addiction to multiple oncogenes can be exploited to prevent ...addiction to multiple oncogenes can...

9
Addiction to multiple oncogenes can be exploited to prevent the emergence of therapeutic resistance Peter S. Choi, Yulin Li, and Dean W. Felsher 1 Division of Oncology, Departments of Medicine, Pathology, and Molecular Imaging, Stanford University School of Medicine, Stanford, CA 94305 Edited by Stephen R. Hann, Vanderbilt University, Nashville, TN, and accepted by the Editorial Board July 2, 2014 (received for review April 2, 2014) Many cancers exhibit sensitivity to the inhibition of a single genetic lesion, a property that has been successfully exploited with oncogene-targeted therapeutics. However, inhibition of single oncogenes often fails to result in sustained tumor regression due to the emergence of therapy-resistant cells. Here, we report that MYC-driven lymphomas frequently acquire activating mutations in β-catenin, including a previously unreported mutation in a splice acceptor site. Tumors with these genetic lesions are highly depen- dent on β-catenin for their survival and the suppression of β-catenin resulted in marked apoptosis causally related to a decrease in Bcl-xL expression. Using a novel inducible inhibitor of β-catenin, we illus- trate that, although MYC withdrawal or β-catenin inhibition alone results in initial tumor regression, most tumors ultimately re- curred, mimicking the clinical response to single-agent targeted therapy. Importantly, the simultaneous combined inhibition of both MYC and β-catenin promoted more rapid tumor regression and successfully prevented tumor recurrence. Hence, we demon- strated that MYC-induced tumors are addicted to mutant β-catenin, and the combined inactivation of MYC and β-catenin induces sus- tained tumor regression. Our results provide a proof of principle that targeting multiple oncogene addicted pathways can prevent therapeutic resistance. oncogene addiction | combination targeted therapy | splice site mutations C ancer cells are highly sensitive to the targeted inhibition of single driver mutations, eliciting a phenomenon known as oncogene addiction(1). The identification of genetic dependencies in multiple tumor types has resulted in the development of sev- eral molecularly targeted therapeutics, including the BCR-ABL kinase inhibitor imatinib for the treatment of chronic myeloge- nous leukemia (CML), the EGFR kinase inhibitor gefitinib for the treatment of nonsmall cell lung cancer (NSCLC), and the BRAF kinase inhibitor vemurafenib for the treatment of advanced melanoma (24). Although these oncogene-targeted agents have provided promising clinical responses, many patients ultimately experience a recurrence of their disease due to the development of drug resistance (46). Thus, it has become evident that mono- therapy with targeted drugs is insufficient for achieving sustained tumor regression. Resistance to targeted therapy can arise through multiple mech- anisms, depending on the tumor type and the targeted oncogenic pathway (7). Cells frequently acquire resistance through mutations in the targeted oncogene itself that disrupt drug binding, as in the case of BCR-ABL and EGFR (8, 5, 6). In addition, resistance to EGFR inhibition in NSCLC and BRAF inhibition in melanoma has been found to occur through a variety of mechanisms that activate downstream signaling proteins or alternative pathways, which can functionally substitute for loss in activity of the targeted oncogene (911). Although significant progress has been made in the identification and inhibition of resistance pathways, it may prove challenging to anticipate and suppress all of the potential mechanisms of resistance for each oncogene-addicted cancer and targeted therapeutic agent. Combination therapy has been successfully applied to prevent resistance in the treatment of infectious diseases such as HIV (12, 13) and tuberculosis (14). In the context of oncogene-targeted therapy for cancer, it has been proposed that a similar strategy, using combinations directed against multiple dependencies, is the most likely to prevent resistance (7). Indeed, mathematical mod- eling indicates that targeting at least two independently re- quired pathways may be sufficient to prevent tumor recurrence (15). However, there exists little experimental evidence directly testing such an approach and it remains unclear which com- binations of targets would be most effective at inducing long- term remissions. MYC is one of the most frequently amplified oncogenes in human cancer (16). In the Eμ-tTA/tetO-MYC conditional mouse model, overexpression of MYC results in the development of aggressive T-cell lymphoma, and MYC inactivation in estab- lished tumors is sufficient to induce tumor regression through processes such as proliferative arrest, cellular senescence, apo- ptosis, and the shutdown of angiogenesis (1719). The extent of regression is dependent on both cell-intrinsic and host-dependent contexts, and in particular, tumors frequently recur following MYC inactivation in the absence of an intact adaptive immune system (20). Recurring tumors restore expression of the MYC transgene or up-regulate expression of endogenous Myc, dem- onstrating that resistance occurs primarily through reactivation of the MYC pathway (21). Thus, MYC oncogene addiction and tumor recurrence in the Eμ-tTA/tetO-MYC lymphoma model resembles the clinical course of human cancers treated with single agent targeted therapy. Here, we demonstrate that the combined inactivation of two oncogene addiction pathways can result in sustained tumor re- gression. Moreover, we describe a previously unidentified splice acceptor site mutation in β-catenin that is associated with MYC- induced lymphomagenesis. Tumors with mutations in β-catenin Significance The rationale of targeting specific genetic dependencies for the treatment of cancer has been validated by the promising clin- ical responses obtained with oncogene-targeted therapies. However, in most cases, the development of resistance remains a major obstacle toward achieving long-term or complete dis- ease remission. Here, we identified mutant β-catenin as a com- mon secondary oncogene addiction pathway in MYC-addicted lymphoma. We demonstrate that, although withdrawal or inhibition of either oncogene is sufficient to induce initial tumor regression, only combined inhibition of both oncogene addiction pathways results in sustained tumor regression. Our results suggest clinical outcomes can be dramatically improved through the simultaneous targeted inhibition of multiple onco- genic addiction pathways. Author contributions: P.S.C., Y.L., and D.W.F. designed research; P.S.C. and Y.L. performed research; P.S.C. and Y.L. analyzed data; and P.S.C. and D.W.F. wrote the paper. The authors declare no conflict of interest. This article is a PNAS Direct Submission. S.R.H. is a guest editor invited by the Editorial Board. 1 To whom correspondence should be addressed. Email: [email protected]. This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. 1073/pnas.1406123111/-/DCSupplemental. E3316E3324 | PNAS | Published online July 28, 2014 www.pnas.org/cgi/doi/10.1073/pnas.1406123111 Downloaded by guest on March 3, 2021

Upload: others

Post on 10-Oct-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Addiction to multiple oncogenes can be exploited to prevent ...Addiction to multiple oncogenes can be exploited to prevent the emergence of therapeutic resistance Peter S. Choi, Yulin

Addiction to multiple oncogenes can be exploited toprevent the emergence of therapeutic resistancePeter S. Choi, Yulin Li, and Dean W. Felsher1

Division of Oncology, Departments of Medicine, Pathology, and Molecular Imaging, Stanford University School of Medicine, Stanford, CA 94305

Edited by Stephen R. Hann, Vanderbilt University, Nashville, TN, and accepted by the Editorial Board July 2, 2014 (received for review April 2, 2014)

Many cancers exhibit sensitivity to the inhibition of a single geneticlesion, a property that has been successfully exploited withoncogene-targeted therapeutics. However, inhibition of singleoncogenes often fails to result in sustained tumor regression dueto the emergence of therapy-resistant cells. Here, we report thatMYC-driven lymphomas frequently acquire activating mutations inβ-catenin, including a previously unreported mutation in a spliceacceptor site. Tumors with these genetic lesions are highly depen-dent on β-catenin for their survival and the suppression of β-cateninresulted in marked apoptosis causally related to a decrease in Bcl-xLexpression. Using a novel inducible inhibitor of β-catenin, we illus-trate that, although MYC withdrawal or β-catenin inhibition aloneresults in initial tumor regression, most tumors ultimately re-curred, mimicking the clinical response to single-agent targetedtherapy. Importantly, the simultaneous combined inhibition ofboth MYC and β-catenin promoted more rapid tumor regressionand successfully prevented tumor recurrence. Hence, we demon-strated that MYC-induced tumors are addicted to mutant β-catenin,and the combined inactivation of MYC and β-catenin induces sus-tained tumor regression. Our results provide a proof of principlethat targeting multiple oncogene addicted pathways can preventtherapeutic resistance.

oncogene addiction | combination targeted therapy | splice site mutations

Cancer cells are highly sensitive to the targeted inhibition ofsingle driver mutations, eliciting a phenomenon known as

“oncogene addiction” (1). The identification of genetic dependenciesin multiple tumor types has resulted in the development of sev-eral molecularly targeted therapeutics, including the BCR-ABLkinase inhibitor imatinib for the treatment of chronic myeloge-nous leukemia (CML), the EGFR kinase inhibitor gefitinib forthe treatment of non–small cell lung cancer (NSCLC), and theBRAF kinase inhibitor vemurafenib for the treatment of advancedmelanoma (2–4). Although these oncogene-targeted agents haveprovided promising clinical responses, many patients ultimatelyexperience a recurrence of their disease due to the development ofdrug resistance (4–6). Thus, it has become evident that mono-therapy with targeted drugs is insufficient for achieving sustainedtumor regression.Resistance to targeted therapy can arise through multiple mech-

anisms, depending on the tumor type and the targeted oncogenicpathway (7). Cells frequently acquire resistance through mutationsin the targeted oncogene itself that disrupt drug binding, as in thecase of BCR-ABL and EGFR (8, 5, 6). In addition, resistance toEGFR inhibition in NSCLC and BRAF inhibition in melanomahas been found to occur through a variety of mechanisms thatactivate downstream signaling proteins or alternative pathways,which can functionally substitute for loss in activity of the targetedoncogene (9–11). Although significant progress has been madein the identification and inhibition of resistance pathways, it mayprove challenging to anticipate and suppress all of the potentialmechanisms of resistance for each oncogene-addicted cancer andtargeted therapeutic agent.Combination therapy has been successfully applied to prevent

resistance in the treatment of infectious diseases such as HIV(12, 13) and tuberculosis (14). In the context of oncogene-targeted

therapy for cancer, it has been proposed that a similar strategy,using combinations directed against multiple dependencies, is themost likely to prevent resistance (7). Indeed, mathematical mod-eling indicates that targeting at least two independently re-quired pathways may be sufficient to prevent tumor recurrence(15). However, there exists little experimental evidence directlytesting such an approach and it remains unclear which com-binations of targets would be most effective at inducing long-term remissions.MYC is one of the most frequently amplified oncogenes in

human cancer (16). In the Eμ-tTA/tetO-MYC conditional mousemodel, overexpression of MYC results in the development ofaggressive T-cell lymphoma, and MYC inactivation in estab-lished tumors is sufficient to induce tumor regression throughprocesses such as proliferative arrest, cellular senescence, apo-ptosis, and the shutdown of angiogenesis (17–19). The extent ofregression is dependent on both cell-intrinsic and host-dependentcontexts, and in particular, tumors frequently recur followingMYC inactivation in the absence of an intact adaptive immunesystem (20). Recurring tumors restore expression of the MYCtransgene or up-regulate expression of endogenous Myc, dem-onstrating that resistance occurs primarily through reactivationof the MYC pathway (21). Thus, MYC oncogene addiction andtumor recurrence in the Eμ-tTA/tetO-MYC lymphoma modelresembles the clinical course of human cancers treated with singleagent targeted therapy.Here, we demonstrate that the combined inactivation of two

oncogene addiction pathways can result in sustained tumor re-gression. Moreover, we describe a previously unidentified spliceacceptor site mutation in β-catenin that is associated with MYC-induced lymphomagenesis. Tumors with mutations in β-catenin

Significance

The rationale of targeting specific genetic dependencies for thetreatment of cancer has been validated by the promising clin-ical responses obtained with oncogene-targeted therapies.However, in most cases, the development of resistance remainsa major obstacle toward achieving long-term or complete dis-ease remission. Here, we identified mutant β-catenin as a com-mon secondary oncogene addiction pathway in MYC-addictedlymphoma. We demonstrate that, although withdrawal orinhibition of either oncogene is sufficient to induce initialtumor regression, only combined inhibition of both oncogeneaddiction pathways results in sustained tumor regression. Ourresults suggest clinical outcomes can be dramatically improvedthrough the simultaneous targeted inhibition of multiple onco-genic addiction pathways.

Author contributions: P.S.C., Y.L., and D.W.F. designed research; P.S.C. and Y.L. performedresearch; P.S.C. and Y.L. analyzed data; and P.S.C. and D.W.F. wrote the paper.

The authors declare no conflict of interest.

This article is a PNAS Direct Submission. S.R.H. is a guest editor invited by the EditorialBoard.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.1406123111/-/DCSupplemental.

E3316–E3324 | PNAS | Published online July 28, 2014 www.pnas.org/cgi/doi/10.1073/pnas.1406123111

Dow

nloa

ded

by g

uest

on

Mar

ch 3

, 202

1

Page 2: Addiction to multiple oncogenes can be exploited to prevent ...Addiction to multiple oncogenes can be exploited to prevent the emergence of therapeutic resistance Peter S. Choi, Yulin

C

β-catenin

β-actin

E

NH2 COOHTransactivation domainArmadillo repeats

...SYLDSGIHSGATTTAPSLSGK...33 37 41 45

P P P P

Exon3

β-catenin protein

T

Asp Asn

31 32 33

Leu Ser

T T T TG G/A A C

*

D32N mutation

A

Thr Ala

40 41 42

Thr Thr

A/G C

*

C C A AC C

T41A mutation

GU AG GU AG GU AG

β-catenin primary transcript

splice donor

Exon1 Exon4Exon3Exon2

splice acceptor

T T T GG/AA C

*

A C

Splice acceptor mutation 2

Exon3splice

acceptor

Splice acceptor mutation 1

Exon3splice

acceptor

T T T GGA/G C

*

A C

Exon1 Exon4Exon3Exon2

Exon1 Exon4Exon2

wildtype transcript

Δexon3 transcript

D

Thymus

β-cateninwildtype

β-cateninmutant

MYC Axin2 Enc1

Rel

ativ

e ex

pres

sion

Rel

ativ

e ex

pres

sion

Rel

ativ

e ex

pres

sion

β-cateninwildtype

β-cateninmutant

β-cateninwildtype

β-cateninmutant

** **

0.1

1

10

0.1

1

10

100

1000

0.1

1

10

100

1000

Primary lymphomas

F

B6678

B6422

B6680

B7647

C1194

C1296

C1369

C1497

C1630

C3598

D0290

β-catenin status: WT WT WT WT WT WT WTT41A G34R Δex3 D32N

Tumor ID:

Lymphoma-derived cell lines

C1194

C1571

C1573

WT WT

B3470

B3760

B4329

B4334

T41AΔex3 Δex3 Δex3 Δex3 Δex3

A

T-cell lymphoma

MYC overexpression

tetOtTA

Cooperating driver mutations

B

19/30

7/30

4/30

β-catenin mutationsNoneMissenseSplice-site

MYC

?????????

Fig. 1. MYC-induced lymphomas frequently harbor stabilizing mutations in β-catenin. (A) In Eμ-tTA/tetO-MYC transgenic mice, T-cell lymphomas are pro-posed to arise as a result of MYC overexpression and alterations in unidentified cooperating pathways. (B) Distribution of β-catenin mutations detected bysequencing of DNA from Eμ-tTA/tetO-MYC primary lymphoma specimens. (C) Western blot analysis for β-catenin protein in primary MYC-induced lymphomasand lymphoma-derived cell lines. β-actin is shown as a loading control. Closed and open triangles denote full-length and Δexon3 β-catenin protein, re-spectively. Tumors with mutant β-catenin are labeled in red. (D) Schematic of β-catenin depicting the phosphorylation residues encoded by exon 3 that arecritical for protein degradation. Also shown are sequence chromatograms of mutations in phosphorylation residues identified in primary MYC-inducedlymphomas. (E) Diagram of conserved splice donor and acceptor sites required for proper splicing of the β-catenin primary transcript. Also shown are se-quence chromatograms of splice acceptor site mutations identified in primary MYC-induced lymphomas. (F) Relative expression levels of transgenic MYC andthe β-catenin/TCF target genes Axin2 and Enc1 across a panel of MYC-induced primary lymphomas with WT (n = 8) or mutant (n = 6) β-catenin, as analyzed byqRT-PCR. Shown are means ± SEM with **P < 0.01.

Choi et al. PNAS | Published online July 28, 2014 | E3317

MED

ICALSC

IENCE

SPN

ASPL

US

Dow

nloa

ded

by g

uest

on

Mar

ch 3

, 202

1

Page 3: Addiction to multiple oncogenes can be exploited to prevent ...Addiction to multiple oncogenes can be exploited to prevent the emergence of therapeutic resistance Peter S. Choi, Yulin

are also highly addicted to this mutant gene product for theirsurvival. We demonstrate that in MYC-induced lymphomas, com-bined addiction to both MYC and β-catenin can be exploited in arational manner to prevent the emergence of therapeutic resistance.

ResultsMYC-Induced Lymphomas Frequently Acquire Stabilizing Mutations inβ-Catenin. Lymphomas initiated by MYC overexpression oftenacquire loss-of-function mutations in tumor suppressor genessuch as p53 and p19ARF (22). However, it remains less wellknown whether MYC overexpression also selects for the acti-vation of additional oncogenic drivers during lymphomagenesis(Fig. 1A). We discovered that a subset of MYC-induced lym-phomas and lymphoma-derived cell lines from the Eμ-tTA/tetO-MYC model expressed elevated levels of β-catenin, with bothfull-length and shorter forms of β-catenin protein detectable byWestern blot (Fig. 1C). To determine the mechanism of β-cateninup-regulation in our panel of MYC-induced lymphomas, weisolated genomic DNA and sequenced the entire β-catenin exon3 region and portions of the upstream and downstream introns.We detected missense mutations in 7 of 30 samples, at or ad-jacent to the phosphorylation sites required for β-catenin deg-radation (Fig. 1D). Interestingly, in 4 of 30 samples, we also

discovered novel mutations at the exon 3 splice acceptor site thatare predicted to result in skipping of exon 3 during processing ofthe primary transcript and an in-frame deletion of the regionrequired for β-catenin degradation (Fig. 1E). Skipping of exon 3was confirmed at the mRNA level by sequencing of β-catenincDNA from lymphoma samples with splice site mutations (Fig.S1). In total, we detected β-catenin mutations in ∼37% of pri-mary lymphomas from Eμ-tTA/tetO-MYC mice (11 of 30; Fig.1B and Table S1). The type of mutation correlated with the sizeof β-catenin protein that was detected, with elevated levels offull-length protein in samples with exon 3 missense mutationsand expression of a shorter-length protein in samples with splicesite mutations (Fig. 1C). Mutations in β-catenin also correlatedwith significant up-regulation of the β-catenin/T-cell factor(TCF) target genes Axin2 (ninefold higher in mutant vs. WT, P <0.01) and Enc1 (21-fold higher in mutant vs. WT, P < 0.001;Fig. 1F) (23, 24). Taken together, our results demonstrate thatmutations leading to stabilization of β-catenin occur frequentlyduring MYC-induced lymphomagenesis.

MYC-Induced Lymphomas Exhibit Addiction to Mutant β-Catenin.Due to the frequency of stabilizing β-catenin mutations in MYC-induced lymphomas, we speculated that β-catenin might be

A

β-catenin

Vinculin

shCo

ntro

l

shRNA:

C D

E

MYC

shβc

at-2

shβc

at-3

shβc

at-4

full-length

Δexon3

B

Time afterinfection

shControl shβcat-2

GFP

Day 1

Day 2

Day 3

Day 41 2 3 4 5 6 7

0

20

40

60

80

100

120

Days after infection

Norm

aliz

ed%

GF P

+

shControlshβcat-2shβcat-3shβcat-4

FshControl shβcat-2 Vector ICAT

β-catenin mutant β-catenin wildtypeβ-catenin mutant β-catenin wildtype

MYC β-catenin Axin2 Enc1 Runx20.0

0.2

0.4

0.6

0.8

1.0

1.2R

elat

ive

mR

NA

expr

essi

on

1 6 1 2 3 4 5 6 1 6 1 2 3 4 5 60

20

40

60

80

100

120

Days after infection

Norm

aliz

ed%

GF P

+

1 6 1 2 3 4 5 6 1 6 1 2 3 4 5 60

20

40

60

80

100

120

Days after infection

Norm

aliz

ed%

GFP

+shControl shβcat-2

Fig. 2. MYC-induced lymphomas exhibit addiction to mutant β-catenin. (A) Western blot analysis of MYC lymphoma cells retrovirally infected with eithercontrol shRNA (shControl) or three independent shRNAs targeting β-catenin (shßcat-2, shßcat-3, and shßcat-4). (B) Relative mRNA expression levels oftransgenic MYC, β-catenin, and three β-catenin target genes (Axin2, Runx2, and Enc1) in MYC lymphoma cells infected with either control or β-catenin shRNA,as analyzed by qRT-PCR. (C) Representative histograms and (D) quantification of GFP fluorescence in MYC lymphoma cells after infection with either controlor β-catenin shRNA coexpressing GFP. (E and F) Quantification of GFP fluorescence over time of MYC lymphoma cell lines mutant or WT for β-catenin andinfected with retrovirus expressing either (E) β-catenin shRNA or (F) the β-catenin inhibitor ICAT. Values represent means ± SD of two cell lines with WTβ-catenin and two cell lines with mutant β-catenin and three independent experiments per cell line. Data are normalized to the percentage of GFP+ cells atday 1 after infection.

E3318 | www.pnas.org/cgi/doi/10.1073/pnas.1406123111 Choi et al.

Dow

nloa

ded

by g

uest

on

Mar

ch 3

, 202

1

Page 4: Addiction to multiple oncogenes can be exploited to prevent ...Addiction to multiple oncogenes can be exploited to prevent the emergence of therapeutic resistance Peter S. Choi, Yulin

required for tumor maintenance. We used a retroviral vectorexpressing β-catenin–specific shRNAs and a GFP reporter toinfect MYC lymphoma cell lines known to be mutant or WT forβ-catenin. Efficient knockdown was observed with three dif-ferent shRNAs targeting β-catenin (Fig. 2A), and suppressionof β-catenin levels resulted in significant depletion of GFP+

cells over time compared with a control nontargeting shRNA(Fig. 2 C and D). Sensitivity to β-catenin knockdown was alsoobserved for an additional 13 MYC lymphoma lines that werecharacterized as mutant for β-catenin (Fig. S2A). In contrast,MYC lymphoma lines identified as WT for β-catenin werecompletely resistant to β-catenin suppression (Fig. 2E), con-firming a strong correlation between β-catenin mutation statusand dependency. Therefore, MYC-induced lymphomas arehighly dependent on continued expression of mutant β-catenin.ICAT (also known as CTNNBIP1) is an endogenously ex-

pressed gene that was first identified in a screen of β-catenininteracting proteins and found to inhibit the interaction betweenβ-catenin and its transcriptional partner TCF (25). Similar toβ-catenin knockdown by shRNA, overexpression of ICAT alsoresulted in specific depletion of MYC lymphoma cells expressingmutant, but not WT, β-catenin (Fig. 2F). In contrast, expressionof dominant-negative TCF4 (TCF4ΔN31) was growth suppres-sive to both β-catenin WT and mutant MYC lymphomas (Fig.S2B). Several reported chemical inhibitors of β-catenin also didnot exhibit selective activity when used to treat either β-cateninWT or mutant MYC lymphomas (Fig. S2C), demonstrating thatthese compounds have significant off-target effects in MYClymphoma cells. Collectively, these results indicate that MYClymphomas are sensitive to inhibition of mutant β-catenin, eitherthrough shRNA-mediated knockdown or by expression of theinhibitory protein ICAT.

Inducible Inhibition of β-Catenin Through a Chemical-Genetic Approach.In lymphoma cells from the Eμ-tTA/tetO-MYC model, expressionof the MYC transgene is driven by the tetracycline-regulatorysystem (tet-system) and is suppressed in the presence of tetracy-cline or tetracycline analogs such as doxycycline (DOX). To sup-press β-catenin activity in a similarly inducible manner, we tookadvantage of a recently developed expression system in whichprotein stability can be controlled by the presence or absence ofa specific chemical ligand (26). A gene of interest can be fused in-frame to a mutant destabilized form of Escherichia coli dihy-drofolate reductase (ecDHFR), resulting in constitutive degrada-tion of the entire protein product. Addition of the DHFR ligandtrimethoprim (TMP) leads to rapid stabilization of the fusionprotein (Fig. 3A). TMP is highly selective for bacterial DHFR andhas favorable pharmacological properties, making the ecDHFR-TMP conditional expression system well suited for both in vitro andin vivo studies in mammalian cells. To generate an inducible in-hibitor of β-catenin, we fused the destabilizing ecDHFR domain(DD) to the N terminus of ICAT (Fig. 3A) and stably expressed theDD-ICAT fusion in a MYC lymphoma cell line. Levels of DD-ICAT were low in the absence of TMP and were rapidly inducedwith addition of TMP (Fig. 3B).We first confirmed orthogonal regulation of MYC and DD-

ICAT by treating cells with DOX alone, TMP alone, or a com-bination of DOX and TMP. MYC levels were only suppressed inthe presence of DOX, whereas DD-ICAT expression was onlyinduced in the presence of TMP (Fig. 3C). Next, we investigatedthe effects of DD-ICAT induction on β-catenin activity. Treat-ment with TMP and stabilization of DD-ICAT resulted in down-regulation of the β-catenin/TCF target genes Axin2, Enc1,Mecom,and Runx2 (Fig. 3D) (23, 24, 27, 28), as well as down-regulation ofa luciferase reporter driven by a 7× repeat of the canonical TCFbinding sequence (Fig. 3E) (29). Our results demonstrate theactivity of a novel inducible inhibitor of β-catenin that can be

temporally controlled and effectively suppresses β-catenin–dependent transcription.

Inhibition of β-Catenin Induces Apoptosis Through Regulation of Bimand Bcl-xL Expression. We next sought to take advantage of theDD-ICAT inducible system to characterize the dependence ofMYC-induced lymphomas on the β-catenin pathway. MYClymphoma cells expressing DD-ICAT were treated with TMP,and levels of cell death over time were measured by staining forthe cell surface apoptotic marker Annexin-V and the DNA dye7-aminoactinomycin D (7-AAD). Inhibition of β-catenin activityvia induced expression of DD-ICAT resulted in an increase inthe total percentage of apoptotic cells, from a basal level of∼13% Annexin-V+ cells before treatment to 64% Annexin-V+

cells after 3 d of treatment with TMP (Fig. 4A). In contrast toMYC inactivation, which induces both cell cycle arrest and ap-optosis (17), we found that β-catenin inhibition had no signifi-cant effect on the percentage of proliferating cells, as determinedby staining for DNA content with propidium iodide (PI) (Fig.4E). As a control, TMP-induced expression of a DD-YFP fusionprotein had no effect on the survival of MYC lymphoma cellsboth in vitro and in vivo (Fig. S3 A–D), demonstrating the lack ofany nonspecific toxicity due to expression of the ecDHFRdestabilizing domain or treatment with TMP.

A

B

α-tubulin

DOX:TMP:

MYC

DD-ICAT

+−−

−−+

++

D

C

Eβ-catenin/TCF target genes

Control DD-ICAT0.0

0.2

0.4

0.6

0.8

1.0

1.2

Rela

tive

luci

fera

seac

tivi ty

(Fire

fly/R

enilla

luci

fera

se)

Destabilizingdomain

DD ICATβ-catenininhibitor

+TMPICATDD

DD-ICATstable

DD-ICATdegraded

LTR

pMSCV-DD-ICAT

α-tubulin

Time of TMP treatment (hours)

DD-ICAT

0 0.5 1 2 3 4 8

βcatTCF

Target genes

ICATDD

βcat

ICATDD

β-cateninoff

−TMP

7xTCF luciferase reporter

Axin2 Enc1 Mecom Runx20.0

0.2

0.4

0.6

0.8

1.0

1.2

Rela

tive

expr

essi

on

– TMP+ TMP

– TMP+ TMP

Fig. 3. Inducible inhibition of β-catenin through a chemical-genetic ap-proach. (A) Schematic outlining the inducible DD-ICAT β-catenin inhibitorconsisting of a degradation domain (DD) and the β-catenin inhibitor ICAT.DD-ICAT is constitutively degraded and becomes stabilized in the presenceof the ligand TMP. Once stabilized, DD-ICAT prevents the interaction be-tween β-catenin and TCF and effectively inhibits β-catenin transcriptionalactivity. (B) Western blot analysis of MYC lymphoma cells expressing DD-ICAT and treated with TMP for 0–8 h. (C) Western blot analysis of MYClymphoma cells expressing DD-ICAT treated with either DOX alone, TMPalone, or both DOX and TMP. (D) Relative expression of β-catenin targetgenes Axin2, Enc1, Mecom/Evi1, and Runx2 in MYC lymphoma cellsexpressing DD-ICAT and treated with either DMSO or TMP for 2 d. (E)Quantification of luciferase signals in MYC lymphoma cells infected witha 7× TCF binding site luciferase reporter and DD-ICAT, treated with eitherDMSO or TMP for 24 h. Values are means ± SD of normalized firefly/renillaluciferase ratios from three independent experiments.

Choi et al. PNAS | Published online July 28, 2014 | E3319

MED

ICALSC

IENCE

SPN

ASPL

US

Dow

nloa

ded

by g

uest

on

Mar

ch 3

, 202

1

Page 5: Addiction to multiple oncogenes can be exploited to prevent ...Addiction to multiple oncogenes can be exploited to prevent the emergence of therapeutic resistance Peter S. Choi, Yulin

To further characterize the mechanism of apoptosis due toinhibition of β-catenin, we collected cells that were treated withTMP for 0, 1, 2, and 3 d and screened the expression of severalpro- and antiapoptotic factors by Western blot (Fig. 4B). In-hibition of β-catenin led to an increase in levels of cleaved cas-pase-3, which correlated with an induction in levels of Bim anda reduction in levels of Bcl-xL at both the mRNA and proteinlevels (Fig. 4 B and C). We observed no changes in the levels ofBirc5, an antiapoptotic factor shown to be a transcriptionaltarget of β-catenin/TCF in colon carcinoma cells (30) (Fig. 4B).In addition, shRNA-mediated knockdown of Bim or ectopicexpression of Bcl-xL was sufficient to significantly dampen theapoptotic response to β-catenin inhibition (Fig. 4D). Theseresults demonstrate that β-catenin is required for the survival ofMYC-induced lymphomas by promoting the expression of Bcl-xLand suppressing the expression of Bim.

Combined Inactivation of MYC and β-Catenin Synergizes in TumorRegression and Is Sufficient to Prevent Tumor Recurrence. As wehave previously shown, MYC-induced lymphomas fail to un-dergo sustained tumor regression upon MYC inactivation in theabsence of an intact host immune system (20). We speculatedthat targeting a secondary addiction pathway in addition to MYCmight be sufficient to prevent tumor recurrence. To test thishypothesis, a cohort of SCID mice was transplanted s.c. withMYC-induced lymphoma cells expressing DD-ICAT and a fireflyluciferase reporter (LUC2). Tumors were allowed to establish,and mice were treated with either DOX alone, TMP alone, ora combination of DOX and TMP. Tumor burden was monitoredover time by bioluminescence imaging (Fig. 5A). Although in-

hibition of a single oncogene induced significant short-term re-gression, all tumors eventually recurred, with resistant tumorcells emerging after ∼2 wk of continuous treatment. Strikingly,the combined inhibition of both MYC and β-catenin resulted inmore rapid tumor regression and long-term recurrence-free sur-vival, despite cessation of treatment after only 16 d (Fig. 5 A–D).Control DD-YFP–expressing tumors developed resistance tocombination treatment with DOX+TMP (Fig. S3E), confirmingthe absence of nonspecific toxicity due to TMP treatment or ex-pression of the DD domain. Our findings demonstrate that theoutgrowth of resistant cells can be effectively suppressed by thecombined inhibition of both MYC and β-catenin.To determine the mechanism of enhanced tumor regression

we observed with combined targeting of MYC and β-catenin, westained tumors treated with DOX alone, TMP alone, or DOX+TMP, for the proliferation marker Ki-67 and the apoptosis markercleaved caspase-3 (Fig. 6). Inactivation of MYC or combined in-activation of both MYC and β-catenin for 4 d resulted in a greaterreduction in Ki-67+ cells compared with inhibition of β-cateninalone (Ki-67+ cells: MYC off = 2.0% and MYC+β-cat off = 0.7%vs. β-cat off = 32.8%, P < 0.001; Fig. 6B). Inhibition of β-cateninor combined inactivation of both MYC and β-catenin resulted inmore apoptosis after 2 d of treatment compared with withdrawalof MYC alone (cleaved caspase-3+ cells: β-cat off = 10.1% andMYC+β-cat off = 8.8% vs. MYC off = 1.7%, P < 0.01; Fig. 6C).Thus, the simultaneous inactivation of MYC and β-catenin pro-motes a greater decrease in proliferation and increase in apoptosiscompared with inhibition of either oncogene alone. Taken to-gether, our results demonstrate that combined inhibition of MYCand β-catenin in MYC-induced lymphomas synergizes to promote

A

Annexin-V

7-A

AD

β-catenin on β-catenin off 1d β-catenin off 2d β-catenin off 3d

3.6% 6.4% 13.7% 14.4%

7.4% 10.3% 29.1% 46.7%

B

DNA content

Cou

ntUntreated β-cat off

(+TMP)MYC off(+DOX)

DD-ICAT

MYC

Cleavedcaspase-3

α-tubulin

Bcl-xL

Bim

0 1 2 3

Days of β-catenin off

Birc5

C

Vector

Bimsh

RNA

Bcl-xL

cDNA

0

10

20

30

40

50

60

%An

nexin

-V+

apop

totic

*****

E

D

0.00.20.40.60.81.01.21.41.6

Days of β-cat off

Rela

tive

mRN

Aex

p res

sion

0 1 2 3

BimmRNA Bcl-xL mRNA

Fig. 4. Mutant β-catenin is required for survival of MYC lymphomas through regulation of apoptotic factors. (A) MYC lymphoma cells expressing DD-ICATwere treated with TMP and levels of apoptosis were tracked by staining for Annexin-V-PE and 7AAD. (B) Western blot analysis for apoptotic proteins in cellstreated with TMP for 0, 1, 2, and 3 d. (C) Levels of proapoptotic Bim and antiapoptotic Bcl-xL mRNA in cells treated with TMP for 0, 1, 2, and 3 d, as determinedby qRT-PCR. (D) Quantification of apoptosis as measured by staining with Annexin-V after treatment with TMP in cells expressing Bim shRNA or Bcl-xL cDNAcompared with empty vector control. All values represent means of at least three independent experiments ± SEM. *P < 0.05, **P < 0.01, ***P < 0.001. (E)MYC lymphoma cells expressing DD-ICAT were either untreated, treated with TMP (1 μM), or treated with DOX for 24 h. Cells were fixed and stained with PIto measure DNA content. Histogram events are gated on viable cells.

E3320 | www.pnas.org/cgi/doi/10.1073/pnas.1406123111 Choi et al.

Dow

nloa

ded

by g

uest

on

Mar

ch 3

, 202

1

Page 6: Addiction to multiple oncogenes can be exploited to prevent ...Addiction to multiple oncogenes can be exploited to prevent the emergence of therapeutic resistance Peter S. Choi, Yulin

tumor regression and is sufficient to prevent the developmentof resistance.

DiscussionThe emergence of therapeutic resistance represents a significantobstacle toward achieving long-term remissions with oncogene-targeted therapy. Although rational combinations of targetedagents offer the greatest potential for preventing tumor re-currence, it remains unclear which specific combinations wouldprovide the greatest therapeutic benefit. Here, we took advan-tage of a transgenic mouse model system in which tumors fre-quently recur after MYC inactivation to investigate the efficacyof simultaneously inhibiting two driver oncogenic events. Wediscovered that MYC-induced lymphomas frequently acquireactivating mutations in a splice acceptor site of β-catenin resultingin its stabilization. In turn, this mutation correlated with markedsensitivity of tumors to β-catenin inhibition. By generating a novelinducible inhibitor of β-catenin, we were able to study the in vivoconsequences of targeting either MYC or β-catenin independentlyvs. combined inhibition of both oncogenes. The inhibition of ei-ther oncogene alone uniformly gives rise to resistance, whereastargeting two oncogenes results in more rapid and sustained tumorregression. Our results specifically identify β-catenin activationthrough a mutation in a splice acceptor site as an additional driverevent associated with MYC-induced lymphomagenesis. Further,we illustrate that cancers can exhibit addiction to multiple onco-genic drivers and that the combined targeting of two independently

required oncogenic dependencies is sufficient to prevent the out-growth of resistant cells.β-catenin is tightly regulated at the posttranslational level

through phosphorylation of residues in exon 3 (Fig. 1D), leadingto ubiquitination and proteasome-mediated degradation (31).Mutations that abrogate β-catenin phosphorylation and result inprotein stabilization have been found in a variety of humancancers (32). We discovered a previously unidentified mecha-nism of β-catenin stabilization through point mutations in theexon 3 splice acceptor site. Although splice site mutations havebeen associated with loss of function in several tumor suppressorgenes, such as TP53, RB1, and BRCA1 (33–36), there have beenrelatively few reports of splicing mutations resulting in the gainof function of an oncogene. Examples to date include deletionsat the intron 10–exon 11 boundary of the receptor tyrosine ki-nase c-KIT that result in loss of an auto-inhibitory region andconstitutive activation of the protein (37, 38), and splice acceptorsite mutations in c-MET that lead to skipping of exon 14 anddeletion of a juxtamembrane domain important for negativeregulation (39). Intragenic deletions in β-catenin that remove allor part of exon 3 have been reported in colorectal carcinoma(40). We conclude that the β-catenin oncogene can also becomeactivated through single nucleotide substitutions occurring at theexon 3 splice acceptor site.Overexpression of MYC due to genomic amplification, trans-

location, or via activation of upstream regulators represents oneof the most common features in human cancer. In addition, the

A

B

MYC offβ-catenin offMYC off + β-catenin off

C

MYC off

Day 0 Day 8 Day 24/26

β-catenin off

MYC off + β-catenin off

0.8

0.6

0.4

0.2

x10 10 (p/s/cm^2/sr)

D

Background

***

MYC offβ-catenin off

MYC off + β-catenin off

**

Minimum residual disease

105

106

107

108

109

1010

1011

Tota

lflu

x( P

hoto

ns/s

ec)

***

MYC off β-cat off MYC off+β-cat off

Treatment stopped

0 10 20 30 40 50 60 70 80 900

20

40

60

80

100

Time after oncogene inactivation (Days)

Recu

rren

ce- fr

e esu

rviva

l (%

)

0 4 8 12 16 20 24 28105

106

107

108

109

1010

1011

1012

60 90Time after oncogene inactivation (Days)

Tota

lflu

x(P

hoto

ns/s

e c)

Fig. 5. Combined targeting of MYC and β-catenin prevents tumor recurrence. (A) MYC lymphoma cells expressing LUC2 and DD-ICAT were transplanteds.c. into SCID recipients. Bioluminescence signal was quantified over time after initiation of treatment to inactivate MYC (+DOX), β-catenin (+TMP), or bothMYC and β-catenin (+DOX/TMP). Values are means ± SEM (DD-ICAT +DOX, n = 7; DD-ICAT +TMP, n = 3; DD-ICAT +DOX/TMP, n = 5). (B) Representativebioluminescence images of tumor-bearing mice at 0, 8, and 24 (+TMP, +DOX/TMP) or 26 (+DOX) d after initiation of treatment. (C) Recurrence-free survival ofmice transplanted with MYC lymphomas and treated with DOX, TMP, or both DOX and TMP as in A. Tumors were scored as recurrences on the day when thebioluminescence signal increased after initial tumor regression. Survival curves were compared by the log-rank test. (D) Quantification of minimum residualdisease in transplanted mice treated with DOX, TMP, or both DOX and TMP. Values are means ± 95% CIs. *P < 0.05, **P < 0.01, ***P < 0.001.

Choi et al. PNAS | Published online July 28, 2014 | E3321

MED

ICALSC

IENCE

SPN

ASPL

US

Dow

nloa

ded

by g

uest

on

Mar

ch 3

, 202

1

Page 7: Addiction to multiple oncogenes can be exploited to prevent ...Addiction to multiple oncogenes can be exploited to prevent the emergence of therapeutic resistance Peter S. Choi, Yulin

WNT/β-catenin pathway is also frequently activated in cancer,due to mutations in β-catenin or loss-of-function mutations innegative regulators such as APC and AXIN2 (32). Interestingly,although MYC is a prominent downstream target of WNT/β-catenin signaling (41), our findings suggest a strong selectivepressure for β-catenin functions independent of MYC in ourlymphoma model. Wnt/β-catenin pathway activation has also beenobserved in models of MYC-driven liver cancer, suggesting MYCand β-catenin can functionally cooperate during tumorigenesis indiverse tissue contexts (42, 43). It has also been reported thatlymphomas initiated by activated β-catenin frequently exhibitoverexpression of MYC due to genomic amplification or rear-rangement and that MYC is required for β-catenin driven lym-phomagenesis (44–46), further supporting a cooperative model forMYC and β-catenin function during tumorigenesis.We found that β-catenin inhibition resulted in the induction of

apoptosis, which closely correlated with decreased levels of theantiapoptotic factor Bcl-xL (Fig. 4 A–C). Ectopic expression ofBcl-XL was also sufficient to completely abrogate the cell deathassociated with loss of β-catenin activity (Fig. 4D). β-catenin hasbeen shown to directly activate the expression of Bcl-xL (45, 47,48), and we found that Bcl-xL is a critical downstream mediatorof the addiction to β-catenin in MYC-induced lymphomas. Thephenotypic consequences of β-catenin inhibition may be explainedby an “oncogenic shock” model, in which the rapid loss of survivalcues such as Bcl-xL and the persistence of proapoptotic signalingresults in an imbalance that favors cell death (49). Interestingly,we observed a synergistic effect on the rate of tumor regression

from the inhibition of both MYC and β-catenin (Fig. 5A) com-pared with inhibition of either oncogene alone. The targeting ofeach oncogene elicited distinct cellular outcomes, with MYC in-activation resulting primarily in cell cycle arrest and β-catenininhibition leading mainly to the induction of apoptosis. The ac-celerated tumor regression upon inactivation of both oncogenescan then be attributed to the collective loss of both proliferationand survival signals. Taken together, these results indicate thatMYC and β-catenin appear to play complementary roles to pro-mote and maintain tumorigenesis.The effectiveness of combination therapy in our model system

may be explained by the low probability of tumor cells with cross-resistance to inhibition of both oncogenes. A similar effect isbelieved to be responsible for the success of other types ofcombination therapy, including highly active antiretroviral ther-apy (HAART) for HIV infection, in which the use of a mixtureof inhibitors targeting different viral enzymes is effective atpreventing the emergence of virions that are resistant to anysingle drug (12, 13). We conclude that the suppression of re-sistance with combination targeted therapy will be most effectiveby selecting targets that are each essential for tumor mainte-nance but are sufficiently independent as to minimize the like-lihood of cross-resistance arising from a single mutation.We provided a proof of concept that the simultaneous tar-

geted inhibition of two mutually required oncogenes may besufficient to prevent the emergence of therapeutic resistance.Additional factors, such as tumor burden and heterogeneity,overall tumor mutation rate, and tumor-host interactions, will

Ki-67

AUntreated

0 2 40

20

40

60

80

100

Days oftreatment:

%Ki

67-p

osi ti

ve

2 4 2 4

MYC off β-cat off MYC andβ-cat off

B CProliferation (Ki-67 staining)

00

2

4

6

8

10

12

14

%C

leav

edca

spas

e-3+

2 4Days oftreatment:

2 4 2 4

MYC off β-cat off MYC andβ-cat off

Apoptosis (Cleaved caspase-3 staining)

DAPI

Cleavedcaspase-3

DAPI

MYC off2 days 4 days 2 days 4 days 2 days 4 days

β-catenin off MYC and β-catenin off

*** *** ****

Fig. 6. Combined inactivation of MYC and β-catenin synergizes to inhibit proliferation and promote apoptosis in vivo. (A) MYC lymphoma cells expressingDD-ICAT were transplanted s.c. into recipient SCID mice. Mice were either left untreated or treated with DOX (MYC off), TMP (β-catenin off), or both DOX andTMP (MYC and β-catenin off) for 2–4 d. Tumors were harvested and stained for the proliferation marker Ki-67 and the apoptotic marker cleaved caspase-3.Nuclei were stained using DAPI. Representative images are shown. (B and C) Quantification of Ki-67 and cleaved caspase-3 staining, in two to three in-dependent tumors from each time point and five fields per tumor. Values represent means ± SEM. **P < 0.01, ***P < 0.001.

E3322 | www.pnas.org/cgi/doi/10.1073/pnas.1406123111 Choi et al.

Dow

nloa

ded

by g

uest

on

Mar

ch 3

, 202

1

Page 8: Addiction to multiple oncogenes can be exploited to prevent ...Addiction to multiple oncogenes can be exploited to prevent the emergence of therapeutic resistance Peter S. Choi, Yulin

likely play a critical role in determining the number and types oftargeted therapies that will be most effective in combination. Inaddition, the ability to target many known and newly discovereddriver events will largely depend on the development of thera-peutics against novel classes of drug targets, such as transcriptionfactors and chromatin regulators. However, as we continue touncover the full landscape of genetic alterations in cancer, theidentification and combined targeting of multiple oncogene ad-diction pathways offers great potential toward preventing re-sistance and improving therapeutic outcomes.

Materials and MethodsTransgenic Mice and Lymphoma Cell Lines. All animal experiments were ap-proved by the Institutional Animal Care and Use Committee at StanfordUniversity. MYC lymphomas and lymphoma-derived cell lines were named forthe identifying number of the mouse from which they were obtained. Celllines were grown in RPMI 1640 media supplemented with 10% (vol/vol) FBS,penicillin/streptomycin, and 50 μM β-mercaptoethanol. MYC was inactivatedby treatment with 20 ng/mL of doxycycline.

Sequencing of β-Catenin. Genomic DNAwas extracted from primary tumors orcell lines using the Qiagen DNeasy kit or by standard phenol-chloroformpurification. A region spanning intron 2 and exon 3 of β-catenin was PCRamplified (see primers in Table S2) and directly sequenced (Sequetech).Mutant peaks were detected by manual inspection of sequencing chroma-tograms and with Mutation Surveyor (Softgenetics). To sequence β-catenincDNA, RNA was first isolated from cells using the Nucleospin RNA II kit(Machery-Nagel), and first-strand cDNA synthesis was carried out with Su-perScript II (Invitrogen) using oligo-dT primers. The 5′ region of the β-catenintranscript was amplified by PCR (see primers in Table S2) and directlysequenced.

Proliferation and Apoptosis Assays. MYC lymphoma cells were infected withretrovirus, and the percentage of GFP+ infected cells was tracked over timeusing a FACScan flow cytometer. Apoptosis was quantified by staining withAnnexin-V-PE and 7-AAD (BD Biosciences). To assess cell cycle status, cellswere first fixed in 70% (vol/vol) ethanol in PBS at −20 °C for at least 12 h.After washing in PBS, cells were resuspended in PI staining solution (PBS/0.1% TritonX-100, 0.2 mg/mL RNase A, and 20 μg/mL PI) and incubated for30 min at room temperature while protected from light. DNA content wasthen measured using a FACScan flow cytometer. All flow cytometric datawere analyzed with Flowjo software (Tree Star Inc.).

Plasmids. For β-catenin shRNA-mediated knockdown, oligos were PCR am-plified and cloned into the XhoI/EcoRI sites of the retroviral LMP vector(RNAi Codex/Openbiosystems). Sequences are listed in Table S3. To generatepMSCV-ICAT-PIG, ICAT was PCR amplified from murine brain cDNA and li-gated into BglII and XhoI sites. To create pMSCV-TCFΔN31-PIG, dominant-negative TCF was PCR amplified from pPGS-CITEneo-TCFΔN31 (obtained

from Karl Sylvester, Stanford University, Stanford, CA) and ligated into BglIIand XhoI sites.

Inducible β-Catenin Inhibitor. The N-terminal ecDHFR-DD-ICAT fusion was firstgenerated using splicing by overlap extension (SOE) PCR. The fused PCRproduct was then ligated into pMSCV-puro (BglII/XhoI). The N-terminalecDHFR-DD-YFP fusion was PCR amplified directly from pBMN-DHFR-(R12YG67S Y100I)-YFP-HA-IRES-HcRed-tandem (obtained from Tom Wandless,Stanford University, Stanford, CA) and ligated into pMSCV-puro (BglII/XhoI).Stabilization of fusion proteins was induced by treatment with 1 μM ofTMP (Sigma).

TCF Luciferase Reporter Assay. The 7TFP lentiviral Wnt reporter (29) wasobtained from Addgene (plasmid 24308) and modified to replace the pu-romycin resistance gene with the neomycin resistance gene, generating7TFN. MYC lymphoma cells expressing renilla luciferase alone or both renillaluciferase and DD-ICAT were stably transduced with the 7TFN reporter. TheDual-Glo Luciferase assay system (Promega) was used to quantify the relativelevels of luciferase activity ± 1 μM TMP. Wnt-responsive firefly luciferasevalues were normalized to control renilla luciferase values.

Tumor Transplantation and Bioluminescence Imaging. Lymphoma cells weretransplanted s.c. into the flank of immunodeficient SCID mice. Tumors weremonitored by calipermeasurement and bioluminescence imaging. Treatmentof mice with DOX, TMP, or DOX+TMP was initiated when tumors reachedsimilar bioluminescent signals. Bioluminescent signal from tumors wasmeasured by i.p. injection of mice with 100 μL of a solution of D-Luciferin (30mg/mL) and imaging with the IVIS Spectrum (Perkin-Elmer). Images werecollected and analyzed using Living Image software (Perkin-Elmer). To in-activate MYC, mice were injected once i.p. with 10 μg of DOX in PBS andmaintained on drinking water containing DOX (100 μg/mL), which wasrefreshed weekly. To induce DD-ICAT or DD-YFP, mice were treated by dailyi.p. injection of 5 mg of pharmaceutical-grade trimethoprim lactate (ABT-590; Alpha Diagnostic International) and maintained on drinking watercontaining a suspension of 1 mg/mL of TMP (Trimethoprim-Sulfamethox-azole suspension; Qualitest Pharmaceuticals), which was refreshed every 3 d.

Immunofluorescence. Tumor-bearing mice were killed at 0, 2, and 4 d afterMYC inactivation, and transplanted tumors were harvested and fixed informalin. Paraffin-embedded sections were immunostained with anti–Ki-67(BD Biosciences, 556003) or anti-cleaved caspase-3 (Cell Signaling Techology,9661). All antibodies are listed in Table S4. Images were obtained on a Nikonmicroscope and analyzed using CellProfiler software (www.cellprofiler.org/).

ACKNOWLEDGMENTS. We thank members of the D.W.F. laboratory forhelpful suggestions and guidance, Tom Wandless (Stanford University) forthe ecDHFR degradation domain constructs, and Karl Sylvester (StanfordUniversity) for the dominant-negative TCF construct. This work was sup-ported by National Institutes of Health (NIH) Training Grant 5 T32 AI07290 (toP.S.C.), NIH Grants R01CA170378 and U54CA149145 (to D.W.F.), and Leukemiaand Lymphoma Society Translational Research Grant R6223-07 (to D.W.F.).

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

2. Druker BJ, et al. (2001) Efficacy and safety of a specific inhibitor of the BCR-ABL ty-rosine kinase in chronic myeloid leukemia. N Engl J Med 344(14):1031–1037.

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

4. Flaherty KT, et al. (2010) Inhibition of mutated, activated BRAF in metastatic mela-noma. N Engl J Med 363(9):809–819.

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

6. Pao W, et al. (2005) Acquired resistance of lung adenocarcinomas to gefitinib or erlotinibis associated with a second mutation in the EGFR kinase domain. PLoS Med 2(3):e73.

7. Glickman MS, Sawyers CL (2012) Converting cancer therapies into cures: Lessons frominfectious diseases. Cell 148(6):1089–1098.

8. Shah NP, et al. (2007) Sequential ABL kinase inhibitor therapy selects for compound drug-resistant BCR-ABL mutations with altered oncogenic potency. J Clin Invest 117(9):2562.

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

10. Nazarian R, et al. (2010) Melanomas acquire resistance to B-RAF(V600E) inhibition byRTK or N-RAS upregulation. Nature 468(7326):973–977.

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

12. Gulick RM, et al. (1997) Treatment with indinavir, zidovudine, and lamivudine inadults with human immunodeficiency virus infection and prior antiretroviral therapy.N Engl J Med 337(11):734–739.

13. Hammer SM, et al. (1997) A controlled trial of two nucleoside analogues plus indinavir inpersons with human immunodeficiency virus infection and CD4 cell counts of 200 per cubicmillimeter or less. AIDS Clinical Trials Group 320 Study Team.N Engl J Med 337(11):725–733.

14. Fox W, Ellard GA, Mitchison DA (1999) Studies on the treatment of tuberculosis un-dertaken by the British Medical Research Council tuberculosis units, 1946-1986, withrelevant subsequent publications. Int J Tuberc Lung Dis 3(10, Suppl 2):S231–S279.

15. Bozic I, et al. (2013) Evolutionary dynamics of cancer in response to targeted com-bination therapy. eLife 2:e00747.

16. Beroukhim R, et al. (2010) The landscape of somatic copy-number alteration acrosshuman cancers. Nature 463(7283):899–905.

17. Felsher DW, Bishop JM (1999) Reversible tumorigenesis by MYC in hematopoieticlineages. Mol Cell 4(2):199–207.

18. Giuriato S, et al. (2006) Sustained regression of tumors upon MYC inactivation re-quires p53 or thrombospondin-1 to reverse the angiogenic switch. Proc Natl Acad SciUSA 103(44):16266–16271.

19. Wu CH, et al. (2007) Cellular senescence is an important mechanism of tumor re-gression upon c-Myc inactivation. Proc Natl Acad Sci USA 104(32):13028–13033.

20. Rakhra K, et al. (2010) CD4(+) T cells contribute to the remodeling of the microen-vironment required for sustained tumor regression upon oncogene inactivation.Cancer Cell 18(5):485–498.

21. Choi PS, et al. (2011) Lymphomas that recur after MYC suppression continue to exhibitoncogene addiction. Proc Natl Acad Sci USA 108(42):17432–17437.

22. Eischen CM, Weber JD, Roussel MF, Sherr CJ, Cleveland JL (1999) Disruption of theARF-Mdm2-p53 tumor suppressor pathway in Myc-induced lymphomagenesis. GenesDev 13(20):2658–2669.

Choi et al. PNAS | Published online July 28, 2014 | E3323

MED

ICALSC

IENCE

SPN

ASPL

US

Dow

nloa

ded

by g

uest

on

Mar

ch 3

, 202

1

Page 9: Addiction to multiple oncogenes can be exploited to prevent ...Addiction to multiple oncogenes can be exploited to prevent the emergence of therapeutic resistance Peter S. Choi, Yulin

23. Fujita M, et al. (2001) Up-regulation of the ectodermal-neural cortex 1 (ENC1) gene,a downstream target of the beta-catenin/T-cell factor complex, in colorectal carci-nomas. Cancer Res 61(21):7722–7726.

24. Jho EH, et al. (2002) Wnt/beta-catenin/Tcf signaling induces the transcription ofAxin2, a negative regulator of the signaling pathway. Mol Cell Biol 22(4):1172–1183.

25. Tago K, et al. (2000) Inhibition of Wnt signaling by ICAT, a novel beta-catenin-interacting protein. Genes Dev 14(14):1741–1749.

26. Iwamoto M, Björklund T, Lundberg C, Kirik D, Wandless TJ (2010) A general chemicalmethod to regulate protein stability in the mammalian central nervous system. ChemBiol 17(9):981–988.

27. Dong YF, Soung Y, Schwarz EM, O’Keefe RJ, Drissi H (2006) Wnt induction of chon-drocyte hypertrophy through the Runx2 transcription factor. J Cell Physiol 208(1):77–86.

28. Wei W, et al. (2011) Biphasic and dosage-dependent regulation of osteoclastogenesisby β-catenin. Mol Cell Biol 31(23):4706–4719.

29. Fuerer C, Nusse R (2010) Lentiviral vectors to probe and manipulate the Wnt signalingpathway. PLoS ONE 5(2):e9370.

30. Zhang T, et al. (2001) Evidence that APC regulates survivin expression: A possiblemechanism contributing to the stem cell origin of colon cancer. Cancer Res 61(24):8664–8667.

31. Clevers H, Nusse R (2012) Wnt/β-catenin signaling and disease. Cell 149(6):1192–1205.32. Polakis P (2000) Wnt signaling and cancer. Genes Dev 14(15):1837–1851.33. Holmila R, Fouquet C, Cadranel J, Zalcman G, Soussi T (2003) Splice mutations in the

p53 gene: Case report and review of the literature. Hum Mutat 21(1):101–102.34. Horowitz JM, et al. (1990) Frequent inactivation of the retinoblastoma anti-oncogene

is restricted to a subset of human tumor cells. Proc Natl Acad Sci USA 87(7):2775–2779.35. Mazoyer S, et al. (1998) A BRCA1 nonsense mutation causes exon skipping. Am J Hum

Genet 62(3):713–715.36. Takahashi T, D’Amico D, Chiba I, Buchhagen DL, Minna JD (1990) Identification of

intronic point mutations as an alternative mechanism for p53 inactivation in lungcancer. J Clin Invest 86(1):363–369.

37. Chen LL, et al. (2005) A mutation-created novel intra-exonic pre-mRNA splice sitecauses constitutive activation of KIT in human gastrointestinal stromal tumors. On-cogene 24(26):4271–4280.

38. Corless CL, et al. (2004) KIT gene deletions at the intron 10-exon 11 boundary in GIstromal tumors. J Mol Diagn 6(4):366–370.

39. Asaoka Y, et al. (2010) Gastric cancer cell line Hs746T harbors a splice site mutationof c-Met causing juxtamembrane domain deletion. Biochem Biophys Res Commun394(4):1042–1046.

40. Iwao K, et al. (1998) Activation of the beta-catenin gene by interstitial deletions in-volving exon 3 in primary colorectal carcinomas without adenomatous polyposis colimutations. Cancer Res 58(5):1021–1026.

41. He TC, et al. (1998) Identification of c-MYC as a target of the APC pathway. Science281(5382):1509–1512.

42. Calvisi DF, Factor VM, Loi R, Thorgeirsson SS (2001) Activation of beta-catenin duringhepatocarcinogenesis in transgenic mouse models: Relationship to phenotype andtumor grade. Cancer Res 61(5):2085–2091.

43. de La Coste A, et al. (1998) Somatic mutations of the beta-catenin gene are frequentin mouse and human hepatocellular carcinomas. Proc Natl Acad Sci USA 95(15):8847–8851.

44. Kaveri D, et al. (2013) β-Catenin activation synergizes with Pten loss and Myc over-expression in Notch-independent T-ALL. Blood 122(5):694–704.

45. Dose M, et al. (2014) β-Catenin induces T-cell transformation by promoting genomicinstability. Proc Natl Acad Sci USA 111(1):391–396.

46. Guo Z, et al. (2007) Beta-catenin stabilization stalls the transition from double-posi-tive to single-positive stage and predisposes thymocytes to malignant transformation.Blood 109(12):5463–5472.

47. Rosenbluh J, et al. (2012) β-Catenin-driven cancers require a YAP1 transcriptionalcomplex for survival and tumorigenesis. Cell 151(7):1457–1473.

48. Xie H, Huang Z, Sadim MS, Sun Z (2005) Stabilized beta-catenin extends thymocytesurvival by up-regulating Bcl-xL. J Immunol 175(12):7981–7988.

49. Sharma SV, et al. (2006) A common signaling cascade may underlie “addiction” to theSrc, BCR-ABL, and EGF receptor oncogenes. Cancer Cell 10(5):425–435.

E3324 | www.pnas.org/cgi/doi/10.1073/pnas.1406123111 Choi et al.

Dow

nloa

ded

by g

uest

on

Mar

ch 3

, 202

1