCG2 and Hedgehog pathway transcript (SMO).
Cyclopamine inhibited SP cell proliferation

ABCG2, ATP-binding cassette G2; ALDH, aldehyde dehydrogenase; SMO, Smoothened; SP, side population.

However, this model has been criticized at both technical and theoretical levels. For example, the use of profoundly immunodeficient non-obese diabetic/severe combined immune deficiency/interleukin-2 knockout mice as a host for human melanoma xenografts shows that more than one in four cells can clonally generate tumours, regardless of surface marker expression.24 However, when Matsui and colleagues repeated these experiments in other solid tumour models, including lung cancer, the distinct cellular phenotype of tumour-initiating cells was preserved.25 These studies suggest that in some highly aggressive tumours, such as melanoma, the stem cell phenotype is a defining feature of the tumour, whereas more differentiated tumours maintain a stem cell hierarchy despite the level of immunodeficiency in xenograft assays.

Phenotypic characterization of CSC

Marker expression is a key element to categorize cancer cells into tumourigenic and nontumourigenic. Cells sorted for expression of CSC markers are passaged through various assays to determine tumourigenicity. One such method is the detection of side-population (SP) phenotypes. SP phenotyping is a functional assay based on the differential ability of the cancer cells to efflux Hoechst 33342 dye, as imparted by the ATP-binding cassette family of transporter proteins present on the cellular membrane.26 The SP assay has the advantage of measuring a functional parameter of the cells. However, the process is difficult to perform on tumour cells from intact clinical samples. Cancer cells as well as many stromal cells may possess dye exclusion properties, which could make the interpretation of results difficult. Moreover, the Hoechst dye is toxic to the cells.27

Aldehyde dehydrogenase (ALDH) activity is an important functional marker of normal and malignant stem/progenitor cells. Through oxidation of retinol to retinoic acid, ALDH is involved in early stem cell development.28 ALDH activity forms the basis of a fluorescence-activated cell sorter-based assay. Initially used to sort haematopoietic stem cells,29 a BIODIPY-aminoacetaldehyde substrate is oxidized intracellularly by ALDH, causing cells to become highly fluorescent. ALDH contributes to drug resistance through detoxification of many cytotoxic agents30 and has been reported as a reliable CSC marker in several tumour types.31,32

Another commonly used strategy is the isolation of CSC by flow cytometry according to the expression of certain surface markers. Commonly used markers include CD133 and CD44. CD133 (prominin-1 or AC133) was originally described in human haematopoietic stem cells33 and has subsequently been used to isolate CSC in many tumour types.14,34,35 CD44 is another transmembrane glycoprotein believed to be activated in a wide range of tumours where it plays a critical role in a variety of cancer cell behaviours including adhesion, migration, invasion and survival.36

LUNG CSC MARKERS

Almost 30 years ago, Carney and colleagues described a rare (<1.5%) population of cells in SCLC and adeno-carcinoma patient samples, with the ability to form colonies in soft agar.37 When inoculated into athymic nude mice, these colonies were able to recapitulate the original SCLC and adenocarcinoma tumours, suggesting ‘stem cell’ characteristic.37 Over the last decade, several investigators have isolated tumourigenic cells from lung cancer using different phenotypic cancer cell characteristics (Table 1).

Ho et al. initially showed that SP cells in NSCLC cancer cell lines were resistant to multiple chemotherapy agents and overexpressed several ATP-binding cassette transporters.17 SP cells also expressed elevated levels of telomerase and showed decreased expression of proliferation marker mini-chromosome maintenance 7, suggesting a quiescent nature.17 SP cells were rich in self-renewal and differentiation potential and were highly tumourigenic in non-obese diabetic/severe combined immune deficiency mice. Subsequently, different researchers investigated several membrane-bound surface markers to identify CSC in lung cancer. Of all the markers explored, CD44 and CD133 gained the most attention.20,38,39 In cell lines from both SCLC and NSCLC, CD133-positive cells generated long-term tumour spheres and differentiated into CD133-negative cells.20In vivo, as few as 1 × 104 CD133-positive cells generated tumours in non-obese diabetic/severe combined immune deficiency mice. CD133-positive cells were also resistant to chemo-therapy and expressed high levels of ATP-binding cassette G2, suggesting an SP phenotype. CD133-positive cells also expressed embryonic stem cell markers (Oct-4 and NANOG),20 and knockdown of Oct-4 by small interfering RNA resulted in reduced clonogenicity and enhanced chemosensitivity of CD133-positive cells.19 Further, treatment of a NSCLC cell line with cisplatin and doxorubicin resulted in enrichment for cells with enhanced tumourigenic potential in severe combined immune deficiency mice. These cells were also enriched for CD133 expression as well as SP phenotype.18 CD133-positive cells also overexpressed adhesion molecules (very late activation antigen-5, intercellular adhesion molecule-1, vascular cell adhesion molecule-1, matrix metalloprotease-2 and matrix metalloprotease-3), suggesting enhanced metastatic potential.19 CD44 expression on the other hand enriched for CSC-like properties in NSCLC cell lines38 but not in SCLC cell lines.39 However, in SCLC cell lines, urokinase-type plasminogen activator positive (CD87) cells were shown to possess multidrug resistance and enhanced clonogenic activity in vitro.40 Another recent study in SCLC cell lines showed that urokinase-type plasminogen activator positive cells are also tumourigenic in athymic nude mice.39

ALDH activity has been found to correlate with stem cell characteristics in lung cancer cell lines. In a study by Jiang et al., ALDH1-positive cells from NSCLC had extensive self-renewal and proliferative potential as well as in vivo tumourigenic potential.22 ALDH1-positive cells also expressed CD133 and showed resistance to common chemotherapy agents. Moreover, high ALDH1 protein expression, unlike other markers, was associated with poor prognosis in early stage NSCLC.22,41 Further, ALDH1-positive cells expressed Notch pathway transcripts, and inhibition of Notch with γ-secretase led to the reduction of ALDH1-positive tumour cells. Additionally, direct inhibition of ALDH protein by small interfering RNA resulted in reduced proliferation and migration in these cells.42

Studies performed to explore prognostic significance of these markers in NSCLC are summarized in Table 2. These findings suggest the existence of a small tumourigenic population of cells in lung cancer, favouring a hierarchical model. Further work is needed to identify more robust markers to define a distinct population of lung CSC.

Table 2.

Clinical studies conducted in NSCLC to study prognostic significance of putative CSC marker

Study Marker Method Population Number of patients Cut-off point Number positive (%) Main results
Jiang et al.22 ALDH IHC Stage 1 NSCLC 148 >10% 29 ALDH-positive patients have worse overall survival
Sullivan et al.41 ALDH1A1
ALDH1A3
CD133
IHC Stage 1 NSCLC 200 NM NM ALDH1A1 but not CD133 and ALDH1A3 are associated with poor clinical outcome
Woo et al.43 CD133 IHC Stage 1 NSCLC 177 17.5% 45 CD133-positive patients have shorter disease-free survival
Salnikov et al.44 CD133 IHC NSCLC all stages 88 Any expression 63 No difference between CD133-positive and negative patients
Li et al.45 ALDH1A1 IHC NSCLC all stages 179 Any expression 45 ALDH1A1 + patients had shorter survival
Dimou et al.46 ALDH IF NSCLC all stages 430 ALDH-positive patients have better prognosis

Variations exist in terms of methodology, patient selection, cut-off point determination as well as in results.

For statistical analysis, patient population is dichotomized into positive and negative cohorts based on appropriate cut-off point on immunohistochemical scoring.

ALDH, aldehyde dehydrogenase; CSC, cancer stem cell; IF, immunofluorescence; IHC, immunohistochemistry; NSCLC, non-small cell lung cancer; —, not reported.

EMBRYONIC STEM CELL PATHWAYS IN LUNG CANCER

Embryonic signalling pathways are fundamental in organogenesis. These pathways include Hedgehog (Hh), Notch, WNT and B lymphoma MLV insertion region 1, together with growth signals from the stem cell niche. All are involved in the renewal of normal stem cells and in maintaining tissue homeostasis.47 Dysregulation of these pathways is believed to be involved in driving CSC activity in a variety of cancers, including lung cancer.48 Therapeutic strategies to target these pathways are summarized in Table 3.

Table 3.

Summary of various agents in clinical development to target embryonic stem cell pathways

Target Agent Population Results Phase of development Reference
Hedgehog (SMO) GDC-0449 Lung cancer cell lines Inhibition of SP+ cells in cell lines Phase II Tian et al.49
Notch (γ-secretase) RO4929097 Advanced NSCLC Safety demonstrated in phase I study
Encouraging activity in some cancers
Phase II Richter et al.63(ASCO abstract)
Hedgehog (SMO) LDE225 Exposure dependent GLI-1 inhibition and disease stabilization in lung adenocarcinoma in phase I study Phase II Tawbi et al.64(ASCO abstract)
Notch DAPT NSCLC cell lines Reduction in ALDH+ tumour cells with cell cycle arrest Preclinical Sullivan et al.41
Notch MRK-003 NSCLC cell lines In vitro and in vivo inhibition of Notch signalling, reduction in proliferation and induction of apoptosis Konishi et al.50
WNT 1 Monoclonal antibody and RNAi NSCLC cell line Induction of apoptosis and growth inhibition in NSCLC cells He et al.51

DAPT = N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester (a γ-secretase inhibitor).

ALDH, aldehyde dehydrogenase; NSCLC, non-small cell lung cancer; SMO, Smoothened; SP, side population; —, not reported.

Hh pathway

The Hh pathway is involved in regulating the proliferation, migration and differentiation of progenitor cells52 and is silenced in most healthy adult tissues. The pathway is activated through the transmembrane domain protein smoothened and subsequent activation of the transcription factors (GLI1, GLI2 and GLI3), which regulate downstream target genes.53 The role of Hh signalling in CSC regulation has been described in many cancers.54,55 There is emerging evidence for Hh–GLI signalling in tumour maintenance in melanoma,56 breast57 and lung cancer.58 Activation of the Hh pathway in SCLC was first demonstrated in 2003.59 Inhibition of Hh signalling with the Veratrum alkaloid cyclopamine, a naturally occurring smoothened antagonist, leads to the loss of tumourigenicity in SCLC.59 These data suggest that deregulation of Hh signalling might result in SCLC through acquisition of neuroendocrine fate of airway progenitor cells in response to paracrine signals from adjacent epithelial cells. In recent data from Park et al., genetically engineered mice revealed that Hh signalling plays an important role in SCLC initiation. Inhibition of Hh signalling, using second-generation smoothened antagonists such as LDE-225 (Novartis), can supplement treatment with chemotherapy to inhibit tumour recurrence.60

Notch pathway

Notch signalling is a highly conserved embryonic pathway in mammals and is frequently involved in cellular fate determination. Activation of Notch signalling through cell-to-cell interaction retains stem cell viability via asymmetric cell division.61 The Notch pathway is involved in the development of normal lung and in determining proximal and distal epithelial cell fate in studies involving knockout mice.62 The role of Notch pathways in normal lung stem cells has been demonstrated in mouse studies involving knockout of Hes1 (hairy and enhancer of split 1), a key downstream target in Notch pathway. Here, suppression of Notch signalling resulted in increased neuroendocrine differentiation but reduced club cells (formerly known as Clara cells).65 Constitutive activation of Notch signalling resulted in delayed differentiation and accumulation of distal airway stem cells.66 The evidence for Notch pathway involvement in lung tumourigenesis comes from studies that showed elevated levels of Notch transcripts in NSCLC cell lines.67,68 Notch activity promoted pro-apoptotic signals through Bim and inhibited anti-apoptotic signals through induction of Survivin.50,67 Blockade of Notch pathway with inhibitors of γ-secretase (a protein complex required for proteolytic cleavage of Notch receptors) resulted in growth inhibition and induction of apoptosis both in vitro and in vivo.50 Further evidence comes from observations that deregulation in Notch signalling is involved in maintenance of oncogenic-Ras-driven cancers.69 Recently, active notch pathway was reported to be required for the clonogenic activity of ALDH-positive cells in NSCLC.41 Blockade of Notch3 by γ-secretase inhibition leading to suppression of clonogenic survival was demonstrated in NSCLC cell lines.68 Restoration of clonogenic survival was achieved after reintroduction of the Notch3 receptor domain.

In human glioma xenografts, inhibition of Notch using γ-secretase inhibitors resulted in diminished tumour growth as well as reduced numbers of the CD133-positive glioma stem cells.70 Some reports have also suggested possible oncogenic mutations in Notch1 receptor in NSCLC.71 However, despite several reports of a possible role for Notch signalling in lung carcinogenesis, there is no clear evidence that this pathway is involved in the maintenance of CSC phenotype. Notch may also have a tumour suppressor effect in squamous epithelia and myeloid malignancies72,73 and an antiangiogenic effect in blood vessels.74 Overall, the Notch signalling pathway is much more complex than currently understood.

In small cell lung cancer, a malignancy characterized by neuroendocrine features, the expression of Notch receptors is rarely seen. Notch signalling, however, is believed to be an attractive anticancer target, not only due to involvement in carcinogenesis and stem cell maintenance but also in tumour angiogenesis75 and immune function.76 Due to positive outcomes in preclinical models, γ-secretase inhibitors were developed, but due to lack of specificity, excessive toxicities especially in the gastrointestinal tract were observed in rodent models.77 More specific inhibitors of Notch transcriptional complex are currently under development and investigation.

WNT/β-CATENIN pathway

Another less understood pathway is WNT/β-CATENIN pathway. Aberrancies in WNT pathway have been linked to lung tumourigenesis.78 Targeting the WNT pathway with monoclonal antibodies resulted in induction of apoptosis in lung cancer cell lines overexpressing WNT-2 protein.79 Further investigation of this pathway in lung CSC is needed in order to determine the therapeutic potential of targeting the WNT pathway.

CONTROVERSIES SURROUNDING THE CSC MODEL IN LUNG CANCER

Although experimental evidence in support of the hierarchical arrangement of cells in lung cancer has accumulated in the last few years, controversies and uncertainties remain. Isolation of tumourigenic cells both in vivo and in vitro relies on specific surface markers that facilitate sorting of cancer cells into phenotypically distinct populations. However, no universal lung CSC marker has yet been discovered, as most of the markers tend to be histotype specific.38,41,80 For example, in many lung cancer samples, CD133 was not detected at all,21,44 and the prognostic role of CD133 expression in lung cancer has also not been clearly established due to discrepant reports.41,43,44 CD44 expression and ALDH activity marks CSC in NSCLC, but the protein expression of both these markers appears to be associated particularly strongly with squamous histology.80,81 Furthermore, the combined effect of these markers in lung CSC is at the moment unclear. Apart from a report where CD133 expression partially overlapped with ALDH activity in NSCLC cell lines,22 there is no convincing data to suggest that the enrichment for one marker also enriches for the others. Instead, it may be possible that these markers play their part independently of each other to represent a distinct subpopulation.82 In order to isolate pure CSC in lung cancer, more robust markers need to be identified.

The sensitivity of these surface markers to accurately delineate tumourigenic population is also being questioned. For example, CD133-negative cells in human lung cancer cell lines may also be tumourigenic, just like their CD133-positive counterparts.83 Similarly, discrepant results were seen with ALDH and SP phenotypes. Both ALDH-positive and ALDH-negative cells were tumourigenic in non-obese diabetic/severe combined immune deficiency mice,84 while non-SP cells retained CSC properties in vitro as well as in xenotransplantation.85 These findings suggest that either other tumour-initiating cells exist independently of these markers, or a phenomenon of ‘cellular plasticity’ exists. The latter, where non-cancer stem/progenitor cells convert to a stem/ progenitor cell phenotype, has recently been demonstrated in experiments conducted by Akunuru et al.82 Using NSCLC cell lines and primary lung adenocarcinoma samples, the authors demonstrated that non-SP, CD133-negative and ALDH-negative cells can generate SP, CD133-positive and ALDH-positive cells under adherent culture conditions.82 This phenomenon closely resembles the hypothesis of ‘phenotypic switching’ described in some other tumour types. Gupta et al. showed in breast cancer cells that different subpopulations of cells exist in phenotypic equilibrium.86 Cells purified for a given phenotype can, over a period of time, switch to an alternate phenotype, thus returning the tumour into an original state of phenotypic equilibrium. These findings are more in keeping with a stochastic model, in which cells under selective pressure are genetically, and therefore phenotypically, more able to form colonies and engraft in mice. However, the phenomenon of cellular plasticity has not been observed in some other tumours. For example, in melanoma, ATP-binding cassette B5-negative cells are unable to generate ATP-binding cassette B5-positive cells.87 In the case of solid tumours like lung cancer, where identification of CSC relies on expression of markers, it then becomes difficult to determine whether the tumour follows stochastic of hierarchical growth. As currently known markers only identify tumourigenic populations to varying extents, the overall accuracy of these markers remains doubtful.

CONCLUSION

The CSC model remains a very important model of cancer biology with major implications for lung cancer. The clinical picture in lung cancer with innate resistance to treatment and high rates of relapse fits this model well. There is considerable experimental data in support of a subpopulation of cancer cells that is capable of resistance to therapeutic strategies and tumour regrowth. Given the high rate of recurrence following therapy in all forms of lung cancer, the capacity to target minimal residual disease populations by blocking their CSC-like activity remains a tantalizing, but as yet theoretical, treatment option. The controversies largely stem from the fact that the markers studied so far are not sufficiently specific or sensitive. Nevertheless, the current identification of markers and pathways is already underpinning some novel developments in therapeutic strategies for patients with lung cancer.

Abbreviations

ALDH

aldehyde dehydrogenase

CSC

cancer stem cell

NSCLC

non-small cell lung cancer

SCLC

small cell lung cancer

SP

side population

Footnotes

The Authors: Muhammad Alamgeer, MRCP, FRACP, is a clinical research fellow in medical oncology with interest in clinical trials, biomarker discovery and cancer stem cells. Craig Peacock, PhD, is a basic and translational cancer researcher focusing on preclinical models of Hedgehog signalling and small cell lung cancer. William Matsui, MD, is a clinician scientist with an interest in multiple myeloma, cancer stem cells, Hedgehog signalling and therapeutic development. Vinod Ganju, FRACP, is a medical oncologist with interest in clinical trials. Neil Watkins, MBBS, PhD, FRACP, is a clinician scientist focusing on basic and preclinical models of cancer, with an interest in stem cells, Hedgehog signalling and epigenetics.

REFERENCES

  • 1.Jemal A, Bray F, Center MM, et al. Global cancer statistics. CA. 2011;61:69–90. doi: 10.3322/caac.20107. [DOI] [PubMed] [Google Scholar]
  • 2.Wu K, House L, Liu W, et al. Personalized targeted therapy for lung cancer. Int. J. Mol. Sci. 2012;13:11471–96. doi: 10.3390/ijms130911471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Kelsey CR, Marks LB, Hollis D, et al. Local recurrence after surgery for early stage lung cancer: an 11-year experience with 975 patients. Cancer. 2009;115:5218–27. doi: 10.1002/cncr.24625. [DOI] [PubMed] [Google Scholar]
  • 4.Wisnivesky JP, Yankelevitz D, Henschke CI. Stage of lung cancer in relation to its size: part 2. Evidence. Chest. 2005;127:1136–9. doi: 10.1378/chest.127.4.1136. [DOI] [PubMed] [Google Scholar]
  • 5.Howlader N, Noon AM, Krapcho M, et al. SEER Cancer Statistics Review, 1975–2009 (Vintage 2009 Populations) National Cancer Institute; Bethesda, MD: 2012. [2 Jan 2013]. based on November 2011 SEER data submission, posted to the SEER web site. Available from URL: http://seer.cancer.gov/csr/1975_ 2009_pops09/ [Google Scholar]
  • 6.Pao W, Miller VA, Politi KA, et al. Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain. PLoS Med. 2005;2:e73. doi: 10.1371/journal.pmed.0020073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Katayama R, Shaw AT, Khan TM, et al. Mechanisms of acquired crizotinib resistance in ALK-rearranged lung cancers. Sci. Transl. Med. 2012;4:120ra17. doi: 10.1126/scitranslmed.3003316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Janne PA, Freidlin B, Saxman S, et al. Twenty-five years of clinical research for patients with limited-stage small cell lung carcinoma in North America. Cancer. 2002;95:1528–38. doi: 10.1002/cncr.10841. [DOI] [PubMed] [Google Scholar]
  • 9.Murray N, Coy P, Pater JL, et al. Importance of timing for thoracic irradiation in the combined modality treatment of limited-stage small-cell lung cancer. The National Cancer Institute of Canada Clinical Trials Group. J. Clin. Oncol. 1993;11:336–44. doi: 10.1200/JCO.1993.11.2.336. [DOI] [PubMed] [Google Scholar]
  • 10.Reya T, Morrison SJ, Clarke MF, et al. Stem cells, cancer, and cancer stem cells. Nature. 2001;414:105–11. doi: 10.1038/35102167. [DOI] [PubMed] [Google Scholar]
  • 11.Lapidot T, Sirard C, Vormoor J, et al. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature. 1994;367:645–8. doi: 10.1038/367645a0. [DOI] [PubMed] [Google Scholar]
  • 12.Al-Hajj M, Wicha MS, Benito-Hernandez A, et al. Prospective identification of tumorigenic breast cancer cells. Proc. Natl. Acad. Sci. U S A. 2003;100:3983–8. doi: 10.1073/pnas.0530291100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Singh SK, Hawkins C, Clarke ID, et al. Identification of human brain tumour initiating cells. Nature. 2004;432:396–401. doi: 10.1038/nature03128. [DOI] [PubMed] [Google Scholar]
  • 14.O'Brien CA, Pollett A, Gallinger S, et al. A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature. 2007;445:106–10. doi: 10.1038/nature05372. [DOI] [PubMed] [Google Scholar]
  • 15.Pastrana E, Silva-Vargas V, Doetsch F. Eyes wide open: a critical review of sphere-formation as an assay for stem cells. Cell Stem Cell. 2011;8:486–98. doi: 10.1016/j.stem.2011.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat. Med. 1997;3:730–7. doi: 10.1038/nm0797-730. [DOI] [PubMed] [Google Scholar]
  • 17.Ho MM, Ng AV, Lam S, et al. Side population in human lung cancer cell lines and tumors is enriched with stem-like cancer cells. Cancer Res. 2007;67:4827–33. doi: 10.1158/0008-5472.CAN-06-3557. [DOI] [PubMed] [Google Scholar]
  • 18.Levina V, Marrangoni AM, DeMarco R, et al. Drug-selected human lung cancer stem cells: cytokine network, tumorigenic and metastatic properties. PLoS ONE. 2008;3:e3077. doi: 10.1371/journal.pone.0003077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Chen YC, Hsu HS, Chen YW, et al. Oct-4 expression maintained cancer stem-like properties in lung cancer-derived CD133-positive cells. PLoS ONE. 2008;3:e2637. doi: 10.1371/journal.pone.0002637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Eramo A, Lotti F, Sette G, et al. Identification and expansion of the tumorigenic lung cancer stem cell population. Cell Death Differ. 2008;15:504–14. doi: 10.1038/sj.cdd.4402283. [DOI] [PubMed] [Google Scholar]
  • 21.Bertolini G, Roz L, Perego P, et al. Highly tumorigenic lung cancer CD133+ cells display stem-like features and are spared by cisplatin treatment. Proc. Natl. Acad. Sci. U S A. 2009;106:16281–6. doi: 10.1073/pnas.0905653106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Jiang F, Qiu Q, Khanna A, et al. Aldehyde dehydrogenase 1 is a tumor stem cell-associated marker in lung cancer. Mol. Cancer Res. 2009;7:330–8. doi: 10.1158/1541-7786.MCR-08-0393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Shi Y, Fu X, Hua Y, et al. The side population in human lung cancer cell line NCI-H460 is enriched in stem-like cancer cells. PLoS ONE. 2012;7:e33358. doi: 10.1371/journal.pone.0033358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Quintana E, Shackleton M, Sabel MS, et al. Efficient tumour formation by single human melanoma cells. Nature. 2008;456:593–8. doi: 10.1038/nature07567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Ishizawa K, Rasheed ZA, Karisch R, et al. Tumor-initiating cells are rare in many human tumors. Cell Stem Cell. 2010;7:279–82. doi: 10.1016/j.stem.2010.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Goodell MA, Brose K, Paradis G, et al. Isolation and functional properties of murine hematopoietic stem cells that are replicating in vivo. J. Exp. Med. 1996;183:1797–806. doi: 10.1084/jem.183.4.1797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Zhong Y, Zhou C, Ma W, et al. Most MCF7 and SK-OV3 cells were deprived of their stem nature by Hoechst 33342. Biochem. Biophys. Res. Commun. 2007;364:338–43. doi: 10.1016/j.bbrc.2007.10.016. [DOI] [PubMed] [Google Scholar]
  • 28.Chute JP, Muramoto GG, Whitesides J, et al. Inhibition of alde-hyde dehydrogenase and retinoid signaling induces the expansion of human hematopoietic stem cells. Proc. Natl. Acad. Sci. U S A. 2006;103:11707–12. doi: 10.1073/pnas.0603806103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Kastan MB, Schlaffer E, Russo JE, et al. Direct demonstration of elevated aldehyde dehydrogenase in human hematopoietic progenitor cells. Blood. 1990;75:1947–50. [PubMed] [Google Scholar]
  • 30.Moreb JS. Aldehyde dehydrogenase as a marker for stem cells. Curr. Stem Cell Res. Ther. 2008;3:237–46. doi: 10.2174/157488808786734006. [DOI] [PubMed] [Google Scholar]
  • 31.Ginestier C, Hur MH, Charafe-Jauffret E, et al. ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell. 2007;1:555–67. doi: 10.1016/j.stem.2007.08.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Matsui W, Huff CA, Wang Q, et al. Characterization of clonogenic multiple myeloma cells. Blood. 2004;103:2332–6. doi: 10.1182/blood-2003-09-3064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Miraglia S, Godfrey W, Yin AH, et al. A novel five-transmembrane hematopoietic stem cell antigen: isolation, characterization, and molecular cloning. Blood. 1997;90:5013–21. [PubMed] [Google Scholar]
  • 34.Richardson GD, Robson CN, Lang SH, et al. CD133, a novel marker for human prostatic epithelial stem cells. J. Cell Sci. 2004;117:3539–45. doi: 10.1242/jcs.01222. [DOI] [PubMed] [Google Scholar]
  • 35.Hess DA, Wirthlin L, Craft TP, et al. Selection based on CD133 and high aldehyde dehydrogenase activity isolates long-term reconstituting human hematopoietic stem cells. Blood. 2006;107:2162–9. doi: 10.1182/blood-2005-06-2284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Marhaba R, Zoller M. CD44 in cancer progression: adhesion, migration and growth regulation. J. Mol. Histol. 2004;35:211–31. doi: 10.1023/b:hijo.0000032354.94213.69. [DOI] [PubMed] [Google Scholar]
  • 37.Carney DN, Gazdar AF, Bunn PA, Jr, et al. Demonstration of the stem cell nature of clonogenic tumor cells from lung cancer patients. Stem Cells. 1982;1:149–64. [PubMed] [Google Scholar]
  • 38.Leung EL, Fiscus RR, Tung JW, et al. Non-small cell lung cancer cells expressing CD44 are enriched for stem cell-like properties. PLoS ONE. 2010;5:e14062. doi: 10.1371/journal.pone.0014062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Qiu X, Wang Z, Li Y, et al. Characterization of sphere-forming cells with stem-like properties from the small cell lung cancer cell line H446. Cancer Lett. 2012;323:161–70. doi: 10.1016/j.canlet.2012.04.004. [DOI] [PubMed] [Google Scholar]
  • 40.Gutova M, Najbauer J, Gevorgyan A, et al. Identification of uPAR-positive chemoresistant cells in small cell lung cancer. PLoS ONE. 2007;2:e243. doi: 10.1371/journal.pone.0000243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Sullivan JP, Spinola M, Dodge M, et al. Aldehyde dehydrogenase activity selects for lung adenocarcinoma stem cells dependent on notch signaling. Cancer Res. 2010;70:9937–48. doi: 10.1158/0008-5472.CAN-10-0881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Moreb JS, Baker HV, Chang LJ, et al. ALDH isozymes downregulation affects cell growth, cell motility and gene expression in lung cancer cells. Mol. Cancer. 2008;7:87. doi: 10.1186/1476-4598-7-87. doi:10.1186/1476-4598-7-87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Woo T, Okudela K, Mitsui H, et al. Prognostic value of CD133 expression in stage I lung adenocarcinomas. Int. J. Clin. Exp. Pathol. 2010;4:32–42. [PMC free article] [PubMed] [Google Scholar]
  • 44.Salnikov AV, Gladkich J, Moldenhauer G, et al. CD133 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. 2010;126:950–8. doi: 10.1002/ijc.24822. [DOI] [PubMed] [Google Scholar]
  • 45.Li X, Wan L, Geng J, et al. Aldehyde dehydrogenase 1A1 possesses stem-like properties and predicts lung cancer patient outcome. J. Thorac. Oncol. 2012;7:1235–45. doi: 10.1097/JTO.0b013e318257cc6d. [DOI] [PubMed] [Google Scholar]
  • 46.Dimou A, Neumeister V, Agarwal S, et al. Measurement of alde-hyde dehydrogenase 1 expression defines a group with better prognosis in patients with non-small cell lung cancer. Am. J. Pathol. 2012;181:1436–42. doi: 10.1016/j.ajpath.2012.06.037. [DOI] [PubMed] [Google Scholar]
  • 47.Karamboulas C, Ailles L. Developmental signaling pathways in cancer stem cells of solid tumors. Biochim. Biophys. Acta. 2013;1830:2481–95. doi: 10.1016/j.bbagen.2012.11.008. [DOI] [PubMed] [Google Scholar]
  • 48.Takebe N, Ivy SP. Controversies in cancer stem cells: targeting embryonic signaling pathways. Clin. Cancer Res. 2010;16:3106–12. doi: 10.1158/1078-0432.CCR-09-2934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Tian F, Mysliwietz J, Ellwart J, et al. Effects of the Hedgehog pathway inhibitor GDC-0449 on lung cancer cell lines are mediated by side populations. Clin. Exp. Med. 2012;12:25–30. doi: 10.1007/s10238-011-0135-8. [DOI] [PubMed] [Google Scholar]
  • 50.Konishi J, Kawaguchi KS, Vo H, et al. Gamma-secretase inhibitor prevents Notch3 activation and reduces proliferation in human lung cancers. Cancer Res. 2007;67:8051–7. doi: 10.1158/0008-5472.CAN-07-1022. [DOI] [PubMed] [Google Scholar]
  • 51.He B, You L, Uematsu K, et al. A monoclonal antibody against Wnt-1 induces apoptosis in human cancer cells. Neoplasia. 2004;6:7–14. doi: 10.1016/s1476-5586(04)80048-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Varjosalo M, Taipale J. Hedgehog: functions and mechanisms. Genes Dev. 2008;22:2454–72. doi: 10.1101/gad.1693608. [DOI] [PubMed] [Google Scholar]
  • 53.Sasaki H, Nishizaki Y, Hui C, et al. Regulation of GLI2 and GLI3 activities by an amino-terminal repression domain: implication of GLI2 and GLI3 as primary mediators of Shh signaling. Development. 1999;126:3915–24. doi: 10.1242/dev.126.17.3915. [DOI] [PubMed] [Google Scholar]
  • 54.Clement V, Sanchez P, de Tribolet N, et al. HEDGEHOG-GLI1 signaling regulates human glioma growth, cancer stem cell self-renewal, and tumorigenicity. Curr. Biol. 2007;17:165–72. doi: 10.1016/j.cub.2006.11.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Peacock CD, Wang Q, Gesell GS, et al. Hedgehog signaling maintains a tumor stem cell compartment in multiple myeloma. Proc. Natl. Acad. Sci. U S A. 2007;104:4048–53. doi: 10.1073/pnas.0611682104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Stecca B, Mas C, Clement V, et al. Melanomas require HEDGEHOG-GLI signaling regulated by interactions between GLI1 and the RAS-MEK/AKT pathways. Proc. Natl. Acad. Sci. U S A. 2007;104:5895–900. doi: 10.1073/pnas.0700776104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Mukherjee S, Frolova N, Sadlonova A, et al. Hedgehog signaling and response to cyclopamine differ in epithelial and stromal cells in benign breast and breast cancer. Cancer Biol. Ther. 2006;5:674–83. doi: 10.4161/cbt.5.6.2906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Yuan Z, Goetz JA, Singh S, et al. Frequent requirement of hedgehog signaling in non-small cell lung carcinoma. Oncogene. 2007;26:1046–55. doi: 10.1038/sj.onc.1209860. [DOI] [PubMed] [Google Scholar]
  • 59.Watkins DN, Berman DM, Burkholder SG, et al. Hedgehog signalling within airway epithelial progenitors and in small-cell lung cancer. Nature. 2003;422:313–7. doi: 10.1038/nature01493. [DOI] [PubMed] [Google Scholar]
  • 60.Park KS, Martelotto LG, Peifer M, et al. A crucial requirement for Hedgehog signaling in small cell lung cancer. Nat. Med. 2011;17:1504–8. doi: 10.1038/nm.2473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Artavanis-Tsakonas S, Rand MD, Lake RJ. Notch signaling: cell fate control and signal integration in development. Science. 1999;284:770–6. doi: 10.1126/science.284.5415.770. [DOI] [PubMed] [Google Scholar]
  • 62.Collins BJ, Kleeberger W, Ball DW. Notch in lung development and lung cancer. Semin. Cancer Biol. 2004;14:357–64. doi: 10.1016/j.semcancer.2004.04.015. [DOI] [PubMed] [Google Scholar]
  • 63.Richter S, McWhirter E, Chen EX, et al. A phase I study of R04929097, an oral gamma secretase inhibitor, in combination with gemcitabine, in patients with advanced solid tumors (PHL-078/CTEP 8575). J. Clin. Oncol. 2012;30(Suppl) Abstr 3082. [Google Scholar]
  • 64.Tawbi HA, Ahnert R, Thomas AL, Shou GY, et al. Phase I study of LDE225 in advanced solid tumors: updated analysis of safety, preliminary efficacy, and pharmacokinetic-pharmacoynamic correlation. J. Clin. Oncol. 2011;29(Suppl) Abstr 3062. [Google Scholar]
  • 65.Ito T, Udaka N, Yazawa T, et al. Basic helix-loop-helix transcription factors regulate the neuroendocrine differentiation of fetal mouse pulmonary epithelium. Development. 2000;127:3913–21. doi: 10.1242/dev.127.18.3913. [DOI] [PubMed] [Google Scholar]
  • 66.Dang TP, Eichenberger S, Gonzalez A, et al. Constitutive activation of Notch3 inhibits terminal epithelial differentiation in lungs of transgenic mice. Oncogene. 2003;22:1988–97. doi: 10.1038/sj.onc.1206230. [DOI] [PubMed] [Google Scholar]
  • 67.Chen H, Thiagalingam A, Chopra H, et al. Conservation of the Drosophila lateral inhibition pathway in human lung cancer: a hairy-related protein (HES-1) directly represses achaete-scute homolog-1 expression. Proc. Natl. Acad. Sci. U S A. 1997;94:5355–60. doi: 10.1073/pnas.94.10.5355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Osanyingbemi-Obidi J, Dobromilskaya I, Illei PB, et al. Notch signaling contributes to lung cancer clonogenic capacity in vitro but may be circumvented in tumorigenesis in vivo. Mol. Cancer Res. 2011;9:1746–54. doi: 10.1158/1541-7786.MCR-11-0286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Weijzen S, Rizzo P, Braid M, et al. Activation of Notch-1 signaling maintains the neoplastic phenotype in human Ras-transformed cells. Nat. Med. 2002;8:979–86. doi: 10.1038/nm754. [DOI] [PubMed] [Google Scholar]
  • 70.Fan X, Matsui W, Khaki L, et al. Notch pathway inhibition depletes stem-like cells and blocks engraftment in embryonal brain tumors. Cancer Res. 2006;66:7445–52. doi: 10.1158/0008-5472.CAN-06-0858. [DOI] [PubMed] [Google Scholar]
  • 71.Westhoff B, Colaluca IN, D'Ario G, et al. Alterations of the Notch pathway in lung cancer. Proc. Natl. Acad. Sci. U S A. 2009;106:22293–8. doi: 10.1073/pnas.0907781106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Nicolas M, Wolfer A, Raj K, et al. Notch1 functions as a tumor suppressor in mouse skin. Nat. Genet. 2003;33:416–21. doi: 10.1038/ng1099. [DOI] [PubMed] [Google Scholar]
  • 73.Klinakis A, Lobry C, Abdel-Wahab O, et al. A novel tumour-suppressor function for the Notch pathway in myeloid leukaemia. Nature. 2011;473:230–3. doi: 10.1038/nature09999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Liu Z, Turkoz A, Jackson EN, et al. Notch1 loss of heterozygosity causes vascular tumors and lethal hemorrhage in mice. J. Clin. Invest. 2011;121:800–8. doi: 10.1172/JCI43114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Phng LK, Gerhardt H. Angiogenesis: a team effort coordinated by notch. Dev. Cell. 2009;16:196–208. doi: 10.1016/j.devcel.2009.01.015. [DOI] [PubMed] [Google Scholar]
  • 76.Radtke F, Fasnacht N, Macdonald HR. Notch signaling in the immune system. Immunity. 2010;32:14–27. doi: 10.1016/j.immuni.2010.01.004. [DOI] [PubMed] [Google Scholar]
  • 77.Imbimbo BP. Therapeutic potential of gamma-secretase inhibitors and modulators. Curr. Top. Med. Chem. 2008;8:54–61. doi: 10.2174/156802608783334015. [DOI] [PubMed] [Google Scholar]
  • 78.Uematsu K, He B, You L, et al. Activation of the Wnt pathway in non small cell lung cancer: evidence of dishevelled overexpression. Oncogene. 2003;22:7218–21. doi: 10.1038/sj.onc.1206817. [DOI] [PubMed] [Google Scholar]
  • 79.You L, He B, Xu Z, et al. Inhibition of Wnt-2-mediated signaling induces programmed cell death in non-small-cell lung cancer cells. Oncogene. 2004;23:6170–4. doi: 10.1038/sj.onc.1207844. [DOI] [PubMed] [Google Scholar]
  • 80.Moreb JS, Zucali JR, Ostmark B, et al. Heterogeneity of aldehyde dehydrogenase expression in lung cancer cell lines is revealed by Aldefluor flow cytometry-based assay. Cytometry B Clin. Cytom. 2007;72:281–9. doi: 10.1002/cyto.b.20161. [DOI] [PubMed] [Google Scholar]
  • 81.Ko YH, Won HS, Jeon EK, et al. Prognostic significance of CD44s expression in resected non-small cell lung cancer. BMC Cancer. 2011;11:340. doi: 10.1186/1471-2407-11-340. doi: 10.1186/1471-2407-11-340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Akunuru S, James Zhai Q, Zheng Y. Non-small cell lung cancer stem/progenitor cells are enriched in multiple distinct phenotypic subpopulations and exhibit plasticity. Cell Death Dis. 2012;3:e352. doi: 10.1038/cddis.2012.93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Meng X, Li M, Wang X, et al. Both CD133+ and CD133- subpopulations of A549 and H446 cells contain cancer-initiating cells. Cancer Sci. 2009;100:1040–6. doi: 10.1111/j.1349-7006.2009.01144.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Ucar D, Cogle CR, Zucali JR, et al. Aldehyde dehydrogenase activity as a functional marker for lung cancer. Chem. Biol. Interact. 2009;178:48–55. doi: 10.1016/j.cbi.2008.09.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Akunuru S, Palumbo J, Zhai QJ, et al. Rac1 targeting suppresses human non-small cell lung adenocarcinoma cancer stem cell activity. PLoS ONE. 2011;6:e16951. doi: 10.1371/journal.pone.0016951. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Gupta PB, Fillmore CM, Jiang G, et al. Stochastic state transitions give rise to phenotypic equilibrium in populations of cancer cells. Cell. 2011;146:633–44. doi: 10.1016/j.cell.2011.07.026. [DOI] [PubMed] [Google Scholar]
  • 87.Schatton T, Murphy GF, Frank NY, et al. Identification of cells initiating human melanomas. Nature. 2008;451:345–9. doi: 10.1038/nature06489. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES