Purpose:

In neurofibromatosis type 1 (NF1) and in highly aggressive malignant peripheral nerve sheath tumors (MPNSTs), constitutively active RAS-GTP and increased MAPK signaling are important in tumorigenesis. Dual specificity phosphatases (DUSPs) are negative regulators of MAPK signaling that dephosphorylate p38, JNK, and ERK in different settings. Although often acting as tumor suppressors, DUSPs may also act as oncogenes, helping tumor cells adapt to high levels of MAPK signaling. We hypothesized that inhibiting DUSPs might be selectively toxic to cells from NF1-driven tumors.

Experimental Design:

We examined DUSP gene and protein expression in neurofibroma and MPNSTs. We used small hairpin RNA (shRNA) to knock down DUSP1 and DUSP6 to evaluate cell growth, downstream MAPK signaling, and mechanisms of action. We evaluated the DUSP inhibitor, (E)-2-benzylidene-3-(cyclohexylamino)-2,3-dihydro-1H-inden-1-one (BCI), in MPNST cell lines and in cell-line and patient-derived MPNST xenografts.

Results:

DUSP1 and DUSP6 are expressed in NF1-deleted tumors. Knockdown of DUSP1 and DUSP6, alone or in combination, reduced MPNST cell growth and led to ERK and JNK hyperactivation increasing downstream TP53 and p-ATM. The DUSP inhibitor, BCI, diminished the survival of NF1-deleted Schwann cells and MPNST cell lines through activation of JNK. In vivo, treatment of an established cell-line xenograft or a novel patient-derived xenograft (PDX) of MPNSTs with BCI increased ERK and JNK activation, caused tumor necrosis and fibrosis, and reduced tumor volume in one model.

Conclusions:

Targeting DUSP1 and DUSP6 genetically or with BCI effectively inhibits MPNST cell growth and promotes cell death, in vitro and in xenograft models. The data support further investigation of DUSP inhibition in MPNSTs.

Translational Relevance

Malignant peripheral nerve sheath tumors (MPNSTs) are devastating, therapy-resistant tumors. Preclinical inhibition of MAPK signaling suppresses tumor growth, but the effects are transient. DUSPs are upregulated in response to elevated MAPK signaling in cancer cells with NF1 deletion, restraining signaling, and helping cells to adapt to oncogenic signals. Here, we demonstrate that neurofibroma and MPNSTs express DUSP1 and DUSP6, and that genetic blockade of these targets inhibits the growth of MPNST cells in vitro. This occurs largely through activation of JNK, which affects p53 and RB, triggering growth arrest and/or cell death. Furthermore, in mice with orthotopic human MPNST xenografts, the DUSP1/6 inhibitor, (E)-2-benzylidene-3-(cyclohexylamino)-2,3-dihydro-1H-inden-1-one (BCI), induces cell death and increases fibrosis in 2 xenograft models, resulting in decreased tumor volume in one model. Our preclinical work supports the development of DUSP1/6 inhibitors for MPNST therapy.

Malignant peripheral nerve sheath tumors (MPNSTs) are devastating chemotherapy- and radiation-resistant soft tissue sarcomas. Rare in the general population, half of MPNSTs arise in patients with the autosomal dominant genetic disorder called neurofibromatosis type 1 (NF1). MPNSTs in patients with NF1 frequently occur in the context of preexisting benign plexiform neurofibroma, and are the leading cause of death in adults with NF1 (1–3). NF1 loss results in activation of RAS proteins that stimulates the MAPK family including ERK1 and ERK2, the JNKs 1, 2, and 3, and the p38 family of kinases (α, β, δ, and γ) resulting in tumor cell growth. However, from gene expression microarray data, we and others have observed that key negative regulators of MAPK signaling are also increased in NF1-deleted tumors (4, 5). Dual specificity phosphatase (DUSP) expression is transcriptionally upregulated by increased ERK activity, providing a negative feedback loop to suppress RAS signaling (6). Within the DUSP family of proteins, the MAP kinase phosphatases (MKP) dephosphorylate tyrosine and serine/threonine residues on ERK, JNK, and p38 (7). DUSPs are subdivided on the basis of cellular localization and substrate specificity. There are over 10 members of this family including DUSP1/MKP-1, which is a nuclear MKP, and DUSP6/MKP-3, which is cytosolic (8). DUSPs substrate specificities depend on the cellular context (8).

Studies have described MAPK negative regulators, including DUSP6, as tumor suppressors. Thus, decreased expression of DUSP genes and proteins can occur in human cancer, including in tumors from pancreas and lung (9, 10). In contrast, DUSPs are upregulated in some leukemia types including pre-B acute lymphoblastic leukemia (ALL) and DUSP1/6 inhibition has antitumor effects in gastric, breast, and leukemia cancer models (11–14). In these cases, it is believed that DUSPs help tumors adapt to excessively high levels of growth factor signals. DUSP inhibition can acutely hyperactivate MAPK signaling, triggering activation of effectors such as p53, ATM, and CHK1/2, which arrest cell growth and/or induce cell death. If NF1-deleted cells with RAS activation depend on DUSPs for growth and survival, then inhibiting DUSP could be effective in NF1 and in the many sporadic tumor types now known to show NF1 loss (15).

Despite advances in understanding MPNST biology, there are no effective available MPNST treatments (2). Some potential targets are growth factor receptors, and the PI3K-AKT-mTOR and Wnt/β-catenin signaling pathways (16, 17). Targeting MEK activity provides growth suppression in certain MPNST models, but effects, when present, are transient (5, 18). Aurora kinase inhibition is cytostatic (19) and with combination therapy may show additive effects (20).

Targeting DUSP is an active area of preclinical development. (E)-2-benzylidene-3-(cyclohexylamino)-2,3-dihydro-1H-inden-1-one (BCI) is a specific allosteric inhibitor of DUSP1/6 (12, 13, 21). Recently, in mouse models of leukemia and gastric cancer, BCI was effective in killing tumor cells in vivo (12–14, 21). We hypothesized that inhibiting DUSPs might be selectively toxic to MPNST cells. In this study, we conducted molecular analyses to better understand the role of DUSP1 and DUSP6 in MPNSTs, using a combination of MPNST cell lines, in vivo cell line xenografts and, a novel patient-derived xenograft (PDX) MPNST model. The results from our studies demonstrate that DUSP1/6 protein expression are important for growth and survival in NF1-deleted MPNSTs and suggest that targeting DUSPs may be a therapeutic option for treating MPNSTs.

Cell lines and drugs

Cell lines included immortalized human Schwann cells (iHSC; ref. 22) with creation and characterization of NF1-deficient HSC1λ cell lines (Williams and colleagues, unpublished). Briefly an early, common, exon of NF1 (exon 10) was targeted to generate indel mutations using CRISPR/Cas9. Those found to be harboring homozygous frameshift mutations resulting in an early termination codon were considered NF1/. Cell lines showing homozygous wild-type (WT) sequences were considered isogenic WT controls. MPNST cell lines, ST8814, S462.TY, STS26T, and 90-8, were obtained as described previously (23, 24). All MPNST cell lines were derived from patients with NF1, except STS26T. Cell lines were maintained in DMEM and 10% FBS and were confirmed negative for Mycoplasma prior to use. Verification of cell lines was performed by short tandem repeat DNA profiling (Genetica DNA Laboratories). Inhibitors used were BCI (Axon Medchem), PD0325901 MEK inhibitor (gift from Kevin M. Shannon, University of California, San Francisco, CA), and JNK inhibitors, JNK-IN-8 (Selleckchem) and SP600125 (LC Laboratories). Drugs were prepared as 10 mmol/L stocks in DMSO and were diluted into media for cell culture treatments. Cell viability was quantified 72 hours after treatment by CellTiter 96 AQueous One Solution Cell Proliferation Assay (MTS; Promega) as per manufacturer's instructions. Combination index data analysis was done using Calcusyn software (Version 1). Combination indices (CI) less than 0.9 indicate synergy, 0.9 to 1.1 indicate additivity, and greater than 1.1 indicate antagonism.

In vitro signaling experiments and TUNEL assays

Cells were serum starved for 16 hours in DMEM media (Life Technologies). MPNST cells were pretreated with inhibitor for 15 to 60 minutes then stimulated with DMEM and 10% FBS (Gemini Bio Products) for 15 to 60 minutes. The iHSCs were stimulated with DMEM-F12 with N2 supplement (Life Technologies) and 10 ng/mL β-Heregulin (β-HRG). Signaling experiments were terminated with a cold PBS wash, and cell lysates were generated for Western blotting. Cell death was analyzed with the In Situ Cell Death Detection Kit (terminal deoxynucleotidyl transferase–mediated dUTP nick end labeling, TUNEL) and TMR red (Roche), by fluorescence microscopy.

Western blotting

Tissues and cells were lysed in a RIPA buffer supplemented with protease and phosphatase inhibitors (Roche). Protein concentrations were determined with the BCA Protein Assay Reagent (Thermo Fisher Scientific). Antibodies used were Dusp1 (Millipore, 07-535), DUSP6 (Abcam, 76310), p38 (Abcam, 7925), p-p38 (Life Technologies, 44684), p53 (Santa Cruz Biotechnology, sc-47698). Antibodies from Cell Signaling Technology were p-DUSP-1 (2857), ERK, p-ERK (4695, 4370), JNK, p-JNK (9252, 4668), c-jun, p-c-jun (2315, 2361), p-p38 (9211), PARP (9542), cyclin D1 (2922), p-ATM (13050), p-RB (retinoblastoma; 9301), and B-actin (5125). Blots were analyzed by densitometry using ImageJ software.

Knockdown experiments

For lentiviral small hairpin RNA (shRNA) infection, MPNST cells at 70% confluence were infected with lentiviral particles containing shRNAs targeting DUSP1 (TRCN0000002516, TRCN0000002517) or DUSP6 (TRCN0000002436, TRCN0000002437) at a multiplicity of infection (MOI) of 5. These were purchased from the Lenti-shRNA Library Core [Cincinnati Children's Hospital Medical Center (CCHMC), Cincinnati, OH; originally from the Sigma Mission Consortium shRNA library]. pLKO.1-puro control was used as the nontargeting (NT) control. Cell lines were infected overnight in the presence of 10 μg/mL polybrene. Stable cell populations were selected with puromycin (2 μg/mL) 48 hours later.

Quantitative PCR

Total RNA was isolated from cells using the RNeasy Kit (Qiagen). The cDNA was made using the High-Capacity cDNA Reverse Transcription Kit (Life Technologies). Real-time PCR was performed on cDNA using SYBR Green (Applied Biosystems). Relative quantification was performed using the ΔΔCt method, n = 6.

Histologic staining

Formalin-fixed, paraffin-embedded tissue blocks were sectioned at 5 μm. Tissue sections were deparaffinized in xylene and rehydrated in ethanol, then water. Sections were stained with hematoxylin and eosin (H&E) or Masson trichrome, in which blue stain indicates collagen. For IHC, sections were subjected to heat-induced antigen retrieval in citrate buffer. Sections were incubated with primary antibody overnight at 4°C and biotinylated secondary antibodies were detected with horseradish peroxidase–conjugated Streptavidin (Elite ABC, Vector Laboratories) treated with DAB substrate (Vector Laboratories). Sections were counterstained with hematoxylin. The primary antibodies used for IHC include Anti-Human Nucleoli Antibody (Abcam, ab190710) and Cleaved Caspase 3 (CC3; Cell signaling Technology No. 9661).

Copy-number alteration data analysis

Copy-number alteration (CNA) data on 51 primary MPNSTs (NF1-associated and sporadic) was from a published GSE3388 dataset [Agilent Human Genome CGH Microarray Kit (4 × 44K); ref. 25]. A circular binary segmentation (CBS) algorithm was applied to the log2 ratios of intensity values from tumor and normal to reduce local noise effects. CBS calculates a likelihood-ratio statistic for each array probe by permutation to locate change points (26). After initial segmentation, smoothing, and postsegmentation normalization processes, CGHcall algorithm (27) was applied to assign each segment an aberration label. We chose the 5-state (amplification, gain, normal, loss, and double loss) prediction design and the probability of each state was calculated, then one state with the highest probability was chosen as a final CNA calling label for each segment.

Affymetrix GeneChip HU133 Plus 2.0 array for patient samples

We reprocessed the published data [Gene Expression Omnibus (GEO) accession #:GSE14038, Affymetrix GeneChip HU133 Plus 2.0] using Bioconductor's affy package (28). Differentially expressed genes between 2 groups were predicted using an adjusted t test after multiple testing (Benjamini–Hochberg) and Bioconductor's limma package (29). ANOVA test was performed using R (v3.3.1).

Mouse and xenograft tumor models

All animal studies were performed according to the guidelines of the Institutional Animal Care and Use Committee of CCHMC. Mouse neurofibroma was obtained from Nf1 fl/fl;Dhh-Cre mice as described previously (30). For the cell line–based xenograft model, S462.TY cells (1.25 × 106) in 30% Matrigel (BD Biosciences) were injected subcutaneously into the flank of 5- to 6-week-old female athymic nude mice. For the PDX MPNST model, biopsied tumor tissue was implanted subcutaneously directly into NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ (NSG) mice. Tumors used for the study were grown in a donor mouse to a volume of approximately 2,500 mm3. The tumor was harvested, cut into 1.5-mm3 pieces, and inserted subcutaneously into the flank of each mouse. Mice bearing F3–F4 grafts were used in this study. Drug dosing was started when tumors reached 150 to 200 mm3. We measured tumors and weighed mice every 3 days. Tumor volume was calculated as follows: L × W2 (π/6), where L is the longest diameter and W is the width. The length of treatment was determined by the time tumors in each vehicle-treated group reached a maximum allowable tumor volume of 2,500 mm3 (21 days for the PDX and 27 days for the S462.TY model).

Statistical analysis

Analyses were performed in GraphPad Prism software version 7.0. Data were mean ± SD or SEM as indicated and were analyzed using ANOVA with Tukey post hoc test or a Student t test, 2-sided. P values <0.05 were considered significant.

Expression of DUSP1 and DUSP6 in neurofibroma and MPNSTs

We used published gene expression profiles to analyze the differential expression of DUSP genes (5). DUSP1 and DUSP6 mRNAs were consistently elevated in neurofibroma and MPNSTs as compared with human Schwann cells, which is consistent with their transcriptional upregulation by mutant NF1 (Fig. 1A). In a genetically engineered mouse model of neurofibroma, we examined DUSP protein levels by immunoblot analysis. DUSP1 was 4× higher in neurofibroma compared with Nf1+/+ (WT) sciatic nerves (Fig. 1B). Anti–p-DUSP1 detects a phosphorylation site known to stabilize the protein (31); p-DUSP1 expression was elevated 7× (Fig. 1B). DUSP6 protein was 13× higher in neurofibroma compared with Nf1+/+ sciatic nerves (Fig. 1B). Compared with WT nerve, neurofibromas consistently showed elevated p-ERK, p-c-jun and decreased p-JNK, with variable levels of p-p38 (Supplementary Fig. S1A). In iHSCs lacking NF1, DUSP1 was 62% higher, p-DUSP1 2× and DUSP6 3× higher than isogenic WT iHSC (Fig. 1C), consistent with the idea that in Schwann cells, NF1 deletion can drive increases in DUSP protein expression. DUSP protein was also elevated in human MPNST cell lines (n = 4), showing an average 30% increase in DUSP1 and an average 3× higher expression of p-DUSP1 compared with iHSC (Fig. 1C). DUSP6 protein was 4 to 15× higher in MPNSTs compared with iHSC, with the highest expression in the NF1-deleted MPNST cell line, ST8814 (Fig. 1C). Compared with iHSC, MPNST cell lines have increased p-ERK with variable levels of p-JNK and p-p38 (Supplementary Fig. S1B).

Figure 1.

Expression of DUSP1 and DUSP6 in neurofibroma and MPNSTs. A, Gene expression microarray showing DUSP transcripts with increased expression in plexiform neurofibroma tumors and MPNST solid tumors versus normal human Schwann cells. The scale bar on right shows mean log2-transformed normalized fold change. B, Immunoblot shows elevated DUSP1, p-DUSP1, and DUSP6 protein in Dhh Cre; Nf1 fl/fl mouse neurofibroma versus normal age-matched WT sciatic nerve (WT nerve). C, Immunoblot shows elevated DUSP1, p-DUSP1 and DUSP6 protein in NF1/ iHSC and MPNST cell lines versus NF1 WT iHSC. β-actin was used as a loading control. D, CNA data analysis of plexiform neurofibroma (pNF), NF1-associated MPNSTs, and sporadic MPNSTs.

Figure 1.

Expression of DUSP1 and DUSP6 in neurofibroma and MPNSTs. A, Gene expression microarray showing DUSP transcripts with increased expression in plexiform neurofibroma tumors and MPNST solid tumors versus normal human Schwann cells. The scale bar on right shows mean log2-transformed normalized fold change. B, Immunoblot shows elevated DUSP1, p-DUSP1, and DUSP6 protein in Dhh Cre; Nf1 fl/fl mouse neurofibroma versus normal age-matched WT sciatic nerve (WT nerve). C, Immunoblot shows elevated DUSP1, p-DUSP1 and DUSP6 protein in NF1/ iHSC and MPNST cell lines versus NF1 WT iHSC. β-actin was used as a loading control. D, CNA data analysis of plexiform neurofibroma (pNF), NF1-associated MPNSTs, and sporadic MPNSTs.

Close modal

DUSP1 and DUSP6 are on human chromosomes 5q35.1 and 12q21.33, respectively. Previous studies of MPNSTs have identified chromosomal gains in these regions (25, 32). We reasoned that during the transformation process, some MPNSTs may genetically alter DUSPs to adapt to increased growth factor signaling. Amplifications of HGF, MET, and PDGFRA genes are present in some MPNSTs, suggesting a role in NF1 tumor progression (33). We analyzed data from human plexiform neurofibroma and primary MPNSTs (NF1-associated and sporadic) to evaluate the genomic status of DUSP1 and DUSP6 (25). We found no copy-number changes in plexiform neurofibroma (Fig. 1D). In NF1-associated MPNSTs, 31.25% of the tumors had copy-number gains in DUSP1 or DUSP6, with 12% having gains in both (Fig. 1D). In sporadic MPNSTs, 34% had gains in DUSP1 or DUSP6 with 5.7% having amplifications of DUSP6 (Fig. 1D). Single-copy losses of DUSP1 or DUSP6 were detected in 25% of NF1-associated MPNSTs with DUSP1 or DUSP6 loss at 8.6% and 5.7% in sporadic MPNSTs (Fig. 1D). Therefore, copy-number changes are not likely to be a major contributor to DUSP1/6 expression in most MPNSTs.

DUSP1 and DUSP6 knockdown decrease MPNST cell growth

We used shRNA to knock down DUSP1 or DUSP6 in the MPNST cell line ST8814. Using 2 specific shRNAs and NT controls, we validated DUSP1/6 knockdown of mRNA and a 55% to 75% decrease in protein (Fig. 2A–D). When DUSP1 was knocked down, levels of DUSP6 did not change and vice versa (Fig. 2C and D). Proliferation was decreased by 28% to 43% on DUSP1 knockdown and 40% to 51% on DUSP6 knockdown (Fig. 2E and F). DUSP1 knockdown cells treated with DUSP6 shRNA showed a further 30% decrease in proliferation (Fig. 2E). DUSP6 knockdown cells treated with DUSP1 shRNA showed a further 20% decrease in proliferation (Fig. 2F). Thus, directly decreasing DUSP1/6 levels, alone or in combination, decreases proliferation of MPNST cells.

Figure 2.

DUSP1 and DUSP6 knockdown decrease cell growth in the NF1/ MPNST cell line, ST8814. A and B, mRNA expression levels in the MPNST cell line, ST8814, after lentiviral shRNA knockdown of DUSP1 or DUSP6. Levels are expressed as normalized to NT shRNA controls. *, P < 0.05; **, P < 0.01 compared with NT control. C, Immunoblots show that DUSP1 protein expression, but not DUSP6, is reduced by day 4 post transduction with 2 DUSP1 shRNAs D, DUSP6 protein expression, but not DUSP1, is reduced by day 4 post transduction with 2 DUSP6 shRNAs. E, shDUSP1 reduces cell proliferation alone and in combination with sh2-DUSP6. F, shDUSP6 reduces cell proliferation alone and in combination with sh2-DUSP1. (*) and (**) represent P < 0.05 and P < 0.01 compared with NT control. Experiments were done twice with n = 6 per experiment.

Figure 2.

DUSP1 and DUSP6 knockdown decrease cell growth in the NF1/ MPNST cell line, ST8814. A and B, mRNA expression levels in the MPNST cell line, ST8814, after lentiviral shRNA knockdown of DUSP1 or DUSP6. Levels are expressed as normalized to NT shRNA controls. *, P < 0.05; **, P < 0.01 compared with NT control. C, Immunoblots show that DUSP1 protein expression, but not DUSP6, is reduced by day 4 post transduction with 2 DUSP1 shRNAs D, DUSP6 protein expression, but not DUSP1, is reduced by day 4 post transduction with 2 DUSP6 shRNAs. E, shDUSP1 reduces cell proliferation alone and in combination with sh2-DUSP6. F, shDUSP6 reduces cell proliferation alone and in combination with sh2-DUSP1. (*) and (**) represent P < 0.05 and P < 0.01 compared with NT control. Experiments were done twice with n = 6 per experiment.

Close modal

DUSP1 and DUSP6 knockdown activates ERK and JNK and regulates cell growth and death

We next tested the consequences of knockdown of DUSP1 or DUSP6 on MAPK signaling in the MPNST cell line, ST8814. In DUSP1 knockdown, 15 to 60 minutes after serum stimulation, p-ERK and p-JNK were 1.75× higher compared with NT controls (Fig. 3A). The phosphorylated JNK substrate p-c-jun was 2× higher, and p-p38 was unchanged (Fig. 3A). In contrast, in DUSP6 knockdown ST8814 cells, p-ERK was only modestly increased (1.3×; Fig. 3B). DUSP6 knockdown resulted in 2.58× higher p-JNK, and its substrate p-c-jun was 2× higher; p-p38 was unchanged compared with NT controls (Fig. 3B). DUSP1 knockdown cells treated with DUSP6 shRNA showed a 26% increase in p-ERK and larger increases in p-JNK and p-c-jun (71% and 56%, respectively) compared with DUSP1 knockdown alone (Fig. 3C). Total protein levels of these markers were unchanged except for c-jun in DUSP6 knockdown, which showed a modest increase (Supplementary Fig. S2A–S2C). We confirmed these results in the S462.TY MPNST cell line (Supplementary Fig. S2D and S2E). DUSP knockdown results in decreased proliferation and increased levels of p-ERK, p-JNK, and p-c-jun (Supplementary Fig. S2D and S2E). These data suggest that in MPNST cells DUSP1/6 play major roles in dephosphorylation of MAPK.

Figure 3.

Activation of ERK and JNK in the NF1/ MPNST cell line ST8814 by shDUSP1 and shDUSP6 knockdown. ST8814 MPNST cells infected with shRNA targeting DUSP1 (sh1-D1; A) or shRNA targeting DUSP6 (sh2-D6; B) were starved overnight in media without serum. Cells were then stimulated with DMEM and 10% FBS for designated minutes (min). Immunoblots were analyzed for p-ERK, p-JNK, p-c-jun, and p-p38 with β-actin as a loading control. C, MAPK signaling effects of DUSP1 shRNA-2 (sh2-D1) alone or in combination with DUSP6 shRNA2 (sh2-D6) in ST8814. D, Immunoblot analysis of TP53, p-ATM, cyclin D1, and PARP in ST8814 cells infected with DUSP1 shRNA-2 (sh2-D1) alone and in combination with DUSP6 shRNA2 (sh2-D6). Immunoblots were quantified by densitometry and expressed as percentages relative to NT2 controls (see text). E, Cell death analyzed with TUNEL fluorescence microscopy (red signal) with nuclei stained with DAPI (blue signal). Arrows indicate dying cells. Scale bar, 25 μm.

Figure 3.

Activation of ERK and JNK in the NF1/ MPNST cell line ST8814 by shDUSP1 and shDUSP6 knockdown. ST8814 MPNST cells infected with shRNA targeting DUSP1 (sh1-D1; A) or shRNA targeting DUSP6 (sh2-D6; B) were starved overnight in media without serum. Cells were then stimulated with DMEM and 10% FBS for designated minutes (min). Immunoblots were analyzed for p-ERK, p-JNK, p-c-jun, and p-p38 with β-actin as a loading control. C, MAPK signaling effects of DUSP1 shRNA-2 (sh2-D1) alone or in combination with DUSP6 shRNA2 (sh2-D6) in ST8814. D, Immunoblot analysis of TP53, p-ATM, cyclin D1, and PARP in ST8814 cells infected with DUSP1 shRNA-2 (sh2-D1) alone and in combination with DUSP6 shRNA2 (sh2-D6). Immunoblots were quantified by densitometry and expressed as percentages relative to NT2 controls (see text). E, Cell death analyzed with TUNEL fluorescence microscopy (red signal) with nuclei stained with DAPI (blue signal). Arrows indicate dying cells. Scale bar, 25 μm.

Close modal

We assessed the effect of DUSP1/6 downregulation on cell-cycle regulation and cell growth markers. Interestingly, upon DUSP1/6 silencing, cyclin D1 levels were reduced despite the high levels of MAPK signaling (Fig. 3D). Although sustained activation of ERK is required for cell-cycle progression, hyperactivation of ERK or JNK signaling can lead to stabilization of p53 that can induce cell-cycle arrest or cell death (34, 35). Indeed, after knockdown of DUSP1, DUSP6, or both, p53 and p-ATM were increased, suggesting that this pathway is active in MPNST cells (Fig. 3D). In addition, knockdown of DUSP1 or DUSP6 induced a 64% increase in PARP cleavage and increased TUNEL+ cells compared with NT controls, indicating the induction of apoptotic cell death (Fig. 3D and E). Collectively, our results identify key pathways by which DUSPs are likely to regulate proliferation and cell death in MPNST cells.

DUSP inhibitor BCI reduces human MPNST cell growth and activates JNK

To verify these results, we tested the effect of the DUSP1/6 inhibitor, BCI, on the growth of the NF1-deleted MPNST cell lines, ST8814 and S462.TY, and on NF1+/+ cell line, iHSC-1λ, and the sporadic MPNST cell line, STS26T. We found that 2 to 4 μmol/L BCI significantly inhibited MPNST cell growth 3 days after treatment (Fig. 4A). These concentrations were similarly effective in leukemia and gastric cancer cell lines (13, 14). The iHSC-1λ and STS26T MPNST cell lines were less sensitive to BCI (Fig. 4A). We evaluated MAPK pathway activation by Western blot analysis. Cells were serum-starved overnight, incubated with 2 μmol/L BCI, and then stimulated with serum-containing media. After 1 hour, p-ERK, p-JNK, p-c-jun, and total c-jun were elevated in the BCI-treated MPNST cell lines, ST8814 and S462.TY, but did not change in iHSC-1λ (Fig. 4B; Supplementary Fig. S3A). There were no detectable differences in p-p38 (Fig. 4B). In contrast, in STS26T cells only p-p38 was increased and cyclin D1 decreased (Supplementary Fig. S3B). Within 24 hours, BCI decreased total PARP and increased cleaved PARP and CC3, indicative of apoptotic cell death in NF1-deficient ST8814 and S462.TY cells (Fig. 4C). Also, there were 1.5 to 2× increases in TP53 and p-ATM in these BCI-treated cells correlating with reduced phospho-RB levels and reduced cell proliferation (Fig. 4C).

Figure 4.

The DUSP inhibitor, BCI, suppresses human MPNST cell growth via JNK activation. A, Cell survival was measured with the MTS assay (OD at 490 nm) after 3 days. Cells were serum starved overnight then treated with BCI in DMEM and 10% FBS. B, Immunoblot analysis of MAPK signaling. MPNST cells were starved overnight, incubated with BCI (2 μmol/L) for 60 minutes, and then stimulated with DMEM and 10% FBS for 1 hour. C, Immunoblot analysis of TP53, p-RB, and p-ATM and PARP cleavage and CC3 in ST8814 and S462.TY MPNST cells 24 hours after treatment with BCI (2 μmol/L). D, ST8814 cells treated with BCI (2 μmol/L) in combination with JNK-IN-8 (0–2.5 μmol/L) and analyzed using the MTS assay. Experiments were done twice with n = 6 per experiment (*, P < 0.05; **, P < 0.01).

Figure 4.

The DUSP inhibitor, BCI, suppresses human MPNST cell growth via JNK activation. A, Cell survival was measured with the MTS assay (OD at 490 nm) after 3 days. Cells were serum starved overnight then treated with BCI in DMEM and 10% FBS. B, Immunoblot analysis of MAPK signaling. MPNST cells were starved overnight, incubated with BCI (2 μmol/L) for 60 minutes, and then stimulated with DMEM and 10% FBS for 1 hour. C, Immunoblot analysis of TP53, p-RB, and p-ATM and PARP cleavage and CC3 in ST8814 and S462.TY MPNST cells 24 hours after treatment with BCI (2 μmol/L). D, ST8814 cells treated with BCI (2 μmol/L) in combination with JNK-IN-8 (0–2.5 μmol/L) and analyzed using the MTS assay. Experiments were done twice with n = 6 per experiment (*, P < 0.05; **, P < 0.01).

Close modal

Given that blocking DUSPs, genetically or pharmacologically, increased p-JNK, we evaluated the possibility that JNK is a critical mediator of decreasing cell proliferation and/or increasing cell death, by testing whether JNK inhibition would alter BCI effects in MPNSTs. In ST8814 MPNST cells treated with the irreversible JNK inhibitor JNK-IN-8 at 0.6 to 2.5 μmol/L, cell proliferation was modestly increased (Supplementary Fig. S3C). These concentrations inhibit JNK activity as indicated by reduced p-c-jun (Supplementary Fig. S3D; ref. 36). Targeting DUSP and JNK in combination reduced the effectiveness of BCI with combination indexes 1.2-3.9 indicating antagonism (Fig. 4D). Similar effects were seen with the JNK inhibitor, SP600125 (Supplementary Fig. S3E and S3F). These data suggest that blocking DUSP phosphatase activity using BCI increases p-JNK–causing effects on MPNST cells. Given that DUSP6 knockdown more effectively hyperactivates MAPKs in MPNST ST8814 cells (Fig. 3B), our data suggest that BCI largely inhibits DUSP6 in NF1-deleted MPNSTs.

In MPNST cells, DUSP inhibition modestly increased phosphorylation of the MEK substrate ERK (Fig. 4B). We tested whether DUSP inhibition alters the effects of MEK inhibition in MPNST cell lines (Supplementary Fig. S4A and S4B). At 0.5 to 2 μmol/L, BCI with 1.25 or 2.5 μmol/L PD0325901, MEK inhibitory concentrations (5), there were no additive effects in ST8814 (CI, 0.9; Supplementary Fig. S4A and S4B) or STS26T (data not shown). In S462.TY, there was evidence of modest synergy (CI, 0.29) over a narrow concentration range (Supplementary Fig. S4A and S4C). The effect of PD0325901 in combination with genetic knockdown of Dusp1/6 had no additive or antagonistic effect on cell proliferation (Supplementary Fig. S4D). Thus, DUSP effects are mediated by JNK and combinations with MEKi may be modestly effective in some MPNST cell lines.

NF1 loss increases efficacy of BCI treatment

Because NF1 deletion in iHSC increased DUSP1 and DUSP6 protein levels (Fig. 1C), we used these cell lines to determine whether BCI would inhibit proliferation in a NF1-dependent manner in Schwann cells. There was a therapeutic window in which NF1-deleted iHSCs were more susceptible compared with WT cells to the antiproliferative effects of BCI (0.25–1 μmol/L; Fig. 5A). The largest difference was at 1 μmol/L (Fig. 5A). DUSP inhibition had lesser effects on iHSC NF1 WT cells, in which the baseline activity of MAPK (p-ERK) was barely detectable (Fig. 5B). Consistent with direct inhibition of DUSPs with shRNA and with BCI in MPNSTs, 1 μmol/L BCI enhanced p-ERK, p-JNK, and p-c-jun signaling in NF1-deleted iHSC after 1 hour, (Fig. 5B; Supplementary Fig. S5A). Constitutive MAPK activity in the iHSC NF1/ cells was strongly increased by treatment with BCI and correlated with increased levels of TP53 and p-ATM and reduced pRB and cyclin D1 (Fig. 5C). Evidence of cell death was variable, with evidence of cleaved PARP but not CC3, suggesting reduced proliferation as the predominate effect in these cells (Fig. 5C). As in MPNST cells, targeting DUSP and JNK in combination diminished the effectiveness of BCI in NF1/ iHSC (CI > 1.3; Fig. 5D; Supplementary Fig. S5B). At these concentrations of JNK-IN-8, p-c-jun was reduced (Supplementary Fig. S5C). Thus, in NF1/ Schwann cells and in MPNST cells with NF1 deletion, DUSPs contributes to cell proliferation and survival. These data suggest that DUSPs act as oncogenes in this context.

Figure 5.

NF1 loss increases efficacy of BCI treatment in Schwann cells. DUSP inhibition with BCI in iHSC WT versus NF1/. A, Cell survival measured with the MTS assay. iHSC WT and NF1/ cells were serum-starved overnight and then treated with BCI (0–4 μmol/L) in DMEM-F12 with N2 supplements and 10 ng/mL β-Heregulin (β-HRG) for 3 days. B, Immunoblot analysis of MAPK signaling after cells were starved overnight, preincubated with BCI (1 μmol/L) for 60 minutes, and then stimulated for 1 hour. C, Immunoblot analysis of cyclin D1, TP53, p-RB, p-ATM, and PARP and CC3 in WT and NF1/ iHSC after 24 hours of treatment with BCI (1 μmol/L). D, Cell survival was measured with the MTS assay. Cells treated with BCI (0.5–1 μmol/L), in combination with JNK-IN-8 (0.3 μmol/L) for 3 days. *, P < 0.05. Experiments were done twice with n = 4–6 per experiment.

Figure 5.

NF1 loss increases efficacy of BCI treatment in Schwann cells. DUSP inhibition with BCI in iHSC WT versus NF1/. A, Cell survival measured with the MTS assay. iHSC WT and NF1/ cells were serum-starved overnight and then treated with BCI (0–4 μmol/L) in DMEM-F12 with N2 supplements and 10 ng/mL β-Heregulin (β-HRG) for 3 days. B, Immunoblot analysis of MAPK signaling after cells were starved overnight, preincubated with BCI (1 μmol/L) for 60 minutes, and then stimulated for 1 hour. C, Immunoblot analysis of cyclin D1, TP53, p-RB, p-ATM, and PARP and CC3 in WT and NF1/ iHSC after 24 hours of treatment with BCI (1 μmol/L). D, Cell survival was measured with the MTS assay. Cells treated with BCI (0.5–1 μmol/L), in combination with JNK-IN-8 (0.3 μmol/L) for 3 days. *, P < 0.05. Experiments were done twice with n = 4–6 per experiment.

Close modal

In vivo effects of DUSP inhibitor BCI in MPNST xenograft models

We tested whether BCI might be a useful therapeutic in vivo using an established cell line–based xenograft model, S462.TY MPNST cells (TY xenograft) engrafted subcutaneously into athymic nude mice. Tumor-bearing mice were injected with BCI 10 mg/kg, i.p., once daily, 5×/week for 27 days. No differences were detectable in tumor volume (Fig. 6A). In spite of the similar tumor volumes, excision of BCI-treated tumors revealed edema associated with encapsulated necrotic tissue that was absent in vehicle-treated mice; areas of necrosis ranged from 8% to 23% of section area (Fig. 6B and C).

Figure 6.

In vivo DUSP inhibition causes tumor necrosis in MPNST xenograft models. A, Tumor volume (mm3) in the S462.TY cell–based xenograft MPNST model dosed with vehicle (Veh) or BCI 10 mg/kg, i.p. once daily, 5×/week for 27 days (n = 9) versus vehicle (n = 6). By day 27, mice required sacrifice. Data points represent mean ± SEM. B, Gross image of S462.TY tumors with necrotic tissue (BCI-treated) compared with vehicle-treated tumors. C, Quantification of tumor necrosis in the S462.TY xenograft MPNST model, expressed as percent total section area. Data points represents mean ± SD. **, P < 0.01, t test. D, Tumor volume (mm3) in the PDX-based xenograft MPNST model dosed with BCI (10 or 25 mg/kg) i.p. once daily, 5×/week for 21 days versus control (n = 5 per group). Data points represent mean ± SEM. *, P < 0.05; ANOVA, Tukey post hoc test). Representative images of H&E-stained and CC3-stained tissue sections in the S462.TY xenograft (E) and PDX xenograft (F). Extensive necrosis (N) and increased CC3 occur compared with vehicle-treated tumors. Scale bar, 100 μm. G, Immunoblot analysis of MAPK signaling markers in S462.TY xenograft model. Mice were dosed with BCI 10 mg/kg, i.p. once daily, 5×/week for 27 days or vehicle and were sacrificed 2 hours after the last dose. H, Immunoblot analysis of MAPK signaling markers in PDX MPNST tumors. Mice were dosed with BCI 10 mg/kg, i.p. once daily, 5×/week for 12 days or vehicle and were sacrificed 2 hours after the last dose.

Figure 6.

In vivo DUSP inhibition causes tumor necrosis in MPNST xenograft models. A, Tumor volume (mm3) in the S462.TY cell–based xenograft MPNST model dosed with vehicle (Veh) or BCI 10 mg/kg, i.p. once daily, 5×/week for 27 days (n = 9) versus vehicle (n = 6). By day 27, mice required sacrifice. Data points represent mean ± SEM. B, Gross image of S462.TY tumors with necrotic tissue (BCI-treated) compared with vehicle-treated tumors. C, Quantification of tumor necrosis in the S462.TY xenograft MPNST model, expressed as percent total section area. Data points represents mean ± SD. **, P < 0.01, t test. D, Tumor volume (mm3) in the PDX-based xenograft MPNST model dosed with BCI (10 or 25 mg/kg) i.p. once daily, 5×/week for 21 days versus control (n = 5 per group). Data points represent mean ± SEM. *, P < 0.05; ANOVA, Tukey post hoc test). Representative images of H&E-stained and CC3-stained tissue sections in the S462.TY xenograft (E) and PDX xenograft (F). Extensive necrosis (N) and increased CC3 occur compared with vehicle-treated tumors. Scale bar, 100 μm. G, Immunoblot analysis of MAPK signaling markers in S462.TY xenograft model. Mice were dosed with BCI 10 mg/kg, i.p. once daily, 5×/week for 27 days or vehicle and were sacrificed 2 hours after the last dose. H, Immunoblot analysis of MAPK signaling markers in PDX MPNST tumors. Mice were dosed with BCI 10 mg/kg, i.p. once daily, 5×/week for 12 days or vehicle and were sacrificed 2 hours after the last dose.

Close modal

We next used a novel PDX MPNST model which was developed by directly implanting patient tumor tissue into mice. The patient tumor had mutations in NF1 and in other genes common in human MPNSTs (CDKN2A/B loss and SUZ12 mutation), Rictor was also amplified. The resulting xenograft MPNSTs maintained characteristics of the original tumor histologically (Supplementary Fig. S6A) and genetically with NF1 and SUZ12 mutations. Grafted tumors remained positive for human nucleoli antigen (Supplementary Fig. S6B). BCI treatment at both doses significantly inhibited the growth of the PDX MPNSTs (Fig. 6D). In tissue sections, both the TY xenograft and the PDX MPNST model showed areas of cell apoptosis (CC3+ cells) and necrotic cell death on histology (TY, 8%–23%; PDX, 2%–17% vs. VEH 1%–2%; Fig. 6E and F). Both models also had areas of fibrosis/collagen deposits as indicated by trichrome staining (Supplementary Fig. S6C).

In the S462.TY xenograft, samples were collected on treatment day 27, 2 hours following the last BCI treatment, and analyzed for MAPK signaling and proliferation/cell death markers. BCI-treated tumors showed 60% to 90% increases in p-ERK, p-JNK, and p-c-jun, 60% lower levels of cyclin D1, and variable levels of TP53 versus controls (Fig. 6G; Supplementary Fig. S6D and S6E). In the PDX model, after 21 days of treatment, there was a trend toward MAPK activation (Supplementary Fig. S6F). Given that signaling data may be confounded by areas of necrosis and collagen/fibrosis, in the PDX model, signaling was analyzed after 12 days of BCI. These BCI-treated tumors had a significant 2.5 to 3.5× increased p-ERK, p-JNK, and p-c-jun, 40% lower levels of cyclin D1, and a 46% increase in TP53 (Fig. 6H; Supplementary Fig. S6G and S6H). Upon discontinuing treatment in the PDX model, tumors showed regrowth and areas of proliferation among areas of necrosis and collagen deposits (Supplementary Fig. S7A). This suggests that BCI affects the MAPK pathway in vivo to transiently inhibit cell proliferation and induce cell death in MPNSTs.

Interestingly, in a separate study using the PDX model, we found that BCI (10 mg/kg, i.p., 5×/week) was as effective as the MEKi PD0325901 (1.5 mg/kg, 5×/week) in inhibiting tumor growth (Supplementary Fig. S7B). We also tested PD0325901 (1.5 mg/kg, 5×/week for 27 days), versus a combination of BCI and PD0325901 (10 mg/kg, 5×/week for 27 days and 1.5 mg/kg, 5×/week for 27 days, respectively) in the TY xenograft model. As described previously, tumor volumes of MEK inhibitor-treated tumors were significantly lower compared with vehicle (Supplementary Fig. S7C). However, there was no significant tumor volume difference between the combination of BCI and PD0325901 versus PD0325901 only (Supplementary Fig. S7C). From histologic analysis, BCI-treated tumors showed more necrotic cell death compared with MEK alone or treatment with the MEKi and DUSPi combination (Supplementary Fig. S7D). Areas of necrosis were also observed in the PDX xenograft model after BCI but not PD0325901 treatment (Supplementary Fig. S7D). Collectively, our data using 2 different in vivo xenograft MPNST models suggests that targeting DUSP1/6 with a single agent, BCI, suppresses MPNST growth and induces cell death.

Novel targets and treatment options for NF1-mediated MPNSTs are urgently needed. DUSPs regulate MAPK signaling by affecting diverse cellular processes including cell proliferation, cell survival, and cell differentiation (37). We posited that understanding how DUSP family proteins affect NF1-mutant MPNSTs would suggest new approaches for MPNST therapy. Our analysis of DUSP1 and DUSP6 shows that both DUSP proteins are overexpressed in MPNSTs compared with normal tissues under basal conditions, in cells and in growing tumors. DUSP1 and DUSP6 may have partially redundant functions in MPNSTs, and it is possible that other DUSPs, including DUSP22, also play roles. DUSP1/6 are upregulated in NF1-mutant neurofibroma, iHSC, and in MPNST cells. In these cell types, DUSPs promote cell proliferation in vitro, as reducing levels of either or both, genetically or with the small-molecule inhibitor BCI, decreases cell proliferation. Each DUSP1 and DUSP6 also protect NF1-mutant MPNST cells from cell death, as silencing DUSP1 or DUSP6 in MPNST cells increased cell death in vitro. In 2 in vivo model systems, BCI, which specifically targets both DUSPs (12, 21) caused necrotic and apoptotic cell death.

In most normal cells, DUSPs are transcriptionally upregulated in response to MAPK signaling, and function as negative feedback regulators to suppress RAS/MAPK signaling. DUSPs are often upregulated in cancers harboring activating mutations in RAS or BRAF (6). The mechanisms underlying DUSP upregulation vary. Patients with pre-B ALL showed reduced CpG methylation of the DUSP6 promoter as compared with normal pre-B cells (13). Gene amplification or increased transcription due to deletion of NF1 and increased MAPK signaling may also play roles. In agreement with data from Courtois-Cox and colleagues (4), we find transcriptional upregulation of DUSPs in NF1 mutant cells. In addition, in 30% of MPNSTs, the DUSP1 or DUSP6 locus, or both, are amplified.

Genetic knockdown of DUSP1 decreased tumorigenesis in orthotopic pancreatic tumors and in non–small cell lung cancer (38, 39). DUSP6 prosurvival effects were also described in chemoresistance screens, in glioblastoma cells, and gastric cancer (14, 40, 41). The utility of the DUSP1/6 inhibitor, BCI, or its analogs, as therapeutics remains to be tested. BCI is a small molecule identified in a zebrafish chemical screen that prevents catalytic phosphatase activity upon substrate binding, contributing to its specificity (21). An allosteric binding site for BCI exists within the phosphatase domains of both DUSP6 and DUSP1, but not in the related DUSP5; activity toward other DUSPs has not been excluded to date (12, 21). BCI demonstrated single-agent efficacy at doses 5 to 40 mg/kg, i.p daily (or twice daily) for up to 1 month in mouse models of gastric cancer and leukemia. These regimens effectively inhibited DUSP1/6 without noted toxicity, as did our doses within this range in MPNST xenografts (12–14). Inhibition of the MEK/ERK pathway combined with DUSP inhibition in vivo did not alter tumor volume or cell death when compared with single agents. Further studies may identify improved dosing regimens and schedules that maximize efficacy.

Previous work showed that inhibiting DUSPs can result in increased phosphorylation of ERK, JNK, and/or p38 MAPKs (13). A predominant role for p-p38 was demonstrated in breast cancer and in therapy-resistant chronic myeloid leukemia (11, 12). In gastric cancer models, DUSP inhibition increased p-ERK, inducing cell death (14). In non–small cell lung cancer, phosphorylated JNK and p38 affected tumor progression upon DUSP1 silencing (39). Indeed, recent DUSP6 knockout studies failed to show expected effects on p-ERK; instead, p-JNK increased (42). These studies highlight the tumor-specific MAPK substrate effects of DUSPs. In MPNST cells, DUSP knockdown or DUSP inhibition with BCI caused increased p-ERK, p-JNK, and phosphorylation of the JNK substrate c-Jun. The finding that blocking JNK with either of 2 unrelated antagonists rescues the effects of BCI suggests that in the NF1-deficient immortalized Schwann cells and MPNST cells we evaluated, DUSP1 and DUSP6 act largely on the JNK arm of the MAPK pathway, supporting the idea that JNK activation plays a critical role in BCI's antitumor effect in MPNSTs.

The duration of MAPK activation is controlled by cell expression of negative regulators of MAPK signaling, including GAPs, such as NF1, and phosphatases such as DUSPs (4). MAPK is known to inhibit growth and promote cell death through altering phosphorylation of RB and ATM, or through TP53 (43). Given that TP53, RB, and ATM are central regulators of the cell cycle, the genetic status of these tumor suppressors may influence sensitivity to DUSP inhibitors. The incidence of mutation of TP53 is 40% in MPNSTs (44). In MPNSTs with WT TP53, DUSP inhibition with acute reactivation of JNK signaling appears to stabilize TP53, enabling the apoptotic pathway as occurs in other settings (34, 35). In ovarian cancer cells, JNK promotes PARP phosphorylation leading to PARP degradation (45). Total and cleaved PARP play important roles in modulation of apoptosis and necrosis. Inhibition of DUSPs and PARP may induce DNA damage and prevent repair, potentially sensitizing radio-resistant MPNST cells to radiation therapy and cell death.

The S462.TY cell line model has a high tumorigenic potential in mice and so is widely used to evaluate therapies for MPNSTs. However, a single model cannot accurately represent the complexity and diversity of MPNST tumors. Models in which patient-derived tissues, including MPNSTs, are directly grafted into immunocompromised mice are increasingly used to test therapeutics (46). We describe a PDX model that contains common cancer-associated mutations found in patient MPNSTs: NF1, CDKN2A/B, SUZ12 (47–49), and retains patient MPNST histologic features. BCI treatment in this model slowed tumor growth and also promoted cell death. With BCI treatment, the differences in tumor volume between the models may have been due to edema, with a delay in clearance of the dying cells and collagen deposits that contribute to tumor volume. Such effects can confound analysis of drug effects on tumor volume, as shown after other drug treatments in sarcoma and xenografted tumors (50). Importantly, in both models, single-agent BCI treatment was sufficient to cause cell death in vivo. For comparison, MEK inhibitor treatment transiently lowers tumor volumes, with no evidence of increased cell death (5).

In NF1-mediated transformation to MPNSTs, tumor cells appear to maintain MAPK activity within a narrow range to enable survival. DUSP inhibition, with increased MAPK signaling through JNK (and possibly other substrates), slows proliferation and can promote cell death. Our data support the development of agents targeting both DUSP1 and DUSP6 for MPNSTs.

D.A. Largaespada is an employee of Surrogen, Inc. and B-MoGen Biotechnologies, reports receiving commercial research grants from Genentech, and holds ownership interest (including patents) in NeoClone Biotechnology, ImmuSoft (previously Discovery Genomics, Inc), B-MoGen Biotechnologies, and Recombinetics, Inc. No potential conflicts of interest were disclosed by the other authors.

Conception and design: A. Ramkissoon, K.B. Williams, D.A. Largaespada, N. Ratner

Development of methodology: A. Ramkissoon, D. Milewski, K.B. Williams, R.L. Williams, K. Choi, N. Ratner

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): A. Ramkissoon, K.E. Chaney, D. Milewski, A.A. Miller, T.V. Kalin, J.G. Pressey, S. Szabo, M. Azam, N. Ratner

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): A. Ramkissoon, K. Choi, A.A. Miller, S. Szabo, M. Azam, N. Ratner

Writing, review, and/or revision of the manuscript: A. Ramkissoon, D. Milewski, K. Choi, J.G. Pressey, N. Ratner

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): R.L. Williams

Study supervision: A. Ramkissoon, M. Azam, N. Ratner

The authors thank Dr. Meenu Kesarwani for providing reagents and critical review of the manuscript, Dr. Tilat Rizvi for advice on IHC, Jonathan Fletcher for tumor dissection assistance, and Kierra Ware for immunoblot assistance. This work was supported by R01 NS086219 (to D.A. Largaespada and N. Ratner), a grant from the Children's Tumor Foundation-Synodos (to D.A. Largaespada), and CancerFree KIDS grants (to A. Ramkissoon). A.A. Miller was supported by a summer fellowship (NIH T35DK060444).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Ducatman
BS
,
Scheithauer
BW
,
Piepgras
DG
,
Reiman
HM
,
Ilstrup
DM
. 
Malignant peripheral nerve sheath tumors. A clinicopathologic study of 120 cases
.
Cancer
1986
;
57
:
2006
21
.
2.
Kim
A
,
Stewart
DR
,
Reilly
KM
,
Viskochil
D
,
Miettinen
MM
,
Widemann
BC
. 
Malignant peripheral nerve sheath tumors state of the science: leveraging clinical and biological insights into effective therapies
.
Sarcoma
2017
;
2017
:
7429697
.
3.
Uusitalo
E
,
Rantanen
M
,
Kallionpaa
RA
,
Poyhonen
M
,
Leppavirta
J
,
Yla-Outinen
H
, et al
Distinctive cancer associations in patients with neurofibromatosis type 1
.
J Clin Oncol
2016
;
34
:
1978
86
.
4.
Courtois-Cox
S
,
Genther Williams
SM
,
Reczek
EE
,
Johnson
BW
,
McGillicuddy
LT
,
Johannessen
CM
, et al
A negative feedback signaling network underlies oncogene-induced senescence
.
Cancer Cell
2006
;
10
:
459
72
.
5.
Jessen
WJ
,
Miller
SJ
,
Jousma
E
,
Wu
J
,
Rizvi
TA
,
Brundage
ME
, et al
MEK inhibition exhibits efficacy in human and mouse neurofibromatosis tumors
.
J Clin Invest
2013
;
123
:
340
7
.
6.
Pratilas
CA
,
Taylor
BS
,
Ye
Q
,
Viale
A
,
Sander
C
,
Solit
DB
, et al
(V600E)BRAF is associated with disabled feedback inhibition of RAF-MEK signaling and elevated transcriptional output of the pathway
.
Proc Natl Acad Sci U S A
2009
;
106
:
4519
24
.
7.
Caunt
CJ
,
Keyse
SM
. 
Dual-specificity MAP kinase phosphatases (MKPs): shaping the outcome of MAP kinase signalling
.
FEBS J
2013
;
280
:
489
504
.
8.
Patterson
KI
,
Brummer
T
,
O'Brien
PM
,
Daly
RJ
. 
Dual-specificity phosphatases: critical regulators with diverse cellular targets
.
Biochem J
2009
;
418
:
475
89
.
9.
Furukawa
T
,
Sunamura
M
,
Motoi
F
,
Matsuno
S
,
Horii
A
. 
Potential tumor suppressive pathway involving DUSP6/MKP-3 in pancreatic cancer
.
Am J Pathol
2003
;
162
:
1807
15
.
10.
Okudela
K
,
Yazawa
T
,
Woo
T
,
Sakaeda
M
,
Ishii
J
,
Mitsui
H
, et al
Down-regulation of DUSP6 expression in lung cancer: its mechanism and potential role in carcinogenesis
.
Am J Pathol
2009
;
175
:
867
81
.
11.
Kaltenmeier
CT
,
Vollmer
LL
,
Vernetti
LA
,
Caprio
L
,
Davis
K
,
Korotchenko
VN
, et al
A tumor cell-selective inhibitor of mitogen-activated protein kinase phosphatases sensitizes breast cancer cells to lymphokine-activated killer cell activity
.
J Pharmacol Exp Ther
2017
;
361
:
39
50
.
12.
Kesarwani
M
,
Kincaid
Z
,
Gomaa
A
,
Huber
E
,
Rohrabaugh
S
,
Siddiqui
Z
, et al
Targeting c-FOS and DUSP1 abrogates intrinsic resistance to tyrosine-kinase inhibitor therapy in BCR-ABL-induced leukemia
.
Nat Med
2017
;
23
:
472
82
.
13.
Shojaee
S
,
Caeser
R
,
Buchner
M
,
Park
E
,
Swaminathan
S
,
Hurtz
C
, et al
Erk negative feedback control enables pre-b cell transformation and represents a therapeutic target in acute lymphoblastic leukemia
.
Cancer Cell
2015
;
28
:
114
28
.
14.
Wu
QN
,
Liao
YF
,
Lu
YX
,
Wang
Y
,
Lu
JH
,
Zeng
ZL
, et al
Pharmacological inhibition of DUSP6 suppresses gastric cancer growth and metastasis and overcomes cisplatin resistance
.
Cancer Lett
2018
;
412
:
243
55
.
15.
Philpott
C
,
Tovell
H
,
Frayling
IM
,
Cooper
DN
,
Upadhyaya
M
. 
The NF1 somatic mutational landscape in sporadic human cancers
.
Hum Genomics
2017
;
11
:
13
.
16.
Patwardhan
PP
,
Surriga
O
,
Beckman
MJ
,
de Stanchina
E
,
Dematteo
RP
,
Tap
WD
, et al
Sustained inhibition of receptor tyrosine kinases and macrophage depletion by PLX3397 and rapamycin as a potential new approach for the treatment of MPNSTs
.
Clin Cancer Res
2014
;
20
:
3146
58
.
17.
Rahrmann
EP
,
Watson
AL
,
Keng
VW
,
Choi
K
,
Moriarity
BS
,
Beckmann
DA
, et al
Forward genetic screen for malignant peripheral nerve sheath tumor formation identifies new genes and pathways driving tumorigenesis
.
Nat Genet
2013
;
45
:
756
66
.
18.
Dodd
RD
,
Mito
JK
,
Eward
WC
,
Chitalia
R
,
Sachdeva
M
,
Ma
Y
, et al
NF1 deletion generates multiple subtypes of soft-tissue sarcoma that respond to MEK inhibition
.
Mol Cancer Ther
2013
;
12
:
1906
17
.
19.
Patel
AV
,
Eaves
D
,
Jessen
WJ
,
Rizvi
TA
,
Ecsedy
JA
,
Qian
MG
, et al
Ras-driven transcriptome analysis identifies aurora kinase A as a potential malignant peripheral nerve sheath tumor therapeutic target
.
Clin Cancer Res
2012
;
18
:
5020
30
.
20.
Currier
MA
,
Sprague
L
,
Rizvi
TA
,
Nartker
B
,
Chen
CY
,
Wang
PY
, et al
Aurora A kinase inhibition enhances oncolytic herpes virotherapy through cytotoxic synergy and innate cellular immune modulation
.
Oncotarget
2017
;
8
:
17412
27
.
21.
Molina
G
,
Vogt
A
,
Bakan
A
,
Dai
W
,
Queiroz de Oliveira
P
,
Znosko
W
, et al
Zebrafish chemical screening reveals an inhibitor of Dusp6 that expands cardiac cell lineages
.
Nat Chem Biol
2009
;
5
:
680
7
.
22.
Li
H
,
Chang
LJ
,
Neubauer
DR
,
Muir
DF
,
Wallace
MR
. 
Immortalization of human normal and NF1 neurofibroma Schwann cells
.
Lab Invest
2016
;
96
:
1105
15
.
23.
DeClue
JE
,
Papageorge
AG
,
Fletcher
JA
,
Diehl
SR
,
Ratner
N
,
Vass
WC
, et al
Abnormal regulation of mammalian p21ras contributes to malignant tumor growth in von Recklinghausen (type 1) neurofibromatosis
.
Cell
1992
;
69
:
265
73
.
24.
Miller
SJ
,
Rangwala
F
,
Williams
J
,
Ackerman
P
,
Kong
S
,
Jegga
AG
, et al
Large-scale molecular comparison of human schwann cells to malignant peripheral nerve sheath tumor cell lines and tissues
.
Cancer Res
2006
;
66
:
2584
91
.
25.
Yang
J
,
Ylipaa
A
,
Sun
Y
,
Zheng
H
,
Chen
K
,
Nykter
M
, et al
Genomic and molecular characterization of malignant peripheral nerve sheath tumor identifies the IGF1R pathway as a primary target for treatment
.
Clin Cancer Res
2011
;
17
:
7563
73
.
26.
Olshen
AB
,
Venkatraman
ES
,
Lucito
R
,
Wigler
M
. 
Circular binary segmentation for the analysis of array-based DNA copy number data
.
Biostatistics
2004
;
5
:
557
72
.
27.
van de Wiel
MA
,
Kim
KI
,
Vosse
SJ
,
van Wieringen
WN
,
Wilting
SM
,
Ylstra
B
. 
CGHcall: calling aberrations for array CGH tumor profiles
.
Bioinformatics
2007
;
23
:
892
4
.
28.
Gautier
L
,
Cope
L
,
Bolstad
BM
,
Irizarry
RA
. 
affy–analysis of Affymetrix GeneChip data at the probe level
.
Bioinformatics
2004
;
20
:
307
15
.
29.
Ritchie
ME
,
Phipson
B
,
Wu
D
,
Hu
Y
,
Law
CW
,
Shi
W
, et al
limma powers differential expression analyses for RNA-sequencing and microarray studies
.
Nucleic Acids Res
2015
;
43
:
e47
.
30.
Wu
J
,
Williams
JP
,
Rizvi
TA
,
Kordich
JJ
,
Witte
D
,
Meijer
D
, et al
Plexiform and dermal neurofibromas and pigmentation are caused by Nf1 loss in desert hedgehog-expressing cells
.
Cancer Cell
2008
;
13
:
105
16
.
31.
Brondello
JM
,
Pouyssegur
J
,
McKenzie
FR
. 
Reduced MAP kinase phosphatase-1 degradation after p42/p44MAPK-dependent phosphorylation
.
Science
1999
;
286
:
2514
7
.
32.
Schmidt
H
,
Taubert
H
,
Meye
A
,
Wurl
P
,
Bache
M
,
Bartel
F
, et al
Gains in chromosomes 7, 8q, 15q and 17q are characteristic changes in malignant but not in benign peripheral nerve sheath tumors from patients with Recklinghausen's disease
.
Cancer Lett
2000
;
155
:
181
90
.
33.
Mantripragada
KK
,
Spurlock
G
,
Kluwe
L
,
Chuzhanova
N
,
Ferner
RE
,
Frayling
IM
, et al
High-resolution DNA copy number profiling of malignant peripheral nerve sheath tumors using targeted microarray-based comparative genomic hybridization
.
Clin Cancer Res
2008
;
14
:
1015
24
.
34.
Fuchs
SY
,
Adler
V
,
Pincus
MR
,
Ronai
Z
. 
MEKK1/JNK signaling stabilizes and activates p53
.
Proc Natl Acad Sci U S A
1998
;
95
:
10541
6
.
35.
Golding
SE
,
Rosenberg
E
,
Neill
S
,
Dent
P
,
Povirk
LF
,
Valerie
K
. 
Extracellular signal-related kinase positively regulates ataxia telangiectasia mutated, homologous recombination repair, and the DNA damage response
.
Cancer Res
2007
;
67
:
1046
53
.
36.
Xie
X
,
Kaoud
TS
,
Edupuganti
R
,
Zhang
T
,
Kogawa
T
,
Zhao
Y
, et al
c-Jun N-terminal kinase promotes stem cell phenotype in triple-negative breast cancer through upregulation of Notch1 via activation of c-Jun
.
Oncogene
2017
;
36
:
2599
608
.
37.
Kidger
AM
,
Keyse
SM.
The regulation of oncogenic Ras/ERK signalling by dual-specificity mitogen activated protein kinase phosphatases (MKPs)
.
Semin Cell Dev Biol
2016
;
50
:
125
32
.
38.
Liu
F
,
Gore
AJ
,
Wilson
JL
,
Korc
M
. 
DUSP1 is a novel target for enhancing pancreatic cancer cell sensitivity to gemcitabine
.
PLoS One
2014
;
9
:
e84982
.
39.
Moncho-Amor
V
,
Ibanez de Caceres
I
,
Bandres
E
,
Martinez-Poveda
B
,
Orgaz
JL
,
Sanchez-Perez
I
, et al
DUSP1/MKP1 promotes angiogenesis, invasion and metastasis in non-small-cell lung cancer
.
Oncogene
2011
;
30
:
668
78
.
40.
MacKeigan
JP
,
Murphy
LO
,
Blenis
J
. 
Sensitized RNAi screen of human kinases and phosphatases identifies new regulators of apoptosis and chemoresistance
.
Nat Cell Biol
2005
;
7
:
591
600
.
41.
Messina
S
,
Frati
L
,
Leonetti
C
,
Zuchegna
C
,
Di Zazzo
E
,
Calogero
A
, et al
Dual-specificity phosphatase DUSP6 has tumor-promoting properties in human glioblastomas
.
Oncogene
2011
;
30
:
3813
20
.
42.
Hsu
WC
,
Chen
MY
,
Hsu
SC
,
Huang
LR
,
Kao
CY
,
Cheng
WH
, et al
DUSP6 mediates T cell receptor-engaged glycolysis and restrains TFH cell differentiation
.
Proc Natl Acad Sci U S A
2018
;
115
:
E8027
-
-36
.
43.
Wada
T
,
Penninger
JM.
Mitogen-activated protein kinases in apoptosis regulation
.
Oncogene
2004
;
23
:
2838
49
.
44.
Brohl
AS
,
Kahen
E
,
Yoder
SJ
,
Teer
JK
,
Reed
DR
. 
The genomic landscape of malignant peripheral nerve sheath tumors: diverse drivers of Ras pathway activation
.
Sci Rep
2017
;
7
:
14992
.
45.
Wang
J
,
Kho
DH
,
Zhou
JY
,
Davis
RJ
,
Wu
GS
. 
MKP-1 suppresses PARP-1 degradation to mediate cisplatin resistance
.
Oncogene
2017
;
36
:
5939
47
.
46.
Castellsague
J
,
Gel
B
,
Fernandez-Rodriguez
J
,
Llatjos
R
,
Blanco
I
,
Benavente
Y
, et al
Comprehensive establishment and characterization of orthoxenograft mouse models of malignant peripheral nerve sheath tumors for personalized medicine
.
EMBO Mol Med
2015
;
7
:
608
27
.
47.
Birindelli
S
,
Perrone
F
,
Oggionni
M
,
Lavarino
C
,
Pasini
B
,
Vergani
B
, et al
Rb and TP53 pathway alterations in sporadic and NF1-related malignant peripheral nerve sheath tumors
.
Lab Invest
2001
;
81
:
833
44
.
48.
De Raedt
T
,
Beert
E
,
Pasmant
E
,
Luscan
A
,
Brems
H
,
Ortonne
N
, et al
PRC2 loss amplifies Ras-driven transcription and confers sensitivity to BRD4-based therapies
.
Nature
2014
;
514
:
247
51
.
49.
Lee
W
,
Teckie
S
,
Wiesner
T
,
Ran
L
,
Prieto Granada
CN
,
Lin
M
, et al
PRC2 is recurrently inactivated through EED or SUZ12 loss in malignant peripheral nerve sheath tumors
.
Nat Genet
2014
;
46
:
1227
32
.
50.
Ran
L
,
Chen
Y
,
Sher
J
,
Wong
EWP
,
Murphy
D
,
Zhang
JQ
, et al
FOXF1 defines the core-regulatory circuitry in gastrointestinal stromal tumor
.
Cancer Discov
2018
;
8
:
234
51
.