Published OnlineFirst October 31, 2019; DOI: 10.1158/0008-5472.CAN-19-1415

Cancer Translational Science Research

YAP1 Mediates Resistance to MEK1/2 Inhibition in Neuroblastomas with Hyperactivated RAS Signaling Grace E. Coggins1,2, Alvin Farrel1,3, Komal S. Rathi1,3, Colin M. Hayes1, Laura Scolaro1, Jo Lynne Rokita1,3,4, and John M. Maris1,5,6

Abstract

Relapsed neuroblastomas are enriched with activating sensitized neuroblastoma cells to trametinib, while overex- mutations of the RAS–MAPK signaling pathway. The MEK1/ pression of constitutively active YAP1 induced trame- 2 inhibitor trametinib delays tumor growth but does not tinib resistance. Mechanistically, significant enhancement of sustain regression in neuroblastoma preclinical models. G1–S cell-cycle arrest, mediated by depletion of /MYCN Recent studies have implicated the Hippo pathway transcrip- and transcriptional output, sensitized RAS-driven neuro- tional coactivator protein YAP1 as an additional driver of blastomas to trametinib following YAP1 deletion. These find- relapsed neuroblastomas, as well as a mediator of trametinib ings underscore the importance of YAP activity in response to resistance in other cancers. Here, we used a highly annotated trametinib in RAS-driven neuroblastomas, as well as the set of high-risk neuroblastoma cellular models to modulate potential for targeting YAP in a trametinib combination. YAP1 expression and RAS pathway activation to test whether increased YAP1 transcriptional activity is a mechanism of Significance: High-risk neuroblastomas with hyperacti- MEK1/2 inhibition resistance in RAS-driven neuroblastomas. vated RAS signaling escape the selective pressure of MEK In NLF (biallelic NF1 inactivation) and SK-N-AS (NRAS inhibition via YAP1-mediated transcriptional reprogramming Q61K) cell lines, trametinib caused a near-complete translo- and may be sensitive to combination therapies targeting both cation of YAP1 protein into the nucleus. YAP1 depletion YAP1 and MEK.

Introduction The development of novel treatments has been hindered by the relative lack of molecularly targetable genomic lesions. Recurrent Neuroblastoma is a malignancy of the developing sympathetic kinase domain gain-of-function mutations in the ALK oncogene nervous system (1–6). Half of all diagnosed neuroblastomas are occur in 8%–15% of all newly diagnosed neuroblastomas (4–8), classified as "high-risk", for which cure rates remain low. Aggres- but may be present in a much larger percentage of relapse sive empiric multimodal therapy, including surgery, chemother- specimens (9–12). Indeed, compared with matched primary apy, radiotherapy, and more recently immunotherapy have tumors, relapsed neuroblastomas have a significantly higher shown incremental improvements in survival rates at the cost of mutational burden, with clonal enrichment in mutations in a host of chronic health comorbidities in survivors. Relapse after RAS–MAPK pathway beyond ALK such as NRAS, KRAS, standard of care remains largely incurable (6, 7). Thus, there is an BRAF, PTPN11, and NF1 (9, 10, 12). Neuroblastoma cellular urgent need for more effective and precise therapies. models with these genetic aberrations have elevated levels of phosphorylated ERK1/2 and are extremely sensitive to the MEK1/2 noncompetitive inhibitor trametinib in vitro, with low 1Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania. 2Department of Systems nanomolar IC50s (9, 13). However, single-agent MEK inhibition Pharmacology and Translational Therapeutics, University of Pennsylvania, Phi- is cytostatic and results only in tumor growth delay in neuroblas- ladelphia, Pennsylvania. 3Department of Biomedical and Health Informatics, toma xenotransplantation models with RAS hyperactiva- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania. 4Center for Data- tion (9, 14, 15), similar to the experience in multiple preclinical Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadel- and clinical settings with single-agent inhibition of MAPK path- 5 phia, Pennsylvania. Department of Pediatrics, Perelman School of Medicine at way–mutated cancers (16–19). For this reason, combination the University of Pennsylvania, Philadelphia, Pennsylvania. 6Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of strategies are being pursued to avoid tumor escape from therapy Pennsylvania, Philadelphia, Pennsylvania. and improve long-term responses. Dual inhibition of MEK1/2 and rational targets, such as BRAF, PI3K/AKT, and CDK4/6, Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). have shown promise in other tumor types, including neuroblastoma (13, 14, 16–20), but in the latter case all xeno- Corresponding Author: John M. Maris, Children's Hospital of Philadelphia, grafts eventually escaped dual MEK and CDK4/6 inhibition (18). Philadelphia, PA 19104. Phone: 215-590-5242; Fax: 267-426-0685; E-mail: [email protected] The is considered tumor suppressive through cytosolic sequestration of the transcriptional coactivator Cancer Res 2019;79:6204–14 protein YAP1 (21–23). Activated YAP1 mediates diverse biologic doi: 10.1158/0008-5472.CAN-19-1415 functions such as organ size, cellular proliferation, and cell 2019 American Association for Cancer Research. survival (24–30). YAP1 dephosphorylation allows translocation

6204 Cancer Res; 79(24) December 15, 2019

Downloaded from cancerres.aacrjournals.org on October 4, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst October 31, 2019; DOI: 10.1158/0008-5472.CAN-19-1415

YAP1 Loss Sensitizes RAS-Driven Neuroblastoma to Trametinib

into the nucleus and interaction with TEAD family and other these experiments. To produce YAP1-knockout pools in SKNAS transcription factors to initiate transcription of a multiple and NLF, cells were transduced with lentivirus for the sgRNA targets (31–37). Several groups have reported that YAP1 may against sequence #1 according to the manufacturer's protocol. For be involved in resistance to trametinib in RAS-driven NLF isogenic cell lines, a second YAP1-knockout pool was pro- cancers (38–42). Recently, increased YAP1 activity was reported duced using lentivirus targeting sequence #2. Two single-cell as a hallmark of relapsed neuroblastoma after intensive chemor- clones were selected from each YAP1-knockout pool and grown adiotherapy (12, 26). In addition, inhibition of YAP1 signaling into stable isogenic cell lines. Antibiotic selection was performed has also been shown to abrogate neuroblastoma metastasis in using 1 mg puromycin (Sigma, #P9620). preclinical models (43). Paradoxically, the YAP1 gene is located The lentiviral YAP-5SA overexpressing plasmid was produced on arm 11q, a region that shows frequent hemizy- by inserting the YAP-5SA sequence from the MYC-YAP-5SA gous deletion, particularly in high-risk neuroblastomas without plasmid (Addgene #33091; ref. 26) into a lentiviral CMV-puro MYCN amplification (44, 45). Here we explore the hypothesis DEST vector (Addgene #39481; ref. 47) using the PCR Cloning that derepression of YAP1 is a critical mediator of resistance to System with Gateway Technology with pDONR221 and Omni- MEK inhibition in neuroblastomas with hyperactivated MAPK MAX2 Competent Cells (Invitrogen #12535029) according to signaling. the manufacturer's recommended protocol. For lentiviral pro- duction, the YAP-5SA lentiviral plasmid was transfected in combination with the pMD2.G VSV-G envelope–expressing Materials and Methods plasmid (Addgene #12260) and psPAX2 lentiviral packaging Cell culture and chemicals plasmid (Addgene #12259). Plasmids were transduced at equi- Human-derived neuroblastoma cell lines were obtained molar concentrations of 3 mmol/L into HEK-293T cells (ATCC, from the Children's Hospital of Philadelphia cell line bank, CRL-3216) using Lipofectamine 3000 (Thermo Fisher Scientific the Children's Oncology Group, and the ATCC (46). Cell #L3000008). Viral supernatant was harvested at 48 hours and lines used included: NLF (RRID:CVCL_E217), SKNAS (RRID: was filtered using a 0.45-mm filter and added to cells with 3 mg CVCL_1700), NB-EBc1 (RRID:CVCL_E218), and SKNFI (RRID: polybrene. Antibiotic selection was performed using 1 mg CVCL_1702). Cell line authentication to confirm genomic puromycin. identity was performed using the GenePrint 24 System (Pro- mega, Guardian Forensic Sciences) every 2 years. Cell lines were Primers continually tested for Mycoplasma contamination after each Sequencing primers to detect mutations in both of the target thaw using the MycoAlert Kit (Cambrex) and were confirmed sequences in the endogenous YAP1 protein TEAD binding to be Mycoplasma negative prior to experimentation. Cells domain were: YAP1_F (50-TAAAGAGAAAGGGGAGGCGG-30) were cultured in RPMI 1640 medium containing 10% FBS, and YAP1_R (50-CCGGGAAGAAAGAAAGGAAGA-30). Primers 2 mmol/L L-Glutamine at 37 Cunder5%CO2 and were for Gateway cloning were designed according to the manufac- maintained at low passage that did not exceed 20 passages. turer's recommendations to remove the YAP-5SA sequence from Trametinib dissolved in DMSO (Cellagen Technologies the MYC-YAP-5SA retroviral plasmid with flanking attB sites. #C4112-5s) was used for in vitro assays, with 0.1% DMSO as Primer sequences were: YAP-5SA_F (50-GGGG ACAAGTTTGTA- a negative control treatment. All cell lines were derived from CAAAAAAGCAGGCTTCACCATGGAACAAAAACTCATCTCA-30) deidentified neuroblastoma patient tumor samples and the and YAP-5SA_R (50-GGGGACCACTTTGTACAAGAAAGCTGG- Children's Hospital of Philadelphia Institutional Review Board GTCCTATAACCATGTAAGAAAGCTTTCTTT-30). agreed with the investigators that this work is not considered human subjects research. Western blotting Protein was isolated from whole-cell lysates using lysis buffer Cell viability assays containing 1 Cell Lysis Buffer (10 from Cell Signaling Tech- Cells were seeded in 96-well cell culture plates at 2,500–4,000 nology, #9803), 2 mmol/L PMSF (Cell Signaling Technology, cells per well depending on growth kinetics. Drug treatments were #8553S), in 100% isopropanol, and 1% phosphatase inhibitor performed in triplicate 24 hours later over a 6-log dose range cocktails 2 (Sigma, #P5726) and 3 (Sigma, #P0044). Protein (0.01–10,000 nmol/L). IC50 values for trametinib were calculated concentration was determined using the Bradford Protein Assay using AUC at 72 hours post-treatment. Cell viability was assessed (Bio-Rad). Approximately 20 mg of protein were run on 4%–15% using CellTiter-Glo (Promega). Cell growth assays were per- gradient Tris-Glycine Gels (Bio-Rad, #5671085) and transferred formed using the IncuCyte Live Cell Analysis System (IncuCyte using the Bio-Rad transfer system. Antibodies used for Western ZOOM, Essen Bioscience) with the 20 objective lens during a blotting include (Cell Signaling Technology, unless otherwise 72-hour treatment. indicated): YAP1 (D8H1X) (1:1,000, #14074), p-YAP1 (S127) (D9W2I) (1:500, #13008S), p-ERK (1:2,000, #4370), ERK CRISPR-Cas9, plasmids, and lentiviral delivery (1:2,000, #4695), b-Actin (1:5,000, #4967S), RB (1:2,000, To produce YAP1-targeting CRISPR-Cas9–knockout cell lines, #9309), p-RB (S807-811) (1:1,000, #9307), PARP (1:1,000, scrambled sgRNA CRISPR/Cas9 All-in-One Lentivirus (ABM #9532), cleaved PARP (1:1,000, #5625S), MYCN (1:2,000, #K011) and the YAP1 sgRNA CRISPR All-in-One Lentivirus Set #9405S), Caspase-3 (1:1,000, #9662), and TATA Box binding (Human) (ABM #K2653115) targeting the YAP1 gene (Accession protein (TBP; 1:1,000, Abcam #ab818). Western blots were visu- Number: NM_1006106.4) were used. Virus with single-guide alized using SuperSignal West Femto Maximum sensitivity sub- RNA (sgRNA) targeting sequence #1 (50-GTGCACGATCT- strate (Thermo Fisher Scientific, #34095) and the FluorChem Q GATGCCCGG-30) and sequence #2 (50-CGCCGTCATGAACCC- chemiluminescent imaging system and FluorChemQ Software CAAGA-30) of the YAP1 TEAD binding domain were selected for v3.4.0 (ProteinSimple).

www.aacrjournals.org Cancer Res; 79(24) December 15, 2019 6205

Downloaded from cancerres.aacrjournals.org on October 4, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst October 31, 2019; DOI: 10.1158/0008-5472.CAN-19-1415

Coggins et al.

RNA isolation and qRT-PCR treatment group. Differentially expressed genes underwent Gene RNA was isolated using the Qiagen miRNEasy Mini Kit (Qia- Ontology analysis using the ToppFun tool from the ToppGene gen). Reverse transcription was performed using the iScript Select Suite and the top 5 ontologies were chosen (52). Gene set cDNA Synthesis Kit (Bio-Rad #1708897). qPCR was performed enrichment analysis (GSEA) was performed using the Molecular using the TaqMan 2X Master Mix (Thermo Fisher Scientific Signatures Database Hallmarks Gene Set collection and run for #4304437) on 384-well plates using the 7900HT Fast Real- 1,000 iterations with a FWER Pcutoff < 0.01. All RNA-sequencing Time PCR Instrument (Applied Biosystems) and the SDS v2.4 data have been deposited in the Gene Expression Omnibus under Software (Applied Biosystems). TaqMan probes (Thermo Fisher accession number GSE130401. Scientific, #4331182) used included: YAP1 (Hs00902712_g1) HPRT1 (Hs02800695_m1), GAPDH (Hs03929097_g1), Statistical analysis CTGF (Hs01026927_g1), CYR61 (Hs00155497_m1), CDK1 Group comparisons were determined with a two-tailed t test (Hs00938777_m1), MCM4 (Hs00907398_m1), MCM6 with a significance cutoff of P < 0.05. Data analysis was performed (Hs00195504_m1), POLA1 (Hs00213524_m1), CCNE1 using GraphPad Prism and R Studio. (Hs01026536_m1), and (Hs00153451_m1).

Flow cytometry Results Samples for cell-cycle analysis were collected after 72 hours of Trametinib causes YAP1 nuclear translocation in RAS-MAPK– trametinib treatment at the IC50 concentration of NLF (20 nmol/ activated neuroblastoma cell lines L) and SKNAS (10 nmol/L). Cells were detached with versene We selected 16 of the 39 cell lines recently profiled and reported (0.02% EDTA in HBSS), washed with PBS þ 1% FBS, fixed for by our group based on YAP1 mRNA expression and mutation approximately 10 seconds by adding ice-cold 70% ethanol drop- status (Fig. 1A; ref. 46). The majority, but certainly not all, of the wise with constant vortexing, and stored at 20C. Cells were lines with mutations in the canonical MAPK pathway showed stained using 1 mL FxCycle Violet (Invitrogen #F10347) per 1 mL YAP1 mRNA and protein expression, but only one of the seven PBS and analyzed using the CytoFLEX LX with 6 lasers (Beckman ALK-mutated lines, and this line (SKNSH) showed robust protein Coulter). Data analysis was performed using the FlowJo v10 expression in the absence of detectable YAP1 mRNA. MYCN software as described previously (14). amplification and 11q copy number alterations for each cell line can be found in Supplementary Table S1. Given that phosphor- RNA sequencing ylation status and subcellular location are inherent to YAP1 Cells were plated in triplicate and treated with 20 nmol/L transcriptional activity, we investigated whether trametinib alters trametinib for 72 hours prior to collection. Cells were lysed on YAP1 nuclear localization in two high YAP1-expressing cell lines, the plate using the QIAzol Lysis Reagent (Qiagen #79306) and NLF and SKNAS. Nuclear and cytoplasmic extracts of NLF and homogenized with Qiashredder Tubes (Qiagen #79654). RNA SKNAS were collected after 72 hours of exposure to trametinib. was then isolated using the RNeasy Mini Kit (Qiagen #74104) We observed a reduction in cytoplasmic phosphorylated YAP1 according to the manufacturer's protocol and quality was deter- across the time course and a concomitant enrichment of nuclear mined using the TapeStation 2200 (Agilent Technologies). All 18 YAP1 (Fig. 1B and C). Together, these data suggest that trametinib samples were of optimal quality and achieved RNA integrity treatment in YAP1-expressing and MAPK-mutant neuroblastoma number (RIN) scores of 10.0. RNA synthetic spike-ins were added models causes depression of the Hippo pathway, resulting in to each sample (48), with Mix A added to the NLF scrambled rapid (days) translocation of YAP1 to the nucleus. / control (sgCon) samples and Mix B added to the NLF YAP1 #1 and #4 samples. Library preparation was done using 1 mg of RNA Loss of YAP1 expression sensitizes neuroblastoma cell lines using the TruSeq Total mRNA Kit with Gold rRNA Removal Mix as to trametinib recommended (Illumina #15031048). All 18 samples were To determine whether YAP1 plays a role in sensitivity to sequenced using v2 chemistry, 2 150 bp, and run on one trametinib in neuroblastoma, we selected two neuroblastoma high-output flow-cell of an Illumina NextSeq 500 instrument. cell lines, NLF (biallelic NF1 inactivation) and SKNAS (NRAS Libraries were demultiplexed, Illumina adapters were trimmed, Q61K), which both harbor endogenous hemizygous deletions of and FASTQ file generated using the Illumina NextSeq Control 11q and thus YAP1 (46). We employed lentiviral CRISPR- Software version 2.02. Cas9 gene editing to produce pools of YAP1-null NLF and SKNAS Raw fastq files (n ¼ 18) from RNA-sequencing data with an cells. Lentivirus containing sgRNA targeted to the YAP1 TEAD average sequencing depth of 22 million reads were aligned to binding domain or a sgCon were used to transduce cells (Sup- human hg19 primary assembly reference genome using the STAR plementary Fig. S1A). We observed incomplete reduction of YAP1 aligner v2.5.3a (49). Gene expression was quantified as fragments mRNA and protein expression in both NLF and SKNAS sgYAP1 per kilobase of transcript per million mapped reads (FPKM) and pools (Supplementary Fig. S1B and S1C). Despite this modest transcript per million using RSEM v1.2.28 normalization and reduction in expression, we next showed that the canonical YAP1 Gencode v23 gene annotation (50). On an average, 88.05% reads target genes CTGF and CYR61 (24) were significantly downregu- were uniquely mapped to the reference genome. Normalization lated in NLF and SKNAS YAP1-depleted cells (Supplementary of RNA expression between samples was performed by analyzing Fig. S1D), suggesting a significant impact on YAP1-mediated the synthetic spike-in standards using Anaquin Software Toolkit transcription. We next sought to determine the impact of trame- distributed by Bioconductor (51). tinib exposure on cell viability in the isogenic pairs differing in Differential expression analysis was performed using the R YAP1 transcriptional activity. We observed that the response of package, DESeq2. Values were log2-transformed and biological these cell lines to trametinib treatment was directly related to the replicates (N ¼ 3) were averaged within each cell line and degree of modulation of YAP1 target genes (Supplementary

6206 Cancer Res; 79(24) December 15, 2019 Cancer Research

Downloaded from cancerres.aacrjournals.org on October 4, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst October 31, 2019; DOI: 10.1158/0008-5472.CAN-19-1415

YAP1 Loss Sensitizes RAS-Driven Neuroblastoma to Trametinib

Figure 1. Trametinib causes nuclear accumulation of unphosphorylated YAP1 protein. A, Expression of YAP1 mRNA (FPKM) across a panel of neuroblastoma cell lines with known RAS–MAPK pathway mutations indicated at the top. Bottom, YAP1 (70 kDa) is expressed in a subset of RAS-driven neuroblastoma cell lines with a b-actin (40 kDa) loading control. B and C, Seventy-two hour trametinib treatment of NLF (20 nmol/L) and SKNAS (10 nmol/L) causes nuclear translocation of YAP1 (70 kDa) protein compared with TBP (40 kDa; B), which was quantified using densitometry (C).

Fig. S1E). Sensitivity to trametinib shifted in both NLF and We next selected for clonal YAP1-null NLF cell lines after serial SKNAS upon YAP1 depletion, with IC50s in SKNAS shifting dilution of CRISPR/Cas9 edited cells and isolated four isogenic from 6.57 nmol/L in sgCon) to 0.81 nmol/L in sgYAP1 (P ¼ clones. Indel mutations were confirmed by Sanger sequencing of 0.0255), as well as in NLF, with IC50s shifting from 15.98 nmol/L genomic DNA, with single nucleotide insertions present in / in sgCon to 7.76 nmol/L in sgYAP1 (P ¼ 0.0019; Supplementary NLF YAP1 lines #1 and #2, and a single nucleotide deletion / Fig. S1E). The growth curves for the sgCon and sgYAP1 lines in NLF YAP1 line #4 (Supplementary Fig. S2). Conversely, / plateau at 35% viability for both NLF and SKNAS, which is NLF YAP1 line #3 showed a mixed population flanking the expected for the control lines due to the cytostatic nature of PAM site. We investigated the effect of YAP1 loss on cellular trametinib. However, it is clear that the modest reduction of growth and observed a modest growth delay of 20% in the YAP1 expression was not sufficient to reduce viability at the NLF sgYAP1 line compared with the sgCon line (Supplementary highest dose of trametinib in neither NLF nor SKNAS sgYAP1 Fig. S3). NLF YAP1 / #2 and #3 mixed clone had comparable lines (Supplementary Fig. S1E). growth rates, but the mixed clone reached a similar confluence as

www.aacrjournals.org Cancer Res; 79(24) December 15, 2019 6207

Downloaded from cancerres.aacrjournals.org on October 4, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst October 31, 2019; DOI: 10.1158/0008-5472.CAN-19-1415

Coggins et al.

sgYAP1. NLF YAP1 / #1 and #4 cells grew at the slowest rate three showed no detectable protein by immunoblotting (Fig. 2A / and only reached to 30%–40% of sgCon confluence. All four and B). The NLF YAP1 #3 mixed clone showed reduced, but NLF YAP / cell lines showed reduced mRNA expression, and detectable, YAP1 protein expression but displayed increased

Figure 2. YAP1 knockout sensitizes neuroblastoma cell lines to trametinib. A, Four isogenic lines were established from the NLF sgYAP1 CRISPR pooled cell line. YAP1 expression is shown for NLF sgCon, sgYAP1 pool, and YAP1/ #1–4(N ¼ 3). B, Immunoblots of NLF sgCon, sgYAP1 pool, and YAP1/ #1–4 for YAP, p-YAP, p-ERK, ERK, and b-actin. C–E, Expression of YAP1 target genes, CTGF and CYR61, in NLF sgCon, sgYAP1 pool, and YAP1/ #1–4.

IC50 curves for trametinib in NLF sgCon, sgYAP1 pool, and YAP1/ #1–4 over a 6-log dose range (D) and a graphical representation of

IC50 values (E) of trametinib (N ¼ 3). Student t test; , P < 0.05; , P < 0.01; , P < 0.001; , P < 0.0001; ns, nonsignificant.

6208 Cancer Res; 79(24) December 15, 2019 Cancer Research

Downloaded from cancerres.aacrjournals.org on October 4, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst October 31, 2019; DOI: 10.1158/0008-5472.CAN-19-1415

YAP1 Loss Sensitizes RAS-Driven Neuroblastoma to Trametinib

phospho-ERK expression. On the basis of the Sanger sequencing three distinct sets: (i) sgCon treated with either DMSO or trame- / results and protein expression, the NLF YAP1 #3 mixed clone tinib (trametinib specific), (ii) sgCon and NLF YAP1 / #4 treated was excluded from subsequent assays. After confirming repression with DMSO (YAP1 / specific), and (iii) sgCon þ DMSO and NLF of CTGF and CYR61 mRNA (Fig. 2C), we determined trametinib YAP1 / #4 þ trametinib (Combination of YAP1 loss and MEK / / IC50 values in the isogenic YAP1 cell lines. All three YAP1 inhibition). Differentially expressed genes in the trametinib- lines were significantly more sensitive to trametinib than NLF specific and YAP1 / -specific groups were subtracted from the / sgCon or NLF sgYAP1 pool, with IC50 values reduced from a trametinib-treated YAP1 gene list (Supplementary Table S5). median of 0.79 to 2.18 nmol/L for the three YAP1 / (P < 0.0001) This final dataset represented the 1,474 differentially expressed versus 7.62 nmol/L for the pooled sgYAP1 (P < 0.0038) compared genes that were unique to the combination of trametinib treat- / with 15.58 nmol/L for the sgCon (Fig. 2D and E). ment in a YAP1 model. analysis of the tran- scripts downregulated within this dataset revealed cell cycle and Constitutively active YAP1 overexpression induces resistance to DNA repair pathways as most significantly enriched (Fig. 4C). trametinib in MAPK pathway–activated neuroblastoma cells GSEA of the 1,474 genes produced only two significantly enriched The YAP1 protein contains five HXRXXS motifs that are recog- gene sets with a family-wise error rate of <0.01: E2F and MYC nized and phosphorylated by LATS1/2 (26). Of these five sites, targets (Fig. 4D). Heatmaps of E2F and MYC target genes show /- phosphorylation of S127 on YAP1 promotes binding with 14-3-3, reduced expression of target genes in NLF YAP1 #1 and #4 which causes cytoplasmic retention of YAP1. Mutating all five compared with sgCon (Fig. 4E). The most striking decrease in serine residues to alanine ablates the LATS1/2 phosphorylation expression occurred with trametinib treatment, particularly in the / sites and yields a constitutively active YAP-5SA protein (26). To NLF YAP1 #4 cell line (Fig. 4E). Importantly, NLF neuroblas- observe the effect of increased YAP activity in response to trame- toma cells do not express MYC but do express MYCN, suggesting tinib, we overexpressed YAP-5SA cDNA in NB-EBc1 (KRAS G12D) that this gene set actually refers to MYCN gene targets. To test this, and SKNFI (NF1 homozygous inactivation), which are both de we performed an additional GSEA using the WEI_MYCN_TAR- novo YAP1 protein null cell lines (Figs. 1A and 3A). Forced high GETS_WITH_E_BOX gene set (Fig. 4F; ref. 53). We confirmed that overexpression of YAP-5SA protein resulted in variable changes in MYCN gene targets are significantly enriched in the list of differ- these cells with different genotypes, in terms of a slight increase in entially expressed genes, with a family-wise error rate of <0.01 and p-YAP1 in the NB-EBc1, and p-ERK in SKNFI. We next confirmed a normalized enrichment score of 3.22. Expression of relevant the upregulation of CTGF and CYR61 in both lines (Fig. 3B cell cycle and DNA replication and repair genes follow a pattern and C). YAP-5SA overexpression induced resistance to trametinib, similar to the E2F and MYC heatmaps (Fig. 4G). Changes in in which cell viability did not reach 50% in either YAP-5SA– expression of E2F1 were more modest, but MYCN expression / overexpressing line compared with the control IC50s in both NB- increases upon YAP1 loss in control-treated NLF YAP1 #1 and EBc1 (73.03 nmol/L; P < 0.001), and SKNFI (16.94 nmol/L; P < #4. In response to trametinib, expression in NLF YAP1 / #1 and 0.0001; Fig. 3D and E). We then forced YAP-5SA overexpression in #4 decreases to similar levels of control and trametinib-treated NLF YAP1 / #1 and #4 cell lines, despite the known limitation NLF sgCon samples. We also confirmed the change in MYCN that the YAP-5SA construct would be recognized and cut by the protein expression, which follows a similar pattern observed in CRISPR-Cas9 machinery. Despite this, we were able to obtain the differential expression results in response to YAP1 loss and modest overexpression of constitutively active YAP1, and a like- trametinib treatment (Supplementary Fig. S5C). In an effort to wise (albeit subtler) induction of relative resistance to trametinib, connect the changes in MYCN expression to YAP–TEAD signaling, partially rescuing the YAP1 / phenotype (Supplementary we identified the conserved DNA-binding motif CATTCC, which Fig. S4A–S4C). is shared by all four TEAD1–4 transcription factors using the online JASPER tool (7th release, 2018 version; Supplementary YAP1 mediates resistance to trametinib in neuroblastoma cells Fig. S6A). We queried the region surrounding the MYCN gene with hyperactivated MAPK signaling through transcriptional locus using Integrated Genomics Viewer and identified CATTCC activation of E2F and MYC(N) sense sequences in the MYCN promoter and the first intron, as To better understand how YAP1 plays a role in trametinib well as an antisense CATTCC sequence in the MYCN promoter sensitivity, we performed RNA sequencing of NLF sgCon and two (Supplementary Fig. S6B). This observation confirms that the isogenic cell lines, NLF YAP1 /- #1 and #4. All three cell lines were TEADs are able to bind at the MYCN locus and the loss of YAP- treated in triplicate with 20 nmol/L trametinib or DMSO for TEAD transcriptional activity upon YAP1 knockout may account 72 hours, at which time, total RNA was isolated (Fig. 4A). After for these changes in MYCN expression. total mRNA sequencing, we confirmed that the biological repli- To validate these RNA-sequencing results, we performed qRT- cates clustered together by principal component analysis (Sup- PCR of five gene targets from Fig. 4G and expression follows the plementary Fig. S5A). We next confirmed that YAP1 and down- expected pattern (Fig. 5A). We also tested this using the SKNAS stream transcriptional targets CTGF, and CYR61 mRNA expres- sgCon and sgYAP1 pooled lines treated with trametinib (or sion was suppressed as predicted in the RNA-sequencing data DMSO), which followed a similar pattern (Fig. 5B). The reduction (Supplementary Fig. S5B). Of note, expression of WWTR1, the of target gene expression was less robust than in the NLF YAP1 / gene encoding the YAP1 paralog TAZ, follows the same trend as isogenic lines likely due to the mosaic YAP1 expression in the YAP1 and its target genes, which confirms that TAZ expression is pooled CRISPR line. Because many of the E2F and MYC target not being upregulated to compensate for YAP1 loss (Supplemen- genes are involved in the cell cycle and DNA replication, we tary Fig. S5B). performed flow cytometry to examine DNA content after 72 hours We next performed three distinct differential expression anal- of trametinib treatment. In response to trametinib, the NLF sgCon yses using the R package DESeq2 (Fig. 4B; Supplementary Tables cells displayed a minor increase in G1 arrest (Fig. 5C). Loss of S2–S4). Differentially expressed genes were identified between YAP1 expression caused a further increase in G1 arrest and an even

www.aacrjournals.org Cancer Res; 79(24) December 15, 2019 6209

Downloaded from cancerres.aacrjournals.org on October 4, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst October 31, 2019; DOI: 10.1158/0008-5472.CAN-19-1415

Coggins et al.

Figure 3. YAP-5SA overexpression induces trametinib resistance in low YAP-expressing neuroblastoma cell lines. A, Immunoblots of NB-EBc1 and SKNFI empty vector and YAP-5SA–overexpressing cells. Immunoblots were probed for p-YAP1 S127 (70 kDa), total YAP1 (70 kDa), phospho-ERK (42 and 44 kDa), total ERK (42 and 44 kDa), and b-actin (40 kDa). B and C, YAP1, CTGF,andCYR61 expression in NB-EBc1 (B) and SKNFI (C) empty vector- and YAP-5SA– overexpressing cells. Relative mRNA expression is represented on a log scale (N ¼ 3). Student t test, , P < 0.05; , P < 0.01; , P < 0.001. D and E, IC50 curves for trametinib between empty vector and YAP-5SA–overexpressing NB-EBc1 (P < 0.001; D) and SKNFI (P < 0.0001; E) cells (N ¼ 3). One-way ANOVA [F(3,74) ¼ 18.69, P < 0.0001] with Sidak multiple comparisons test.

greater increase in G1 arrest upon trametinib treatment. In the NLF impairing the proliferative capacity of neuroblastoma cell lines /- YAP1 #4, which had the most significant decrease in YAP1 (Fig. 5E). target gene expression, we observed that 90% of the cells were arrested at G1-phase in response to trametinib (Fig. 5C). These data were verified in the SKNAS pooled cells, but to a lesser Discussion degree as expected (Fig. 5D). We further investigated whether Relapsed neuroblastomas remain largely incurable, but recent or not the combination of YAP1 loss and trametinib treatment insight into relapse-specific mutations provides an opportunity to causes apoptosis. We did not observe increases in cleaved PARP develop targeted therapies (9, 12). Hyperactivation of the RAS or cleaved caspase 3 in the YAP1 / cell lines treated with or pathway is a common finding in relapsed neuroblastomas, sug- without trametinib (Supplementary Fig. S5C). From these data, gesting this contributes to resistance to standard up front che- we propose that trametinib induces a change in cellular sig- moradiotherapy. MEK inhibition shows cytostasis and eventual naling that causes a reduction in YAP1 protein phosphorylation tumor outgrowth in neuroblastoma preclinical models, and induces YAP1 nuclear translocation, where it can promote highlighting the need to identify combination therapies for this the transcription of E2F and MYCN target genes. In the absence subset of patients. of nuclear YAP1, trametinib treatment induces a significant Here, we identify enhanced activation of Hippo pathway reduction in E2F and MYCN target gene expression. As a protein YAP1 as a cellular adaptation to MEK1/2 inhibition in consequence, we have shown G0–G1 cell-cycle arrest, thus RAS-driven neuroblastomas. We show that while only a subset

6210 Cancer Res; 79(24) December 15, 2019 Cancer Research

Downloaded from cancerres.aacrjournals.org on October 4, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst October 31, 2019; DOI: 10.1158/0008-5472.CAN-19-1415

YAP1 Loss Sensitizes RAS-Driven Neuroblastoma to Trametinib

Figure 4. Increased trametinib sensitivity upon YAP1 loss is due to loss of E2F and MYC target gene expression. A, Workflow of RNA-sequencing experiment. NLF sgCon, YAP1/ #1 and #4 were treated in triplicate with either DMSO or 20 nmol/L trametinib for 72 hours and total RNA was isolated. B, Venn diagram showing shared genes among three differential expression analyses: (i) trametinib-specific: sgCon DMSO versus sgCon Tram, (ii) YAP1/ #4-specific: sgCon þ DMSO versus YAP1/ #4 þ DMSO, and (iii) YAP1/- #4 þ trametinib: sgCon þ DMSO versus YAP1/ #4 þ trametinib. C, Top 5 gene ontologies represented among the 1,474 / / unique trametinib-treated YAP1 #4 genes. D, GSEA of the 1,474 unique trametinib-treated YAP1 #4 genes with a FWER Pcutoff < 0.01. E, Heatmaps of FPKM values normalized by row for each gene represented in the E2F and MYC target gene sets. F, GSEA of the 1,474 unique trametinib-treated YAP1/ #4 genes against the WEI_MYCN_TARGETS_WITH_E_BOX gene set. G, FPKM values among all six groups for a subset of E2F and MYC target genes.

www.aacrjournals.org Cancer Res; 79(24) December 15, 2019 6211

Downloaded from cancerres.aacrjournals.org on October 4, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst October 31, 2019; DOI: 10.1158/0008-5472.CAN-19-1415

Coggins et al.

Figure 5. Trametinib treatment of YAP1/

cells causes G1 cell-cycle arrest. A and B, Expression of CDK1, MCM4, MCM6, POLA1, and CCNE1 in NLF sgCon and YAP1/ #1 and 4(N ¼ 3; A) and SKNAS sgCon and sgYAP1 (N ¼ 3; B). Cells were treated with DMSO or trametinib (NLF, 20 nmol/L; SKNAS, 10 nmol/L). C and D, Cell-cycle analysis of NLF sgCon and YAP1/ #1–4(N ¼ 3; C) and SKNAS sgCon and sgYAP1 (N ¼ 3; D) treated with DMSO or trametinib (NLF, 20 nmol/L; SKNAS, 10 nmol/L) for 72 hours. Flow cytometry was performed to detect the proportion of cells

present in G1-, S-, and G2-phase. E, Proposed mechanism of inhibiting MEK1/2 signaling and YAP1 activity in RAS-driven neuroblastoma. Dot, phosphorylation. Student t test; , P < 0.05; , P < 0.01; , P < 0.001; , P < 0.0001; ns, nonsignificant.

of RAS-driven neuroblastoma cell lines express detectable YAP1 explain the cytostatic effects of MEK inhibition in RAS-driven protein, short-term exposure to trametinib induces the trans- neuroblastoma. This finding also supports the purported clin- location of unphosphorylated "active" YAP1 into the nucleus. ical relevance of YAP1 in this disease, as neuroblastomas have The exact mechanism causing the reduction in YAP1 protein been shown to acquire increased YAP1 transcriptional activity phosphorylation, as well as the mechanism for nuclear trans- upon relapse (12). location, remains to be defined. The latter may be a result of Our findings show that in cells with YAP1 edited out, E2F and reduced phosphorylated YAP1, although actin stress fiber for- MYCN target gene sets were downregulated when MEK1/2 was mation has been reported to cause nuclear translocation in inhibited. This result provides additional biological value to the response to BRAF inhibitor resistance (54). Therefore, there importance of the Hippo pathway in conferring resistance to may be multiple mechanisms involved in the YAP1 protein RAS–MAPK pathway inhibition. Because of the low MYC expres- dynamics in response to MEK inhibition. In the YAP1-expres- sion in NLF cells, we demonstrated that MYCN gene targets were sing neuroblastoma cell lines, we discovered that YAP1 protein differentially expressed and that MYCN expression increased in expression levels were directly related to trametinib sensitivity. response to YAP1 loss but decreased when combined with MEK In YAP1-expressing cell lines, genetic depletion of YAP1 expres- inhibition. TEAD4 has been reported to bind to a consensus site in sion sensitized to trametinib, while overexpression of consti- the MYCN promoter and function in a YAP1-independent man- tutively active YAP1 induced trametinib resistance in neuro- ner in neuroblastoma cells (55). It is possible that the absence of blastoma cell lines with undetectable YAP1. This observation YAP1 may allow the TEAD to initiate an alternate gene may be clinically useful, as YAP1 transcriptional activity may expression program. However, we observed that this effect is lost

6212 Cancer Res; 79(24) December 15, 2019 Cancer Research

Downloaded from cancerres.aacrjournals.org on October 4, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst October 31, 2019; DOI: 10.1158/0008-5472.CAN-19-1415

YAP1 Loss Sensitizes RAS-Driven Neuroblastoma to Trametinib

when combined with MEK inhibition. Alternatively, MYCN has Authors' Contributions been shown to be regulated by E2F proteins in neuroblasto- Conception and design: G.E. Coggins, J.L. Rokita, J.M. Maris ma (56), which may indicate E2F1 target gene expression as the Development of methodology: G.E. Coggins, J.L. Rokita, J.M. Maris primary cause of the gene expression changes causing the Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): G.E. Coggins, C.M. Hayes, J.L. Rokita, J.M. Maris observed G cell-cycle arrest. The exact mechanism causing E2F 1 Analysis and interpretation of data (e.g., statistical analysis, biostatistics, gene target expression to decrease remains unclear. YAP and TEAD computational analysis): G.E. Coggins, A. Farrel, K.S. Rathi, J.M. Maris have been reported to cooperate with E2F by ChIP analyses to Writing, review, and/or revision of the manuscript: G.E. Coggins, A. Farrel, coordinate cell-cycle gene expression (38). The loss of both MEK- L. Scolaro, J.L. Rokita, J.M. Maris activated and YAP-activated E2F-related gene expression may Administrative, technical, or material support (i.e., reporting or organizing contribute to the differential gene expression observed in data, constructing databases): G.E. Coggins, J.M. Maris Study supervision: G.E. Coggins, J.L. Rokita, J.M. Maris response to MEK1/2 inhibition and YAP1 depletion. Recent literature has also shown that BRAF inhibitor resistance can Acknowledgments induce YAP-activated E2F-related cell-cycle gene expression in an This work was supported in part by NIH grants R35 CA220500 and actin-dependent manner (54). Here, we present data suggesting a P01CA217959 (to J.M. Maris), 1F31CA220844-01A1 (to G.E. Coggins) similar effect may occur in the context of MEK inhibition in and T32GM008076 (to G.E. Coggins), grants from Cookies for Kids neuroblastomas with RAS activation. Cancer (to G.E. Coggins and J.M. Maris), the Press On Foundation This study has important clinical implications because combi- (to J.M. Maris), the Giulio D'Angio Endowed Chair (to J.M. Maris), natorial inhibition of MEK1/2 and YAP1 signaling could be an and an Alex's Lemonade Stand Foundation Young Investigator Award (toJ.L.Rokita).WearegratefultoTim Mercer for providing RNA sequins effective combination to circumvent cellular reprogramming. for RNA sequencing and to the Jefferson Cancer Genomics Laboratory While no Hippo pathway modulating drugs are currently been for library preparation and next-generation sequencing. pQCXIH-Myc- tested in the clinic, there is increasing interest within academia and YAP-5SA was a gift from Kunliang Guan (Addgene plasmid # 33093; industry to develop inhibitors of YAP1 activity (57). It is important http://n2t.net/addgene:33093; RRID:Addgene_33093). pLenti CMV to note that the clinical relevance of the combination of YAP1 and Puro DEST (w118-1) was a gift from Eric Campeau & Paul Kaufman MEK inhibition in neuroblastoma would be limited to tumors (Addgene plasmid # 17452; http://n2t.net/addgene:17452; RRID: Addgene_17452). that both harbor RAS–MAPK pathway mutations and express de novo YAP1 ( and/or induced by MEK inhibition). As inhibitors The costs of publication of this article were defrayed in part by the of YAP1 activity are developed, our data support the development payment of page charges. This article must therefore be hereby marked of combined MEK1/2 and YAP1 inhibition for neuroblastomas advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate with hyperactivated MAPK signaling. this fact.

Disclosure of Potential Conflicts of Interest Received May 6, 2019; revised August 23, 2019; accepted October 16, 2019; No potential conflicts of interest were disclosed. published first October 31, 2019.

References 1. Hoehner JC, Gestblom C, Hedborg F, Sandstedt B, Olsen L, Pahlman S. A 12. Schramm A, Koster J, Assenov Y, Althoff K, Peifer M, Mahlow E, et al. developmental model of neuroblastoma: differentiating stroma-poor Mutational dynamics between primary and relapse neuroblastomas. tumors' progress along an extra-adrenal chromaffin lineage. Lab Invest Nat Genet 2015;47:872–7. 1996;75:659–75. 13. Rader J, Russell MR, Hart LS, Nakazawa MS, Belcastro LT, Martinez D, et al. 2. Park JR, Eggert A, Caron H. Neuroblastoma: biology, prognosis, and Dual CDK4/CDK6 inhibition induces cell-cycle arrest and senescence in treatment. Hematol Oncol Clin North Am 2010;24:65–86. neuroblastoma. Clin Cancer Res 2013;19:6173–82. 3. Maris JM, Matthay KK. Molecular biology of neuroblastoma. J Clin Oncol 14. Hart LS, Rader J, Raman P, Batra V, Russell MR, Tsang M, et al. Preclinical 1999;17:2264–79. therapeutic synergy of MEK1/2 and CDK4/6 inhibition in neuroblastoma. 4. Maris JM, Hogarty MD, Bagatell R, Cohn SL. Neuroblastoma. Lancet 2007; Clin Cancer Res 2017;23:1785–96. 369:2106–20. 15. Umapathy G, Guan J, Gustafsson DE, Javanmardi N, Cervantes-Madrid D, 5. Park JR, Bagatell R, London WB, Maris JM, Cohn SL, Mattay KK, et al. Djos A, et al. MEK inhibitor trametinib does not prevent the growth of Children's oncology group's 2013 blueprint for research: neuroblastoma. anaplastic lymphoma kinase (ALK)-addicted neuroblastomas. Sci Signal Pediatr Blood Cancer 2013;60:985–93. 2017. doi: 10.1126/scisignal.aam7550. 6. Maris JM. Recent advances in neuroblastoma. N Engl J Med 2010;362: 16. Lugowska I, Kosela-Paterczyk H, Kozak K, Rutkowski P. Trametinib: a MEK 2202–11. inhibitor for management of metastatic melanoma. Onco Targets Ther 7. Cohn SL, Pearson AD, London WB, Monclair T, Ambros PF, Brodeur GM, 2015;8:2251–9. et al. The International Neuroblastoma Risk Group (INRG) classification 17. Lito P, Saborowski A, Yue J, Solomon M, Joseph E, Gadal S, et al. system: an INRG task force report. J Clin Oncol 2009;27:289–97. Disruption of CRAF-mediated MEK activation is required for effec- 8. Mosse YP, Laudenslager M, Longo L, Cole KA, Wood A, Attiyeh EF, et al. tive MEK inhibition in KRAS mutant tumors. Cancer Cell 2014;25: Identification of ALK as a major familial neuroblastoma predisposition 697–710. gene. Nature 2008;455:930–5. 18. Lito P, Rosen N, Solit DB. Tumor adaptation and resistance to RAF 9. Eleveld TF, Oldridge DA, Bernard V, Koster J, Colmet Daage L, Diskin SJ, inhibitors. Nat Med 2013;19:1401–9. et al. Relapsed neuroblastomas show frequent RAS-MAPK pathway 19. Zhao Y, Adjei AA. The clinical development of MEK inhibitors. Nat Rev Clin mutations. Nat Genet 2015;47:864–71. Oncol 2014;11:385–400. 10. Padovan-Merhar OM, Raman P, Ostrovnaya I, Kalletla K, Rubnitz KR, 20. YaoZ,TorresNM,TaoA,GaoY,LuoL,LiQ,etal.BRAFmutantsevade Sanford EM, et al. Enrichment of targetable mutations in the relapsed ERK-dependent feedback by different mechanisms that determine neuroblastoma genome. PLoS Genet 2016;12:e1006501. their sensitivity to pharmacologic inhibition. Cancer Cell 2015;28: 11. Schleiermacher G, Javanmardi N, Bernard V, Leroy Q, Cappo J, Rio Frio T, 370–83. et al. Emergence of new ALK mutations at relapse of neuroblastoma. J Clin 21. Huang J, Wu S, Barrera J, Matthews K, Pan D. The Hippo signaling Oncol 2014;32:2727–34. pathway coordinately regulates cell proliferation and apoptosis by

www.aacrjournals.org Cancer Res; 79(24) December 15, 2019 6213

Downloaded from cancerres.aacrjournals.org on October 4, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst October 31, 2019; DOI: 10.1158/0008-5472.CAN-19-1415

Coggins et al.

inactivating Yorkie, the Drosophila homolog of YAP. Cell 2005;122: 39. Lin L, Sabnis AJ, Chan E, Olivas V, Cade L, Pazarentzos E, et al. The Hippo 421–34. effector YAP promotes resistance to RAF- and MEK-targeted cancer ther- 22. Dong J, Feldmann G, Huang J, Wu S, Zhang N, Comerford SA, et al. apies. Nat Genet 2015;47:250–6. Elucidation of a universal size-control mechanism in Drosophila and 40. ShaoDD,XueW,KrallEB,BhutkarA,PiccioniF,WangX,etal.KRAS mammals. Cell 2007;130:1120–33. and YAP1 converge to regulate EMT and tumor survival. Cell 2014;158: 23. Overholtzer M, Zhang J, Smolen GA, Muir B, Li W, Sgroi DC, et al. 171–84. Transforming properties of YAP, a candidate oncogene on the chromo- 41. Hong X, Nguyen HT, Chen Q, Zhang R, Hagman Z, Voorhoeve PM, et al. some 11q22 amplicon. Proc Natl Acad Sci U S A 2006;103:12405–10. Opposing activities of the Ras and Hippo pathways converge on regulation 24. Chan SW, Lim CJ, Chen L, Chong YF, Huang C, Song H, et al. The Hippo of YAP protein turnover. EMBO J 2014;33:2447–57. pathway in biological control and cancer development. J Cell Physiol 42. Slemmons KK, Crose LE, Rudzinski E, Bentley RC, Linardic CM. Role of the 2011;226:928–39. YAP oncoprotein in priming ras-driven rhabdomyosarcoma. PLoS One 25. Chen Q, Zhang N, Xie R, Wang W, Cai J, Choi KS, et al. Homeostatic control 2015;10:e0140781. of Hippo signaling activity revealed by an endogenous activating mutation 43. Seong BK, Fathers KE, Hallett R, Yung CK, Stein LD, Mouaaz S, et al. A in YAP. Genes Dev 2015;29:1285–97. metastatic mouse model identifies genes that regulate neuroblastoma 26. Zhao B, Wei X, Li W, Udan RS, Yang Q, Kim J, et al. Inactivation of YAP metastasis. Cancer Res 2017;77:696–706. oncoprotein by the Hippo pathway is involved in cell contact inhibition 44. Attiyeh EF, London WB, Mosse YP, Wang Q, Winter C, Khazi D, et al. and tissue growth control. Genes Dev 2007;21:2747–61. Chromosome 1p and 11q deletions an doutcomes in neuroblastoma. 27. Pan D. The Hippo signaling pathway in development and cancer. Dev Cell N Engl J Med 2005;353:2243–53. 2010;19:491–505. 45. Mlakar V, Mlakar SJ, Lopez G, Maris JM, Ansari M, Gumy-Pause F. 11q 28. Yu FX, Zhao B, Panupinthu N, Jewell JL, Lian I, Wang LH, et al. Regulation deletion in neuroblastoma: a review of biological and clinical implica- of the Hippo-YAP pathway by G-protein-coupled signaling. Cell tions. Mol Cancer 2017;16:114. 2012;150:780–91. 46. Harenza JL, Diamond MA, Adams RN, Song MM, Davidson HL, Hart LS, 29. Wu S, Huang J, Dong J, Pan D. Hippo encodes a Ste-20 family protein et al. Transcriptomic profiling of 39 commonly-used neuroblastoma cell kinase that restricts cell proliferation and promotes apoptosis in conjunc- lines. Sci Data 2017;4:170033. tion with salvador and warts. Cell 2003;114:445–56. 47. Campeau E, Ruhl VE, Rodier F, Smith CL, Rahmberg BL, Fuss JO, et al. A 30. Wei X, Shimizu T, Lai ZC. Mob as tumor suppressor is activated by versatile viral system for expression and depletion of proteins in mam- Hippo kinase for growth inhibition in Drosophila. EMBO J 2007;26: malian cells. PLoS One 2009;4:e6529. 1772–81. 48. Hardwick SA, Chen WY, Wong T, Deveson IW, Blackburn J, Andersen SB, 31. Lei QY, Zhang H, Zhao B, Zha ZY, Bai F, Pei XH, et al. TAZ promotes cell et al. Spliced synthetic genes as internal controls in RNA sequencing proliferation and epithelial-mesenchymal transition and is inhibited by experiments. Nat Methods 2016;13:792–8. the Hippo pathway. Mol Cell Biol 2008;28:2426–36. 49. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. STAR: 32. Steinhardt AA, Gayyed MF, Klein AP, Dong J, Maitra A, Pan D, et al. ultrafast universal RNA-seq aligner. Bioinformatics 2013;29:15–21. Expression of Yes-associated protein in common solid tumors. 50. Li B, Dewey CN. RSEM: accurate transcript quantification from RNA-Seq Hum Pathol 2008;39:1582–9. data with or without a reference genome. BMC Bioinformatics 2011;12: 33. Chen L, Chan SW, Zhang X, Walsh M, Lim CJ, Hong W, et al. Structural 323. basis of YAP recognition by TEAD4 in the Hippo pathway. Genes Dev 51. Wong T, Deveson IW, Hardwick SA, Mercer TR. ANAQUIN: a software 2010;24:290–300. toolkit for the analysis of spike-in controls for next generation sequencing. 34. Liu-Chittenden Y, Huang B, Shim JS, Chen Q, Lee SJ, Anders RA, et al. Bioinformatics 2017;33:1723–4. Genetic and pharmacological disruption of the TEAD-YAP complex sup- 52. Chen J, Bardes EE, Aronow BJ, Jegga AG. ToppGene suite for gene list presses the oncogenic activity of YAP. Genes Dev 2012;26:1300–5. enrichment analysis and candidate gene prioritization. Nucleic Acids Res 35. Cottini F, Hideshima T, Xu C, Sattler M, Dori M, Agnelli L, et al. Rescue of 2009;37:W305–11. Hippo coactivator YAP1 triggers DNA damage-induced apoptosis in 53. Wei JS, Song YK, Durinck S, Chen QR, Cheuk AT, Tsang P, et al. The MYCN hematological cancers. Nat Med 2014;20:599–606. oncogene is a direct target of miR-34a. Oncogene 2008;27:5204–13. 36. Adler JJ, Johnson DE, Heller BL, Bringman LR, Ranahan WP, Conwell 54. Kim MH, Kim J, Hong H, Lee SH, Lee JK, Jung E, et al. Actin remodeling MD, et al. Serum deprivation inhibits the transcriptional co-activator confers BRAF inhibitor resistance to melanoma cells through YAP/TAZ YAP and cell growth via phosphorylation of the 130-kDa isoform of activation. EMBO J 2016;35:462–78. angiomotin by the LATS1/2 protein kinases. Proc Natl Acad Sci U S A 55. Rajbhandari P, Lopez G, Capdevila C, Salvatori B, Yu J, Rodriguez-Barrueco 2013;110:17368–73. R, et al. Cross-cohort analysis identifies a TEAD4-MYCN positive feedback 37. Rayego-Mateos S, Rodrigues-Diez R, Morgado-Pascual JL, Rodrigues loop as the core regulatory element of high-risk neuroblastoma. Diez RR, Mas S, Lavoz C, et al. Connective tissue growth factor is a Cancer Discov 2018;8:582–99. new ligand of epidermal growth factorreceptor.JMolCellBiol2013;5: 56. Strieder V, Lutz W. E2F proteins regulate MYCN expression in neuroblas- 323–35. tomas. J Biol Chem 2003;278:2983–9. 38. Kapoor A, Yao W, Ying H, Hua S, Liewen A, Wang Q, et al. Yap1 activation 57. Chan P, Han X, Zheng B, DeRan M, Yu J, Jarugumilli GK, et al. Autop- enables bypass of oncogenic Kras addiction in pancreatic cancer. Cell 2014; almitoylation of TEAD proteins regulates transcriptional output of the 158:185–97. Hippo pathway. Nat Chem Biol 2016;12:282–9.

6214 Cancer Res; 79(24) December 15, 2019 Cancer Research

Downloaded from cancerres.aacrjournals.org on October 4, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst October 31, 2019; DOI: 10.1158/0008-5472.CAN-19-1415

YAP1 Mediates Resistance to MEK1/2 Inhibition in Neuroblastomas with Hyperactivated RAS Signaling

Grace E. Coggins, Alvin Farrel, Komal S. Rathi, et al.

Cancer Res 2019;79:6204-6214. Published OnlineFirst October 31, 2019.

Updated version Access the most recent version of this article at: doi:10.1158/0008-5472.CAN-19-1415

Supplementary Access the most recent supplemental material at: Material http://cancerres.aacrjournals.org/content/suppl/2019/10/31/0008-5472.CAN-19-1415.DC1

Cited articles This article cites 56 articles, 18 of which you can access for free at: http://cancerres.aacrjournals.org/content/79/24/6204.full#ref-list-1

Citing articles This article has been cited by 9 HighWire-hosted articles. Access the articles at: http://cancerres.aacrjournals.org/content/79/24/6204.full#related-urls

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at Subscriptions [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://cancerres.aacrjournals.org/content/79/24/6204. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from cancerres.aacrjournals.org on October 4, 2021. © 2019 American Association for Cancer Research.