+ All Categories
Home > Documents > Genetic modifiers of EGFR dependence in non-small cell lung cancer

Genetic modifiers of EGFR dependence in non-small cell lung cancer

Date post: 30-Nov-2023
Category:
Upload: independent
View: 0 times
Download: 0 times
Share this document with a friend
6
Genetic modifiers of EGFR dependence in non-small cell lung cancer Tanaz Sharifnia a,b,c , Victor Rusu a,d , Federica Piccioni a , Mukta Bagul a , Marcin Imielinski a,b,c , Andrew D. Cherniack a , Chandra Sekhar Pedamallu a,c , Bang Wong a , Frederick H. Wilson a,c , Levi A. Garraway a,c , David Altshuler a,d,e , Todd R. Golub a , David E. Root a , Aravind Subramanian a , and Matthew Meyerson a,b,c,1 a Broad Institute of Harvard and MIT, Cambridge, MA 02142; Departments of b Pathology and d Genetics and Medicine, Harvard Medical School, Boston, MA 02115; c Department of Medical Oncology, DanaFarber Cancer Institute, Boston, MA 02115; and e Department of Molecular Biology and Diabetes Unit, Massachusetts General Hospital, Boston, MA 02114 Edited by Bert Vogelstein, Johns Hopkins University, Baltimore, MD, and approved November 18, 2014 (received for review June 29, 2014) Lung adenocarcinomas harboring activating mutations in the epidermal growth factor receptor (EGFR) represent a common mo- lecular subset of non-small cell lung cancer (NSCLC) cases. EGFR mutations predict sensitivity to EGFR tyrosine kinase inhibitors (TKIs) and thus represent a dependency in NSCLCs harboring these alterations, but the genetic basis of EGFR dependence is not fully understood. Here, we applied an unbiased, ORF-based screen to identify genetic modifiers of EGFR dependence in EGFR-mutant NSCLC cells. This approach identified 18 kinase and kinase-related genes whose overexpression can substitute for EGFR in EGFR- dependent PC9 cells, and these genes include seven of nine Src family kinase genes, FGFR1, FGFR2, ITK, NTRK1, NTRK2, MOS, MST1R, and RAF1. A subset of these genes can complement loss of EGFR activity across multiple EGFR-dependent models. Unbiased gene-expression profiling of cells overexpressing EGFR bypass genes, together with targeted validation studies, reveals EGFR- independent activation of the MEK-ERK and phosphoinositide 3-kinase (PI3K)-AKT pathways. Combined inhibition of PI3K-mTOR and MEK restores EGFR dependence in cells expressing each of the 18 EGFR bypass genes. Together, these data uncover a broad spectrum of kinases capable of overcoming dependence on EGFR and underscore their convergence on the PI3K-AKT and MEK-ERK signaling axes in sustaining EGFR-independent survival. epidermal growth factor receptor | non-small cell lung cancer | ORF T he term oncogene addictionhas been used to describe the phenomenon whereby tumor cells exhibit singular reliance on an oncogene or oncogenic pathway for their survival, despite the accumulation of multiple genetic lesions (1). In non-small cell lung cancer (NSCLC), this principle is perhaps best exem- plified with the finding that epidermal growth factor receptor (EGFR) mutations predict response to EGFR tyrosine kinase inhibitors (TKIs) gefitinib and erlotinib, and thus represent a de- pendency in the subset of tumors harboring these alterations (26). However, though EGFR-mutant NSCLCs typically respond dra- matically to EGFR TKIs, clinical responses are not universal, even within this genetically defined cohort, with the rate of objective response estimated to be 71% (5, 6). Furthermore, the over- whelming majority of patients who initially respond to EGFR inhibitors ultimately develop resistance to therapy (7). A deeper understanding of the genetic underpinnings of EGFR addiction, and how EGFR-mutant cells can overcome reliance on EGFR, may improve clinical outcomes. Here, we have applied an unbiased screening approach to iden- tify genetic modifiers of EGFR dependence in NSCLC. Mounting evidence supports the existence of several genetic modifiers of EGFR dependence in EGFR-mutant NSCLC, which can reduce the degree to which these tumors rely on EGFR and thereby contribute to EGFR TKI resistance (8). Examples include ampli- fication of the MET receptor tyrosine kinase (RTK) (9), activation of the NF-κB pathway (8), amplification of the HER2 (ERBB2) RTK (10), amplification of the CRKL gene (11), and activation of the AXL kinase (12). Notably, MET bypass can be reciprocally achieved via EGFR activation in MET-dependent cells (13), and analogous examples of reciprocal kinase switching have been reported in other kinase-driven cancer models (14, 15). These and other findings suggest that compensatory kinase switching may be a more general way in which oncogene-dependent cancers over- come reliance on their primary driver kinase (14, 16), but the full- range of kinases capable of mediating EGFR bypass has not been systematically studied. Recent advances in large-scale functional genetic libraries have made it possible to query a wide range of genetic perturbations for their ability to modulate specific cellular phenotypes in mammalian systems (17, 18). Using the model of EGFR-mutant, erlotinib- sensitive NSCLC cells, we have performed a systematic ORF-based screen to identify kinase and kinase-related genes whose over- expression can complement loss of EGFR activity in an EGFR- dependent context. Our findings indicate broad potential for EGFR substitution in the setting of EGFR dependence, with compensatory mechanisms commonly conferring EGFR-independent activation of the PI3K-AKT and MEK-ERK signaling pathways. Importantly, this approach has recovered known mechanisms of erlotinib resistance as well as identified novel mediators of EGFR bypass in EGFR-mutant NSCLC. These data support the idea that the EGFR-dependent state can be redundantly driven by diverse genetic inputs that commonly converge on shared downstream signaling nodes. Results An ORF-Based Screen Identifies 18 Genes Whose Expression Can Substitute for EGFR in an EGFR-Dependent NSCLC Cell Line. To identify genes that complement loss of EGFR activity in the Significance Non-small cell lung cancers (NSCLCs) harboring mutations in the epidermal growth factor receptor (EGFR) gene are often singularly reliant on EGFR activity for tumor cell survival, but the genetic basis for this dependence is not fully understood. In this study, we have performed a screen to identify a spectrum of kinase genes whose overexpression can overcome NSCLC cellsreliance on EGFR. Using both unbiased and targeted approaches, we demonstrate that these genes commonly by- pass dependence on EGFR through reactivation of downstream signaling pathways. Author contributions: T.S., A.S., and M.M. designed research; T.S. and M.B. performed research; V.R., B.W., F.H.W., L.A.G., D.A., T.R.G., D.E.R., and A.S. contributed new re- agents/analytic tools; T.S., V.R., F.P., M.B., M.I., A.D.C., and C.S.P. analyzed data; F.P. supervised screening; B.W. visualized data; F.H.W. and L.A.G. guided anaplastic lym- phoma kinase studies; and T.S. and M.M. wrote the paper. Conflict of interest statement: M.M. was a consultant and received research support from Novartis, and receives research support from Bayer. M.M., L.A.G., and T.R.G. are equity holders in, and consultants for, Foundation Medicine. This article is a PNAS Direct Submission. 1 To whom correspondence should be addressed. Email: matthew_meyerson@dfci. harvard.edu. This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. 1073/pnas.1412228112/-/DCSupplemental. www.pnas.org/cgi/doi/10.1073/pnas.1412228112 PNAS | December 30, 2014 | vol. 111 | no. 52 | 1866118666 GENETICS
Transcript

Genetic modifiers of EGFR dependence in non-smallcell lung cancerTanaz Sharifniaa,b,c, Victor Rusua,d, Federica Piccionia, Mukta Bagula, Marcin Imielinskia,b,c, Andrew D. Cherniacka,Chandra Sekhar Pedamallua,c, Bang Wonga, Frederick H. Wilsona,c, Levi A. Garrawaya,c, David Altshulera,d,e,Todd R. Goluba, David E. Roota, Aravind Subramaniana, and Matthew Meyersona,b,c,1

aBroad Institute of Harvard and MIT, Cambridge, MA 02142; Departments of bPathology and dGenetics and Medicine, Harvard Medical School, Boston,MA 02115; cDepartment of Medical Oncology, Dana–Farber Cancer Institute, Boston, MA 02115; and eDepartment of Molecular Biology and Diabetes Unit,Massachusetts General Hospital, Boston, MA 02114

Edited by Bert Vogelstein, Johns Hopkins University, Baltimore, MD, and approved November 18, 2014 (received for review June 29, 2014)

Lung adenocarcinomas harboring activating mutations in theepidermal growth factor receptor (EGFR) represent a common mo-lecular subset of non-small cell lung cancer (NSCLC) cases. EGFRmutations predict sensitivity to EGFR tyrosine kinase inhibitors(TKIs) and thus represent a dependency in NSCLCs harboring thesealterations, but the genetic basis of EGFR dependence is not fullyunderstood. Here, we applied an unbiased, ORF-based screen toidentify genetic modifiers of EGFR dependence in EGFR-mutantNSCLC cells. This approach identified 18 kinase and kinase-relatedgenes whose overexpression can substitute for EGFR in EGFR-dependent PC9 cells, and these genes include seven of nine Srcfamily kinase genes, FGFR1, FGFR2, ITK, NTRK1, NTRK2, MOS,MST1R, and RAF1. A subset of these genes can complement lossof EGFR activity across multiple EGFR-dependent models. Unbiasedgene-expression profiling of cells overexpressing EGFR bypassgenes, together with targeted validation studies, reveals EGFR-independent activation of the MEK-ERK and phosphoinositide3-kinase (PI3K)-AKT pathways. Combined inhibition of PI3K-mTORand MEK restores EGFR dependence in cells expressing each ofthe 18 EGFR bypass genes. Together, these data uncover a broadspectrum of kinases capable of overcoming dependence on EGFRand underscore their convergence on the PI3K-AKT and MEK-ERKsignaling axes in sustaining EGFR-independent survival.

epidermal growth factor receptor | non-small cell lung cancer | ORF

The term “oncogene addiction” has been used to describe thephenomenon whereby tumor cells exhibit singular reliance

on an oncogene or oncogenic pathway for their survival, despitethe accumulation of multiple genetic lesions (1). In non-smallcell lung cancer (NSCLC), this principle is perhaps best exem-plified with the finding that epidermal growth factor receptor(EGFR) mutations predict response to EGFR tyrosine kinaseinhibitors (TKIs) gefitinib and erlotinib, and thus represent a de-pendency in the subset of tumors harboring these alterations (2–6).However, though EGFR-mutant NSCLCs typically respond dra-matically to EGFR TKIs, clinical responses are not universal, evenwithin this genetically defined cohort, with the rate of objectiveresponse estimated to be ∼71% (5, 6). Furthermore, the over-whelming majority of patients who initially respond to EGFRinhibitors ultimately develop resistance to therapy (7). A deeperunderstanding of the genetic underpinnings of EGFR addiction,and how EGFR-mutant cells can overcome reliance on EGFR, mayimprove clinical outcomes.Here, we have applied an unbiased screening approach to iden-

tify genetic modifiers of EGFR dependence in NSCLC. Mountingevidence supports the existence of several genetic modifiers ofEGFR dependence in EGFR-mutant NSCLC, which can reducethe degree to which these tumors rely on EGFR and therebycontribute to EGFR TKI resistance (8). Examples include ampli-fication of the MET receptor tyrosine kinase (RTK) (9), activationof the NF-κB pathway (8), amplification of the HER2 (ERBB2)RTK (10), amplification of the CRKL gene (11), and activation ofthe AXL kinase (12). Notably, MET bypass can be reciprocally

achieved via EGFR activation in MET-dependent cells (13), andanalogous examples of reciprocal kinase switching have beenreported in other kinase-driven cancer models (14, 15). These andother findings suggest that compensatory kinase switching may bea more general way in which oncogene-dependent cancers over-come reliance on their primary driver kinase (14, 16), but the full-range of kinases capable of mediating EGFR bypass has not beensystematically studied.Recent advances in large-scale functional genetic libraries have

made it possible to query a wide range of genetic perturbations fortheir ability to modulate specific cellular phenotypes in mammaliansystems (17, 18). Using the model of EGFR-mutant, erlotinib-sensitive NSCLC cells, we have performed a systematic ORF-basedscreen to identify kinase and kinase-related genes whose over-expression can complement loss of EGFR activity in an EGFR-dependent context. Our findings indicate broad potential for EGFRsubstitution in the setting of EGFR dependence, with compensatorymechanisms commonly conferring EGFR-independent activation ofthe PI3K-AKT andMEK-ERK signaling pathways. Importantly, thisapproach has recovered knownmechanisms of erlotinib resistance aswell as identified novel mediators of EGFR bypass in EGFR-mutantNSCLC. These data support the idea that the EGFR-dependentstate can be redundantly driven by diverse genetic inputs thatcommonly converge on shared downstream signaling nodes.

ResultsAn ORF-Based Screen Identifies 18 Genes Whose Expression CanSubstitute for EGFR in an EGFR-Dependent NSCLC Cell Line. Toidentify genes that complement loss of EGFR activity in the

Significance

Non-small cell lung cancers (NSCLCs) harboring mutations inthe epidermal growth factor receptor (EGFR) gene are oftensingularly reliant on EGFR activity for tumor cell survival, butthe genetic basis for this dependence is not fully understood. Inthis study, we have performed a screen to identify a spectrumof kinase genes whose overexpression can overcome NSCLCcells’ reliance on EGFR. Using both unbiased and targetedapproaches, we demonstrate that these genes commonly by-pass dependence on EGFR through reactivation of downstreamsignaling pathways.

Author contributions: T.S., A.S., and M.M. designed research; T.S. and M.B. performedresearch; V.R., B.W., F.H.W., L.A.G., D.A., T.R.G., D.E.R., and A.S. contributed new re-agents/analytic tools; T.S., V.R., F.P., M.B., M.I., A.D.C., and C.S.P. analyzed data; F.P.supervised screening; B.W. visualized data; F.H.W. and L.A.G. guided anaplastic lym-phoma kinase studies; and T.S. and M.M. wrote the paper.

Conflict of interest statement: M.M. was a consultant and received research support fromNovartis, and receives research support from Bayer. M.M., L.A.G., and T.R.G. are equityholders in, and consultants for, Foundation Medicine.

This article is a PNAS Direct Submission.1To whom correspondence should be addressed. Email: [email protected].

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1412228112/-/DCSupplemental.

www.pnas.org/cgi/doi/10.1073/pnas.1412228112 PNAS | December 30, 2014 | vol. 111 | no. 52 | 18661–18666

GEN

ETICS

setting of EGFR dependence, we performed an ORF over-expression screen in an EGFR-dependent NSCLC cell line in thepresence of erlotinib. A library of 589 ORFs encoding humankinases and kinase-related proteins [Center for Cancer SystemsBiology (CCSB)/Broad Institute Kinase ORF Collection] (17,18) was expressed via lentiviral transduction in the EGFR-mutant NSCLC cell line, PC9, which is sensitive to EGFR TKIswith an IC50 of ∼30 nM (19). ORF-expressing PC9 cells weretreated with 300 nM erlotinib, 3 μM erlotinib, or DMSO for 72 hbefore being assayed for cell viability. Experimental ORFs werescreened alongside positive control EGFR ORFs encoding theT790M “gatekeeper” mutation, which can promote resistance toEGFR TKIs (20), in cis with a canonical EGFR activating mu-tation [EGFR-Δ(E746-A750)-T790M and EGFR-L858R-T790M](21). Additional positive controls included ORFs encoding acti-vating alleles of several MAPK family members.Cell viability under screening conditions was very low; the me-

dian relative viability for all experimental ORFs was 12% at the300-nM dose and 8% at the 3-μM dose (Fig. 1). Overexpression of19 ORFs, of the 589 tested, led to a significant increase in viabilityof erlotinib-treated PC9 cells, with viability of at least 39% in 300nM erlotinib and at least 31% in 3 μM erlotinib (Fig. 1). The geneswhose ectopic expression led to greater viability in the presence oferlotinib include known modifiers of EGFR dependence (AXL,ERBB2, CRKL) as well as genes and gene families that are newlyidentified candidate complementation partners of EGFR inEGFR-mutant lung cancer cells, including eight of the nine Srcfamily kinase members, as well as FGFR1 and FGFR2; ITK;MST1R; MOS; NTRK1 and NTRK2; and RAF1. The 19 geneswhose expression led to apparent EGFR substitution in the primaryscreen were selected for validation and functional characterization.cDNA vectors corresponding to the 19 candidate EGFR by-

pass genes were sequence-verified and confirmed to expressprotein (Fig. S1A). PC9 cells overexpressing each of these ORFs

were assayed for erlotinib sensitivity across multiple drug dosesusing 72-h growth inhibition assays (Fig. 2A). For comparison,the erlotinib-resistant EGFR-mutant positive controls EGFR-Δ(E746-A750)-T790M (henceforth EGFR-ex19del-T790M) andEGFR-L858R-T790M were tested in parallel. Ectopic expressionof 18 of the 19 candidate bypass ORFs was capable of reducingerlotinib sensitivity relative to cells transduced with an inert ORF(Fig. 2A). This reduced sensitivity corresponded to a >twofold to>400-fold shift in IC50 values (Table S1). One primary screeninghit, YES1, failed to bypass EGFR dependence in our validationstudies, leaving seven of nine Src family kinase genes confirmed toconfer the complementation phenotype (Fig. 2A and Table S1);and notably, this gene scored nearest to the cutoff we used toselect ORFs for validation (Fig. 1). We noted that the knownEGFR bypass gene MET did not score in our primary screen,likely due to failure of theMET expression vector to express METprotein (Fig. S1B). Overall, we estimate that the vast majority ofscreened ORFs express protein in PC9 cells (Fig. S1 C and D).The 18 genes capable of bypassing EGFR dependence in PC9

cells were used to transduce four additional EGFR-mutantNSCLC cell line models: the EGFR TKI-sensitive modelsHCC827, HCC2935, and H3255 (22), and the partially sensitivemodel HCC2279 (22, 23). Transduced cells were treated with 3μM erlotinib and assayed for cell viability after 72 h (Fig. S2 A–Eand Fig. 2B), and the effects of the 18 ORFs across all fivemodels tested were grouped by gene and cell line (Fig. 2B). In-terestingly, two classes of EGFR modifiers emerged from thisanalysis: one set of genes (CRKL, SRC, RAF1, FRK, BLK, andHCK, in addition to the erlotinib-resistant EGFR-ex19del-T790M mutant) whose overexpression could universally ornearly universally reduce EGFR dependence across the modelstested (Fig. 2B, red cluster), and a second set of genes whoseeffects varied across different cell line models (Fig. 2B, bluecluster). Overexpression of variable-effect cluster members LCK,FGR, LYN, and MST1R, and ITK, for example, each leads toincreased cell viability in the presence of erlotinib in three of thefive models tested (Fig. 2B, blue cluster). It is possible that thediffering effects of the genes in the variable-effect cluster onEGFR dependence can be attributed to the transcriptionalheterogeneity of these cell lines, which fall in distinct clusterswhen comparing expression profiles among lung cancer cell lines(Fig. S3), as well as to varying strengths of gene-specific effects.Importantly, the clinically validated EGFR TKI resistance-mediating genes AXL and ERBB2 each modify EGFR dependencein only a subset of the models tested, suggesting that the effects ofclinically relevant modifiers of EGFR dependence can also varyacross different EGFR-mutant models. Together, these dataidentify 18 modifiers of EGFR dependence in PC9 cells, of whicha subset can universally complement loss of EGFR activity acrossmultiple EGFR-dependent contexts.Interestingly, in investigating whether EGFR bypass ORFs are

specific to EGFR TKIs, we observed that overexpression ofseveral EGFR bypass genes, including AXL, CRKL, ERBB2,MST1R, RAF1, and several Src family kinases, can overcome sen-sitivity to the anaplastic lymphoma kinase (ALK) inhibitor TAE684in EML4-ALK-positive H3122 NSCLC cells (Fig. S4A). In contrast,none of the EGFR bypass genes caused appreciable changes incellular response to the chemotherapy drug cisplatin in PC9 cells(Fig. S4B). These findings suggest that the kinases identified in thisscreen do not universally mediate drug resistance, but they mayhave a more general role in modifying oncogene dependencein NSCLC.

Inhibition of Bypass Kinases Restores Erlotinib Sensitivity, andCoinhibition Is Associated with Phospho-AKT Attenuation for a Subsetof EGFR Bypass Genes. To enable the most inclusive analysis ofEGFR bypass genes, all 18 EGFR modifiers identified in PC9cells were carried forward for further study. First, because themodifier genes were predominantly kinases, we sought to de-termine whether the observed bypass phenotypes were kinase-dependent, as well as to verify the specificity of ORF activity, by

Percentage of control, 300 nM erlotinib

Per

cent

age

of c

ontro

l, 3

μM e

rlotin

ib

YES1

Experimental ORFsMutant EGFR controlsMutant MAPK pathway controlsNegative controlsNo virus controls

LCK

ERBB2

FGFR1FRK

NTRK2RAF1

MOSITK

LY N AXLMST1R

NTRK1FGFR2

CRKL BLKSRC

HCK

FGR

KRASV12

HRASV12

MEKDD

0 20 40 60 80 100

0

20

10

40

60

80

30

50

70

90

Fig. 1. An ORF-based screen identifies 18 kinase and kinase-related modi-fiers of EGFR dependence. Screening results for PC9 cells transduced witha library of kinase ORFs, treated with 300 nM or 3 μM erlotinib or vehicle, thenassayed for cell viability after 72 h using CellTiter-Glo. Experimental ORFs(green points) were screened alongside EGFR-ex19del-T790M and EGFR-L858R-T790M positive controls (yellow points); activating alleles of the MAPK family(orange points); inert gene controls (red points); and no virus controls (bluepoints). Data are expressed as percent viability relative to vehicle. Nineteencandidate modifiers of EGFR dependence were selected for validation.

18662 | www.pnas.org/cgi/doi/10.1073/pnas.1412228112 Sharifnia et al.

testing whether enzymatic inhibition of bypass-promoting kinasescould restore EGFR dependence. PC9 cells transduced with ki-nase ORFs were treated with erlotinib alone, the relevant kinaseinhibitor (where available) alone, or their combination for 72 h,then assayed for cell viability (Fig. 3A and Fig. S5 A–G). Inhibitorstested included dasatinib (targeting the Src family kinases); XL880(targeting AXL and MST1R); NVP-BGJ398 (BGJ398; targetingFGFR1 and FGFR2); lestaurtinib (targeting NTRK1 andNTRK2); lapatinib (targeting ERBB2); BMS-509744 (targetingITK); and AZ628 (targeting RAF1). The on-target activity ofthese inhibitors was confirmed for a select number of ORFs (Fig.S5 H–M).Cells expressing Src family kinase genes could indeed be

resensitized to erlotinib when cotreated with dasatinib, whereasthese cells were not similarly sensitive to either agent alone (Fig.3A and Fig. S5A). In contrast, resensitization to combinederlotinib/dasatinib treatment was not achieved in cells expressingEGFR-ex19del-T790M. Gene-specific rescue of erlotinib sensi-tivity was similarly observed for AXL; MST1R; FGFR1 andFGFR2; NTRK1 and NTRK2; ERBB2; ITK; and RAF1 uponcotreatment with their respective inhibitors (Fig. 3A and Fig. S5B–G). Combination treatment was also associated with increasedlevels of apoptosis in cells expressing bypass genes, comparedwith treatment with either inhibitor alone (Fig. S6). Together,these data suggest that the EGFR bypass phenotype induced bythese ORFs is specific to the encoded gene and requires thekinase activity of the expressed protein.A few kinase inhibitors showed somewhat different patterns of

growth inhibition or rescue. Lestaurtinib alone has some activityin LACZ- and EGFR-ex19del-T790M-transduced cells (Fig. 3Aand Fig. S5D), although only NTRK1- and NTRK2-expressingcells exhibit enhanced sensitivity under combination treatment.We also observed that AZ628 treatment only modestly resensi-tizes RAF1-transduced cells to erlotinib (Fig. 3A and Fig. S5G),consistent with the finding that RAF1 overexpression is itselfa mechanism of resistance to RAF inhibition (18).Next, because EGFR TKI treatment in EGFR-mutant cells

typically elicits down-regulation of the PI3K-AKT and MEK-ERK signaling pathways (2), we asked if rescue of EGFR de-pendence using combination treatment was associated with

a distinct PI3K-AKT and/or MEK-ERK signaling profile. ORF-expressing cells were treated with erlotinib, the relevant kinaseinhibitor, or their combination for 6 h, and assayed for down-stream signaling activation (Fig. 3 B–H). LACZ-expressing cellsundergo the expected down-regulation of phospho-EGFR, -AKT,and -ERK1/2 in the presence of erlotinib, in contrast to EGFR-ex19del-T790M-expressing cells (Fig. 3B). Cells transduced withSrc family kinases, however, maintain AKT phosphorylation in thepresence of 100 nM erlotinib, and this effect is reversed only uponcotreatment with dasatinib. Selective attenuation of phospho-AKT under combination treatment was similarly observed in cellstransduced with ERBB2, ITK, NTRK1, NTRK2, and very modestlywith FGFR1, FGFR2, AXL, and MST1R (Fig. 3 C–G).Selective down-regulation of phospho-AKT was not observed

in RAF1-expressing cells cotreated with erlotinib/AZ628 (Fig.3H), although as mentioned above, this combination was notsufficient to fully sensitize these cells (Fig. S5G). Combined erloti-nib/AZ628 treatment also fails to abolish phospho-ERK activationin these cells (Fig. 3H). Indeed, AZ628 treatment paradoxicallyinduces phospho-RAF1 activation in RAF1-expressing cells (Fig.S5M), consistent with previous findings using RAF inhibitors inother RAFWT models (24).

EGFR Bypass Genes Commonly Induce Similar Transcriptional Effects,Which Are Anticorrelated to Those Induced by MEK and PI3KInhibitors. Given that a subset of kinase genes can substitute forEGFR in PC9 cells, we were interested in identifying signalingpathways that were potentially common to overexpression ofthese genes. A hypothesis-driven analysis described above in-dicated a role for activation of the AKT pathway for a subset ofORFs; however, an unbiased systematic approach might moreclearly address whether EGFR bypass genes act through commonor divergent pathways. We reasoned that a high-dimensionaltranscriptional readout would provide an unbiased approach toaddress this question. We assembled reagents for the 18 EGFRbypass genes (positive-phenotype genes) as well as 19 kinase genesthat had failed to modify EGFR dependence in the primary screen(negative-phenotype genes). PC9 cells were transduced with thesevectors and controls for 72 h, then subjected to 24-h erlotinib

A B

PC

9

HC

C22

79

HC

C29

35

H32

55

HC

C82

7

ERBB2FGFR1ITKNTRK2MOSAXLFGFR2NTRK1MST1RLYNFGRLCKHCKBLKFRKRAF1SRCCRKLT790M

ERBB2

FGR

FRK0

50

100

150

ITK

LCK

AXL 0

50

100

150

LYN

MST1R NTRK1 NTRK2

SRC-4 -3 -2 -1 0 1

YES1-4 -3 -2 -1 0 1

LACZLUCIFERASEEGFR L858R T790MEGFR ex19del T790MExperimental ORF

Log μM erlotinib

Per

cent

age

of c

ontro

l FGFR2

BLK

HCK

FGFR1

MOS

RAF1-4 -3 -2 -1 0 1

0

50

100

150

0

50

100

150

0

50

100

150

0

50

100

150

0

50

100

150

CRKL

P ≥ 0.05 or no increase0.01 < P < 0.050.001 < P < 0.010.0001 < P < 0.001P < 0.0001

Increased viability under erlotinib treatment, relative to LACZ

LACZ

Fig. 2. Validation of the primary screen. (A) PC9cells expressing candidate EGFR bypass ORFs orcontrols were treated with erlotinib and assayedfor cell viability after 72 h using CellTiter-Glo. Dataare expressed as percent viability relative to vehi-cle-treated cells and represent the mean ± SD offour replicates. Graphs with identical control curvesreflect experiments performed in parallel on thesame day. (B) Heat map summarizing effects ofbypass ORFs validated in A across five EGFR-mutantcell line models. The 18 EGFR bypass genes vali-dated in PC9 cells were used for validation studiesin HCC2279, HCC2935, H3255, and HCC827 cells(Fig. S2). Grayscale values represent increasinglysignificant levels of ORF-induced erlotinib resis-tance relative to LACZ controls (black, 0; white, +4).Data were generated from cells treated with 1 μM(PC9) or 3 μM erlotinib (HCC2279, HCC2935, H3255,HCC827) (A and Fig. S2). Dendrograms in the rowand column margins represent groupings of ORFsand cell lines, respectively. ORFs segregate accord-ing to whether their EGFR bypass-promotingeffects are universal or nearly universal (red cluster)or variable (blue cluster) across the cell lines tested.T790M denotes EGFR-ex19del-T790M.

Sharifnia et al. PNAS | December 30, 2014 | vol. 111 | no. 52 | 18663

GEN

ETICS

treatment. Gene expression profiles for each ORF were used tocompute signatures as described in SI Materials and Methods.Unsupervised hierarchical clustering of these signatures yielded

two distinct clusters (Fig. 4A). Intriguingly, profiles segregatedlargely in accordance with positive vs. negative phenotypes; onesubcluster was comprised solely of 12 bypass-associated profilesand the two EGFR double-mutant controls (Fig. 4A, red). Giventhat this cluster contained almost all bypass-associated geneprofiles and none of the negative-phenotype profiles, we chose tofocus our subsequent analyses on the profiles represented in thispositive-phenotype cluster.To address whether the expression profiles of cells transduced

with bypass-associated ORFs could yield insights into their biologicaleffects, we used a large expression-profiling resource created by theLibrary of Integrated Network-Based Cellular Signatures (LINCS)program (www.lincscloud.org). This catalog was generated fromdiverse human cell lines treated with a large number of geneticand chemical perturbagens. Gene expression signatures from cellsexpressing each of the 12 genes in the positive-phenotype cluster(excluding controls) were independently used to query LINCS.We first sought to identify LINCS ORFs whose transcriptional

effects positively correlate with query ORFs, to potentially ad-dress whether EGFR bypass genes act through shared or distinctpathways. When considering the top ∼3% of positively corre-lated ORFs (Fig. 4B), query ORF signatures commonly correlatewith LINCS signatures generated using the same ORF or fromother EGFR bypass ORFs (Fig. 4B, magenta cells). Notably,none of the cell lines profiled in the LINCS dataset is EGFRmutant, and most are not of a lung lineage. Together with thehierarchical clustering, these data suggest that a major subset ofEGFR bypass genes induce similar transcriptional effects, which donot appear to be restricted to an EGFR-mutant cellular context.The LINCS database also contains over 34,000 gene expres-

sion profiles from cells treated with 3,103 compounds. Wequeried LINCS for chemical perturbations whose transcriptionaleffects were anticorrelated with those of query genes, hypothe-sizing that such perturbations could represent compound classeswith potential to reverse ORF-mediated effects. Among the top0.7% anticorrelated compounds (Fig. 5A, blue bars), 13 of 21(>61%) could be classified as either MEK or PI3K inhibitors(Fig. 5A, Lower Left). Along with the third-most abundant target,SRC, these were the only recurrent drug targets represented atthe top of this list (Fig. S7A). These findings indicated that the

transcriptional states induced by our panel of query genes wereopposed to those induced by PI3K or MEK inhibition, andsuggested that either or both of these pathways could be reac-tivated in ORF-expressing cells.

EGFR Modifier Genes Commonly Reactivate AKT and/or ERK1/2Signaling, and PI3K-mTOR and MEK Coinhibition Restores EGFRDependence. Given that PI3K and MEK inhibitor treatment sig-natures were anticorrelated with those of our positive-phenotypequery genes (Fig. 5A), we asked whether EGFR bypass genesreactivate the PI3K-AKT or MEK-ERK pathways. PC9 cells

A

B

LACZT790MBLKFGRFRKHCKLCK LYNSRC

LACZT790MAXLMST1R

LACZT790MFGFR1FGFR2LACZT790MNTRK1NTRK2LACZT790MERBB2

LACZT790MRAF1

erlot

inib

kinas

e inh

ibito

r

erlot

inib

+ kin

ase

in

hibito

r

dasa

tinib

XL8

80B

GJ3

98le

stau

rtini

bla

patin

ibA

Z628

AUC (Global)

Min Max

Total EGFR

Total AKT

Total ERK 1/2

Phospho-EGFR

Phospho-ERK 1/2

Phospho-AKT

LACZ T790M BLK

dasatinib (100 nM)

FGR

FRK HCK LCK LYN

erlotinib (100 nM)

SRC

Total EGFR

Total AKT

Total ERK 1/2

Phospho-EGFR

Phospho-ERK 1/2

Phospho-AKT

XL880 (100 nM)erlotinib (100 nM)

T790M

Total EGFR

Total AKT

Total ERK 1/2

Phospho-EGFR

Phospho-ERK 1/2

Phospho-AKT

BGJ398 (100 nM)erlotinib (100 nM)

LACZ AXL MST1R

T790M LACZ FGFR1 FGFR2

Total EGFR

Total AKT

Total ERK 1/2

Phospho-EGFR

Phospho-ERK 1/2

Phospho-AKT

lestaurtinib (100 nM)erlotinib (100 nM)

T790M LACZ NTRK1 NTRK2

Total EGFR

Total AKT

Total ERK 1/2

Phospho-EGFR

Phospho-ERK 1/2

Phospho-AKT

lapatinib (100 nM)erlotinib (100 nM)

T790M LACZ ERBB2

Total EGFR

Total AKT

Total ERK 1/2

Phospho-EGFR

Phospho-ERK 1/2

Phospho-AKT

BMS-509744 (1 μM) erlotinib (1 μM)

T790M LACZ ITK

Total EGFR

Total AKT

Total ERK 1/2

Phospho-EGFR

Phospho-ERK 1/2

Phospho-AKT

dasatinib (100 nM)erlotinib (100 nM)

C

D

E

F

G

Total EGFR

Total AKT

Total ERK 1/2

Phospho-EGFR

Phospho-ERK 1/2

Phospho-AKT

AZ628 (1 μM) erlotinib (1 μM)

T790M LACZ RAF1HLACZT790MITKB

MS

-50

9744

Fig. 3. Kinase inhibition of bypass-promoting kina-ses restores erlotinib sensitivity, and coinhibition isassociated with phospho-AKT attenuation for a sub-set of ORFs. (A) Inhibitor sensitivity of PC9 cellsexpressing indicated EGFR bypass-associated kinasesand controls following treatment with erlotinib, therelevant kinase inhibitor, or their combination for72 h. Cell viability was assayed with CellTiter-Glo, andthe resultant dose–response curves (Fig. S5) wereused to generate area under curve (AUC) values,plotted. Darker/lighter squares represent smaller/larger AUC values. (B–H) Immunoblot analysis of PC9cells expressing bypass-associated kinases and con-trols under combination drug treatment. Transducedcells were treated with indicated doses of erlotinib,the relevant kinase inhibitor, or a combination for 6h. Cells were incubated with 0.5% serum media 18 hbefore and during drug/DMSO treatment. Kinaseinhibitors tested included (B) dasatinib, (C) XL880,(D) BGJ398, (E) lestaurtinib, (F) lapatinib, (G) BMS-509744, and (H) AZ628. T790M denotes EGFR-ex19del-T790M.

A

B

CR

KL

BLK

EG

FR ex19del T790M

EG

FR L858R

T790MFG

RH

CK

FRK

NTR

K1

SR

CITKLC

KM

OS

RA

F1N

TRK

2FG

FR1

FGFR

2JA

K1

MA

PK

AP

K3

STK

40LY

N

AX

LC

AM

K1D

CD

K5R

1M

AG

I1LU

CIFE

RA

SE

LAC

ZN

LKP

RK

AR

2AS

TYK

1M

ST1R

ITGB

1BP

3M

AP

K13

FER

CK

S1B

TAO

K3

CS

NK

2A1

ER

BB

2P

RK

CB

LOC

652799P

DP

K1

TES

K2

CCCC

BLK CRKL FGR FRK HCK ITK LCK MOS NTRK1 NTRK2 RAF1 SRCSRC WWTR1 FGR TNFRSF1A LYN RAF1 LYN RAF1 RAF1 RAF1 RAF1 LYN

RARG FADD LYN LYN SRC CARD9 SRC IGF2 SRC MAGEB6 DDIT3 WWTR1RAF1 FOS CRK SRC TIRAP FGR IFNG PITPNA BCL10 FOS MECP2 RAF1IFNG RELB GPR139 RAF1 BCL10 CRK FGR CCND1 LYN NFE2L2 GRPR FGR

TIRAP ZNF238 SRC CD40 LPAR1 SUOX GPR139 NQO2 IGF2 ZNF238 TP53 GPBAR1FGR LPAR1 RAF1 BCL10 GPR139 FOS IFNB1 EIF4A2 TP53 ZRANB2 GPBAR1 SRCELK3 CRK MAGEB6 CRK MAGEB6 FAS RAF1 PSMB10 RBM6 GPBAR1 ITGAE NFE2L2

VPS28 RAF1 TIRAP IFNG LTBR CCND1 TIRAP MECP2 FADD NDUFA13 GPR119 GPR139MAGEB6 CCND1 RELB FGR CD40 GPBAR1 MAGEB6 IL10 IFNG MECP2 WWTR1 LPAR1PRDM1 SNAI3 FADD RELB FGR GPR139 CTBP1 CHRAC1 ADORA2A IL6R AKT1 PRKACACDO1 SRC TNFRSF10B TIRAP RAF1 PPP1CA NFE2L2 RASD1 PSMB10 FOXA3 ZNF669 CRKCRK FGR TRAF2 GABPB2 CRK BRAF LPAR1 GRPR SQRDL PLK1 SMAD7 GPR119

Query Signature

Pos

tivel

y-co

rrel

ated

OR

Fs

Fig. 4. EGFR bypass genes commonly induce similar transcriptional effects.(A) Unsupervised hierarchical clustering of PC9 cells overexpressing bypass-mediating ORFs (+); kinase ORFs unable to confer EGFR bypass (−); andcontrols (C). Twelve bypass-mediating ORFs displaying membership in a sin-gle cluster (positive-phenotype cluster, red) were used to query LINCS. (B)LINCS ORFs whose signatures most positively correlate with those of positive-phenotype cluster genes. Each of the 12 positive-phenotype cluster geneswas used to independently query LINCS. The top ∼3% positively correlatedORFs are listed in descending order. Query ORFs commonly correlate moststrongly with the same ORF and/or other EGFR bypass genes (magenta).

18664 | www.pnas.org/cgi/doi/10.1073/pnas.1412228112 Sharifnia et al.

overexpressing EGFR bypass genes were treated with erlotinibfor 6 h and profiled for activation of EGFR, AKT, and ERK1/2(Fig. 5B). Indeed, persistent activation of AKT and/or ERK1/2signaling under erlotinib treatment is a common feature of themajority of EGFR bypass genes, with sustained phospho-AKTmost prominently associated with BLK, FGR, FRK, HCK, LCK,SRC, ITK, and CRKL; and sustained phospho-ERK1/2 mostprominently associated with RAF1, BLK, ITK, MOS, AXL, andCRKL. Other ORFs, such as those encoding FGFR family genes,appear to display very modest activation of one or both of thesepathways, which may also be reflective of their more modestrescue phenotype (Fig. 2A). We also observed that cells ex-pressing several kinases (BLK, LCK, FGR, and FRK) maintainEGFR phosphorylation in the presence of erlotinib; we foundthat overexpression of these and other Src family kinases in PC9cells can also modify a specific tyrosine residue, Y845, on EGFRthat is known to be the major site for c-Src-mediated phos-phorylation (Fig. S8A) (25).We similarly tested a representative subset of EGFR bypass

ORFs in H3255 cells for their ability to reactivate downstreampathways (Fig. S8B). Consistent with our findings in PC9 cells,we observed varying degrees of phospho-ERK1/2 activationunder erlotinib treatment among kinases that can bypass EGFRin this cell line. In contrast, we noted that several of the ORFsthat fail to promote erlotinib resistance in H3255 cells do notreactivate ERK1/2, although it was impossible to exclude thepossibility of ERK1/2 reactivation by LYN and NTRK2. Theobserved differences in phospho-ERK1/2 activation in this cellline potentially explain the sometimes varied resistance-inducingeffects of ORFs in different EGFR-mutant models.To determine whether reactivation of these signal transducers

is required for ORF-induced bypass, PC9 cells transduced withEGFR bypass genes were treated with erlotinib; the dual PI3K-mTOR inhibitor NVP-BEZ235 (BEZ235); the MEK inhibitorAZD6244; or their combinations, and assayed for cell viabilityafter 72 h (Fig. 5C). Drug doses of BEZ235 (500 nM) andAZD6244 (2.5 μM) were determined empirically to be the lowestdoses capable of maximally inhibiting AKT and ERK1/2 phos-phorylation (Fig. S7 B and C), inhibit the activity of their targetsalone and in each of the combinations tested (Fig. S7D), and arein similar ranges to those used by other investigators in EGFR-mutant cell lines (26). Combined BEZ235/AZD6244 treatmentwas sufficient to restore sensitivity to erlotinib across all ORF-

expressing cell lines, with sensitivity often enhanced relative toerlotinib treatment in LACZ-expressing cells. In cases where at leastpartial restoration of erlotinib sensitivity could be achieved usingeither BEZ235 or AZD6244, resensitization was always potentiatedby combining these two agents. Together, these data are consistentwith the concept that reactivation of AKT and ERK1/2 signalingcommonly underlies ORF-mediated EGFR bypass.

DiscussionWe report the use of a systematic ORF-based screening ap-proach to identify the spectrum of kinases capable of bypassingEGFR dependence in EGFR-mutant NSCLC cells. To ourknowledge, this represents the first cDNA-based genetic comple-mentation screen for loss of EGFR in an EGFR-mutant model.This approach has identified known as well as previously un-identified drivers of EGFR bypass in EGFR-mutant NSCLC. Moregenerally, it has revealed the breadth of kinases with compensatorypotential in the setting of EGFR oncogene addiction: 18 genes forPC9 cells, including both tyrosine and serine/threonine kinases.Among the newly identified drivers of EGFR bypass in EGFR-

mutant NSCLC, Src family kinases were highly represented,comprising over one-third of screening hits. The finding that Srcfamily kinases are sufficient to modify EGFR dependence is inline with considerable evidence linking EGFR activity with thisfamily: c-Src itself has been characterized extensively with re-spect to its cooperative relationship with EGFR (27), and theintroduction of dominant-active c-Src can reduce the inhibitoryeffects of erlotinib in head and neck squamous cell carcinomamodels (28). Src family kinase activation has been observed incetuximab-resistant colorectal adenocarcinoma and NSCLCsquamous cell carcinoma in vitro models (29, 30), and it has beenreported that CRIPTO1-mediated EGFR TKI resistance inNSCLC is Src-dependent (31). Furthermore, a recent reportdescribed increased expression and activation of Src, mediatedby integrin activation, in EGFR TKI-resistant lung adenocarci-noma models (32). The present study extends these findings todemonstrate that overexpression of SRC is itself sufficient tobypass EGFR dependence in EGFR-dependent NSCLC, andthat this function is shared by seven of the nine family members.Similarly, we noted that fibroblast growth factor receptor

(FGFR) family kinase genes FGFR1 and FGFR2, though notpreviously recognized as sufficient for EGFR bypass, nonethelessalign with previous work describing up-regulation of FGFR1 as

Total EGFR

Total AKT

Total ERK 1/2

Phospho-EGFR

Phospho-ERK 1/2

Phospho-AKT

0 0.01

0.1

1 10

0 0.01

0.1

1 10

0 0.01

0.

1 1 10

LACZ EGFR ex19del T790M BLK

erlotinib (μM) 0 0.01

0.

1 1 10

CRKL

ERBB2 FGFR1 FGFR2 FGR FRK

HCK ITK LCK LYN MOS

MST1R NTRK1 NTRK2 SRC

0 0.01

0.

1 1 10

AXL

RAF1

0 0.01

0.1

1 10

0 0.01

0.1

1 10

0 0.01

0.

1 1 10

0 0.01

0.

1 1 10

0 0.01

0.

1 1 10

0 0.01

0.1

1 10

0 0.01

0.1

1 10

0 0.01

0.

1 1 10

0 0.01

0.

1 1 10

0 0.01

0.

1 1 10

0 0.01

0.1

1 10

0 0.01

0.1

1 10

0 0.01

0.

1 1 10

0 0.01

0.

1 1 10

0 0.01

0.

1 1 10

Per

cent

age

of c

ontro

l

B

C

erl BEZ

erl/B

EZAZD

erl/A

ZD

BEZ/AZD

erl/B

EZ/AZDerl BEZ

erl/B

EZAZD

erl/A

ZD

BEZ/AZD

erl/B

EZ/AZDerl BEZ

erl/B

EZAZD

erl/A

ZD

BEZ/AZD

erl/B

EZ/AZD

0

50

100

0

50

100

0

50

100

0

50

100

0

50

100

A

Median normalized rank0 0.2-0.2 0.8-0.8 1-1

-0.965-1

0.6-0.6 0.4-0.4

MEK

All

PI3KSRC

Recurrent targetclasses

Negatively correlated Positively correlated

Total EGFR

Total AKT

Total ERK 1/2

Phospho-EGFR

Phospho-ERK 1/2

Phospho-AKT

erlotinib (μM)

Total EGFR

Total AKT

Total ERK 1/2

Phospho-EGFR

Phospho-ERK 1/2

Phospho-AKT

erlotinib (μM)

Total EGFR

Total AKT

Total ERK 1/2

Phospho-EGFR

Phospho-ERK 1/2

Phospho-AKT

erlotinib (μM)

erl BEZ

erl/B

EZAZD

erl/A

ZD

BEZ/AZD

erl/B

EZ/AZD

NTRK1 NTRK2 RAF1 SRC

LCK LYN MOS MST1R

FGR FRK HCK ITK

CRKL ERBB2 FGFR1 FGFR2

LACZ EGFR ex19del T790M AXL BLK

Fig. 5. EGFR bypass genes commonly reactivateAKT and/or ERK1/2 signaling, and PI3K-mTOR andMEK coinhibition restores EGFR dependence. (A)Barcode plot displaying LINCS compounds rankedby the correlations of their signatures to those ofpositive-phenotype cluster ORFs. The most posi-tively/negatively correlated compounds approachranks of 1 and −1, respectively. The 0.7% mostnegatively correlated compounds (blue portion ofbarcode plot) include several inhibitors targetingMEK, PI3K, and SRC (Fig. S7A). Each bar representsa chemical reagent. A single reagent targeting bothPI3K and SRC is denoted with an asterisk. (B) Im-munoblot analysis of PC9 cells overexpressing theindicated ORFs and treated with erlotinib for 6 h.Cells were incubated with 0.5% serum media 18 hbefore and during drug/DMSO treatment. (C) Cellviability of PC9 cells overexpressing the indicatedORFs following treatment with 100 nM erlotinib(erl), 500 nM of BEZ235 (BEZ), 2.5 μM of AZD6244(AZD), or their combinations for 72 h. Cell viabilitywas assayed with CellTiter-Glo. Data are expressedas percent viability relative to vehicle-treated cellsand represent the mean ± SD of four replicates.

Sharifnia et al. PNAS | December 30, 2014 | vol. 111 | no. 52 | 18665

GEN

ETICS

well as the ligand FGF2 in gefitinib-resistant NSCLC models, withconcomitant dependency on the FGFR pathway (33, 34), and withother studies implicating FGF ligands in TKI resistance (15, 35).Other genes identified, including the neurotrophic tyrosine

kinase receptor (NTKR) family kinases NTRK1 and NTRK2, theTec family kinase ITK, MST1R, and the serine/threonine kinasesMOS and RAF1, have not previously been appreciated to modifyEGFR dependence in EGFR-mutant lung cancer cells, and thusunderscore the power of this screening approach in identifyingnovel mediators of bypass for a given dependency.Identifying the spectrum of kinases capable of EGFR bypass

is of considerable clinical interest given that patients withEGFR-mutant NSCLCs almost invariably acquire resistance toEGFR TKIs (7); a large fraction (30%) of acquired resistancecases are driven by unknown mechanism(s) (36); and becausemounting evidence suggests that activation of alternative driverkinases, such as MET, represents a common route by whichkinase-driven cancers acquire resistance to therapy (9, 12, 18).A systematic study of kinase-driven EGFR bypass may revealthe scope of potential kinases switches, and whether they actthrough common or divergent pathways in sustaining EGFR-independent survival. Our findings suggest that the diversekinases capable of replacing EGFR in PC9 cells uniformlyconverge upon downstream pathways.More generally, our observation that a large number of kinase

inputs can redundantly sustain cancer cell survival is consistentwith recent reports describing broad potential for growth factor-mediated inhibitor resistance in several tumor dependency models

(14, 15), with the finding that coactivation of multiple RTKs inglioblastoma cells overcomes reliance on any one RTK for down-stream signaling activation (16), and with the identification of ninekinase-related genes whose overexpression can overcome RAF in-hibition in BRAF-mutant melanoma cells (18). Taken together, ourfinding that a diverse set of kinases can redundantly drive theEGFR-dependent state may thus represent a more general featureof signal transduction in oncogene-dependent cancers.

Materials and MethodsKinase ORF Screen. Screening was performed using a kinase ORF library of 589ORFs (CCSB/Broad Institute Kinase ORF Collection) (17, 18), along with the con-trols displayed in Fig. 1. Transduced PC9 cells were treated with 3 μM erlotinib,300 nM erlotinib, or DMSO, and cell viability was assayed 3 d later using CellTiter-Glo (Promega). Additional details are described in SI Materials and Methods.

Additional Materials and Methods. Additional materials and methods, in-cluding cell culture and reagents; screen validation and drug sensitivity assays;immunoblotting; gene expression profiling; and LINCS analysis, are describedin SI Materials and Methods.

ACKNOWLEDGMENTS. We thank the Broad Genetic Perturbation Platformfor generation of the ORF library; the Broad Connectivity Map team forgeneration of the Library of Integrated Network-Based Cellular Signatures(LINCS) dataset and technical assistance; V. Dancik for LINCS compound identi-fication; and J. Chmielecki, H. Greulich, E. Stover, Z. Tothova, and X. Wu forhelpful discussions. Support for this work was provided by National Cancer In-stitute Grants R01CA116020, R01CA109038, and P50CA090578 (to M.M.) andNational Institutes of Health Common Fund Grant 5U54HG006093 (to T.R.G.).

1. Weinstein IB (2002) Cancer. Addiction to oncogenes—the Achilles heal of cancer.Science 297(5578):63–64.

2. Paez JG, et al. (2004) EGFR mutations in lung cancer: correlation with clinical responseto gefitinib therapy. Science 304(5676):1497–1500.

3. Lynch TJ, et al. (2004) Activating mutations in the epidermal growth factor receptorunderlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med350(21):2129–2139.

4. Pao W, et al. (2004) EGF receptor gene mutations are common in lung cancers from“never smokers” and are associated with sensitivity of tumors to gefitinib and erlo-tinib. Proc Natl Acad Sci USA 101(36):13306–13311.

5. Mok TS, et al. (2009) Gefitinib or carboplatin-paclitaxel in pulmonary adenocarci-noma. N Engl J Med 361(10):947–957.

6. Rosell R, et al.; Spanish Lung Cancer Group (2009) Screening for epidermal growthfactor receptor mutations in lung cancer. N Engl J Med 361(10):958–967.

7. Engelman JA, Settleman J (2008) Acquired resistance to tyrosine kinase inhibitorsduring cancer therapy. Curr Opin Genet Dev 18(1):73–79.

8. Bivona TG, et al. (2011) FAS and NF-κB signalling modulate dependence of lungcancers on mutant EGFR. Nature 471(7339):523–526.

9. Engelman JA, et al. (2007) MET amplification leads to gefitinib resistance in lungcancer by activating ERBB3 signaling. Science 316(5827):1039–1043.

10. Takezawa K, et al. (2012) HER2 amplification: A potential mechanism of acquiredresistance to EGFR inhibition in EGFR-mutant lung cancers that lack the second-siteEGFRT790M mutation. Cancer Discov 2(10):922–933.

11. Cheung HW, et al. (2011) Amplification of CRKL induces transformation and epi-dermal growth factor receptor inhibitor resistance in human non-small cell lungcancers. Cancer Discov 1(7):608–625.

12. Zhang Z, et al. (2012) Activation of the AXL kinase causes resistance to EGFR-targetedtherapy in lung cancer. Nat Genet 44(8):852–860.

13. Bachleitner-Hofmann T, et al. (2008) HER kinase activation confers resistance to METtyrosine kinase inhibition in MET oncogene-addicted gastric cancer cells. Mol CancerTher 7(11):3499–3508.

14. Harbinski F, et al. (2012) Rescue screens with secreted proteins reveal compensatorypotential of receptor tyrosine kinases in driving cancer growth. Cancer Discov 2(10):948–959.

15. Wilson TR, et al. (2012) Widespread potential for growth-factor-driven resistance toanticancer kinase inhibitors. Nature 487(7408):505–509.

16. Stommel JM, et al. (2007) Coactivation of receptor tyrosine kinases affects the re-sponse of tumor cells to targeted therapies. Science 318(5848):287–290.

17. Yang X, et al. (2011) A public genome-scale lentiviral expression library of humanORFs. Nat Methods 8(8):659–661.

18. Johannessen CM, et al. (2010) COT drives resistance to RAF inhibition through MAPkinase pathway reactivation. Nature 468(7326):968–972.

19. Yeo WL, et al. (2010) Erlotinib at a dose of 25 mg daily for non-small cell lung cancerswith EGFR mutations. J Thorac Oncol 5(7):1048–1053.

20. Kobayashi S, et al. (2005) EGFR mutation and resistance of non-small-cell lung cancerto gefitinib. N Engl J Med 352(8):786–792.

21. Sharma SV, Bell DW, Settleman J, Haber DA (2007) Epidermal growth factor receptormutations in lung cancer. Nat Rev Cancer 7(3):169–181.

22. Sos ML, et al. (2009) Predicting drug susceptibility of non-small cell lung cancers basedon genetic lesions. J Clin Invest 119(6):1727–1740.

23. Lu Y, Liang K, Li X, Fan Z (2007) Responses of cancer cells with wild-type or tyrosinekinase domain-mutated epidermal growth factor receptor (EGFR) to EGFR-targetedtherapy are linked to downregulation of hypoxia-inducible factor-1alpha. MolCancer 6:63.

24. Hatzivassiliou G, et al. (2010) RAF inhibitors prime wild-type RAF to activate theMAPK pathway and enhance growth. Nature 464(7287):431–435.

25. Biscardi JS, et al. (1999) c-Src-mediated phosphorylation of the epidermal growthfactor receptor on Tyr845 and Tyr1101 is associated with modulation of receptorfunction. J Biol Chem 274(12):8335–8343.

26. Faber AC, et al. (2009) Differential induction of apoptosis in HER2 and EGFR addictedcancers following PI3K inhibition. Proc Natl Acad Sci USA 106(46):19503–19508.

27. Ishizawar R, Parsons SJ (2004) c-Src and cooperating partners in human cancer. CancerCell 6(3):209–214.

28. Stabile LP, et al. (2013) c-Src activation mediates erlotinib resistance in head and neckcancer by stimulating c-Met. Clin Cancer Res 19(2):380–392.

29. Lu Y, et al. (2007) Epidermal growth factor receptor (EGFR) ubiquitination as amechanism of acquired resistance escaping treatment by the anti-EGFR monoclonalantibody cetuximab. Cancer Res 67(17):8240–8247.

30. Wheeler DL, et al. (2009) Epidermal growth factor receptor cooperates with Src familykinases in acquired resistance to cetuximab. Cancer Biol Ther 8(8):696–703.

31. Park KS, et al. (2014) CRIPTO1 expression in EGFR-mutant NSCLC elicits intrinsic EGFR-inhibitor resistance. J Clin Invest 124(7):3003–3015.

32. Kanda R, et al. (2013) Erlotinib resistance in lung cancer cells mediated by in-tegrinbeta1/Src/Akt-driven bypass signaling. Cancer Res 73(20):6243–6253.

33. Ware KE, et al. (2013) A mechanism of resistance to gefitinib mediated by cellularreprogramming and the acquisition of an FGF2-FGFR1 autocrine growth loop. On-cogenesis 2:e39.

34. Terai H, et al. (2013) Activation of the FGF2-FGFR1 autocrine pathway: A novelmechanism of acquired resistance to gefitinib in NSCLC. Mol Cancer Res 11(7):759–767.

35. Ware KE, et al. (2010) Rapidly acquired resistance to EGFR tyrosine kinase inhibitors inNSCLC cell lines through de-repression of FGFR2 and FGFR3 expression. PLoS ONE5(11):e14117.

36. Sequist LV, et al. (2011) Genotypic and histological evolution of lung cancers ac-quiring resistance to EGFR inhibitors. Sci Transl Med 3(75):75ra26.

18666 | www.pnas.org/cgi/doi/10.1073/pnas.1412228112 Sharifnia et al.


Recommended