5
Platinum Priority Brief Correspondence Editorial by XXX on pp. x–y of this issue A Functional Genetic Screen Identifies the Phosphoinositide 3-kinase Pathway as a Determinant of Resistance to Fibroblast Growth Factor Receptor Inhibitors in FGFR Mutant Urothelial Cell Carcinoma Liqin Wang a,y , Tonc ´i S ˇ usˇtic ´ a,y , Rodrigo Leite de Oliveira a,y , Cor Lieftink a , Pasi Halonen a , Marieke van de Ven b , Roderick L. Beijersbergen a , Michel M. van den Heuvel c , Rene ´ Bernards a, *, Michiel S. van der Heijden a,c, * a Division of Molecular Carcinogenesis, Cancer Genomics Netherlands, The Netherlands Cancer Institute, Amsterdam, The Netherlands; b Mouse Clinic Intervention Unit, The Netherlands Cancer Institute, Amsterdam, The Netherlands; c Division of Medical Oncology, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands E U R O P E A N U R O L O G Y X X X ( 2 0 1 6 ) X X X X X X ava ilable at www.sciencedirect.com journa l homepage: www.europea nurology.com Article info Article history: Accepted January 6, 2017 Associate Editor: James Catto Keywords: FGFR Bladder cancer PI3K Synergy Abstract Activating mutations and translocations of the FGFR3 gene are commonly seen in urothelial cell carcinoma (UCC) of the bladder and urinary tract. Several fibroblast growth factor receptor (FGFR) inhibitors are currently in clinical development and response rates appear promising for advanced UCC. A common problem with targeted therapeutics is intrinsic or acquired resistance of the cancer cells. To find potential drug targets that can act synergistically with FGFR inhibition, we performed a synthetic lethality screen for the FGFR inhibitor AZD4547 using a short hairpin RNA library targeting the human kinome in the UCC cell line RT112 (FGFR3-TACC3 translocation). We identified multiple members of the phosphoinositide 3-kinase (PI3K) pathway and found that inhibition of PIK3CA acts synergistically with FGFR inhibitors. The PI3K inhibitor BKM120 acted synergistically with inhibition of FGFR in multiple UCC and lung cancer cell lines having FGFR mutations. Consistently, we observed an elevated PI3K-protein kinase B pathway activity resulting from epidermal growth factor receptor or Erb-B2 receptor tyrosine kinase 3 reactivation caused by FGFR inhibition as the underlying molecular mechanism of the synergy. Our data show that feedback pathways activated by FGFR inhibition converge on the PI3K pathway. These findings provide a strong rationale to test FGFR inhibitors in combination with PI3K inhibitors in cancers harboring genetic activation of FGFR genes. # 2017 European Association of Urology. Published by Elsevier B.V. All rights reserved. y These authors contributed equally. * Corresponding authors. Department of Medical Oncology, Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands. Tel. +31 20 512 6245; Fax: +31 20 512 2572 (M.S. van der Heijden); Department of Medical Oncology, Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amster- dam, The Netherlands. Tel. +31 20 512 1952 (R. Bernards). E-mail addresses: [email protected] (R. Bernards), [email protected] (M.S. van der Heijden). EURURO-7215; No. of Pages 5 Please cite this article in press as: Wang L, et al. A Functional Genetic Screen Identifies the Phosphoinositide 3-kinase Pathway as a Determinant of Resistance to Fibroblast Growth Factor Receptor Inhibitors in FGFR Mutant Urothelial Cell Carcinoma. Eur Urol (2017), http://dx.doi.org/10.1016/j.eururo.2017.01.021 http://dx.doi.org/10.1016/j.eururo.2017.01.021 0302-2838/# 2017 European Association of Urology. Published by Elsevier B.V. All rights reserved.

E UROPEAN UROLOGY XXX (2016) XXX–XXX - EU …eu-acme.org/europeanurology/upload_articles/van der h.pdf · E UROPEAN UROLOGY XXX (2016) XXX–XXX ava ilableat journa l homepage:

Embed Size (px)

Citation preview

EURURO-7215; No. of Pages 5

Platinum Priority – Brief CorrespondenceEditorial by XXX on pp. x–y of this issue

A Functional Genetic Screen Identifies the Phosphoinositide

3-kinase Pathway as a Determinant of Resistance to Fibroblast

Growth Factor Receptor Inhibitors in FGFR Mutant Urothelial Cell

Carcinoma

Liqin Wang a,y, Tonci Sustic a,y, Rodrigo Leite de Oliveira a,y, Cor Lieftink a, Pasi Halonen a,Marieke van de Ven b, Roderick L. Beijersbergen a, Michel M. van den Heuvel c, Rene Bernards a,*,Michiel S. van der Heijden a,c,*

a Division of Molecular Carcinogenesis, Cancer Genomics Netherlands, The Netherlands Cancer Institute, Amsterdam, The Netherlands; b Mouse Clinic

Intervention Unit, The Netherlands Cancer Institute, Amsterdam, The Netherlands; c Division of Medical Oncology, Antoni van Leeuwenhoek Hospital,

Amsterdam, The Netherlands

E U R O P E A N U R O L O G Y X X X ( 2 0 1 6 ) X X X – X X X

ava i lable at www.sc iencedirect .com

journa l homepage: www.europea nurology.com

Article info

Article history:Accepted January 6, 2017

Associate Editor:

James Catto

Keywords:

FGFR

Bladder cancer

PI3K

Synergy

Abstract

Activating mutations and translocations of the FGFR3 gene are commonly seen inurothelial cell carcinoma (UCC) of the bladder and urinary tract. Several fibroblastgrowth factor receptor (FGFR) inhibitors are currently in clinical development andresponse rates appear promising for advanced UCC. A common problem with targetedtherapeutics is intrinsic or acquired resistance of the cancer cells. To find potential drugtargets that can act synergistically with FGFR inhibition, we performed a syntheticlethality screen for the FGFR inhibitor AZD4547 using a short hairpin RNA librarytargeting the human kinome in the UCC cell line RT112 (FGFR3-TACC3 translocation).We identified multiple members of the phosphoinositide 3-kinase (PI3K) pathway andfound that inhibition of PIK3CA acts synergistically with FGFR inhibitors. The PI3Kinhibitor BKM120 acted synergistically with inhibition of FGFR in multiple UCC andlung cancer cell lines having FGFR mutations. Consistently, we observed an elevatedPI3K-protein kinase B pathway activity resulting from epidermal growth factor receptoror Erb-B2 receptor tyrosine kinase 3 reactivation caused by FGFR inhibition as theunderlying molecular mechanism of the synergy. Our data show that feedback pathwaysactivated by FGFR inhibition converge on the PI3K pathway. These findings provide astrong rationale to test FGFR inhibitors in combination with PI3K inhibitors in cancersharboring genetic activation of FGFR genes.

# 2017 European Association of Urology. Published by Elsevier B.V. All rights reserved.

y These authors contributed equally.* Corresponding authors. Department of Medical Oncology, Division of Molecular Carcinogenesis,The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands. Tel. +3120 512 6245; Fax: +31 20 512 2572 (M.S. van der Heijden); Department of Medical Oncology, Divisionof Molecular Carcinogenesis, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amster-dam, The Netherlands. Tel. +31 20 512 1952 (R. Bernards).

[email protected] (R. Bernards), [email protected] (M.S. van der Heijden).

E-mail addresses: r.bern

Please cite this article in press as: Wang L, et al. A Functional Genetic Screen Identifies the Phosphoinositide 3-kinase Pathway as aDeterminant of Resistance to Fibroblast Growth Factor Receptor Inhibitors in FGFR Mutant Urothelial Cell Carcinoma. Eur Urol(2017), http://dx.doi.org/10.1016/j.eururo.2017.01.021

http://dx.doi.org/10.1016/j.eururo.2017.01.0210302-2838/# 2017 European Association of Urology. Published by Elsevier B.V. All rights reserved.

E U R O P E A N U R O L O G Y X X X ( 2 0 1 6 ) X X X – X X X2

EURURO-7215; No. of Pages 5

Fibroblast growth factor receptors (FGFRs) are activated in a

subset of urothelial cell carcinoma (UCC), most commonly

by FGFR3 mutation or overexpression of FGFR1 [1]. Clinical

trials with FGFR inhibitors (FGFRi) are currently ongoing in

UCC. Initial results with BGJ398 showed encouraging

response rates [2], though information on the durability

of these responses is currently lacking. Akin to other

molecularly-targeted therapies, resistance is likely to be a

(A) (

(C)

AZD4547

RT

Ctrl.

(E)

0 50 100 150 2000

25

50

75

100

Time (h)

Con

fluen

ce (

%)

Untreated RT112 cells

80nM AZD4547 (FGFR inhibitor)

654321012345678

0 2 4log10(baseMeanA)

log

2Fo

ldC

han

ge

G

AZD4547 vs untreated

shR

NA

ab

un

dan

ce o

f th

e tr

eate

d/u

ntr

eate

d s

amp

les

Average read count of each shRNA in the untreated sample

Fig. 1 – PIK3CA suppression enhances sensitivity to fibroblast growth factor rec(80 nM) and viability was followed using an incucyte assay. Error bars represenRNA (shRNA) screen for enhancers of AZD4547 sensitivity. Human kinome shRNthen left untreated (Ctrl.) for 10 d or treated with 30 nM AZD4547 for 14 d. Aftechain reaction and their abundance was quantified by deep sequencing. (C) Repfrom the shRNA screen. The y-axis shows log2 of the fold change of shRNA abulog10 of the average read count of each shRNA in the untreated sample. shPIK3the presence of at least three independent shRNAs in three biological screen rewas measured by PIK3CA protein levels by western blot. HSP90 protein expressindependent shPIK3CA vectors are indicated by colony formation assay in 50 nM

Please cite this article in press as: Wang L, et al. A Functional GenetDeterminant of Resistance to Fibroblast Growth Factor Receptor

(2017), http://dx.doi.org/10.1016/j.eururo.2017.01.021

major concern. Resistance to FGFR inhibition was observed

in vitro (Fig. 1A, Supplementary materials and methods):

the FGFR-TACC3 translocated cell line RT112 responded

initially, but cells quickly adapted to AZD4547, an inhibitor

of FGFR1, FGFR2, and FGFR3 (weaker activity against

FGFR4). Resistance to targeted therapies often develops

through feedback activation of additional signaling path-

ways: one such example is synergy between FGFRi with

B)

(D)

112

Kinome shRNA library

Ctrl. AZD4547

Culture to select

Recovered bar codes

HSP90

PIK3CA

RT112

eneSymbolAKT3EGFRPIK3CAPIK3R1

eptor (FGFR) inhibition in UCC. (A) RT112 cells were treated with AZD4547t four biological replicates. (B) Outline of synthetic lethality short hairpinA library polyclonal virus was produced to infect RT112 cells, which werer selection, shRNA inserts from both arms were recovered by polymeraseresentation of the relative abundance of the shRNA barcode sequences

ndance of the treated and untreated samples. The x-axis indicates theCA, shPIK3R1, shAKT3, and shEGFR identified as the top hit according toplicates. (D) The level of knockdown of PIK3CA by three different shRNAsion is used for normalization. (E) The functional phenotypes of

AZD4547. The cells were fixed, stained, and photographed after 10 d.

ic Screen Identifies the Phosphoinositide 3-kinase Pathway as aInhibitors in FGFR Mutant Urothelial Cell Carcinoma. Eur Urol

Fig. 2 – Functional and biochemical interaction between fibroblast growth factor receptor (FGFR) and phosphoinositide 3-kinase (PI3K) inhibition inurothelial cell carcinoma. (A,B) Synergistic response of RT112 and JMSU1 to combinations of FGFR (AZD4547) and PI3K (BKM120) inhibitors. RT112 andJMSU1 cells were cultured in increasing concentration of FGFR inhibitor AZD4547 alone, PI3K inhibitor BKM120 (0.5 mM) alone, or their combination.

E U R O P E A N U R O L O G Y X X X ( 2 0 1 6 ) X X X – X X X 3

EURURO-7215; No. of Pages 5

Please cite this article in press as: Wang L, et al. A Functional Genetic Screen Identifies the Phosphoinositide 3-kinase Pathway as aDeterminant of Resistance to Fibroblast Growth Factor Receptor Inhibitors in FGFR Mutant Urothelial Cell Carcinoma. Eur Urol(2017), http://dx.doi.org/10.1016/j.eururo.2017.01.021

E U R O P E A N U R O L O G Y X X X ( 2 0 1 6 ) X X X – X X X4

EURURO-7215; No. of Pages 5

inhibitors of epidermal growth factor receptor (EGFR)

[3]. Feedback mechanisms that render cells insensitive to

kinase inhibition often occur through activation of other

kinases. We screened a short hairpin RNA (shRNA) library

targeting all 518 human kinases and 17 additional kinase-

related genes to find genes whose inhibition enhances the

sensitivity to FGFRi AZD4547 in FGFR3 mutant UCC (Fig. 1B).

RT112 cells were infected with a lentiviral library contain-

ing some 5000 shRNAs and cultured in the absence or

presence of AZD4547 for 14 d. Cells were then harvested,

DNA was isolated, and the relative abundance of shRNA

vectors was measured by deep sequencing. The read counts

were normalized and analyzed with DESeq2 (https://

bioconductor.org/packages/release/bioc/html/DESeq2.

html) to identify shRNAs and their corresponding target

genes that show significant depletion in the presence of

AZD4547 (Supplementary Tables 1 and 2). We observed

multiple components of the PI3K signaling pathway

including the catalytic component PIK3CA, the regulatory

subunit PIK3R1, and the downstream target AKT3 (Fig. 1C).

In addition, we found EGFR for which synergy with FGFR

inhibition has been described [3]. Because of the presence of

several components of the PI3K pathway, we decided to

focus our validation on the central node, the catalytic

subunit of PI3K, PIK3CA. Three different shRNAs targeting

PIK3CA were significantly depleted in the drug-treated

group compared with the control. This suggested that

suppression of PIK3CA synergizes with FGFRi in FGFR3

mutant bladder cancer. To validate this finding, we infected

RT112 cells with each of these three shRNAs targeting

PIK3CA and treated with or without AZD4547 for 2 wk. All

three shRNAs against PIK3CA induce efficient knockdown of

PIK3CA protein expression as determined by western blot

analysis (Fig. 1D). Parental RT112 cells did not significantly

respond to FGFR inhibition or PIK3CA suppression alone, but

knockdown of PIK3CA strongly enhanced the response to

the FGFRi AZD4547 (Fig. 1E).

We next investigated whether pharmaceutical inhibition

of the PI3K pathway was synergistic with FGFR inhibition

using BKM120, a pan-PI3K inhibitor with modest antitumor

activity in cancer patients, as a single agent. We found that

RT112 (FGFR3-TACC3 translocation) [4] and JMSU1 (FGFR1

amplification) [5] were not significantly inhibited by

BKM120 or AZD4547 monotherapy. However, synergy

was observed with the combination in long-term in vitro

colony formation assays (Figs. 2 A and 2B), incucyte

proliferation assay (Supplementary Figs 1A and 1B), and

The cells were fixed, stained, and photographed after 14 d. (C,D) Biochemical acells were harvested after 24 h of drug treatment. Phosphorylated-AKT (p-AKTFRS2), total FRS2 (FRS2), phosphorylated-ERK (p-ERK), and total ERK (ERK) weFGFR inhibitor (100 nM, 7 d) treated RT112. AZD4547 induced feedback activatreactivation of EGFR and ERBB3 in association with elevated PI3K-AKT signalin50 mM AZD4547. Phosphorylated-EGFR (p-EGFR), total EGFR (EGFR), phosphoryFGFR3), phosphorylated-AKT (p-AKT), total AKT (AKT), phosphorylated-FRS2 (p(ERK) were measured. b-actin served as a loading control. (G) FGFR inhibitor (suppresses tumor growth in RT112 xenograft model. (H) FGFR altered tumors

upregulate other RTKs that result in enhanced signaling through the PI3K-AKTPI3K inhibitor eliminates the PI3K-AKT activity and synergistically kills cancerc-PARP = poly (adenosine diphosphate-ribose) polymerase; ERBB3 = Erb-B2 receRTK = receptor tyrosine kinases.* p < 0.01 analysis of variance.** p < 0.0001.

Please cite this article in press as: Wang L, et al. A Functional GenetDeterminant of Resistance to Fibroblast Growth Factor Receptor

(2017), http://dx.doi.org/10.1016/j.eururo.2017.01.021

synergy assays (Supplementary Fig. 1C). Moreover, bio-

chemical analysis indicated that the combination resulted

in the induction of cleaved poly (adenosine diphosphate-

ribose) polymerase, a hallmark of apoptosis (Figs. 2C and

2D). To expand our results to another cancer type with FGFR

alterations, we tested this drug combination in two

independent FGFR1 amplified lung cancer cell line models:

H520 (squamous cell carcinoma) and DMS114 (small cell

carcinoma) [6]. As observed in the UCC lines, these two lung

cancer cell models showed apoptosis in response to the

drug combination, but not to either drug alone (Supple-

mentary Figs. 1D–G). Of note, similar results were recently

obtained for ponatinib in combination with mechanistic

target of rapamycin inhibition in NSCLC cells [7].

To address the molecular mechanism underlying the

synergy between PI3K and FGFR inhibition, we further

analyzed signaling pathways in AZD4547-treated RT112

cells using receptor tyrosine kinase phosphorylation blots

(Fig. 2E). We found that FGFR inhibition induced a feedback

mechanism to activate Erb-B2 receptor tyrosine kinase

3 and to a lesser extent EGFR. As a consequence, PI3K-AKT

signaling was activated, thereby presumably blunting the

effects of FGFRi (Fig. 2F). Feedback activation of receptor

tyrosine kinases (EGFR and ligand-associated Erb-B2

receptor tyrosine kinase 2/Erb-B2 receptor tyrosine kinase

3) [3,8] has been previously reported. Our findings provide a

strong rationale that feedback signaling converges on the

PI3K pathway, but not on the BRAF-MEK-ERK pathway

(Fig. 2F), and resistance could be counteracted by PI3K

inhibitors, in order to enhance the FGFRi sensitivity.

To test the combination treatment in vivo, we engrafted

RT112 bladder cancer cells into NMRI-nu mice. When

tumors reached approximately 100 mm3, mice were

randomized into different cohorts and treated with vehicle,

AZD4547, BKM120, or the drug combination. As shown in

Figure 2G, treatment with the single drugs AZD4547 or

BKM120 resulted in limited tumor growth inhibition.

However, treatment with the combination of AZD4547

and BKM20 resulted in persistent suppression of tumor

growth throughout the duration of the experiment.

Immunohistochemistry staining of the tumors at the end

of the experiment (Supplementary Fig. 1I) shows the

combination suppressed tumor proliferation (Ki67) and

inducted apoptosis (cleaved caspase 3).

In two FGFR3-activated cell lines, MGHU3 and SW780,

synergy was not observed (Supplementary Figs. 1H and 1J).

MGHU3 cells carry an activating mutation in the AKT1

nalysis of combination of FGFR and PI3K inhibitors. RT112 and JMSU1), total AKT (AKT), cleaved-PARP (c-PARP), phosphorylated-FRS2 (p-re measured. HSP90 served as a loading control. (E) RTK blot analysis ofion of EGFR and ERBB3. f, Biochemical analysis indicated theg, but not MAPK signaling after treating RT112 cells for 1 wk withlated-ERBB3 (p-ERBB3), total HER3 (HER3), phosphorylated-FGFR3 (p--FRS2), total FRS2 (FRS2), phosphorylated-ERK (p-ERK), and total ERKAZD4547) in combination with PI3K inhibitor (BKM120) significantlyinitially respond to FGFR inhibition. However, the tumor cells are able to

pathway, leading to drug resistance. Combining FGFR inhibition with a cells.ptor tyrosine kinase 3; MAPK = mitogen-activated protein kinase;

ic Screen Identifies the Phosphoinositide 3-kinase Pathway as aInhibitors in FGFR Mutant Urothelial Cell Carcinoma. Eur Urol

E U R O P E A N U R O L O G Y X X X ( 2 0 1 6 ) X X X – X X X 5

EURURO-7215; No. of Pages 5

(E17K) gene, causing pathway activation downstream of

PIK3CA [9]. Indeed, these cells were highly sensitive to

mechanistic target of rapamycin inhibition (AZD8055). This

finding suggests that comprehensive genetic profiling

remains relevant before initiating combination treatment,

in order to establish the best possible treatment. SW780 cells

carry a translocation, FGFR3-BAIAP2L1 [4]. This translocation

has not been found in other cancers and the mechanism of

FGFR activation is unknown, though cells appear to be

dependent on FGFR signaling [10]. Other resistance path-

ways could therefore be active in this cell line.

In conclusion, our data show that resistance pathways to

FGFR inhibition often converge on the PI3K pathway and

provide a strong rationale to treat FGFR3-altered UCC with a

combination of FGFR and PI3K inhibitors (Fig. 2H). Muta-

tions in the PIK3CA gene, which can cooccur with FGFR

mutations, would also be targeted by this strategy. These

results may apply to other cancer types as well, for example

squamous NSCLC.

Author contributions: Michiel S. van der Heijden had full access to all the

data in the study and takes responsibility for the integrity of the data and

the accuracy of the data analysis.

Study concept and design: Wang, Sustic, de Oliveira, van den Heuvel,

Bernards, van der Heijden.

Acquisition of data: Wang, Sustic, de Oliveira, van de Ven.

Analysis and interpretation of data: Wang, Sustic, de Oliveira, Halonen,

Lieftink, Beijersbergen, van den Heuvel, Bernards, van der Heijden.

Drafting of the manuscript: Wang, Beijersbergen, van der Heijden.

Critical revision of the manuscript for important intellectual content: Wang,

Sustic, de Oliveira, Lieftink, Halonen, Beijersbergen, van den Heuvel,

Bernards, van der Heijden.

Statistical analysis: Halonen, Lieftink, Beijersbergen.

Obtaining funding: van der Heijden, de Oliveira, Beijersbergen.

Administrative, technical, or material support: van de Ven.

Supervision: Beijersbergen, Beijersbergen, van der Heijden.

Other: None.

Financial disclosures: Michiel S. van der Heijden certifies that all conflicts

of interest, including specific financial interests and relationships and

affiliations relevant to the subject matter or materials discussed in the

manuscript (eg, employment/affiliation, grants or funding, consultan-

cies, honoraria, stock ownership or options, expert testimony, royalties,

or patents filed, received, or pending), are the following: This work was

supported by grants from the Dutch Cancer Society and the Netherlands

Please cite this article in press as: Wang L, et al. A Functional GenetiDeterminant of Resistance to Fibroblast Growth Factor Receptor I(2017), http://dx.doi.org/10.1016/j.eururo.2017.01.021

Organization for Scientific Research to van der Heijden (Veni program)

and Cancer Genomics Netherlands; de Oliveira was supported by an

EMBO long-term fellowship.

Funding/Support and role of the sponsor: None.

Appendix A. Supplementary data

Supplementary data associated with this article can be

found, in the online version, at http://dx.doi.org/10.1016/j.

eururo.2017.01.021.

References

[1] Cancer Genome Atlas Research Network. Comprehensive molecular

characterization of urothelial bladder carcinoma. Nature

2014;507:315–22.

[2] Nogova L, Sequist LV, Perez Garcia JM, et al. Evaluation of BGJ398, a

fibroblast growth factor receptor 1-3 kinase inhibitor, in patients

with advanced solid tumors harboring genetic alterations in fibro-

blast growth factor receptors: results of a global phase I, dose-

escalation and dose-expansion study. J Clin Oncol 2016:

JCO2016672048.

[3] Herrera-Abreu MT, Pearson A, Campbell J, et al. Parallel RNA inter-

ference screens identify EGFR activation as an escape mechanism in

FGFR3-mutant cancer. Cancer Discov 2013;3:1058–71.

[4] Williams SV, Hurst CD, Knowles MA. Oncogenic FGFR3 gene fusions

in bladder cancer. Hum Mol Genet 2013;22:795–803.

[5] Pearson A, Smyth E, Babina IS, et al. High-level clonal FGFR ampli-

fication and response to FGFR inhibition in a translational clinical

trial. Cancer Discov 2016;6:838–51.

[6] Weiss J, Sos ML, Seidel D, et al. Frequent and focal FGFR1 amplifi-

cation associates with therapeutically tractable FGFR1 dependency

in squamous cell lung cancer. Sci Transl Med 2010;2:62ra93.

[7] Singleton KR, Hinz TK, Kleczko EK, et al. Kinome RNAi screens reveal

synergistic targeting of MTOR and FGFR1 pathways for treatment of

lung cancer and HNSCC. Cancer Res 2015;75:4398–406.

[8] Wang J, Mikse O, Liao RG, et al. Ligand-associated ERBB2/3 activa-

tion confers acquired resistance to FGFR inhibition in FGFR3-de-

pendent cancer cells. Oncogene 2015;34:2167–77.

[9] Davies BR, Guan N, Logie A, et al. Tumors with AKT1E17K mutations

are rational targets for single agent or combination therapy with

AKT inhibitors. Mol Cancer Ther 2015;14:2441–51.

[10] Wu YM, Su F, Kalyana-Sundaram S, et al. Identification of target-

able FGFR gene fusions in diverse cancers. Cancer Discov

2013;3:636–47.

c Screen Identifies the Phosphoinositide 3-kinase Pathway as anhibitors in FGFR Mutant Urothelial Cell Carcinoma. Eur Urol