<<

Published OnlineFirst February 24, 2015; DOI: 10.1158/0008-5472.CAN-14-2785 Therapeutics, Targets, and Chemical Biology Research

PDK1 and SGK3 Contribute to the Growth of BRAF-Mutant and Are Potential Therapeutic Targets Marzia Scortegagna1, Eric Lau1, Tongwu Zhang2, Yongmei Feng1, Chris Sereduk3, Hongwei Yin3, Surya K. De1, Katrina Meeth4,5, James T. Platt4,5, Casey G. Langdon4,5, Ruth Halaban4, Maurizio Pellecchia1, Michael A. Davies6, Kevin Brown2, David F. Stern5, Marcus Bosenberg4,5, and Ze'ev A. Ronai1

Abstract

Melanoma development involves members of the AGC screen, pan-PI3K inhibition synergized with PDK1 inhibition to family, including AKT, PKC, and, most recently, PDK1, as eluci- suppress growth, suggesting that focused blockade of dated recently in studies of Braf::Pten mutant melanomas. Here, we PDK1/PI3K signaling might offer a new therapeutic modality for report that PDK1 contributes functionally to skin pigmentation wild-type PTEN tumors. We also noted that responsiveness to and to the development of melanomas harboring a wild-type PDK1 inhibition associated with decreased expression of pigmen- PTEN genotype, which occurs in about 70% of human melano- tation and increased expression of cytokines and inflamma- mas. The PDK1 substrate SGK3 was determined to be an important tory genes, suggesting a method to stratify patients with melanoma mediator of PDK1 activities in melanoma cells. Genetic or phar- for PDK1-based therapies. Overall, our work highlights the poten- macologic inhibition of PDK1 and SGK3 attenuated melanoma tial significance of PDK1 as a therapeutic target to improve mel- growth by inducing G1 phase cell-cycle arrest. In a synthetic lethal anoma treatment. Cancer Res; 75(7); 1–14. 2015 AACR.

Introduction Using this model, we showed that genetic inactivation or pharmacologic inhibition of PDK1 delays melanoma develop- Approximately 70% of melanomas with mutated BRAF also ment and metastasis. However, wild-type (WT) PTEN is exhibit the inactivation of the tumor suppressor PTEN, result- expressed in a sizable fraction (70%) of BRAF-mutant human ing in constitutive activation of the PI3K signaling pathway (1). melanomas (1, 6, 7), and the role of PDK1 in the progression of Phosphoinositide-dependent kinase 1 (PDK1), an immediate such melanomas is unknown. Here, we used genetic and downstream of PI3K, is a master kinase able to phos- pharmacologic models to show that PDK1 plays an even more phorylate more than 20 members of the AGC kinase family, significant role in the development of WT Pten, BrafV600E mouse which includes PKA, AKT, PKC, p70S6k, and SGK (2, 3). The and human melanomas, compared with the Pten-deficient, relationship between PDK1 and PTEN was first revealed by the BrafV600E melanomas. demonstration that the lethality of Pten deficiency in flies was Although studies in several cancer types suggest that AKT is the rescued by deletion of Pdk1, establishing PDK1 as the main main downstream effector of the PI3K/PDK1 signaling pathway, downstream effector of PI3K (4). Recently, we investigated the increasing evidence indicates that additional factors are equally role of PDK1 in melanomas using a mouse model in which important (8–10). For example, the overexpression of AKT in expression of mutated BRAF (BRAFV600E) and deletion of Pten is PDK1-knockout (KO) cancer cells was demonstrated to be insuf- conditionally and specifically activated in melanocytes (5). ficient to restore the malignant phenotype (11). The 3 isoforms of the SGK family of AGC , SGK1, SGK2, and SGK3, are also activated by the PI3K/PDK1 signaling 1Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California. 2Division of Cancer Epidemiology and Genetics, Laboratory pathway. SGKs exhibit similar substrate specificity to AKT, and of Translational Genomics, NCI, Bethesda, Maryland. 3Cancer and Cell both kinases influence the activity of involved in cell Biology Division, The Translational Genomics Research Institute growth, survival, and migration (12, 13). Several studies have (TGen), Phoenix, Arizona. 4Department of Dermatology, Yale Univer- sity, School of Medicine, New Haven, Connecticut. 5Department of demonstrated important roles for the SGK isoforms in PI3K Pathology,Yale University, School of Medicine, New Haven, Connecti- signaling in both physiologic and pathologic conditions. SGK1 6 cut. Melanoma Medical Oncology, MD Anderson Cancer Center, and SGK3 are ubiquitously expressed, whereas SGK2 is restrict- Houston, Texas. ed to the kidney, pancreas, liver, and brain. Given the function Note: Supplementary data for this article are available at Cancer Research of SGK1 and SGK3 in cell proliferation and survival, it is not Online (http://cancerres.aacrjournals.org/). surprising that they have been shown to be involved in the Correspondence Author: Ze'ev A. Ronai, Sanford-Burnham Medical Research growth of several (14, 15). However, their contribution Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037. Phone: 858-646-3185; in melanoma remains unclear. Fax: 815-366-8003; E-mail: [email protected] In this study, we identify SGKs as key mediators of PDK1 doi: 10.1158/0008-5472.CAN-14-2785 activity in melanoma and demonstrate the importance of the 2015 American Association for Cancer Research. PDK1/SGK signaling axis in the growth of PTEN WT melanomas.

www.aacrjournals.org OF1

Downloaded from cancerres.aacrjournals.org on October 5, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst February 24, 2015; DOI: 10.1158/0008-5472.CAN-14-2785

Scortegagna et al.

AB C PDK1 WT PDK1 WT PDK1 KO PDK1 WT PDK1 KO

PDK1 KO D

PDK1 WT PDK1 KO E F 5 100 4 PDK1 WT 80 3 PDK1KO

2 60 1

# Tumor/mouse 0 40

Percent survival 20 O 0 PDK1 WT PDK1 K 0 20406080 Days G H

PDK1 WT PDK1 KO PDK1 WT PDK1 KO

I

PDK1 WT PDK1 KO K WT KO P-PDK1

P-AKT 308

P-FOXO3

P-GSK3β

P-PRAS40 J P-RSK 15 PDK1 WT PDK1 KO P-P70S6K

10 P-NDRG1

* P-PKC 5 AKT % S100 positive cells β-Actin PDK1 WT PDK1 KO

OF2 Cancer Res; 75(7) April 1, 2015 Cancer Research

Downloaded from cancerres.aacrjournals.org on October 5, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst February 24, 2015; DOI: 10.1158/0008-5472.CAN-14-2785

PDK1 and SGK3 in BRAF-Mutant Melanoma

We also demonstrate that PI3K inhibitor can synergize with PDK1 silencing and transfection inhibitors in suppressing melanoma growth and point to possible shRNAs for mouse Pdk1, Sgk1,andSgk3 were purchased from means for the stratification of human BRAFV600E PTEN WT tumors Sigma. Viral particles were produced in HEK293T cells trans- for PDK-targeted therapies. fected with the plasmid of interest and appropriated packaging plasmids using Jet Prime (Polyplus transfection). Target cells Materials and Methods were infected with viral particles by spinoculation in the pres- ence of polybrene (4 mg/mL; Sigma). Stable clones were estab- Primary melanoma cells and human melanoma cell lines lished by growth in media containing puromycin (1 mg/mL; Murine melanoma cells Sanford Burnham Melanoma A2 InVivoGen). (SBM-A2) and SBM-A3 were derived from primary cutaneous þ/þ / lesions from Braf ::Pten ::Cdkn2a mice. Tumors were cut Western blotting. in small parts and digested with collagenase (10 mg/mL; Sigma) Cells were harvested and lysed in RIPA buffer [50 mmol/L fi m for 1 hour at 37 C and ltered through a 100- m nylon cell Tris-HCl, pH 7.5, 150 mmol/L NaCl, 1% Triton X-100, 0.1% strainer (BD Falcon). Cells were resuspended in DMEM supple- SDS, 0.1% sodium deoxycholate, 1 mmol/L EDTA, 1 mmol/L mented with 10% FBS and penicillin/streptomycin and incubated fl sodium orthovanadate, 1 mmol/L phenylmethylsulfonyl uor- at 37 C. Once established, cell lines were passaged twice before ide (PMSF), 10 mg/mL aprotinin, and 10 mg/mL leupeptin]. Cell use in experiments. The mouse YUMM1.5 and YUMM1.9 and the lysates were subjected to SDS-PAGE and the proteins were human 501MEL and UACC903 cell lines were maintained in transferred onto nitrocellulose membranes (Osmonics Inc.). DMEM medium supplemented with 10% FBS and penicillin/ Membranes were incubated with primary antibodies for 18 streptomycin. Low-passage human melanoma cells YUHEF, hours at 4C, washed, and then incubated with secondary YUSIK, YUMAC, YUGASP, and YUROB were obtained from the antibody conjugated with fluorescent dye. After processing, Tissue Resource Core of the Yale SPORE in and all cell membranes were analyzed usingtheOdysseyImagingSystem lines were used at or under passage 25 as has been described (16). (Amersham Biosciences). These melanoma cells were maintained in OptiMEM supplemen- ted with 5% FBS and 1% penicillin/streptomycin. Information on Immunofluorescence microscopy the genotype and origin of the cell lines used is shown in Sections of skin and lymph nodes prepared as described above Supplementary Table S2. were deparaffinized,rehydrated,washedin PBS,and incubated with Dako Block for 30 minutes at room temperature. Antigen T2 Activation of the Tyr::CreER transgene retrieval for S100 immunostaining was performed by incubation in 4-Hydroxytamoxifen (4-HT) was prepared at 50 mg/mL in citrate buffer (pH 6.0) in a decloaking chamber (Biocare Medical). m Antibodies were diluted in Dako antibody diluent at 1:500 and DMSO and 10 L was applied to the dorsal skin on postnatal days 1, 3, and 5 using a small paintbrush. incubated with sections overnight at 4 C. Secondary antibodies conjugated to Alexa Fluor 594 (Molecular Probes) were diluted to 1: 400 and incubated with sections for 1 hour at room temperature. Histologic analyses Nuclei were counterstained with SlowFade Gold anti-fade reagent fi Tumors sections were xed overnight in Z-Fix (buffered zinc containing 40,6-diamidino-2-phenylindole (DAPI; Vector). formalin fixative, Anatech) at 4C. Sections were then washed twice with PBS and processed for paraffin embedding. Paraffin qRT-PCR analysis blocks were sliced at 5 mm and sections were stained with Total RNA was extracted using a miniprep kit (Sigma) and hematoxylin and eosin (H&E). digested with DNase I. cDNA was synthesized using oligo(dT) and random hexamer primers, and qPCR was performed on Antibodies and reagents biological triplicates using SYBR Green. Amplification of histone The following antibodies were purchased from Cell Signaling H3.3A served as an internal control. The PCR primers were Technologies: pNDRG1, pPDK1, pAKT308, pAKT473, pGSK3b designed using Primer3 and their specificity was checked using (Ser 9), pPRAS40 (Thr 246), pFOXO3a (Thr 32), pERK1/2, AKT, BLAST. The PCR products were limited to 100 to 200 base pairs. 0 FOXO3a, pP70S6K (Thr 389), pS6K (Ser 235/236), PRAS40, Primer sequences were: mouse H3.3a:forward,5-aagcagactgcccg- 0 0 0 GSK3b, ERK1/2, SGK3, cyclin D1, PTEN, P70S6K, and S6. caaat-3 and reverse, 5 -ggcctgtaacgatgaggtttc-3 ;mousePdk1:for- 0 0 0 Antibodies against b-actin, SGK1, PKC, and tubulin were pur- ward, 5 -acgccctgaagacttcaagtttg-3 and reverse, 5 -gccagttctcgggc- 0 0 0 chased from Santa Cruz Biotechnology. 4-HT and antibodies caga-3 ;mouseSgk1:forward,5-cgtccgaacgggacaacat-3 and 0 0 0 against S100 were purchased from Sigma. Synthesis of the reverse, 5 -gtccaccgtccggtcatac-3 ;mouseSgk3: forward, 5 - 0 0 0 PDK1 inhibitor GSK2334470 was performed as previously tcccagctctgacgaacaca-3 and reverse, 5 -tcaaactctgcgtatctcctga-3 ; 0 0 0 described (5). mouse Dct: forward, 5 -gtcctccactcttttacagacg-3 and reverse, 5 -

Figure 1. þ þ þ þ Loss of PDK1 inhibits the onset of melanoma development and delays metastasis. A and B, representative images of PDK1 WT (BrafV600E::Cdkn2a / ::Pten / ::Pdk1 / )or þ þ PDK1 KO (BrafV600E::Cdkn2a / ::Pten / ::Pdk1 / ) mice 21 days after administration of 4-HT. C and D, representative images of the dorsal skin (C) and lymph nodes (D) from PDK1 WT and PDK1 KO mice 38 days after systemic administration of 4-HT. E, quantification of tumors in PDK1 WT and PDK1 KO mice (n ¼ 12 and 8, respectively). F, Kaplan–Meier survival curves of PDK1 WT and PDK1 KO mice (n ¼ 16 and 18, respectively). P < 0.001 by log-rank (Mantel–Cox) test. G and H, H&E-stained (G) and S100-immunostained (H) skin sections from PDK1 WT and PDK1 KO mice 38 days after 4-HT administration. Bars, 100 mm. I and J, H&E-stained (I) and S100 þ (J)-immunostained lymph nodes from PDK1 WT and PDK1 KO mice 38 days after administration of 4-HT. Bars, 100 mm. Graph shows mean SEM of S100 cells from three animals per genotype. , P < 0.005. K, Western blot analysis of the indicated proteins in primary melanoma cultures derived from PDK1 WT and PDK1 KO mice.

www.aacrjournals.org Cancer Res; 75(7) April 1, 2015 OF3

Downloaded from cancerres.aacrjournals.org on October 5, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst February 24, 2015; DOI: 10.1158/0008-5472.CAN-14-2785

Scortegagna et al.

attcggttgtgaccaatgggt-30;mouseTyr:forward,50-tctggacct- aprotinin 10 mg/mL]. Lysates were transferred at volumes of cagttccccttc-30 and reverse, 50-aacttacagtttccgcagttga-30;mouseIl6: 25 to 30 mL into a PCR 96-well plate. Ten microliters of 4 SDS/ forward, 50-ctgcaagagacttccatccag-30 and reverse, 50-agtggtataga- 2-ME sample buffer (35% glycerol, 8% SDS, 0.25 mol/L Tris- caggtctgttgg-3; mouse Mmp3: forward, 50-ggcctggaacagtcttggc-30 HCl, pH 6.8; with 10% b-mercaptoethanol added before use) and reverse, 50-tgtccatcgttcatcatcgtca-30;mouseZeb1:forward,50- was added to each sample well. The plates were covered accgccgtcatttatcctgag-30 and reverse, 50-catctggtgttccgttttcatca-30. and incubated for 5 minutes at 95C and then centrifuged for 1 minute at 2,000 rpm. Samples were applied to RPPA slides as fi Quanti cation of lymph node metastasis previously described (19). Immunofluorescent staining of S100 in lymph node sections was performed as described above. S100-positive tumor cells in Synthetic lethal screen for PDK1 inhibitor combination the sections were quantified by first scanning at 20 magnifica- Interactions between PDKi (GSK2334470) and 45 other agents tion with the Aperio ScanScope FL system (Aperio Technologies) were determined in a "one versus many" screen at the Yale Center and then analyzing cell numbers using the Area Quantification FL for Molecular Discovery. A master 384-well plate was set up algorithm (version 11, Aperio Technologies). The algorithm was manually, incorporating 4 or 8 dilutions of 45 test agents to yield tuned using a preset procedure and the subsequent macro was a range from 10 mmol/L to 1 nmol/L final in the experiments. The saved and applied to all slides. master plates also included multiple negative control wells (0.2% DMSO vehicle) and staurosporine-positive "kill" controls for a Colony formation assay final 10 mmol/L staurosporine. The test agents consisted of 2,4- Five hundred tumor cells were plated into each well of a 6-well dinitrophenol, 4u8c, 17-DMAG, 4485, atorvastatin, AZD-2014, plate and incubated for 7 to 12 days. Viable colonies were stained AZD-6244, BI-D1870, BKM-120, bortezomib, brefeldin A, carfil- with crystal violet (Sigma-Aldrich) and the plates were imaged. zomib, cerulenin, EMD638683, EX 527, FASN211, fatostatin A, The colony numbers and intensity were determined using ImageJ FCCP, GGTI-298, GSK650394A, GSK690693, GSK2334470, software. Each experiment was performed at least 3 times. GSK2606416, homoharringtonine, metformin, oligomycin, PF429242, phenformin, piperlongumine, rosiglitazone, SB in vivo Animal studies and experiments 204990, SBRI108610, SBRI108634, SBRI108684, SBRI108692, All mouse experiments were performed under the guidelines SBRI108734, SBRI108740, simvastatin, SRT1720, STA-4783, STF- of the Institutional Animal Care and Use Committee (IACUC) 083010, T0070907, , tunicamycin, and . A V600E of Sanford-Burnham Medical Research Institute. Braf :: total of 750 to 1,000 cells per 16 mL medium per well were / þ/þ / Cdkn2a ::Pten and Pdk1 mice were generated as previ- dispensed into 384-well plates using a Thermo multidrop dis- ously described (17, 18). Cohorts of at least 6 animals per group penser. After incubation overnight to allow cell attachment, a pin were used in each of the experimental groups. tool was used to transfer 20 nL of compounds from each well of the master plate into the 384-well test plates containing cells. Three-dimensional growth assay Next, 4 mL of either 0.1% DMSO or GSK233470 to yield final 2 or Cells were induced to form spheroids using the hanging drop 10 mmol/L were added. Cells were incubated an additional 3 days, method. The cells were plated at 200 cells per 20 mL per well in a and growth was quantified by CellTiterGlo (Promega) to read out Nunc-60 well microwell MiniTray. The trays were covered, ATP accumulation, which correlates well with viable cell number. inverted, and incubated at 37 Cinahumidified 5% CO 2 Additivity and superadditivity were calculated using the Bliss incubator for 5 days. Spheroids from wells containing single independence model and area under the curve (AUC) calcula- spheroids were transferred to a 48-well plate coated with 1% tions. To avoid overweighting the high concentration points, the low-melting-point agarose. Compounds or DMSO vehicle were natural log concentrations are used to calculate AUC. AUC cal- added, and images of spheroids were captured every 48 hours culation is performed using the function "auc" from the R library for 8 days using an Olympus IX-71 microscope equipped with a "MESS," using the linear type option. This function approximates camera. The relative spheroid areas were measured using Ima- the area under a set of points by summing the trapezoidal geJ64 software. segments between adjacent points. Normalization of the AUC Flow cytometric cell-cycle analysis calculation is performed by dividing the calculated AUC by the Cell lines were seeded in 6-well tissue culture plates at 1 105 width of the log concentration range for each plated agent to cells per well, incubated overnight, and then treated with shRNAs. correct for differences in concentration ranges. Cells were harvested by trypsinization, fixed in 70% ethanol in PBS Statistical analysis at 20 C, and then stored until further use. For analysis, cells were All data except survival curves were analyzed by the unpaired t washed once in PBS and incubated in cell-cycle staining buffer (60 test. Kaplan–Meier survival curves were compiled using Prism mg/mL propidium iodide and 0.15 mg/mL RNase A; Sigma) for 20 software (GraphPad) and statistical significance was assessed minutes. Biological triplicates of 10,000 cells (within the G –G 1 2 using the log-rank (Mantel–Cox) test. P < 0.05 was considered gates) per sample were collected for each experiment, and the data statistically significant. were analyzed using FlowJo software (TreeStar).

Reverse-phase protein array analysis Results The cells were washed twice in ice-cold PBS, then lysed in 30 PDK1 deletion promotes tumor suppression and reduces the V600E / mL of reverse-phase protein array (RPPA) lysis buffer [1% Triton pigmentation of Braf ::Cdkn2a mice harboring WT Pten X-100, 50 nmol/L HEPES (pH 7.4), 150 nmol/L NaCl, 1.5 Our previous studies demonstrated that the genetic inacti- V600E / / fi nmol/L MgCl2, 1 mmol/L EGTA, 100 nmol/L NaF, 10 nmol/L vation of Pdk1 in Braf ::Cdkn2a ::Pten animals signi - NaPPi, 10% glycerol, 1 nmol/L PMSF, 1 nmol/L Na3VO4,and cantly delays melanoma development and effectively inhibits

OF4 Cancer Res; 75(7) April 1, 2015 Cancer Research

Downloaded from cancerres.aacrjournals.org on October 5, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst February 24, 2015; DOI: 10.1158/0008-5472.CAN-14-2785

PDK1 and SGK3 in BRAF-Mutant Melanoma

Figure 2. shRNA-mediated inhibition of PDK1 or SGK3 suppresses of target proteins, expression of cyclin D1, and growth of melanoma cells. A, Western blot analysis of the indicated proteins in primary melanoma cultures derived from BrafV600E::Cdkn2a / ::PTEN / mice (YUMM1.5 and YUMM1.9) or þ þ BrafV600E::Cdkn2a / ::PTEN / mice (SBM-A2 and SBM-A3). B, Western blot analysis of the indicated proteins from YUMM1.5 and SBM-A2 melanoma cells expressing control, Sgk1-, Pdk1-,orSgk3-targeted shRNA. C and D, colony formation assay of SBM-A2 (C) and YUMM1.5 (D) cultures expressing the indicated shRNAs. Individual wells shown are representative of three experiments with triplicate cultures. The graph represents the quantification of colony-forming ability compared with control shRNA. Error bars, SEM. E, representative images of YUMM1.5 spheroids expressing the indicated shRNAs. Relative spheroid sizes were quantified using ImageJ software (NIH) and are presented as the mean SEM of 6 spheroids per group.

www.aacrjournals.org Cancer Res; 75(7) April 1, 2015 OF5

Downloaded from cancerres.aacrjournals.org on October 5, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst February 24, 2015; DOI: 10.1158/0008-5472.CAN-14-2785

Scortegagna et al.

þ þ metastasis (5). Because about 70% of human melanomas harbor Cdkn2a / ::Pten / mice. In these mice, the expression of WT PTEN, we sought to determine the effects of Pdk1 deletion on BrafV600E is induced specifically in melanocytes by the adminis- the development and progression of melanoma in BrafV600E:: tration of the estrogen analog 4-HT (5). BrafV600E::Cdkn2a / ::

Figure 3. Pharmacologic inhibition of PDK1 or SGK3 inhibits target and melanoma . A, Western blot analysis of the indicated proteins in YUMM1.5, YUMM1.9, SBM-A2, and SBM-A3 cells treated for 24 hours with DMSO (vehicle), 10 mmol/L GSK2334470 (PDK1i), or 10 mmol/L GSK650394 (SGKi). B and C, spheroid formation by YUMM1.5 (B) and YUMM1.9 (C) cells incubated with 5 or 20 mmol/L inhibitors. Spheroid volumes were quantified on the indicated days as described for Fig. 2. Results are the mean SEM of 6 spheroids per group.

OF6 Cancer Res; 75(7) April 1, 2015 Cancer Research

Downloaded from cancerres.aacrjournals.org on October 5, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst February 24, 2015; DOI: 10.1158/0008-5472.CAN-14-2785

PDK1 and SGK3 in BRAF-Mutant Melanoma

Figure 4. Inhibition of PDK1 or SGK3 induces cell-cycle arrest in G1. Cell-cycle distribution of SBM-A3 and YUMM1.5 cells stably expressing control shRNA (shSCR) or Pdk1-orSgk3-targeted shRNAs. Left, percentage of cells in the indicated cell-cycle phases. Values are the mean SEM of biologic triplicates. Right, representative FACS profiles of the DNA content (propidium iodide staining) of cells expressing the indicated shRNAs. Targeted , P < 0.05; , P < 0.001; , P < 0.0001.

Pdk1 / (PDK1 KO) mice systemically treated with 4-HT on We next investigated some of the key postnatal days 1, 3, and 5 exhibited markedly reduced pigmen- pathways regulated by PDK1. For this, we established early- tation on day 21 after 4-HT administration compared with passage (1–2) cell cultures of tumors from BrafV600E::Cdkn2a / þ þ þ þ þ þ þ þ similarly treated BrafV600E::Cdkn2a / ::Pdk1 / (Pdk1 WT) mice ::Pten / ::Pdk1 / and BrafV600E::Cdkn2a / ::Pten / ::Pdk1 / (Fig. 1A and B). Consistent with these observations, heavy pig- mice, which enabled the analysis of AGC kinase pathway com- mentation and melanomagenesis was observed in the skin (Fig. ponents. As expected, the absence of PDK1 resulted in a marked 1C) and lymph nodes (Fig. 1D) of PDK1 WT mice whereas it was reduction in the phosphorylation of downstream target proteins significantly reduced in the PDK1 KO mice. Examination of mice such as AKT, FOXO3a, GSK3b, PRAS40, RSK, p70S6K, and the on day 38 after 4-HT administration revealed that 7 of 12 (40%) SGK1 substrate NDRG1 (Fig. 1K). PDK1 WT mice contained tumors, whereas tumors were unde- Collectively, these data establish that PDK1 plays an important tectable in the PDK1 KO mice (Fig. 1E). Moreover, the mean role in the development of melanoma in BrafV600E/WT Pten mice, survival time of the PDK1 KO mice was significantly prolonged in addition to its reported role in the BrafV600E mice in which Pten compared with that of the PDK1 WT mice (76 vs. 42 days; P < is inactivated or deleted. 0.001; Fig. 1F). Notably, only a single administration of 4-HT was required to observe the effects of PDK1 deletion on melanoma- SGK1 and SGK3 are key mediators of PDK1-dependent genesis in the BrafV600E::Cdkn2a / animals (Supplementary melanomagenesis Fig. S1A). Under these conditions, tumor development was The results shown above identify several potential PDK1 sub- delayed (50 days) but still markedly reduced in the PDK1 KO strates that might be required for melanoma development in Pdk1 (2 of 8 mice) compared with the PDK1 WT (12/14 mice) mice WT mice, including the transcription factor FOXO3a, which was (Supplementary Fig. S1). also identified in our earlier study (5). Notably, FOXO3a is The marked effects of Pdk1 deletion on melanomagenesis in regulated by phosphorylation of the same phosphoacceptor sites BrafV600E::Cdkn2a / were confirmed by immunohistochemi- by either AKT or SGK. To determine whether SGK1 or SGK3 plays a cal and immunofluorescent evaluation of the skin. Consistent role in PDK1-dependent melanoma development, we performed with these effects, we observed the presence of significantly shRNA-mediated knockdown (KD) of Pdk1, Sgk1,orSgk3 in reduced pigmented, hyperproliferative foci within the skin two melanoma lines each derived from BrafV600E::Cdkn2a / :: sections (Fig. 1G) and lymph nodes (Fig. 1I) from Pdk1 KO Pten / mice (lines YUMM1.5 and YUMM1.9) and BrafV600E:: þ þ compared with Pdk1 WT mice. Similarly, immunofluorescence Cdkn2a / ::Pten / mice (lines SBM-A2 and SBM-A3) and exam- staining for S100, a marker for both melanotic and amela- ined the effects of KD on the expression and phosphorylation of notic melanomas, revealed substantially more tumor cells key PDK1 signaling components (Fig. 2A and Supplementary within the skin (Fig. 1H) and more extensive invasion of the Fig. S2A–S2D). As expected, the degree of AKT, FOXO3a, and lymph nodes (Fig. 1J) of Pdk1 WT compared with Pdk1 KO PRAS40 phosphorylation was notably lower in the PTEN WT mice. tumor cells, reflecting PTEN activity (Fig. 2A). The degree of the

www.aacrjournals.org Cancer Res; 75(7) April 1, 2015 OF7

Downloaded from cancerres.aacrjournals.org on October 5, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst February 24, 2015; DOI: 10.1158/0008-5472.CAN-14-2785

Scortegagna et al.

A 2 040 20 0 –20 Color key Value

YUHEF

YUSIK

YUMAC

YUGASP

YUROB

B BTZ 1 nmol/L BTZ 4 nmol/L BTZ 4 nmol/L BTZ 4 nmol/L

mol/L mol/L mol/L mol/L mol/L mol/L mol/L mol/L mol/L μmol/L μ μ μmol/L μmol/L μmol/L μmol/L

DMSO PDK1i 0.1PDK1i μmol/L 1 μmol/LPDK1i 5 μmol/LBTZ 1 nmol/LPDK1i 0.1 PDK1iμ 1 μPDK1imol/L 5 DMSO PDK1i 0.1PDK1i μ 1 μ PDK1i 5 μ BTZ 4 nmol/LPDK1i 0.1 PDK1iμ 1 PDK1i 5 μ DMSO PDK1i 0.1 μmol/LPDK1i 1 μmol/LPDK1i 5 BTZ 4 nmol/LPDK1i 0.1PDK1i μmol/L 1 μPDK1i 5 μmol/L DMSO PDK1i 0.1 μPDK1imol/L 1 μPDK1imol/L 5 BTZ 4 nmol/LPDK1i 0.1PDK1i 1 PDK1i 5 P-AKT 473

P-FOXO3

P-S6

ATF4

CHOP Cleav. casp.3

β-Tubulin

S6

AKT

FOXO3

PARP UACC903 cells Mel-501 cells

SBM-A2 cells YUMM1.9 cells

C PDK1i 1 μmol/L DMSO PDK1i 1 μmol/L BTZ 200 nmol/L BTZ 200 nmol/L 120 DMSO Day 1

80 PDK1i 1 μmol/L

BTZ 200 nmol/L Day 7 Relative area Relative 40 PDK1i 1 μmol/L BTZ 200 nmol/L

Day 1 3 5 7

OF8 Cancer Res; 75(7) April 1, 2015 Cancer Research

Downloaded from cancerres.aacrjournals.org on October 5, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst February 24, 2015; DOI: 10.1158/0008-5472.CAN-14-2785

PDK1 and SGK3 in BRAF-Mutant Melanoma

KD for Pdk1, Sgk1 and Sgk3 was largely efficient (Supplementary trations, whereas the effect of PDK1i was more pronounced at Fig. S2A–S2D). PDK1 KD effectively attenuated phosphorylation lower concentrations (Fig. 3B and C). These results suggest that of FOXO3a, NDRG1, S6, P70S6K, and 4EBP1 in both Pten WT and the role of PDK1 in melanoma growth is mediated, at least in part, mutant cell lines (Fig. 2B and Supplementary Fig. S2E). In contrast, by SGK. KD of Sgk1 abolished phosphorylation of NDRG1 but had min- imal effects on p70S6K, FOXO3a, PRAS40, and 4EBP1 and KD of Inhibition of SGK3 or PDK1 causes melanoma cell-cycle arrest Sgk3 reduced phosphorylation of 4EBP1, P70S6K, and S6 but had at G1 no effect on NDRG1 (Fig. 2B and Supplementary Fig. S2E). To determine the mechanism(s) by which genetic or pharma- Consistent with recent reports that SGK3 and p70S6K regulate cological inhibition of SGK3 suppresses melanoma growth, we cyclin D1 expression and cell growth (20), we observed reduced performed FACS analysis to determine the proportion of cells cyclin D1 expression after KD of PDK1 and SGK3 but not KD of in each phase of the cell cycle after KD of PDK1 or SGK3.We SGK1 (Fig. 2B and Supplementary Fig. S2E). The effects of SGK3 KD found that the expression of either shSgk3 or shPdk1 led to the on p70S6K phosphorylation and cyclin D1 protein expression in accumulation of BrafV600E::Cdkn2a / ::Pten / and BrafV600E:: fi fi / þ/þ melanoma cells were con rmed using an independent Sgk3-spec c Cdkn2a ::Pten cells in G1 (Fig. 4), and this finding was shRNA (Supplementary Fig. S2G). These results demonstrate that confirmed using an independent Sgk3-specific shRNA (Supple- the effects of PDK1 inhibition are closely mimicked by SGK3 mentary Fig. S3). These results indicate that the observed changes inhibition, raising the possibility that this protein might mediate in cell growth following the inhibition of SGK3 and PDK1 activity the effects of PDK1 on growth of mutant Braf, Pten WT melanomas. are attributed to cell-cycle arrest at the G1 phase. To test this further, we examined colony formation and 3- dimensional (3D) growth of YUMM1.5 (Pten / ) and SBM-A2 PDK1 inhibition synergizes with and PI3K/mTOR þ þ (Pten / ) cell lines expressing control or Pdk1-, Sgk1-, and Sgk3- inhibition to attenuate melanoma growth targeted shRNA. We found that clonal growth was virtually Combination therapies are often more potent inhibitors of abolished by KD of Pdk1 or Sgk3 but minimally affected by Sgk1 tumor growth and can also suppress the growth of tumors KD (Fig. 2C, Fig. 2D). Notably, KD of all 3 proteins reduced resistant to the individual therapies. Therefore, we next screened melanoma sphere formation and growth to some extent, with for pharmacologic inhibitors that might act synergistically with Pdk1 KD the most effective (90% inhibition vs. cells expressing PDK1i in suppressing melanoma growth. For this, we tested the scrambled shRNA), followed by Sgk3 KD (70% inhibition) and PDK1 inhibitor (GSK2334470) in combination with 45 known Sgk1 KD (55% inhibition; Fig. 2E). These effects were confirmed inhibitors, including modulators of MAPK and PI3K activity, using independent Sgk1-orSgk3-targeted shRNAs (50% inhi- endoplasmic reticulum (ER) stress, metabolic stress, and of reac- bition of sphere formation over 7 days; Supplementary Fig. S2F, tive oxygen species (Fig. 5A). To ensure coverage of multiple S2H, and S2I). These data demonstrate that SGK3 inhibition subtypes, we tested melanoma lines including BRAF mutant, effectively inhibits colony formation and 3D growth of BRAFV600E HRAS mutant, NRAS mutant, and WT for BRAF, HRAS, and NRAS. PTEN WT and PTEN-null melanomas. Cell growth was analyzed after treatment with 2 or 10 mmol/L of the PDK1 inhibitor over a broad range of doses of the second Pharmacologic inhibition of SGK3 inhibits melanoma cell agent (Supplementary Fig. S4). Determination of synergy was growth made by monitoring inhibition of combination therapy minus We next asked whether pharmacologic inhibition of PDK1 and Bliss additivity of individual agent responses. This assessment SGK would phenocopy the genetic inactivation with shRNA. To revealed that 50% greater effect was identified compared with this end, we treated the BrafV600E::Cdkn2a / ::Pten / and what would have been predicted by additivity in 24 conditions. þ þ BrafV600E::Cdkn2a / ::Pten / cell cultures with either the SGK Among the compounds showing potent synergistic activity inhibitor (SGKi) GSK650394 (21) or the PDK1 inhibitor (PDK1i) with GSK2334470 in suppressing growth were the pan-PI3K GSK2334470 (22) for 24 hours and then examined PDK1-depen- inhibitor BKM120 and the proteasome inhibitors bortezomib dent signaling pathway components. Confirming the expected and carfilzomib (Fig. 5A and Supplementary Fig. S4). In agree- effects, PDK1i reduced the phosphorylation of all of its down- ment with the effects on cell growth, we found that the addition of stream components, whereas SGKi reduced NDRG1 and S6 1 or 4 nmol/L bortezomib further inhibited phosphorylation of phosphorylation but did not affect the upstream components key PDK1 substrates compared with inhibition of PDK1 alone FOXO3a and PRAS40 (Fig. 3A). We next examined the effects of (AKT, FOXO3a, and S6 phosphorylation compared with PDK1 pharmacologic SGKi on sphere formation of YUMM1.5 cells inhibition alone; Fig. 5B) and concomitantly further activated ER incubated with the PDK1i and SGKi. Both inhibitors were effec- stress (ATF4 and CHOP induction) and (cleaved cas- tive and essentially abolished sphere formation at high concen- pase-3 and PARP; Fig. 5B). A similar synergistic effect of the PDK1i

Figure 5. Combination screen of PDK1i against 45 candidate test agents. A, heat map of model-free AUC of GSK2334470 at each of the two concentrations tested with unsupervised clustering of test agents (rows) and cell lines (columns). Red dots mark bortezomib, carfilzomib, and BKM-120 combinations with 10 mmol/L GSK2334470. Dilutions of test agents were combined with either 0.1% DMSO vehicle, 2 mmol/L GSK2334470, or 10 mmol/L GSK2334470 and incubated with cell lines and assayed after 3 days with CellTiterGlo as described in Materials and Methods. Cell lines tested were YUROB, YUHEF (WT BRAF, NRAS), YUMAC, YUSIK (BRAF), YUGASP (NRAS). B, Western blot analysis of the indicated proteins in mouse melanoma cells SBM-A2 and YUMM1.9 and human melanoma cell lines UACC903 and Mel501 treated for 24 hours with 0.1, 1, or 5 mmol/L of GSK2334470 (PDK1i) in the presence or absence of the bortezomib (BTZ) at 1 nmol/L (SBM-A2) or 4 nmol/L (YUMM1.9, UACC903, Mel501). C, growth of YUMM1.5 spheroids treated with the indicated concentrations of GSK2334470 (PDK1i) and bortezomib alone or in combination. Relative areas were calculated as described for Fig. 2. Values are the mean SEM of 6 spheroids per group.

www.aacrjournals.org Cancer Res; 75(7) April 1, 2015 OF9

Downloaded from cancerres.aacrjournals.org on October 5, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst February 24, 2015; DOI: 10.1158/0008-5472.CAN-14-2785

Scortegagna et al.

OF10 Cancer Res; 75(7) April 1, 2015 Cancer Research

Downloaded from cancerres.aacrjournals.org on October 5, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst February 24, 2015; DOI: 10.1158/0008-5472.CAN-14-2785

PDK1 and SGK3 in BRAF-Mutant Melanoma

and bortezomib was observed with the human melanoma cell Stratification of melanoma to PDK1 inhibition lines UACC903 and Mel501, although the degree of caspase-3 To determine whether sensitivity of melanoma to PDK1i is cleavage was not detectable in the UACC903 cells, possibly due to associated with a specific gene signature, we assessed the effects of the higher level of AKT in these cells (Fig. 5B). Notably, the PDK1i on 19 melanoma cell lines for which and synergistic effect on apoptosis of YUMM1.5 cells following co- genomic data are available. The PDK1i 50% inhibitory treatment of bortezomib and PDK1 inhibitors was seen in 3D concentration (IC50) for cell growth and 50% effective concen- cultures only when higher concentrations of bortezomib were tration (EC50) were used to segregate the cell lines (Fig. 7A). The administered (200 nmol/L in 3D cultures, compared with 4 seven most resistant (UACC1940, 2994, 2512, 1120, 1118, 558, nmol/L in the 2D cultures; Fig. 5C). The latter might be attributed and 2641) and the seven most sensitive (UACC903, 2331, 612, to the higher expression of antiapoptotic genes in cells grown in 2496, 647, 952, and 3337) cell lines were then selected for 3D cultures, as previously reported for cells and Ingenuity Pathway Analysis (IPA) to identify the genes and path- mesothelioma cell lines (23, 24). ways most significantly affected by PDK1i (P < 0.05). This analysis The pan-PI3K inhibitor BKM120 (25, 26) was also found to identified 1,178 differentially expressed genes, including multiple synergize with the PDK1i in reducing AKT, FOXO3a, PRAS40, components of the TIF2 nuclear co-regulator and mTOR signaling and GSK3b phosphorylation in both the PTEN WT and mutant pathways that were specifically associated with sensitivity to melanoma cells (Fig. 6A). Notably, the combination of PDK1i. Further refinement of this assessment to include only BKM120 with PDK1i induced cell death in the PTEN WT but genes showing 10-fold differential expression clearly distin- not in the PTEN-mutant melanoma cells, measured by cleaved guished 2 clusters of genes associated with sensitivity and resis- caspase-3 levels (Fig. 6A). Nevertheless, the combination of tance to PDK1i (Fig. 7B). Sensitivity to PDK1i was associated with BKM120 and PDK1i attenuated cell growth in the PTEN- the reduced expression of several pigmentation genes (DCT, mutant melanoma cells, as reflected by the reduced expression PMEL, MelanA) and elevation of number of cytokines and immu- of cyclin D1 (Fig. 6A). These observations indicated that the nomodulators, including IL8, IL1B, IL6, Serpin1, Serpin2, and response of melanoma cells to combined inhibition of PI3K/ PDGF (Fig. 7B). These findings were confirmed by qPCR. PDK1 depends on the degree of PTEN/AKT signaling and PDK1i-treated YUMM1.5 cells showed a reduction in DCT and determines the susceptibility to undergo cytostatic or cytotoxic TYR mRNA and an increase in IL6 mRNA compared with untreat- response. ed cells (Fig. 7C). Consistent with these results, canonical pathway We have further assessed the effect of the PDKi and BKM120 analysis showed that sensitivity of melanoma cells to PDK1i was combination on human melanoma harboring PTEN WT or associated with decreased expression of MITF, the key upstream mutant genotypes. Consistent with the finding in the mouse regulator of pigmentation genes (P ¼ 1.6E-18). Integrin signaling melanoma lines, this combination was effective in inhibiting genes were also strongly associated with PDK1i sensitivity (P ¼ the key AKT/PDK1 signaling in both melanoma lines, albeit 7.1E-4). Analysis of mRNA levels confirmed reduced expression of with greater effectiveness on the PTEN WT–derived cells. Tyr and Dct, 2 pigmentation genes in both Pten WT and mutant Cell death program, monitored by caspase-3 cleavage, was melanoma cells derived from the Pdk1 KD tumors (Supplemen- induced in the Pten WT (SBM-A2 and Mel501) but not Pten- tary Fig. S6). Likewise, lack of Pdk1 in these melanomas also mutant (YUMM1.5 and UACC903) melanomas by the attenuated the expression of epithelial–mesenchymal transition BKM120 and PDK1i inhibitor combination (Fig. 6B). When (EMT)-related genes (28), Zeb1 and Mmp3 (Supplementary assessed in 3D growth, this combination elicited cytostatic Fig. S6). Analysis of 3 melanoma lines revealed slight differences effect, limiting, albeit significantly, the growth of these tumors (i.e., effect on Tyr level in YUMM1.9 and on MMP3 in YUMM1.5 cells (Fig. 6C). cells was limited, compared with changes seen in the other 2 The combined effect of PDK1 and PI3K inhibitors on the cultures), pointing to heterogeneity among the cell types used human melanoma cell lines UACC903 was analyzed by RPPA here. These findings points to a number of genes that could be using a panel of 172 antibodies directed to components of further explored as markers for the stratification of patients with major signaling pathways (19, 27). This analysis revealed that melanoma for therapy by PDK1 inhibition. combined PDK1 and PI3K inhibition elicited an additive effect, which was reflected in the degree of decreased phosphorylation Discussion of key components along the PDK1 pathway, including S6, A number of pathways are known to contribute to melanoma b AKT,p70S6K,aswellastuberin,mTOR,andGSK3 ,pointing development and progression, including the MAPK signaling to effect of this combination also on mTOR and GSK3 path- pathway, which is deregulated in more than 70% of melanomas ways (Supplementary Table S1, Fig. 5A, and Supplementary (NRAS and BRAF ), and the PI3K/AKT signaling path- Fig. S5). way, which is deregulated in more than 50% of these tumors, fi These ndings points for the effectiveness of combined PI3K/ in part due to genetic mutations and in part due to altered PDK1 inhibition, which attenuates the growth of melanoma cells, post translational modifications (29, 30). Using genetic and phar- with a notably greater effect on the PTEN WT cultures. macologic inhibitors, we previously demonstrated the importance

Figure 6. PDK1 and PI3K/mTOR inhibitors synergize to suppress melanoma cell growth. A, Western blot analysis of the indicated proteins in YUMM1.9 and SBM-A2 cells treated for 24 hours with 0.1, 1, or 5 mmol/L of GSK2334470 (PDK1i) in the presence or absence of 1 or 3 mmol/L of the dual PI3K/mTOR inhibitor BKM120. B, Western blot analysis of the indicated proteins in the human melanoma cell lines UACC903 and Mel501 treated with 0.1, 1, or 5 mmol/L of GSK2334470 (PDK1i) in thepresenceorabsenceof1or3mmol/L of BKM120. C, growth of YUMM1.5 spheroids treated with the indicated concentrations of GSK2334470 (PDK1i) and BKM120 alone or in combination. Relative areas were calculated as described for Fig. 2. Values are the mean SEM of 6 spheroids per group.

www.aacrjournals.org Cancer Res; 75(7) April 1, 2015 OF11

Downloaded from cancerres.aacrjournals.org on October 5, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst February 24, 2015; DOI: 10.1158/0008-5472.CAN-14-2785

Scortegagna et al.

Figure 7. Stratification of human melanomas by sensitivity to PDK1 inhibition. A, the mean IC50 for GSK2334470 inhibition of growth of 19 human melanoma cell lines was used to segregate cell lines into sensitivity or resistance to the inhibitor. B, heat map of the results of the IPA showing the genes/pathways most significantly altered (P < 0.05; fold change > 1.5) by GSK2334470 (1,178 differentially expressed genes). C, qPCR analysis of Dct, Tyr,andIl6 mRNA in YUMM1.5 cells treated with DMSO or GSK2334470 at 5 mmol/L for 24 hours (Dct and Tyr)orat10mmol/L for 6 hours (Il6). Values are the mean SEM of biologic triplicates. , P < 0.05; , P < 0.01.

of the master AGC kinase—PDK1—for the development and human melanomas. Significantly, we found that PDK1 actually metastasis of BrafV600E Pten / melanomas (5). Here, we extend plays a more pronounced role in PTEN WT than PTEN-mutant those findings to show that PDK1 also plays an important role in genotypes, a finding that is expected because of the preserved melanomas harboring WT PTEN, which includes about 70% of PTEN activity, which harnesses AKT and related PI3K

OF12 Cancer Res; 75(7) April 1, 2015 Cancer Research

Downloaded from cancerres.aacrjournals.org on October 5, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst February 24, 2015; DOI: 10.1158/0008-5472.CAN-14-2785

PDK1 and SGK3 in BRAF-Mutant Melanoma

components. Together, our findings further our appreciation knowledge gained from mechanistic and clinical studies. As a for the impact of PDK1 to the AGC kinase landscape. result, future clinical management options will be more extensive The significance of our observations is highlighted in syn- and are likely to include modulators of pathways that are inde- thetic lethal screens by the identification of pan-PI3K inhibitor pendent of the MAPK signaling axis. Our data substantiate the that effectively synergize with PDK1i, particularly in PTEN importance of PDK1 and one of its downstream substrates, SGK3, WT melanomas, suggesting that the focused targeting of the for melanoma development and progression and further suggest PI3K–PDK1 signaling axis might represent a novel therapeutic that inhibitors of this signaling pathway might be useful for the modality for PTEN WT melanomas. A similar approach with development of clinical management options for melanoma. combination MEKi and BRAFi has produced promising results in ongoing clinical trials (31). Notably, our data point to a gene Disclosure of Potential Conflicts of Interest signature that distinguishes between PDK1i-sensitive melano- M.A. Davies reports receiving commercial research grants from Glaxo- mas (which express low levels of pigmentation genes associated SmithKline, AstraZeneca, Merck, , Myriad, Sanofi-Aventis, and with MITF signaling) and PDK1i-resistant melanomas (which Oncothyreon and is a consultant/advisory board member for GlaxoSmithKline, express inflammation-related genes, including IL6 and IL3); Novartis, Genentech, and Sanofi-Aventis. No potential conflicts of interests this signature could potentially be used for the stratification of were disclosed by the other authors. patients for PDK1-targeted therapies. Given the recent finding that low MITF expression correlates with resistance of melano- Authors' Contributions mas to BRAFi (32), it would be of interest to further examine Conception and design: M. Scortegagna, H. Yin, D.F. Stern, M. Bosenberg, the possibility that targeting the PDK1 pathway might also be Z.A. Ronai Development of methodology: M. Scortegagna, C. Sereduk, H. Yin, S.K. De, efficient for inhibiting the resistance phenotype. Notably, fi D.F. Stern, M. Bosenberg PDK1-de cient melanomas also exhibited reduced ZEB1 and Acquisition of data (provided animals, acquired and managed patients, MMP3 expression, pointing to PDK1 role in control of the EMT, provided facilities, etc.): M. Scortegagna, E. Lau, C. Sereduk, C.G. Langdon, explaining the reduced metastasis observed in vivo and atten- R. Halaban, M. Pellecchia, M. Bosenberg uated growth in 3D in culture. Of interest, our results reveal that Analysis and interpretation of data (e.g., statistical analysis, biostatistics, inhibition of either PDK1 or SGK3 decreased the phosphory- computational analysis): M. Scortegagna, E. Lau, T. Zhang, C. Sereduk, H. Yin, J.T. Platt, M.A. Davies, K. Brown, D.F. Stern, M. Bosenberg, Z.A. Ronai lation of 4-EBP1, pointing to possible role of these AGC kinases Writing, review, and/or revision of the manuscript: M. Scortegagna, E. Lau, in the regulation of CAP-dependent translation. Consistent T. Zhang, H. Yin, M.A. Davies, D.F. Stern, M. Bosenberg, Z.A. Ronai with these observations, eIF4F, another component of this Administrative, technical, or material support (i.e., reporting or organizing translation initiation complex, was recently linked with the data, constructing databases): M. Scortegagna, T. Zhang, K. Meeth, R. Halaban, resistance to anti-BRAF and anti-MEK therapies in BRAF-mutant Z.A. Ronai melanoma, colon, and thyroid cancer cells (33). Study supervision: M. Scortegagna, Z.A. Ronai Other (synthesis of the compound): S.K. De PDK1 inhibitors that are suitable for use in clinical trials have not yet been developed. Although the PDK1i used in our study (GSK2334470) is quite specific and exhibits excellent properties Acknowledgments for work with cultured cells (22, 34), it is unsuitable for further The authors thank Yale SPORE in Skin Cancer and TGEN for cell lines used in the present study. They also thank Dario Alessi for providing the conditional preclinical or clinical development. Other PDK1 inhibitors are Pdk1 / mice and Vince Hearing for the kind gift of the Tyrp1 antibodies and currently being developed, which might exhibit acceptable safety members of the Ronai laboratory for extensive discussions. profiles and be eligible for further advanced clinical evaluation. An alternative approach to inhibiting PDK1 is to identify and Grant Support target one or more downstream PDK1 substrates that are crucial Z.A. Ronai gratefully acknowledges support from the NCI (CA179170 and for melanoma development and progression. In this regard, we CA128814), the Hervey Family Non-endowment Fund at The San Diego report here that the genetic and pharmacologic inhibition of Foundation, and the Melanoma Research Foundation. SGK3 largely phenocopies the effects of PDK1 inhibition on The costs of publication of this article were defrayed in part by the payment of melanoma cells, including growth arrest in G1 phase. Our findings page charges. This article must therefore be hereby marked advertisement in are consistent with earlier reports that pointed to the role of SGK3 accordance with 18 U.S.C. Section 1734 solely to indicate this fact. in melanoma (8). The repertoire of potential targets for melanoma therapy is Received September 23, 2014; revised December 15, 2014; accepted January expanding considerably, thanks in large part to the extensive 20, 2015; published OnlineFirst February 24, 2015.

References 1. Hodis E, Watson IR, Kryukov GV, Arold ST, Imielinski M, Theurillat JP, et al. and inhibits metastasis of Braf(V600E)::Pten(/) melanoma. A landscape of driver mutations in melanoma. Cell 2012;150:251–63. 2014;33:4330–9. 2. Mora A, Komander D, van Aalten DM, Alessi DR. PDK1, the master 6. Davies MA, Stemke-Hale K, Lin E, Tellez C, Deng W, Gopal YN, et al. regulator of AGC kinase signal transduction. Semin Cell Dev Biol 2004;15: Integrated molecular and clinical analysis of AKT activation in metastatic 161–70. melanoma. Clin Cancer Res 2009;15:7538–46. 3. Pearce LR, Komander D, Alessi DR. The nuts and bolts of AGC protein 7. Zhou XP, Gimm O, Hampel H, Niemann T, Walker MJ, Eng C. Epigenetic kinases. Nat Rev Mol Cell Biol 2010;11:9–22. PTEN silencing in malignant melanomas without PTEN mutation. Am J 4. Rintelen F, Stocker H, Thomas G, Hafen E. PDK1 regulates growth through Pathol 2000;157:1123–8. Akt and S6K in Drosophila. Proc Natl Acad Sci U S A 2001;98:15020–5. 8. Vasudevan KM, Barbie DA, Davies MA, Rabinovsky R, McNear CJ, Kim JJ, 5. Scortegagna M, Ruller C, Feng Y, Lazova R, Kluger H, Li JL, et al. Genetic et al. AKT-independent signaling downstream of oncogenic PIK3CA muta- inactivation or pharmacological inhibition of Pdk1 delays development tions in human cancer. Cancer Cell 2009;16:21–32.

www.aacrjournals.org Cancer Res; 75(7) April 1, 2015 OF13

Downloaded from cancerres.aacrjournals.org on October 5, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst February 24, 2015; DOI: 10.1158/0008-5472.CAN-14-2785

Scortegagna et al.

9. Sato S, Fujita N, Tsuruo T. Involvement of 3-phosphoinositide-dependent 22. Najafov A, Sommer EM, Axten JM, Deyoung MP, Alessi DR. Characteri- -1 in the MEK/MAPK signal transduction pathway. J Biol zation of GSK2334470, a novel and highly specific inhibitor of PDK1. Chem 2004;279:33759–67. Biochem J 2011;433:357–69. 10. Zeng X, Xu H, Glazer RI. Transformation of mammary epithelial cells by 3- 23. Yang TM, Barbone D, Fennell DA, Broaddus VC. Bcl-2 family proteins phosphoinositide-dependent protein kinase-1 (PDK1) is associated with contribute to apoptotic resistance in lung cancer multicellular spheroids. the induction of protein kinase Calpha. Cancer Res 2002;62:3538–43. Am J Respir Cell Mol Biol 2009;41:14–23. 11. Gagliardi PA, di Blasio L, Orso F, Seano G, Sessa R, Taverna D, et al. 3- 24. Barbone D, Ryan JA, Kolhatkar N, Chacko AD, Jablons DM, Sugarbaker DJ, phosphoinositide-dependent kinase 1 controls breast tumor growth in a et al. The Bcl-2 repertoire of mesothelioma spheroids underlies acquired kinase-dependent but Akt-independent manner. Neoplasia 2012;14: apoptotic multicellular resistance. Cell Death Dis 2011;2:e174. 719–31. 25. Burger MT, Pecchi S, Wagman A, Ni ZJ, Knapp M, Hendrickson T, et al. 12. Bruhn MA, Pearson RB, Hannan RD, Sheppard KE. Second AKT: the rise of Identification of NVP-BKM120 as a potent, selective, orally bioavailable SGK in cancer signalling. Growth Factors 2010;28:394–408. class I PI3 kinase inhibitor for treating cancer. ACS Med Chem Lett 2011; 13. Tessier M, Woodgett JR. Serum and glucocorticoid-regulated protein 2:774–9. kinases: variations on a theme. J Cell Biochem 2006;98:1391–407. 26. Aziz SA, Jilaveanu LB, Zito C, Camp RL, Rimm DL, Conrad P, et al. Vertical 14. Xu J, Wan M, He Q, Bassett RL Jr, Fu X, Chen AC, et al. SGK3 is associated targeting of the phosphatidylinositol-3 kinase pathway as a strategy for with estrogen receptor expression in breast cancer. Breast Cancer Res Treat treating melanoma. Clin Cancer Res 2010;16:6029–39. 2012;134:531–41. 27. Park ES, Rabinovsky R, Carey M, Hennessy BT, Agarwal R, Liu W, et al. 15. Sommer EM, Dry H, Cross D, Guichard S, Davies BR, Alessi DR. Elevated Integrative analysis of proteomic signatures, mutations, and drug respon- SGK1 predicts resistance of breast cancer cells to Akt inhibitors. Biochem J siveness in the NCI 60 cancer cell line set. Mol Cancer Ther 2010;9:257–67. 2013;452:499–508. 28. Caramel J, Papadogeorgakis E, Hill L, Browne GJ, Richard G, Wierinckx A, 16. Tworkoski K, Singhal G, Szpakowski S, Zito CI, Bacchiocchi A, Muthusamy et al. A switch in the expression of embryonic EMT-inducers drives the V, et al. Phosphoproteomic screen identifies potential therapeutic targets in development of malignant melanoma. Cancer Cell 2013;24:466–80. melanoma. Mol Cancer Res 2011;9:801–12. 29. Parsons R. Human cancer, PTEN and the PI-3 kinase pathway. Semin Cell 17. Dankort D, Curley DP, Cartlidge RA, Nelson B, Karnezis AN, Damsky WE Jr, Dev Biol 2004;15:171–6. et al. Braf(V600E) cooperates with Pten loss to induce metastatic mela- 30. Correia NC, Girio A, Antunes I, Martins LR, Barata JT. The multiple layers of noma. Nat Genet 2009;41:544–52. non-genetic regulation of PTEN tumour suppressor activity. Eur J Cancer 18. Lawlor MA, Mora A, Ashby PR, Williams MR, Murray-Tait V, Malone L, et al. 2014;50:216–25. Essential role of PDK1 in regulating cell size and development in mice. 31. Abdel-Wahab O, Klimek VM, Gaskell AA, Viale A, Cheng D, Kim E, et al. EMBO J 2002;21:3728–38. Ef ficacy of intermittent combined RAF and MEK inhibition in a patient 19. Tibes R, Qiu Y, Lu Y, Hennessy B, Andreeff M, Mills GB, et al. Reverse phase with concurrent BRAF- and NRAS-mutant malignancies. Cancer Discov protein array: validation of a novel proteomic technology and utility for 2014;4:538–45. analysis of primary leukemia specimens and hematopoietic stem cells. Mol 32. Van Allen EM, Mouw KW, Kim P, Iyer G, Wagle N, Al-Ahmadie H, et al. Cancer Ther 2006;5:2512–21. Somatic ERCC2 mutations correlate with cisplatin sensitivity in muscle- 20. Wang Y, Zhou D, Chen S. SGK3 is an androgen-inducible kinase promoting invasive urothelial carcinoma. Cancer Discov 2014;4:1140–53. prostate cancer cell proliferation through activation of p70 S6 kinase and 33. Boussemart L, Malka-Mahieu H, Girault I, Allard D, Hemmingsson O, up-regulation of cyclin D1. Mol Endocrinol 2014;28:935–48. Tomasic G, et al. eIF4F is a nexus of resistance to anti-BRAF and anti-MEK 21. Sherk AB, Frigo DE, Schnackenberg CG, Bray JD, Laping NJ, Trizna W, et al. cancer therapies. Nature 2014;513:105–9. Development of a small-molecule serum- and glucocorticoid-regulated 34. Najafov A, Shpiro N, Alessi DR. Akt is efficiently activated by PIF-pocket- kinase-1 antagonist and its evaluation as a prostate cancer therapeutic. and PtdIns(3,4,5)P3-dependent mechanisms leading to resistance to Cancer Res 2008;68:7475–83. PDK1 inhibitors. Biochem J 2012;448:285–95.

OF14 Cancer Res; 75(7) April 1, 2015 Cancer Research

Downloaded from cancerres.aacrjournals.org on October 5, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst February 24, 2015; DOI: 10.1158/0008-5472.CAN-14-2785

PDK1 and SGK3 Contribute to the Growth of BRAF-Mutant Melanomas and Are Potential Therapeutic Targets

Marzia Scortegagna, Eric Lau, Tongwu Zhang, et al.

Cancer Res Published OnlineFirst February 24, 2015.

Updated version Access the most recent version of this article at: doi:10.1158/0008-5472.CAN-14-2785

Supplementary Access the most recent supplemental material at: Material http://cancerres.aacrjournals.org/content/suppl/2015/02/25/0008-5472.CAN-14-2785.DC1

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 Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://cancerres.aacrjournals.org/content/early/2015/03/20/0008-5472.CAN-14-2785. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from cancerres.aacrjournals.org on October 5, 2021. © 2015 American Association for Cancer Research.