Oncogene (2015) 34, 5771–5780 © 2015 Macmillan Publishers Limited All rights reserved 0950-9232/15 www.nature.com/onc

REVIEW Lineage factors and differentiation states in lung cancer progression

WKC Cheung1 and DX Nguyen1,2

Lung cancer encompasses a heterogeneous group of malignancies. Here we discuss how the remarkable diversity of major lung cancer subtypes is manifested in their transforming cell of origin, oncogenic dependencies, phenotypic plasticity, metastatic competence and response to therapy. More specifically, we review the increasing evidence that links this biological heterogeneity to the deregulation of cell lineage-specific pathways and the transcription factors that ultimately control them. As determinants of pulmonary epithelial differentiation, these poorly characterized transcriptional networks may underlie the etiology and biological progression of distinct lung cancers, while providing insight into innovative therapeutic strategies.

Oncogene (2015) 34, 5771–5780; doi:10.1038/onc.2015.85; published online 30 March 2015

INTRODUCTION intermediates during this process.5 In the post-natal lungs, AT2 cells 4,6 Pathologists have long been familiar with the existence of various also acquire progenitor-like functions to generate AT1 cells. lung cancer histological subtypes and the challenges in accurately Following severe injury and inflammation, distal epithelial regen- 7,8 classifying them. Thoracic cancer can be divided into the following eration can also occur from putative lineage-negative stem cells. two major histotypes: small cell lung cancer (SCLC) and non-small Having compiled a working catalog of pulmonary epithelial cell lung cancer (NSCLC). Most lung cancer patients are diagnosed stem/progenitor lineages, recent studies have attempted to with NSCLC, which can be further subclassified into lung determine which of these cells are targets for transformation in adenocarcinoma (LUAD), lung squamous cell carcinoma (LUSC) various lung cancers by using genetically engineered mouse and large cell carcinoma. Updated histological subclasses of LUAD models (GEMMs).9 Several GEMMs exploit cell type-specific have been proposed for diagnosis and treatment.1 As revealed by promoters, such as surfactant C (SPC) in AT2 cells, club recent genome-wide studies, individual lung tumors also possess cell 10-kDa protein (CC10) in club cells and calcitonin -related highly diverse genomes, which further underscore the biological peptide in pulmonary neuroendocrine (NE) cells, to express complexity of these diseases. Untangling the relationship between commonly seen in lung cancer patients in a lineage- lung tumor histotype, phenotype and molecular heterogeneity specific manner.10 In sum, both the oncogenic and represents a critical barrier in improving the clinical outcome epithelial cell type in which it is expressed can influence which of thoracic malignancies, which collectively account for most lung cancer subtypes will form in mice. This topic has been cancer-related deaths to date.2 extensively reviewed elsewhere,11–14 and recent updates are discussed here (Figure 1). LUNG CANCER: PLEIOTROPIC ORIGINS AND DRIVERS Current views on the cellular origins of different lung cancers are Small cell lung cancer shaped by our understanding of normal pulmonary development Because SCLC arises in the central airways and expresses NE and homeostasis. The lung epithelium arises from the ventral side markers, it has long been postulated that this lung cancer type of the anterior foregut endoderm, where primary lung buds are originates from pulmonary NE cells. These progenitors are rare 15 formed.3 Following extensive branching of the proximal conducting and usually cluster as NE bodies in the bronchioles. Most human airways including the trachea, bronchi and bronchioles, cells at the SCLCs harbor inactivating mutations in the prototypical tumor 16 distal branch tips differentiate into alveolar type 1 (AT1) and 2 suppressors TP53 and the retinoblastoma 1 (RB1) . By using (AT2) cells, which constitute the gas-exchanging alveoli. In the cell type-specific conditional knockouts of these two tumor developing and adult lungs, multiple regional epithelial cell types suppressors,17 independent groups have confirmed that pulmonary – can serve as pools of progenitor cells. In the trachea and main NE cells are the predominant cells of origin of SCLC.18 20 bronchi, basal cells give rise to secretory and ciliated cells of the Interestingly, inactivation of Tp53 and Rb1 in a subset of AT2 luminal layer, whereas in bronchiolar epithelium, club cells cells, but not in club cells, may also lead to SCLC at reduced (formerly known as Clara cells) can self-renew and generate efficiency.18 The progression of Tp53/Rb1 null SCLC can be ciliated cells. In the distal airway, AT1 and AT2 cells arise directly accelerated by the additional loss of Pten, which has recently from a bipotent progenitor during embryogenesis.4 A recent been proposed as a frequent, but late genetic event during SCLC single-cell transcriptome analysis identified several maturation tumorigenesis.20,21

1Department of Pathology, Pathology and Cancer Center, Yale University School of Medicine, New Haven, CT, USA and 2Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA. Correspondence: Dr DX Nguyen, Department of Pathology, Pathology and Cancer Center, Yale University School of Medicine, PO Box 208023, New Haven, CT 06520-8023, USA. E-mail: [email protected] Received 18 December 2014; revised 13 February 2015; accepted 16 February 2015; published online 30 March 2015 Transcription control of lineage states in lung cancer WKC Cheung and DX Nguyen 5772

Figure 1. Known and predicted influence of the cell of origin vs oncogenic mutation in the formation of different lung cancer subtypes. BADJ, bronchoalveolar duct junction; CGRP, calcitonin gene-related peptide; PNEC, pulmonary neuroendocrine cell.

Lung squamous cell carcinoma bronchioles can also occur if the Tp53 is + 30 LUSC expresses basal cell markers (including KRT5, p63 and ) concomitantly deleted in CC10 cells. Some of these effects may and frequently occurs in the proximal airway. Therefore, it has depend on distinct oncogenic activities of variant Kras mutations. been proposed that LUSC arises from basal progenitors. Until Inflammatory responses may also enhance the overall transforming 27 recently, the lack of LUSC GEMM has precluded the identification effects of associated genotypes/cell phenotypes. By contrast, + of its cellular origin, and several attempts have been made to mutant Egfr expression in CC10 cells can give rise to LUAD that 31 model this malignancy. Although rarely mutated in human LUSC, rarely metastasizes. Moreover, the cell surface antigenic profile of tumor-initiating cells varies when comparing LUADs driven by inactivation of the tumor suppressor Stk11 (also known as Lkb1)in 32 Kras-mutant mice can generate LUSC, but also creates a wide Egfr and Kras mutations. Altogether, the experimental evidence spectrum of NSCLCs including mixed adenosquamous carcinoma to date implicates a combination of unique genetic and and large cell carcinoma.22 In addition, IKKα kinase inactivation in environmental contexts within the distal airway that is required mice generates spontaneous LUSC exclusively, albeit at a relatively for LUAD initiation. The function of other driver mutations in LUAD low efficiency.23 The comprehensive catalog of genomic altera- might be equally variable across regional epithelial cell lineages, tions in LUSC identified recurrent PTEN mutations.24 Concomitant contributing even further to the heterogeneous nature of this inactivation of Lkb1 and Pten leads to LUSC in GEMM with 100% NSCLC subtype. penetrance.25 Notably, the resulting tumors resemble many histological and transcriptional features of the human LUSC basal SWITCHING PATHS TO PROGRESS subtype. Given this result, it is expected that the cellular origin(s) fi of LUSC will soon be identified using conditional knockouts of Following tumor initiation from speci c cell types, lung cancers Lkb1 and Pten in GEMM. can seemingly adopt various aberrant differentiation states. In solid epithelial tumors, one of the most extensively studied manifestations of this phenomenon is epithelial-to-mesenchymal Lung adenocarcinoma transition (EMT). EMT is a developmental process that is not only Among different lung cancer subtypes, the cellular origins of linked to tumor cell invasion but also to oncogenesis and tumor LUAD are well studied owing to the availability of multiple initiation.33 In human lung cancers, clinical evidence for EMT is GEMMs. Several of these rely on the spatial and temporal seen in a fraction of LUADs that have acquired resistance to induction of an oncogenic Kras-mutant allele. In humans, KRAS tyrosine kinase inhibitors.34–36 Independent of EMT, lung cancer is most frequently mutated in tissues of endodermal origin, histopathological variations may correlate with cell lineage states including the lung epithelium.26 Independent groups have shown that are unique or selective for the airways. For instance, at the that Kras-driven murine LUAD preferentially arises from AT2 cells, genomic level, resected human NSCLCs can be distinguished by consistent with its predominant site of diagnosis at the distal profiles associated with different stages of airway and the immunohistochemical staining of alveolar markers pulmonary development.37 Despite arising from the alveolar – in early stage patient tumors.27 30 In contrast, Kras activation epithelium, LUADs can convert to SCLCs, the latter being a NE driven by the CC10 , presumably in club cells, cancer more typically located in the central lungs.38 LUAD bronchioalveolar stem cells and a small percentage of AT2 cells, conversion to an ‘SCLC-like’ phenotype is associated with drug leads to hyperplasia at the bronchoalveolar duct junction, but not resistance.35,39–41 Furthermore, a significant proportion of high- frank LUAD.28 In principle, tumor progression in the terminal grade LUADs histologically resembles LUSCs, which express

Oncogene (2015) 5771 – 5780 © 2015 Macmillan Publishers Limited Transcription control of lineage states in lung cancer WKC Cheung and DX Nguyen 5773 markers of proximal airway basal cells. Mixed adenosquamous lungs, Rb is required for NE cell fate, while the specification of tumors retain epithelial markers, yet are more invasive and have a other airway cell types can occur in the absence of Rb due to worse prognosis.42–44 Within the LUAD subtype specifically, a compensation from other pocket .62 This selective gene module of normal alveolar differentiation stratifies tumors requirement for RB1 during mammalian lung development is with distinct grades, biological properties and clinical outcome.45 consistent with the NE origins of SCLC and the potent SCLC The transcriptomic portrait of lung epithelial specification is phenotype generated via Rb and mutations in GEMMs. ultimately reflected in the nomenclature adopted by The Cancer 24,46 Genome Atlas to subclassify human LUADs and LUSCs. This Basal factors in LUSC remarkable molecular heterogeneity seen between and within fl In LUSCs, Sry-related HMG box 2 (SOX2) is one of the most human lung cancer subtypes may re ect the relative abundance frequently amplified and overexpressed genes.24,63–65 SOX2 of various progenitor cell lineages in a tumor mass. Instead of fi encodes for a TF best known for its ability to reprogram being xed over time, tumor histopathology can also deviate due embryonic stem cells. During normal development, however, to the capacity of a tumor-initiating cell population to dediffer- SOX2 is essential for basal cell commitment in the proximal entiate or transdifferentiate relative to its original lineage. 66 fi airways. As an oncogene, SOX2 is required for the survival of Signi cantly, molecular aberrations or extracellular signals can LUSC cells, but its aberrant expression alone is not sufficient for directly drive this phenotypic plasticity. transformation.65 A combination of genetic alterations, such as the In GEMMs, lineage-tracing experiments have demonstrated that loss of Lkb1,67 might be required for Sox2-driven LUSC in mice. Kras-transformed AT2 cells self-renew without additional changes fi 4 SOX2 is usually co-ampli ed with the basal lineage TF p63 at in cell fate, at least up until the formation of adenomas. By 63 + − 3q26. Their interaction and colocalization at target contrast, Kras-mutant CC10 hyperplasias can switch to a CC10 / 68 + gene loci support oncogenesis instead of pluripotency. Within SPC -like phenotype to develop into LUAD in the terminal the same amplicon as SOX2 is PRKCI, which encodes for protein 27 fl bronchioles. This may be induced by local in ammation or kinase Cι that phosphorylates SOX2, leading to the activation of oncogenic RAS-mediated AT2 differentiation, a lineage state that Sonic hedgehog (Shh) signaling and LUSC self-renewal.69 is more conducive to LUAD progression. Phenotypic conversion of In contrast to its role in promoting LUSCs, high levels of Sox2 in AT2-derived LUADs is seen when mutating the tumor suppressor CC10+ cells restrict the formation of LUAD in the bronchioles by Stk11 in conjunction with Kras, which generates tumors that suppressing Notch activation.70 transdifferentiate into LUSCs.22,47 LUSC metaplasia from LUADs is regulated by conserved developmental signals including the Hippo/Yap48 and the transforming growth factor/SMAD Alveolar factors in LUAD pathways.49 Another developmental program required for respira- In distal epithelial cells, a different SOX family member, SOX9, tion is the canonical WNT/β-catenin pathway.50–52 Mutations in marks the branch tips of early lungs and functions downstream of components of this pathway such as APC, GSK3B and CTNNB1 are tyrosine kinase signaling to suppress premature alveolar 53 differentiation.71,72 SOX9 is overexpressed in human LUAD and its detected in 15% of human LUADs. Multiple studies report 73,74 frequent overexpression of various WNT pathway components expression correlates with poor patient survival. Induction of and epigenetic silencing of WNT pathway antagonists in human Sox9 expression has been observed in murine LUADs harboring 54 fi activating mutations in Kras and β-catenin, and this overexpres- LUADs. In GEMM, lung-speci c expression of a constitutively 56,57 active mutant β-catenin is not sufficient to initiate LUAD, but sion is associated with high-grade tumors. Moreover, SOX9 is preferentially upregulated in human KRAS mutant adenomas and may increase the pool of progenitor cells available for 75 transformation.55,56 Deletion of the WNT antagonist APC coop- is also induced by Notch activation. 57 NK2 1 (NKX2-1), also known as thyroid TF-1, is erates with Kras to induce dedifferentiated invasive LUADs. 76 Moreover, transcriptional activation of the WNT/TCF effector LEF1 expressed in AT2 cells and a subset of bronchiolar cells. NKX2-1 58,59 is essential for lung morphogenesis and alveolar cell is a marker and mediator of LUAD metastasis to the brain. The 77 mechanism by which WNT/TCF promotes LUAD may involve both differentiation. Thyroid TF-1 protein is a biomarker of thymic the expansion of the cell of origin and epigenetic reprograming of cancers and LUADs. About 15% of LUADs harbor NKX2-1 amplification78–80 that correlates with poor outcome81,82 and is transformed cells into a primitive state that has increased 80,83 metastatic potential. required for tumor cell viability. NKX2-1 can support pro- tumorigenic signaling downstream of mutant epidermal growth factor receptor (EGFR)84 and is required for EGFR-mediated LINEAGE TRANSCRIPTION FACTORS IN CONTEXT transformation in vivo.85 Paradoxically, NKX2-1 expression also correlates with better outcome, as its expression is frequently Underlying the diverse pathways, molecular perturbations and fi differentiation states in lung cancers are transcription factors (TFs) suppressed in high-grade LUADs with wild-type/non-ampli ed NKX2-1.81,82,86,87 Importantly, in murine LUAD models, wild-type and cofactors whose expression and activity can be restricted for 85,88 certain cell lineages.3,60 As will be discussed below, the genes Nkx2-1 inhibits LUAD progression driven by Kras. This tumor- suppressive effect is closely linked to epithelial differentiation encoding several such lineage TFs can be directly mutated, fi 85 activated or silenced in particular lung cancer subtypes, suggesting states, as Nkx2-1 haploinsuf ciency promotes mucinous LUAD and activation of enteric lineage markers that are normally that they are context-dependent drivers of tumor progression and 88 phenotypic heterogeneity. repressed in alveolar cells. Repression of Nkx2-1 is also required for stochastic dissemination and metastasis by tumors with Kras and p53 mutations.86 NE factors in SCLC The contradictory role of certain lineage TFs likely depends on Inactivating mutations in RB1 are ubiquitous to all SCLCs.16 The the epigenetic context in which they function. One determinant of extended pRb family of pocket proteins interacts with a myriad of such contexts is the relative abundance and accessibility of DNA binding TFs and cofactors required for cell cycle progression, different cofactors in a given progenitor cell. The levels and self-renewal, , senescence and .61 dependencies of cofactors can be initially dictated by cell lineage Although components of the pRB pathway are frequently states (for example, tumor cell of origin or stage) and further inactivated across many cancers, the strong selection for dysregulated by somatic alterations (for example, mutation, gene mutations in RB1 exclusively within SCLC suggests that pRb itself amplification and DNA methylation). NKX2-1, for instance, can is a lineage-specific tumor suppressor. Remarkably, in the murine interact with multiple DNA binding transcriptional repressors or

© 2015 Macmillan Publishers Limited Oncogene (2015) 5771 – 5780 Transcription control of lineage states in lung cancer WKC Cheung and DX Nguyen 5774 activators to expand or limit the range of target genes.89 Wild-type The oncogenic function of GATA6 may depend on its amplification levels of NKX2-1 maintain alveolar differentiation and inhibit or other cooperating gene amplifications103 and availability of proliferation by restricting the activity and genomic target loci of interacting TF partners, such as SP1,105 KLF5 and GATA4.104 FOXA1/288 and AP1 factors,85 respectively. In this regulatory Elevated levels of various GATAs correlate with EMT in a murine network, decreases in NKX2-1 causes derepression of genes that LUAD model.108 On the other hand, expression of GATAs is mediate cell proliferation,90 EMT,91 motility92 and resistance to generally silenced in a significant proportion of human anoikis.86 Conversely, NKX2-1 amplification is often accompanied LUADs.46,109 In a subset of high-grade LUADs, GATA6 is co- by overexpression of the lineage TF FOXA1.93 In this latter context, repressed with other alveolar TFs with whom it may cooperate to 45 which includes aberrantly high levels of NKX2-1 and FOXA1, both inhibit LUAD metastasis. Hypermethylation and mutation of 109 TFs can cooperatively access and transactivate de novo genes GATA6 itself are uncommon in LUADs, but, as in other cancers, 110 required for cancer cell survival.93 Thus, genes that would alternative mechanisms may contribute to its downregulation. otherwise not be targeted by physiological levels of a given The homeodomain only protein X (HOPX), reported as a marker 5,6 lineage TF can be co-opted for malignant progression under of AT1 cells, is an atypical homedomain protein that is essential aberrant epigenetic settings (Figure 2). for lung maturation. Although Hopx lacks a DNA binding domain, it acts downstream of Nkx2-1 and Gata6 to modulate alveolar- specific gene expression in a histone-deacetylase-dependent An expanding network of alveologenic tumor suppressors 111 manner. HOPX exhibits tumor inhibitory effects in a wide range In addition to NKX2-1, several other alveolar specifying TFs can of cancers.112–116 In LUADs, HOPX expression correlates tightly limit LUAD progression. Many of these factors genetically and with NKX2-1,93 and it can restrain tumor cell invasion and physically cooperate with one another to regulate the expression metastasis.45 Moreover, ectopic HOPX expression can induce of prototypical alveolar differentiation markers, including surfac- senescence and is preferentially silenced via DNA methylation in tant proteins. This suggests that the transcriptional network that the LUAD subtype, where decreased HOPX levels correlate with controls alveologenesis is broadly linked to tumor suppression in poor clinical outcome.117 Reducing both HOPX and GATA6 in LUAD the LUAD subtype. cells not only reverses alveolar identity but also activates the The GATA family proteins are DNA binding TFs that can prime expression of squamous basal cell markers and pro-invasive target promoters for repression or activation in specific tissues.94 genes.45 Thus, HOPX and its cooperating TFs may function GATA6 is abundantly expressed in the distal lungs where it is similarly to NKX2-1 by controlling latent differentiation programs required for lung branching morphogenesis and alveolar specifi- that limit overall metastatic competence.118 cation, and it restricts the expansion of adult progenitor cells at CCAAT binding protein-α (C/EBPα) is a family member – the bronchoalveolar duct junction.95 97 Gata6 directly binds to of basic TFs. C/EBPα is required for perinatal AT2 Nkx2-1 and induces the expression of surfactant proteins in AT2 differentiation and surfactant gene expression.119,120 In the adult – – cells.98 100 Similar to NKX2-1, GATA6 has both oncogenic101 105 lungs, loss of C/EBPα impairs epithelial regeneration.121 Although and tumor-suppressive effects106,107 in a variety of cancers. rarely mutated in lung cancer,122 C/EBPα is often suppressed by

Figure 2. Epigenetic and cellular context dictates the effect of lineage TFs in lung cancer. Given a specific cellular origin, lung cancer progression can be driven by repression of a lineage TF and its tumor-suppressive gene targets. Conversely, amplification of a lineage TF may activate oncogenic genes following de novo TF complex formation at novel target promoters. The tumor-suppressive and oncogenic activities may be adopted by the same TF in different epigenetic contexts. These contexts are defined by the overall abundance and availability of TFs and their partnering cofactors. TF thresholds may be dependent on cooperating mutations and different cellular origins (not depicted).

Oncogene (2015) 5771 – 5780 © 2015 Macmillan Publishers Limited Transcription control of lineage states in lung cancer WKC Cheung and DX Nguyen 5775 DNA hypermethylation,123 and its re-expression was shown to to target promoters.154 has also been suggested to broadly inhibit the proliferation and survival of human NSCLC cells.124 amplify gene transcription in cancer cells.155 In resected human In mice, Cebpa is dispensable for spontaneous LUAD initiation, LUADs, MYC overexpression is linked to increased DNA methyla- however, its loss accelerates stress-induced LUAD formation, tion of known tumor suppressor genes.46 Activation of other indicative of a tumor-suppressive role in response to environ- specific C-myc targets in human LUADs correlates with perturba- mental stimuli.125 Further underlying the complexity of respiratory tions in lineage TF networks and poor clinical outcome.45,156,157 TF networks, C/EBPα binds to NKX2-1126 and Cebpa expression is Consistent with these in silico analysis of human tumors, Myc also regulated downstream of Nkx2-1 during development.119 overexpression from SPC+ AT2 progenitors in mice is sufficient to FOXA1 and FOXA2, previously termed hepatocyte nuclear factor induce macrometastases that have undergone a mixed lineage 3α/β, belong to the subfamily of forkhead box TFs. FOXA1/2 have switch.158 Finally, human mutations in the MYC pathway are similar temporal and spatial expression patterns in developing mutually exclusive to SMARCA4 (or BRG1),148,159 which encodes a lungs. With partially overlapping functions, they have critical roles major component of the SWI/SNF chromatin-remodeling in lung morphogenesis and alveolar differentiation.127,128 Both complex160 and is a mediator of SCLC NE differentiation.148 FOXA1/2 are binding partners of NKX2-1 in LUADs,88,129 and they Therefore, despite its multiple functions, the major consequence can activate latent epithelial differentiation genes when NKX2-1 is of MYC activation in lung cancers may be to epigenetically suppressed.88 FOXA2 also cooperates with GATA6 to regulate the reprogram specific lineage states. expression of cell adhesion genes in the developing lung epithelium.130 In both normal epithelial cells and lung cancer LINEAGE PLASTICITY AND METASTASIS: CAUSE OR cells, FOXA2 induces the expression of C/EBPα119 and vice versa.131 CONSEQUENCE? Similar to other alveolar TFs, FOXA2 silencing is observed in LUADs through promoter hypermethylation.132 Finally, overexpression of Epithelial lineage plasticity has been associated with cancer FOXA2 in human NSCLC cells inhibits cell growth, invasion and metastasis. As core mediators of cell fate, lineage TFs may EMT.131,133 therefore regulate genes required for lung cancer cell dissemina- The runt-related TFs (RUNX) are required for the development tion and distant organ colonization. However, considering the of several mammalian tissues and are dysregulated in epithelial diverse functional targets of these TFs, is their capacity to regulate cancers.134 In the murine lungs, Runx3 is expressed by distal metastasis directly connected to the lineage state in a given cell? epithelial cells including AT2 cells, and its targeted loss causes In lung cancer, this remains an open question, but some insight alveolar hyperplasia.135,136 RUNX3 is yet another alveolar gene may be drawn from recent studies. noted to be methylated at a significantly high frequency in lung The overexpression of EMT inducing TFs in epithelial cancer malignancies.137 RUNX3 methylation status is an independent cells is often interpreted as a marker of increased cell invasiveness 161 prognostic factor in NSCLC patients138 and is associated with and metastasis. Overexpression of EMT inducing TFs has been 162 chemoresistance.139 Other modes of RUNX3 loss of function, such observed in various models of metastatic LUAD. However, this as EZH2-mediated H3K27 trimethylation140 and MDM2-mediated is confounded by the fact that EMT promoting TFs also target ubiquitination,141 have been reported in other cancers, but have genes that are independently required for other cellular functions such as cell survival, proliferation, DNA damage responses and yet to be explored in LUAD. In GEMMs, heterozygous loss of Runx3 163,164 causes pre-neoplastic lung adenoma.135,136 Notably, Kras activa- self-renewal. In some carcinoma models, EMT is neither fi tion only results in non-mucinous LUADs, whereas Runx3 loss suf cient nor required for cell invasion, while disseminating cancer cooperates with mutant Kras to initiate both mucinous and non- cells that undergo the reverse process, mesenchymal-to-epithelial mucinous LUADs.142 This further suggests that deregulation of cell transition, have a greater advantage for tumor re-initiation and fate-determining TFs can drive transition to different lung tumor metastatic colonization. Intriguingly, some studies suggest that subtypes. The molecular connection between RUNX3 and other mesenchymal carcinoma cells may not invade beyond the local tumor, but rather enhance the dissemination of other epithelial prototypical alveolar TFs is not yet known, but its ability to 165,166 abrogate the ARF-p53 pathway by interacting with the bromodo- cell populations with tumor re-initiating capacity. An main protein BRD2 implicates an epigenetic modulatory role.142 analogous phenomenon has been described in GEMMs of SCLCs, where a fraction of malignant cells can transition from their NE origins into non-NE cells that express mesenchymal genes.167 MYC revisited Although these non-NE cells are clonally related to the rest of the The MYC family proto-oncogenes, which C-myc (Myc), SCLC, they do not metastasize. On the other hand, their presence N-myc and L-myc, regulate numerous biological processes. in the tumor mass enhances the ability of the NE lineage-positive Although not restricted to pulmonary lineages, the MYC proteins cells to spread. Thus, modulation of lineage states by TFs may are nonetheless potent transcriptional regulators of lung epithelial indirectly influence metastatic potential by controlling intratu- differentiation. N-myc is expressed at high levels in the distal moral clonal dynamics, presumably via paracrine signals (Figure 3). airway epithelium of mice143 and is controlled by Wnt/β-catenin An alternative mechanistic consequence of tumor metaplasia is signaling.52 Loss of N-myc and C-myc can delay lung branching that it may lead to the activation of lineage gene programs that morphogenesis.144,145 C-myc is amplified in human LUAD.46,146 All directly control the metastatic capacity of a given cell. The three MYC members are frequently amplified in SCLC,16,147 but not activation of latent basal epithelial markers, such as cytokeratins in LUSC.24 Moreover, recent genomic studies have identified loss- (K), has been reported in more invasive tumor cells. For example, of-function mutations in MGA and MAX, two genes that encode for cells at the invasive border of luminal breast tumors express the MYC inhibitory proteins.46,148 In human lung cancers, alterations in basal epithelial marker K14 and basal TF p63. These invasive basal MGA, MAX and MYC are mutually exclusive,46,148 indicating a more cells arise from the well-differentiated luminal cells and are pervasive activation of the MYC pathway via mutations than was responsible for collective tumor cell invasion in a manner that is initially appreciated. independent of EMT.168 In liver cancer, the biliary/progenitor In GEMMs, expression of Myc in SPC+ or CC10+ cells induces marker K19 is expressed in a subset of hepatocellular carcinomas LUAD.149,150 In addition, Myc-induced LUAD requires Kras with a high incidence of microvascular invasion and metastasis.169 activating mutation,150 which can be substituted by Notch1 Similarly, LUAD cells may acquire invasion potential by down- overexpression.151 Myc is also required for tumor maintenance in regulating alveolar differentiation genes and upregulating the a variety of Ras pathway-initiated NSCLC models.152,153 MYC can squamous basal cell markers K6A/B.45 Importantly, knockdown of activate or repress specific genes by recruiting different cofactors these cytokeratins in their respective disease models inhibits tumor

© 2015 Macmillan Publishers Limited Oncogene (2015) 5771 – 5780 Transcription control of lineage states in lung cancer WKC Cheung and DX Nguyen 5776

Figure 3. Hypothetical mechanisms by which cell lineage plasticity and heterogeneity promote lung cancer metastasis. (a) Epithelial lineage- positive malignant cells, such as NE cells in SCLC, constitute the majority of distant metastasis, but their dissemination is enhanced via non-cell autonomous signals from mesenchymal transitioned tumor cells in the primary tumor. (b) LUAD cells may disseminate via a classical EMT/ mesenchymal-to-epithelial transition model. (c) Alternatively, LUAD cells may undergo multiple switches in the expression of airway epithelial lineage-specific markers that promote metastasis via cell autonomous mechanisms.

cell invasiveness, suggesting an unexpected direct role for basal/ kinase and BCL-Xl.182 In contrast, LUADs that have undergone biliary epithelial markers in malignant invasion and metastasis. lineage transition become drug refractory, even when those treatments target their driver oncogenes. At the same time, such lineage transitions may have foreseeable shifts in signaling HARNESSING LINEAGE STATES FOR THERAPY dependencies. For example, the survival of tyrosine kinase The TFs discussed here are but a few examples linking the inhibitor-resistant EGFR mutant tumor cells, which are more molecular control of cell lineage fate to lung cancer progression. mesenchymal like, requires activation of the receptor tyrosine fi Despite the broad biological signi cance of these observations, kinase AXL,34,183 which can be inhibited with small molecules. their clinical and therapeutic impacts remain underexplored. Part of Given the fact that lung cancers can adopt various differentia- the reason for this is the intrinsic technical barriers in pharmaco- tion states, new biological insight may be revealed by compre- logically manipulating pathways or TFs that are not only tissue fi fi fi hensively de ning their underlying transcriptional circuits. These speci c but also likely to have dosage- and stage-speci c effects circuits may in turn modify the oncogenic dependencies of a during tumor progression. Nevertheless, exploiting aberrant tumor given tumor, providing novel therapeutic opportunities. Major differentiation states or lineage TFs for therapy has been shown to challenges remain in directly inducing or reverting these lineage- be clinically beneficial in leukemia and neuroblastoma170 and pre- specific pathways at different stages of disease progression. clinically feasible in melanoma,171,172 glioma,173,174 nasopharyngeal 175 176 Nevertheless, several lung cancer differentiation states, defined carcinoma and rhabdomyosarcoma. Driving the tumors toward more differentiated states may not only delay their growth either transcriptionally or histologically, may already be used as but also potentially increase their sensitivity to commonly used both prognostic and predictive biomarkers. Moving forward, a fi cytotoxic agents. systematic search for ef cacious agents tailored not only to the Several novel compounds have recently been explored to oncogenic drivers of various lung cancer subtypes but also to target the expression or activity of TFs. For example, drugs the cell lineage of their malignant compartment, holds the that inhibit the chromatin-remodeling bromodomain and promise of abating metastatic progression. extraterminal proteins are being actively investigated to target the lineage-dependent activity of TFs in multiple cancers, including LUAD.177,178 Another potential approach is to use covalent inhibitors of cyclin-dependent kinase 7, which preferen- CONFLICT OF INTEREST tially mediates the expression of super-enhancer associated The authors declare no conflict of interest. genes.179,180 Because such genes include oncogenic MYC and NE TFs, this class of transcription targeting drugs may be particularly effective against SCLC.181 Assessing the lineage state of the bulk tumor may have more ACKNOWLEDGEMENTS fi immediate clinical bene ts for anticipating dynamic therapeutic We thank all members of the Nguyen laboratory for their discussions and critical responses. This is relevant in the treatment of KRAS mutant LUADs, reviews of the manuscript. We apologize for omitting some examples and primary which are highly heterogeneous. Notably, a subset of KRAS mutant references due to space constraints. Our research is funded by grants from Uniting LUADs that are well differentiated and retain epithelial markers Against Lung Cancer (to WKCC) and the National Cancer Institute (1R01CA166376, respond specifically to a combination of drugs that inhibit MAP 1R21CA170537 and 1R01CA191489 to DXN).

Oncogene (2015) 5771 – 5780 © 2015 Macmillan Publishers Limited Transcription control of lineage states in lung cancer WKC Cheung and DX Nguyen 5777 REFERENCES 31 Politi K, Zakowski MF, Fan PD, Schonfeld EA, Pao W, Varmus HE. Lung 1 Travis WD, Brambilla E, Riely GJ. New pathologic classification of lung cancer: adenocarcinomas induced in mice by mutant EGF receptors found in human relevance for clinical practice and clinical trials. J Clin Oncol 2013; 31: lung cancers respond to a tyrosine kinase inhibitor or to down-regulation of the 992–1001. receptors. Genes Dev 2006; 20: 1496–1510. 2 Siegel R, Ma J, Zou Z, Jemal A. Cancer statistics, 2014. CA Cancer J Clin 2014; 32 Curtis SJ, Sinkevicius KW, Li D, Lau AN, Roach RR, Zamponi R et al. Primary tumor 64:9–29. genotype is an important determinant in identification of lung cancer 3 Morrisey EE, Hogan BL. Preparing for the first breath: genetic and cellular propagating cells. Cell Stem Cell 2010; 7:127–133. mechanisms in lung development. Dev Cell 2010; 18:8–23. 33 Tsai JH, Yang J. Epithelial-mesenchymal plasticity in carcinoma metastasis. Genes 4 Desai TJ, Brownfield DG, Krasnow MA. Alveolar progenitor and stem cells in lung Dev 2013; 27: 2192–2206. development, renewal and cancer. Nature 2014; 507:190–194. 34 Zhang Z, Lee JC, Lin L, Olivas V, Au V, LaFramboise T et al. Activation of the AXL 5 Treutlein B, Brownfield DG, Wu AR, Neff NF, Mantalas GL, Espinoza FH et al. kinase causes resistance to EGFR-targeted therapy in lung cancer. Nat Genet Reconstructing lineage hierarchies of the distal lung epithelium using single-cell 2012; 44:852–860. RNA-seq. Nature 2014; 509:371–375. 35 Sequist LV, Waltman BA, Dias-Santagata D, Digumarthy S, Turke AB, Fidias P et al. 6 Barkauskas CE, Cronce MJ, Rackley CR, Bowie EJ, Keene DR, Stripp BR et al. Genotypic and histological evolution of lung cancers acquiring resistance to Type 2 alveolar cells are stem cells in adult lung. J Clin Invest 2013; 123: EGFR inhibitors. Sci Transl Med 2011; 3: 75ra26. 3025–3036. 36 Uramoto H, Shimokawa H, Hanagiri T, Kuwano M, Ono M. Expression of selected 7 Zuo W, Zhang T, Wu DZ, Guan SP, Liew AA, Yamamoto Y et al. p63(+)Krt5(+) gene for acquired drug resistance to EGFR-TKI in lung adenocarcinoma. Lung distal airway stem cells are essential for lung regeneration. Nature 2015; 517: Cancer 2011; 73:361–365. 616–620. 37 Borczuk AC, Gorenstein L, Walter KL, Assaad AA, Wang L, Powell CA. Non-small- 8 Vaughan AE, Brumwell AN, Xi Y, Gotts JE, Brownfield DG, Treutlein B et al. cell lung cancer molecular signatures recapitulate lung developmental Lineage-negative progenitors mobilize to regenerate lung epithelium after pathways. Am J Pathol 2003; 163:1949–1960. major injury. Nature 2015; 517: 621–625. 38 Watson WL, Berg JW. Oat cell lung cancer. Cancer 1962; 15:759–768. 9 Meuwissen R, Berns A. Mouse models for human lung cancer. Genes Dev 2005; 39 Alam N, Gustafson KS, Ladanyi M, Zakowski MF, Kapoor A, Truskinovsky AM et al. 19: 643–664. Small-cell carcinoma with an epidermal growth factor receptor mutation in a 10 Rawlins EL, Perl AK. The a'MAZE"ing world of lung-specific transgenic mice. Am J never-smoker with gefitinib-responsive adenocarcinoma of the lung. Clin Lung Respir Cell Mol Biol 2012; 46: 269–282. Cancer 2010; 11:E1–E4. 11 Blanpain C. Tracing the cellular origin of cancer. Nat Cell Biol 2013; 15:126–134. 40 Popat S, Wotherspoon A, Nutting CM, Gonzalez D, Nicholson AG, O'Brien M. 12 Hanna JM, Onaitis MW. Cell of origin of lung cancer. J Carcinog 2013; 12:6. Transformation to "high grade" neuroendocrine carcinoma as an acquired drug 13 Sutherland KD, Berns A. Cell of origin of lung cancer. Mol Oncol 2010; 4:397–403. resistance mechanism in EGFR-mutant lung adenocarcinoma. Lung Cancer 2013; 14 Chen Z, Fillmore CM, Hammerman PS, Kim CF, Wong KK. Non-small-cell lung 80:1–4. cancers: a heterogeneous set of diseases. Nat Rev Cancer 2014; 14:535–546. 41 Watanabe S, Sone T, Matsui T, Yamamura K, Tani M, Okazaki A et al. Transfor- 15 van Meerbeeck JP, Fennell DA, De Ruysscher DK. Small-cell lung cancer. Lancet mation to small-cell lung cancer following treatment with EGFR tyrosine kinase 2011; 378: 1741–1755. inhibitors in a patient with lung adenocarcinoma. Lung Cancer 2013; 82: 16 Peifer M, Fernández-Cuesta L, Sos ML, George J, Seidel D, Kasper LH et al. 370–372. Integrative genome analyses identify key somatic driver mutations of small-cell 42 Filosso PL, Ruffini E, Asioli S, Giobbe R, Macri L, Bruna MC et al. Adenosquamous lung cancer. Nat Genet 2012; 44: 1104–1110. lung carcinomas: a histologic subtype with poor prognosis. Lung Cancer 2011; 17 Meuwissen R, Linn SC, Linnoila RI, Zevenhoven J, Mooi WJ, Berns A. Induction of 74:25–29. small cell lung cancer by somatic inactivation of both Trp53 and Rb1 in a 43 Cooke DT, Nguyen DV, Yang Y, Chen SL, Yu C, Calhoun RF. Survival comparison conditional mouse model. Cancer Cell 2003; 4:181–189. of adenosquamous, squamous cell, and adenocarcinoma of the lung after 18 Sutherland KD, Proost N, Brouns I, Adriaensen D, Song JY, Berns A. Cell of origin lobectomy. Ann Thorac Surg 2010; 90: 943–948. of small cell lung cancer: inactivation of Trp53 and Rb1 in distinct cell types of 44 Nakagawa K, Yasumitu T, Fukuhara K, Shiono H, Kikui M. Poor prognosis after adult mouse lung. Cancer Cell 2011; 19: 754–764. lung resection for patients with adenosquamous carcinoma of the lung. 19 Park KS, Liang MC, Raiser DM, Zamponi R, Roach RR, Curtis SJ et al. Characterization Ann Thorac Surg 2003; 75: 1740–1744. of the cell of origin for small cell lung cancer. Cell Cycle 2011; 10:2806–2815. 45 Cheung WK, Zhao M, Liu Z, Stevens LE, Cao PD, Fang JE et al. Control of 20 Song H, Yao E, Lin C, Gacayan R, Chen MH, Chuang PT. Functional character- alveolar differentiation by the lineage transcription factors GATA6 and ization of pulmonary neuroendocrine cells in lung development, injury, and HOPX inhibits lung adenocarcinoma metastasis. Cancer Cell 2013; 23: tumorigenesis. Proc Natl Acad Sci USA 2012; 109: 17531–17536. 725–738. 21 McFadden DG, Papagiannakopoulos T, Taylor-Weiner A, Stewart C, Carter SL, 46 Cancer Genome Atlas Research N. Comprehensive molecular profiling of lung Cibulskis K et al. Genetic and clonal dissection of murine small cell lung adenocarcinoma. Nature 2014; 511:543–550. carcinoma progression by genome sequencing. Cell 2014; 156: 1298–1311. 47 Han X, Li F, Fang Z, Gao Y, Li F, Fang R et al. Transdifferentiation of lung 22 Ji H, Ramsey MR, Hayes DN, Fan C, McNamara K, Kozlowski P et al. LKB1 mod- adenocarcinoma in mice with Lkb1 deficiency to squamous cell carcinoma. ulates lung cancer differentiation and metastasis. Nature 2007; 448:807–810. Nat Commun 2014; 5: 3261. 23 Xiao Z, Jiang Q, Willette-Brown J, Xi S, Zhu F, Burkett S et al. The pivotal role of 48 Gao Y, Zhang W, Han X, Li F, Wang X, Wang R et al. YAP inhibits squamous IKKalpha in the development of spontaneous lung squamous cell carcinomas. transdifferentiation of Lkb1-deficient lung adenocarcinoma through ZEB2- Cancer Cell 2013; 23:527–540. dependent DNp63 repression. Nat Commun 2014; 5: 4629. 24 Cancer Genome Atlas Research N. Comprehensive genomic characterization of 49 Ischenko I, Liu J, Petrenko O, Hayman MJ. Transforming growth factor-beta squamous cell lung cancers. Nature 2012; 489:519–525. signaling network regulates plasticity and lineage commitment of lung 25 Xu C, Fillmore CM, Koyama S, Wu H, Zhao Y, Chen Z et al. Loss of Lkb1 and Pten cancer cells. Cell Death Differ 2014; 21: 1218–1228. leads to lung squamous cell carcinoma with elevated PD-L1 expression. Cancer 50 Goss AM, Tian Y, Tsukiyama T, Cohen ED, Zhou D, Lu MM et al. Wnt2/2b and Cell 2014; 25: 590–604. beta-catenin signaling are necessary and sufficient to specify lung progenitors in 26 Quinlan MP, Quatela SE, Philips MR, Settleman J. Activated Kras, but not Hras or the foregut. Dev Cell 2009; 17:290–298. Nras, may initiate tumors of endodermal origin via stem cell expansion. Mol Cell 51 Mucenski ML, Wert SE, Nation JM, Loudy DE, Huelsken J, Birchmeier W et al. Biol 2008; 28: 2659–2674. beta-Catenin is required for specification of proximal/distal cell fate during lung 27 Mainardi S, Mijimolle N, Francoz S, Vicente-Dueñas C, Sánchez-García I, Barbacid M. morphogenesis. J Biol Chem 2003; 278: 40231–40238. Identification of cancer initiating cells in K-Ras driven lung adenocarcinoma. Proc 52 Shu W, Guttentag S, Wang Z, Andl T, Ballard P, Lu MM et al. Wnt/beta-catenin Natl Acad Sci USA 2014; 111:255–260. signaling acts upstream of N-myc, BMP4, and FGF signaling to regulate 28 Xu X, Rock JR, Lu Y, Futtner C, Schwab B, Guinney J et al. Evidence for type II cells proximal-distal patterning in the lung. Dev Biol 2005; 283: 226–239. as cells of origin of K-Ras-induced distal lung adenocarcinoma. Proc Natl Acad Sci 53 Ding L, Getz G, Wheeler DA, Mardis ER, McLellan MD, Cibulskis K et al. Somatic USA 2012; 109: 4910–4915. mutations affect key pathways in lung adenocarcinoma. Nature 2008; 455: 29 Lin C, Song H, Huang C, Yao E, Gacayan R, Xu SM et al. Alveolar type II cells 1069–1075. possess the capability of initiating lung tumor development. PLoS One 2012; 7: 54 Stewart DJ. Wnt signaling pathway in non-small cell lung cancer. J Natl Cancer e53817. Inst 2014; 106: djt356. 30 Sutherland KD, Song JY, Kwon MC, Proost N, Zevenhoven J, Berns A. Multiple 55 Reynolds SD, Zemke AC, Giangreco A, Brockway BL, Teisanu RM, Drake JA et al. cells-of-origin of mutant K-Ras-induced mouse lung adenocarcinoma. Proc Natl Conditional stabilization of beta-catenin expands the pool of lung stem cells. Acad Sci USA 2014; 111: 4952–4957. Stem Cells 2008; 26: 1337–1346.

© 2015 Macmillan Publishers Limited Oncogene (2015) 5771 – 5780 Transcription control of lineage states in lung cancer WKC Cheung and DX Nguyen 5778 56 Pacheco-Pinedo EC, Durham AC, Stewart KM, Goss AM, Lu MM, Demayo FJ et al. 82 Tang X, Kadara H, Behrens C, Liu DD, Xiao Y, Rice D et al. Abnormalities of the Wnt/β-catenin signaling accelerates mouse lung tumorigenesis by imposing an TITF-1 lineage-specific oncogene in NSCLC: implications in lung cancer patho- embryonic distal progenitor phenotype on lung epithelium. J Clin Invest 2011; genesis and prognosis. Clin Cancer Res 2011; 17: 2434–2443. 121: 1935–1945. 83 Tanaka H, Yanagisawa K, Shinjo K, Taguchi A, Maeno K, Tomida S et al. 57 Sánchez-Rivera FJ, Papagiannakopoulos T, Romero R, Tammela T, Bauer MR, Lineage-specific dependency of lung adenocarcinomas on the lung development Bhutkar A et al. Rapid modelling of cooperating genetic events in cancer regulator TTF-1. Cancer Res 2007; 67: 6007–6011. through somatic genome editing. Nature 2014; 516: 428–431. 84 Yamaguchi T, Yanagisawa K, Sugiyama R, Hosono Y, Shimada Y, Arima C et al. 58 Nguyen DX, Chiang AC, Zhang XH, Kim JY, Kris MG, Ladanyi M et al. WNT/TCF NKX2-1/TITF1/TTF-1-Induced ROR1 is required to sustain EGFR survival signaling signaling through LEF1 and HOXB9 mediates lung adenocarcinoma metastasis. in lung adenocarcinoma. Cancer Cell 2012; 21:348–361. Cell 2009; 138:51–62. 85 Maeda Y, Tsuchiya T, Hao H, Tompkins DH, Xu Y, Mucenski ML et al. Kras(G12D) 59 Bleckmann A, Siam L, Klemm F, Rietkotter E, Wegner C, Kramer F et al. Nuclear and Nkx2-1 haploinsufficiency induce mucinous adenocarcinoma of the lung. LEF1/TCF4 correlate with poor prognosis but not with nuclear beta-catenin in J Clin Invest 2012; 122: 4388–4400. cerebral metastasis of lung adenocarcinomas. Clin Exp Metastasis 2013; 30: 86 Winslow MM, Dayton TL, Verhaak RG, Kim-Kiselak C, Snyder EL, Feldser DM et al. 471–482. Suppression of lung adenocarcinoma progression by Nkx2-1. Nature 2011; 473: 60 Maeda Y, Dave V, Whitsett JA. Transcriptional control of lung morphogenesis. 101–104. Physiol Rev 2007; 87:219–244. 87 Berghmans T, Paesmans M, Mascaux C, Martin B, Meert AP, Haller A et al. Thyroid 61 Burkhart DL, Sage J. Cellular mechanisms of tumour suppression by the 1--a new prognostic factor in lung cancer: a meta-analysis. retinoblastoma gene. Nat Rev Cancer 2008; 8: 671–682. Ann Oncol 2006; 17:1673–1676. 62 Wikenheiser-Brokamp KA. Rb family proteins differentially regulate distinct 88 Snyder EL, Watanabe H, Magendantz M, Hoersch S, Chen TA, Wang DG et al. cell lineages during epithelial development. Development 2004; 131: Nkx2-1 represses a latent gastric differentiation program in lung adenocarci- 4299–4310. noma. Mol Cell 2013; 50:185–199. 63 Bass AJ, Watanabe H, Mermel CH, Yu S, Perner S, Verhaak RG et al. SOX2 is an 89 Yamaguchi T, Hosono Y, Yanagisawa K, Takahashi T. NKX2-1/TTF-1: an enigmatic amplified lineage-survival oncogene in lung and esophageal squamous cell oncogene that functions as a double-edged sword for cancer cell survival and carcinomas. Nat Genet 2009; 41: 1238–1242. progression. Cancer Cell 2013; 23: 718–723. 64 Hussenet T, Dali S, Exinger J, Monga B, Jost B, Dembele D et al. SOX2 is 90 Kim IJ, Quigley D, To MD, Pham P, Lin K, Jo B et al. Rewiring of human lung cell an oncogene activated by recurrent 3q26.3 amplifications in human lung lineage and mitotic networks in lung adenocarcinomas. Nat Commun 2013; 4: squamous cell carcinomas. PLoS One 2010; 5: e8960. 1701. 65 Lu Y, Futtner C, Rock JR, Xu X, Whitworth W, Hogan BL et al. Evidence that SOX2 91 Saito RA, Watabe T, Horiguchi K, Kohyama T, Saitoh M, Nagase T et al. Thyroid overexpression is oncogenic in the lung. PLoS One 2010; 5: e11022. transcription factor-1 inhibits transforming growth factor-beta-mediated 66 Que J, Luo X, Schwartz RJ, Hogan BL. Multiple roles for Sox2 in the developing epithelial-to-mesenchymal transition in lung adenocarcinoma cells. Cancer Res and adult mouse trachea. Development 2009; 136: 1899–1907. 2009; 69:2783–2791. 67 Mukhopadhyay A, Berrett KC, Kc U, Clair PM, Pop SM, Carr SR et al. Sox2 92 Hosono Y, Yamaguchi T, Mizutani E, Yanagisawa K, Arima C, Tomida S et al. cooperates with Lkb1 loss in a mouse model of squamous cell lung cancer. MYBPH, a transcriptional target of TTF-1, inhibits ROCK1, and reduces cell Cell Rep 2014; 8:40–49. motility and metastasis. EMBO J 2012; 31:481–493. 68 Watanabe H, Ma Q, Peng S, Adelmant G, Swain D, Song W et al. SOX2 and p63 93 Watanabe H, Francis JM, Woo MS, Etemad B, Lin W, Fries DF et al. Integrated colocalize at genetic loci in squamous cell carcinomas. J Clin Invest 2014; 124: cistromic and expression analysis of amplified NKX2-1 in lung adenocarcinoma 1636–1645. identifies LMO3 as a functional transcriptional target. Genes Dev 2013; 27: 69 Justilien V, Walsh MP, Ali SA, Thompson EA, Murray NR, Fields AP. The 197–210. PRKCI and SOX2 oncogenes are coamplified and cooperate to activate 94 Molkentin JD. The zinc finger-containing transcription factors GATA-4, -5, and -6. Hedgehog signaling in lung squamous cell carcinoma. Cancer Cell 2014; 25: Ubiquitously expressed regulators of tissue-specific gene expression. J Biol Chem 139–151. 2000; 275: 38949–38952. 70 Xu X, Huang L, Futtner C, Schwab B, Rampersad RR, Lu Y et al. The cell of origin 95 Zhang Y, Goss AM, Cohen ED, Kadzik R, Lepore JJ, Muthukumaraswamy K et al. and subtype of K-Ras-induced lung tumors are modified by Notch and Sox2. A Gata6-Wnt pathway required for epithelial stem cell development and airway Genes Dev 2014; 28: 1929–1939. regeneration. Nat Genet 2008; 40: 862–870. 71 Chang DR, Martinez Alanis D, Miller RK, Ji H, Akiyama H, McCrea PD et al. Lung 96 Yang H, Lu MM, Zhang L, Whitsett JA, Morrisey EE. GATA6 epithelial branching program antagonizes alveolar differentiation. Proc Natl Acad regulates differentiation of distal lung epithelium. Development 2002; 129: Sci USA 2013; 110: 18042–18051. 2233–2246. 72 Rockich BE, Hrycaj SM, Shih HP, Nagy MS, Ferguson MA, Kopp JL et al. Sox9 plays 97 Keijzer R, van Tuyl M, Meijers C, Post M, Tibboel D, Grosveld F et al. The multiple roles in the lung epithelium during branching morphogenesis. Proc Natl transcription factor GATA6 is essential for branching morphogenesis and Acad Sci USA 2013; 110: E4456–E4464. epithelial cell differentiation during fetal pulmonary development. Development 73 Zhou CH, Ye LP, Ye SX, Li Y, Zhang XY, Xu XY et al. Clinical significance of SOX9 in 2001; 128: 503–511. human non-small cell lung cancer progression and overall patient survival. J Exp 98 Shaw-White JR, Bruno MD, Whitsett JA. GATA-6 activates transcription of thyroid Clin Cancer Res 2012; 31: 18. transcription factor-1. J Biol Chem 1999; 274: 2658–2664. 74 Jiang SS, Fang WT, Hou YH, Huang SF, Yen BL, Chang JL et al. Upregulation of 99 Liu C, Glasser SW, Wan H, Whitsett JA. GATA-6 and thyroid transcription factor-1 SOX9 in lung adenocarcinoma and its involvement in the regulation of cell directly interact and regulate surfactant protein-C gene expression. J Biol Chem growth and tumorigenicity. Clin Cancer Res 2010; 16:4363–4373. 2002; 277: 4519–4525. 75 Capaccione KM, Hong X, Morgan KM, Liu W, Bishop JM, Liu L et al. Sox9 mediates 100 Bruno MD, Korfhagen TR, Liu C, Morrisey EE, Whitsett JA. GATA-6 activates Notch1-induced mesenchymal features in lung adenocarcinoma. Oncotarget transcription of surfactant protein A. J Biol Chem 2000; 275: 1043–1049. 2014; 5: 3636–3650. 101 Kwei KA, Bashyam MD, Kao J, Ratheesh R, Reddy EC, Kim YH et al. 76 Ikeda K, Clark JC, Shaw-White JR, Stahlman MT, Boutell CJ, Whitsett JA. Gene Genomic profiling identifies GATA6 as a candidate oncogene amplified in structure and expression of human thyroid transcription factor-1 in respiratory pancreatobiliary cancer. PLoS Genet 2008; 4: e1000081. epithelial cells. J Biol Chem 1995; 270: 8108–8114. 102 Zhong Y, Wang Z, Fu B, Pan F, Yachida S, Dhara M et al. GATA6 activates Wnt 77 Minoo P, Hamdan H, Bu D, Warburton D, Stepanik P, deLemos R. TTF-1 regulates signaling in pancreatic cancer by negatively regulating the Wnt antagonist lung epithelial morphogenesis. Dev Biol 1995; 172:694–698. Dickkopf-1. PLoS One 2011; 6: e22129. 78 Weir BA, Woo MS, Getz G, Perner S, Ding L, Beroukhim R et al. Characterizing the 103 Lin L, Bass AJ, Lockwood WW, Wang Z, Silvers AL, Thomas DG et al. cancer genome in lung adenocarcinoma. Nature 2007; 450:893–898. Activation of GATA binding protein 6 (GATA6) sustains oncogenic lineage- 79 Kendall J, Liu Q, Bakleh A, Krasnitz A, Nguyen KC, Lakshmi B et al. Oncogenic survival in ;esophageal adenocarcinoma. Proc Natl Acad Sci USA 2012; 109: cooperation and coamplification of developmental transcription factor genes in 4251–4256. lung cancer. Proc Natl Acad Sci USA 2007; 104: 16663–16668. 104 Chia NY, Deng N, Das K, Huang D, Hu L, Zhu Y et al. Regulatory crosstalk between 80 Kwei KA, Kim YH, Girard L, Kao J, Pacyna-Gengelbach M, Salari K et al. Genomic lineage-survival oncogenes KLF5, GATA4 and GATA6 cooperatively promotes profiling identifies TITF1 as a lineage-specific oncogene amplified in lung cancer. gastric cancer development. Gut 2014; e-pub ahead of print 22 July 2014; Oncogene 2008; 27: 3635–3640. doi:10.1136/gutjnl-2013-306596. 81 Barletta JA, Perner S, Iafrate AJ, Yeap BY, Weir BA, Johnson LA et al. Clinical 105 Belaguli NS, Aftab M, Rigi M, Zhang M, Albo D, Berger DH. GATA6 promotes significance of TTF-1 protein expression and TTF-1 gene amplification in lung colon cancer cell invasion by regulating urokinase plasminogen activator gene adenocarcinoma. J Cell Mol Med 2009; 13: 1977–1986. expression. Neoplasia 2010; 12: 856–865.

Oncogene (2015) 5771 – 5780 © 2015 Macmillan Publishers Limited Transcription control of lineage states in lung cancer WKC Cheung and DX Nguyen 5779 106 Capo-chichi CD, Roland IH, Vanderveer L, Bao R, Yamagata T, Hirai H et al. 130 Jimenez FR, Lewis JB, Belgique ST, Wood TT, Reynolds PR. Developmental lung Anomalous expression of epithelial differentiation-determining GATA factors in expression and transcriptional regulation of claudin-6 by TTF-1, Gata-6, ovarian tumorigenesis. Cancer Res 2003; 63: 4967–4977. and FoxA2. Respir Res 2014; 15:70. 107 Kamnasaran D, Qian B, Hawkins C, Stanford WL, Guha A. GATA6 is an 131 Halmos B, Bassères DS, Monti S, D'Aló F, Dayaram T, Ferenczi K et al. astrocytoma tumor suppressor gene identified by gene trapping of mouse A transcriptional profiling study of CCAAT/enhancer binding protein targets glioma model. Proc Natl Acad Sci USA 2007; 104: 8053–8058. identifies hepatocyte nuclear factor 3 beta as a novel tumor suppressor in 108 Yang Y, Ahn YH, Gibbons DL, Zang Y, Lin W, Thilaganathan N et al. The Notch lung cancer. Cancer Res 2004; 64: 4137–4147. ligand Jagged2 promotes lung adenocarcinoma metastasis through a miR-200- 132 Basseres DS, D'Alò F, Yeap BY, Löwenberg EC, Gonzalez DA, Yasuda H et al. dependent pathway in mice. J Clin Invest 2011; 121: 1373–1385. Frequent downregulation of the transcription factor Foxa2 in lung cancer 109 Guo M, Akiyama Y, House MG, Hooker CM, Heath E, Gabrielson E et al. Hyper- through epigenetic silencing. Lung Cancer 2012; 77:31–37. methylation of the GATA genes in lung cancer. Clin Cancer Res 2004; 10: 133 Tang Y, Shu G, Yuan X, Jing N, Song J. FOXA2 functions as a suppressor of tumor 7917–7924. metastasis by inhibition of epithelial-to-mesenchymal transition in human lung 110 Caslini C, Capo-chichi CD, Roland IH, Nicolas E, Yeung AT, Xu XX. Histone cancers. Cell Res 2011; 21: 316–326. modifications silence the GATA transcription factor genes in ovarian cancer. 134 Ito Y, Bae SC, Chuang LS. The RUNX family: developmental regulators in cancer. Oncogene 2006; 25: 5446–5461. Nat Rev Cancer 2015; 15:81–95. 111 Yin Z, Gonzales L, Kolla V, Rath N, Zhang Y, Lu MM et al. Hop functions down- 135 Lee KS, Lee YS, Lee JM, Ito K, Cinghu S, Kim JH et al. Runx3 is required for the stream of Nkx2.1 and GATA6 to mediate HDAC-dependent negative regulation differentiation of lung epithelial cells and suppression of lung cancer. Oncogene of pulmonary gene expression. Am J Physiol Lung Cell Mol Physiol 2006; 291: 2010; 29: 3349–3361. L191–L199. 136 Lee JM, Shin JO, Cho KW, Hosoya A, Cho SW, Lee YS et al. Runx3 is a crucial 112 Yamashita K, Kim MS, Park HL, Tokumaru Y, Osada M, Inoue H et al. HOP/ regulator of alveolar differentiation and lung tumorigenesis in mice. Differ- OB1/NECC1 promoter DNA is frequently hypermethylated and involved in entiation 2011; 81:261–268. tumorigenic ability in esophageal squamous cell carcinoma. Mol Cancer Res 137 Liang Y, He L, Yuan H, Jin Y, Yao Y. Association between RUNX3 promoter 2008; 6:31–41. methylation and non-small cell lung cancer: a meta-analysis. J Thorac Dis 2014; 6: 113 Yamaguchi S, Asanoma K, Takao T, Kato K, Wake N. Homeobox gene HOPX is 694–705. epigenetically silenced in human uterine endometrial cancer and suppresses 138 Yanagawa N, Tamura G, Oizumi H, Kanauchi N, Endoh M, Sadahiro M et al. estrogen-stimulated proliferation of cancer cells by inhibiting serum Promoter hypermethylation of RASSF1A and RUNX3 genes as an independent response factor. Int J Cancer 2009; 124: 2577–2588. prognostic prediction marker in surgically resected non-small cell lung cancers. 114 Waraya M, Yamashita K, Katoh H, Ooki A, Kawamata H, Nishimiya H et al. Cancer Lung Cancer 2007; 58: 131–138. specific promoter CpG Islands hypermethylation of HOP homeobox (HOPX) 139 Zheng Y, Wang R, Song HZ, Pan BZ, Zhang YW, Chen LB. Epigenetic gene and its potential tumor suppressive role in pancreatic carcinogenesis. downregulation of RUNX3 by DNA methylation induces docetaxel chemoresistance BMC Cancer 2012; 12: 397. in human lung adenocarcinoma cells by activation of the AKT pathway. Int J 115 Ooki A, Yamashita K, Kikuchi S, Sakuramoto S, Katada N, Kokubo K et al. Potential Biochem Cell Biol 2013; 45: 2369–2378. utility of HOP homeobox gene promoter methylation as a marker of tumor 140 Fujii S, Ito K, Ito Y, Ochiai A. Enhancer of zeste homologue 2 (EZH2) down- aggressiveness in gastric cancer. Oncogene 2010; 29: 3263–3275. regulates RUNX3 by increasing histone H3 methylation. J Biol Chem 2008; 283: 116 Katoh H, Yamashita K, Waraya M, Margalit O, Ooki A, Tamaki H et al. Epigenetic 17324–17332. silencing of HOPX promotes cancer progression in colorectal cancer. Neoplasia 141 Chi XZ, Kim J, Lee YH, Lee JW, Lee KS, Wee H et al. Runt-related transcription 2012; 14: 559–571. factor RUNX3 is a target of MDM2-mediated ubiquitination. Cancer Res 2009; 69: 117 Chen Y, Yang L, Cui T, Pacyna-Gengelbach M, Petersen I. HOPX is methylated 8111–8119. and exerts tumour suppressive function through Ras-induced senescence in 142 Lee YS, Lee JW, Jang JW, Chi XZ, Kim JH, Li YH et al. Runx3 inactivation is a crucial human lung cancer. J Pathol 2015; 235:397–407. early event in the development of lung adenocarcinoma. Cancer Cell 2013; 24: 118 Watanabe H, Meyerson M. Hopping between differentiation states in lung 603–616. adenocarcinoma. Cancer Cell 2013; 23:707–709. 143 Okubo T, Knoepfler PS, Eisenman RN, Hogan BL. Nmyc plays an essential role 119 Martis PC, Whitsett JA, Xu Y, Perl AK, Wan H, Ikegami M. C/EBPalpha is required during lung development as a dosage-sensitive regulator of progenitor cell for lung maturation at birth. Development 2006; 133:1155–1164. proliferation and differentiation. Development 2005; 132: 1363–1374. 120 Basseres DS, Levantini E, Ji H, Monti S, Elf S, Dayaram T et al. Respiratory failure 144 Moens CB, Auerbach AB, Conlon RA, Joyner AL, Rossant J. A targeted mutation due to differentiation arrest and expansion of alveolar cells following lung- reveals a role for N-myc in branching morphogenesis in the embryonic specific loss of the transcription factor C/EBPalpha in mice. Mol Cell Biol 2006; 26: mouse lung. Genes Dev 1992; 6:691–704. 1109–1123. 145 Cohen JC, Scott DK, Miller J, Zhang J, Zhou P, Larson JE. Transient in utero 121 Sato A, Xu Y, Whitsett JA, Ikegami M. CCAAT/enhancer binding protein-α knockout (TIUKO) of C-MYC affects late lung and intestinal development in regulates the protease/antiprotease balance required for bronchiolar epithelium the mouse. BMC Dev Biol 2004; 4:4. regeneration. Am J Respir Cell Mol Biol 2012; 47:454–463. 146 Iwakawa R, Kohno T, Kato M, Shiraishi K, Tsuta K, Noguchi M et al. MYC 122 Costa DB, Dayaram T, D'Alo F, Wouters BJ, Tenen DG, Meyerson M et al. C/EBP amplification as a prognostic marker of early-stage lung adenocarcinoma alpha mutations in lung cancer. Lung Cancer 2006; 53:253–254. identified by whole genome copy number analysis. Clin Cancer Res 2011; 17: 123 Tada Y, Brena RM, Hackanson B, Morrison C, Otterson GA, Plass C. Epigenetic 1481–1489. modulation of tumor suppressor CCAAT/enhancer binding protein alpha activity 147 Iwakawa R, Takenaka M, Kohno T, Shimada Y, Totoki Y, Shibata T et al. Genome- in lung cancer. J Natl Cancer Inst 2006; 98:396–406. wide identification of genes with amplification and/or fusion in small cell 124 Halmos B, Huettner CS, Kocher O, Ferenczi K, Karp DD, Tenen DG. lung cancer. Genes Cancer 2013; 52:802–816. Down-regulation and antiproliferative role of C/EBPalpha in lung cancer. Cancer 148 Romero OA, Torres-Diz M, Pros E, Savola S, Gomez A, Moran S et al. MAX Res 2002; 62: 528–534. inactivation in small cell lung cancer disrupts MYC-SWI/SNF programs and is 125 Sato A, Yamada N, Ogawa Y, Ikegami M. CCAAT/enhancer-binding protein-α synthetic lethal with BRG1. Cancer Discov 2014; 4: 292–303. suppresses lung tumor development in mice through the p38α MAP kinase 149 Tran PT, Fan AC, Bendapudi PK, Koh S, Komatsubara K, Chen J et al. Combined pathway. PLoS One 2013; 8: e57013. Inactivation of MYC and K-Ras oncogenes reverses tumorigenesis in lung 126 Tomita T, Kido T, Kurotani R, Iemura S, Sterneck E, Natsume T et al. CAATT/ adenocarcinomas and lymphomas. PLoS One 2008; 3: e2125. enhancer-binding proteins alpha and delta interact with NKX2-1 to synergisti- 150 Allen TD, Zhu CQ, Jones KD, Yanagawa N, Tsao MS, Bishop JM. Interaction cally activate mouse secretoglobin 3A2 gene expression. J Biol Chem 2008; 283: between MYC and MCL1 in the genesis and outcome of non-small-cell 25617–25627. lung cancer. Cancer Res 2011; 71: 2212–2221. 127 Wan H, Kaestner KH, Ang SL, Ikegami M, Finkelman FD, Stahlman MT et al. Foxa2 151 Allen TD, Rodriguez EM, Jones KD, Bishop JM. Activated Notch1 induces regulates alveolarization and goblet cell hyperplasia. Development 2004; 131: lung adenomas in mice and cooperates with Myc in the generation of lung 953–964. adenocarcinoma. Cancer Res 2011; 71: 6010–6018. 128 Wan H, Dingle S, Xu Y, Besnard V, Kaestner KH, Ang SL et al. Compensatory roles 152 Soucek L, Whitfield JR, Sodir NM, Massó-Vallés D, Serrano E, Karnezis AN et al. of Foxa1 and Foxa2 during lung morphogenesis. J Biol Chem 2005; 280: Inhibition of Myc family proteins eradicates KRas-driven lung cancer in mice. 13809–13816. Genes Dev 2013; 27:504–513. 129 Minoo P, Hu L, Xing Y, Zhu NL, Chen H, Li M et al. Physical and functional 153 Juan J, Muraguchi T, Iezza G, Sears RC, McMahon M. Diminished WNT -4 interactions between homeodomain NKX2.1 and winged helix/forkhead FOXA1 β-catenin -4 c-MYC signaling is a barrier for malignant progression of in lung epithelial cells. Mol Cell Biol 2007; 27:2155–2165. BRAFV600E-induced lung tumors. Genes Dev 2014; 28:561–575.

© 2015 Macmillan Publishers Limited Oncogene (2015) 5771 – 5780 Transcription control of lineage states in lung cancer WKC Cheung and DX Nguyen 5780 154 Adhikary S, Eilers M. Transcriptional regulation and transformation by Myc 170 Reynolds CP, Lemons RS. Retinoid therapy of childhood cancer. Hematol Oncol proteins. Nat Rev Mol Cell Biol 2005; 6: 635–645. Clin North Am 2001; 15:867–910. 155 Lin CY, Lovén J, Rahl PB, Paranal RM, Burge CB, Bradner JE et al. 171 Saez-Ayala M, Montenegro MF, Sanchez-Del-Campo L, Fernandez-Perez MP, Transcriptional amplification in tumor cells with elevated c-Myc. Cell 2012; 151: Chazarra S, Freter R et al. Directed phenotype switching as an effective anti- 56–67. melanoma strategy. Cancer Cell 2013; 24: 105–119. 156 Sinha S, Adler AS, Field Y, Chang HY, Segal E. Systematic functional character- 172 Konieczkowski DJ, Johannessen CM, Abudayyeh O, Kim JW, Cooper ZA, Piris A ization of cis-regulatory motifs in human core promoters. Genome Res 2008; 18: et al. A melanoma cell state distinction influences sensitivity to MAPK pathway 477–488. inhibitors. Cancer Discov 2014; 4: 816–827. 157 Wong DJ, Liu H, Ridky TW, Cassarino D, Segal E, Chang HY. Module map of stem 173 Rohle D, Popovici-Muller J, Palaskas N, Turcan S, Grommes C, Campos C et al. cell genes guides creation of epithelial cancer stem cells. Cell Stem Cell 2008; 2: An inhibitor of mutant IDH1 delays growth and promotes differentiation of 333–344. glioma cells. Science 2013; 340: 626–630. 158 Rapp UR, Korn C, Ceteci F, Karreman C, Luetkenhaus K, SerafinVet al. MYC is a 174 Campos B, Wan F, Farhadi M, Ernst A, Zeppernick F, Tagscherer KE et al. metastasis gene for non-small-cell lung cancer. PLoS One 2009; 4: e6029. Differentiation therapy exerts antitumor effects on stem-like glioma cells. 159 Medina PP, Romero OA, Kohno T, Montuenga LM, Pio R, Yokota J et al. Frequent Clin Cancer Res 2010; 16: 2715–2728. BRG1/SMARCA4-inactivating mutations in human lung cancer cell lines. 175 Yan M, Zhang Y, He B, Xiang J, Wang ZF, Zheng FM et al. IKKalpha restoration via Hum Mutat 2008; 29:617–622. EZH2 suppression induces nasopharyngeal carcinoma differentiation. Nat 160 Wilson BG, Roberts CW. SWI/SNF nucleosome remodellers and cancer. Nat Rev Commun 2014; 5: 3661. Cancer 2011; 11:481–492. 176 Tremblay AM, Missiaglia E, Galli GG, Hettmer S, Urcia R, Carrara M et al. The Hippo 161 Brabletz T. To differentiate or not--routes towards metastasis. Nat Rev Cancer transducer YAP1 transforms activated satellite cells and is a potent effector of 2012; 12: 425–436. embryonal rhabdomyosarcoma formation. Cancer Cell 2014; 26:273–287. 162 Gibbons DL, Lin W, Creighton CJ, Rizvi ZH, Gregory PA, Goodall GJ et al. 177 Lockwood WW, Zejnullahu K, Bradner JE, Varmus H. Sensitivity of human lung Contextual extracellular cues promote tumor cell EMT and metastasis by adenocarcinoma cell lines to targeted inhibition of BET epigenetic signaling regulating miR-200 family expression. Genes Dev 2009; 23: 2140–2151. proteins. Proc Natl Acad Sci U S A 2012; 109: 19408–19413. 163 Brabletz T. EMT and MET in metastasis: where are the cancer stem cells? Cancer 178 Shimamura T, Chen Z, Soucheray M, Carretero J, Kikuchi E, Tchaicha JH et al. Cell 2012; 22: 699–701. Efficacy of BET bromodomain inhibition in Kras-mutant non-small cell 164 Puisieux A, Brabletz T, Caramel J. Oncogenic roles of EMT-inducing transcription lung cancer. Clin Cancer Res 2013; 19: 6183–6192. factors. Nat Cell Biol 2014; 16:488–494. 179 Kwiatkowski N, Zhang T, Rahl PB, Abraham BJ, Reddy J, Ficarro SB et al. Targeting 165 Tsuji T, Ibaragi S, Hu GF. Epithelial-mesenchymal transition and cell cooperativity transcription regulation in cancer with a covalent CDK7 inhibitor. Nature 2014; in metastasis. Cancer Res 2009; 69: 7135–7139. 511:616–620. 166 Celià-Terrassa T, Meca-Cortés O, Mateo F, de Paz AM, Rubio N, Arnal-Estapé A 180 Chipumuro E, Marco E, Christensen CL, Kwiatkowski N, Zhang T, Hatheway CM et al. Epithelial-mesenchymal transition can suppress major attributes et al. CDK7 inhibition suppresses super-enhancer-linked oncogenic transcription of human epithelial tumor-initiating cells. J Clin Invest 2012; 122: in MYCN-driven cancer. Cell 2014; 159: 1126–1139. 1849–1868. 181 Christensen CL, Kwiatkowski N, Abraham BJ, Carretero J, Al-Shahrour F, Zhang T 167 Calbo J, van Montfort E, Proost N, van Drunen E, Beverloo HB, Meuwissen R et al. et al. Targeting transcriptional addictions in small cell lung cancer with a A functional role for tumor cell heterogeneity in a mouse model of small cell covalent CDK7 inhibitor. Cancer Cell 2014; 26: 909–922. lung cancer. Cancer Cell 2011; 19:244–256. 182 Corcoran RB, Cheng KA, Hata AN, Faber AC, Ebi H, Coffee EM et al. Synthetic 168 Cheung KJ, Gabrielson E, Werb Z, Ewald AJ. Collective invasion in breast lethal interaction of combined BCL-XL and MEK inhibition promotes tumor cancer requires a conserved basal epithelial program. Cell 2013; 155: regressions in KRAS mutant cancer models. Cancer Cell 2013; 23:121–128. 1639–1651. 183 Byers LA, Diao L, Wang J, Saintigny P, Girard L, Peyton M et al. An epithelial- 169 Govaere O, Komuta M, Berkers J, Spee B, Janssen C, de Luca F et al. Keratin 19: a mesenchymal transition gene signature predicts resistance to EGFR and PI3K key role player in the invasion of human hepatocellular carcinomas. Gut 2014; inhibitors and identifies Axl as a therapeutic target for overcoming EGFR inhi- 63: 674–685. bitor resistance. Clin Cancer Res 2013; 19: 279–290.

Oncogene (2015) 5771 – 5780 © 2015 Macmillan Publishers Limited