<<

Published OnlineFirst July 15, 2013; DOI: 10.1158/1535-7163.MCT-13-0189

Molecular Cancer Tools and Technologies Therapeutics

NF1 Deletion Generates Multiple Subtypes of Soft-Tissue Sarcoma That Respond to MEK Inhibition

Rebecca D. Dodd1, Jeffrey K. Mito2, William C. Eward3, Rhea Chitalia1, Mohit Sachdeva1, Yan Ma1, Jordi Barretina5,6,7, Leslie Dodd4, and David G. Kirsch1,2

Abstract Soft-tissue sarcomas are a heterogeneous group of tumors arising from connective tissue. Recently, mutations in the neurofibromin 1 (NF1) were identified in multiple subtypes of human soft-tissue sarcomas. To study the effect of NF1 inactivation in the initiationandprogressionof distinct sarcoma subtypes, we have developed a novel mouse model of temporally and spatially restricted NF1-deleted sarcoma. To generate primary sarcomas, we inject adenovirus containing Cre recombinase into NF1flox/flox; Ink4a/Arfflox/flox mice at two distinct orthotopic sites: intramuscularly or in the sciatic nerve. The mice develop either high-grade myogenic sarcomas or malignant peripheral nerve sheath tumor (MPNST)-like tumors, respectively. These tumors reflect the histologic properties and spectrum of sarcomas found in patients. To explore the use of this model for preclinical studies, we conducted a study of mitogen-activated protein kinase (MAPK) pathway inhibition with the MEK inhibitor PD325901. Treatment with PD325901 delays tumor growth through decreased cyclin D1 mRNA and cell proliferation. We also examined the effects of MEK inhibition on the native tumor stroma and find that PD325901 decreases VEGFa expression in tumor cells with a corresponding decrease in microvessel density. Taken together, our results use a primary tumor model to show that sarcomas can be generated by loss of NF1 and Ink4a/Arf, and that these tumors are sensitive to MEK inhibition by direct effects on tumor cells and the surrounding microenvironment. These studies suggest that MEK inhibitors should be further explored as potential sarcoma therapies in patients with tumors containing NF1 deletion. Mol Cancer Ther; 12(9); 1906–17. 2013 AACR.

Introduction Recent genomic studies have identified novel mutations Soft-tissue sarcomas are an aggressive and heteroge- in a diverse spectrum of human soft tissue sarcomas that neous group of tumors that arise from connective tissue, may reveal new molecular targets for therapy (2). One of including the muscle, nerve sheath, blood vessel, fat, and the most frequently mutated gene described is neurofi- fibrous tissue. These tumors are fatal in 30% of patients (1), bromin 1 (NF1), a tumor suppressor that functions as a and the standard treatment of wide resection and radia- negative regulator of the Ras pathway. Although the loss tion carries considerable morbidity among survivors. of NF1 is well characterized in malignant peripheral nerve sheath tumors (MPNST), NF1 mutations in other soft- tissue sarcomas have only been recently identified. Using genomic sequencing and copy number analysis, NF1 Authors' Affiliations: Departments of 1Radiation Oncology, 2Pharmacol- 3 4 mutations were identified in myxofibrosarcoma (10.5%; ogy and Cancer Biology, Orthopaedic Surgery, and Pathology, Duke ¼ ¼ University Medical Center, Durham, North Carolina; 5The Broad Institute of n 4 of 38) and pleomorphic liposarcoma (8%; n 2 of 24) Harvard and MIT, Cambridge; 6Department of Medical Oncology, and (3). Similar studies have identified genomic loss of the NF1 7Center for Cancer Discovery, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts in 15% of pediatric embryonal rhabdomyosarcomas (n ¼ 4 of 26; ref. 4). This deletion was mutually exclusive Note: Supplementary data for this article are available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/). with Ras gene mutations. The identification of novel mutations in sarcomas with complex karyotypes provides Current address for J. Barretina: Novartis Institutes for Biomedical new opportunities to model human sarcomas and to Research, Cambridge, MA 02139; current address for L. Dodd, Department of Pathology and Laboratory Medicine, University of North Carolina at investigate new therapies for this difficult to treat disease. Chapel Hill, Chapel Hill, NC 27599. In addition to identifying NF1 mutations in spontane- Corresponding Author: David G. Kirsch, Duke University Medical Center, ous sarcomas in the general population, patients with Box 91006, Durham, NC 27708. Phone: 919-681-8605; Fax: 919-681- neurofibromatosis type 1 (NF1) are at increased risk for 1867; E-mail: [email protected] developing aggressive soft-tissue sarcomas. NF1 affects 1 doi: 10.1158/1535-7163.MCT-13-0189 in 3,000 live births and is caused by germline inactivation 2013 American Association for Cancer Research. of a single allele of the NF1 gene (5). The most common

1906 Mol Cancer Ther; 12(9) September 2013

Downloaded from mct.aacrjournals.org on September 26, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst July 15, 2013; DOI: 10.1158/1535-7163.MCT-13-0189

A Novel Mouse Model of NF1-Deleted Soft-Tissue Sarcoma

sarcoma afflicting patients with NF1 is MPNST, with a Importantly, this model is able to directly assess the lifetime risk of 8% to 13% (5). Patients with NF1 are also at effects of MEK inhibition on native tumor stroma, as risk for developing non-neurogenic sarcomas, including these primary tumors develop in immunocompetent rhabdomyosarcoma and undifferentiated pleomorphic mice with native vasculature. By showing the ability sarcoma (UPS), with a 3% to 6% lifetime risk (6–8). Most of these tumors to respond to MEK inhibition, we lay a malignant NF1-deleted tumors have inactivation of either foundation for future studies using this model to test the or Ink4a/Arf tumor suppressor genes, with more efficacy of other drugs for NF1-deficient sarcomas in the than 50% of MPNSTs expressing wild-type p53 and homo- preclinical setting. zygous deletion of Ink4a/Arf (9). Deletion of NF1 alone is not sufficient to cause high-grade sarcomas, as tissue- Materials and Methods specific deletion of NF1 with either Krox20-Cre or Mouse sarcoma model Dhh-Cre results in neurofibromas (10, 11). Similarly, dele- All mouse work was conducted in accordance with tion of either Ink4a or Arf alone is not sufficient for Duke University Institutional Animal Care and Use formation of neurofibromas or MPNSTs on an NF1 het- Committee approved protocols. The NF1flox/flox mice þ erozygous background from either NF1 / ; Ink4a / (11) and Ink4a/Arfflox/flox mice (26) have been previously þ mice (12) or NF1 / ; Arf / mice (13). In addition, we reported. Primers sequences used to show recombination have previously reported that conditional deletion of of the appropriate alleles are found in Supplementary Ink4a/Arf alone by adenovirus-expressing Cre recombi- Table S1 or have been previously reported (11). Tumors nase (Ad-Cre) injection is insufficient for sarcoma initia- were generated by injection of Ad-Cre (University of tion (14). Deletion of both Ink4a and Arf in an NF1 Iowa, Iowa City, IA; Vector core) into the lower left leg of þ heterozygous background (NF1 / ; Ink4a/Arf / ) gen- NF1flox/flox; Ink4a/Arfflox/flox compound mice as erated sporadic MPNSTs in 26% of mice (12). The most previously described (14). For sciatic nerve injections, the widely used NF1-dependent mouse sarcoma model is the nerve was exposed by an incision in the surrounding þ þ NF1 / ; p53 / cis mouse, which develops NF1- and p53- muscle, followed by injection of 25 mL Ad-Cre into the null sarcomas throughout the body following LOH (15, nerve. Tumor volumes were calculated using the formula 16). Despite the prevalence of NF1 mutations in soft-tissue V ¼ (p L W H)/6, with L, W, and H representing sarcomas, the role of NF1 in the development of soft- the length, width, and height of the tumor in mm, tissue sarcomas, particularly in the background of wild- respectively. type p53 alleles, has not been fully explored in a primary mouse model of soft-tissue sarcoma. Therefore, we de- Histology and immunostaining signed a study to test whether deletion of both NF1 and for immunohistochemistry included: S100 Ink4a/Arfcould generate high-grade soft-tissue sarcomas (DakoCytomation), MyoD1 (DakoCytomation), pERK in an NF1 wild-type background. (Cell Signaling Technology), pS6 (Cell Signaling Technol- Genetically engineered mouse models have greatly ogy), Ki67 (BD Pharmagen), CD31 (BD Pharmagen), advanced our understanding of sarcomas (14, 15, 17–19) pVEGFR-2 (Cell Signaling Technology), and MECA-32 and have served as a preclinical platform for therapies (BD Pharmagen). All immunostaining was conducted (20–25). However, for many models, mice develop sar- with citrate-based antigen retrieval, with the exception comasatmultiplesites,whichcanmaketumordetec- of MECA-32 and pVEGFR2, which were conducted with tion and assessment of treatment response challenging. EDTA-based retrieval. Terminal deoxynucleotidyl trans- Here, we developed a novel mouse model of temporally ferase–mediated dUTP nick end labeling (TUNEL) stain- and spatially restricted NF1-deleted sarcoma to increase ing used the In Situ Cell Death Detection Kit, TMR Red our understanding of NF1-mutant tumors. Using Cre- (Sigma), as per the manufacturer’s instructions. To visu- loxP technology, tumors are initiated through Cre- alize mast cells, slides were stained with toluidine blue mediated deletion of conditional NF1 and Ink4a/Arf solution (0.02% toluidine blue in 1% NaCl, pH 2.2) for 2 floxed alleles (NF1flox/flox;Ink4a/Arfflox/flox). These mice minutes, followed by two washes in distilled water and develop both high-grade myogenic sarcomas and three washes in 100% ethanol. MPNST-like tumors, based on the site of Cre injection. These tumors reflect the histologic properties and spec- PD325901 inhibitor treatment trum of sarcomas found in patients. Because sarcomas Treatment started when tumors were between 200 and in this model develop in a spatially and temporally 300 mm3. Mice receiving vehicle alone were treated with restricted manner, this modelisparticularlyusefulfor NMP 10% (1-methyl-2-pyrrolidone)/PEG300 90% (Fluka) testing therapeutic drug responses, and we have used orally 5 days per week. PD325901 (ref. 27; Sigma) was this model as a preclinical platform to assess new dissolved in NMP 10%/PEG300 90%. Mice were treated therapies for patients with NF1-deleted sarcomas. We with 5 mg/kg of PD325901 orally 5 days per week from the have identified mitogen-activated protein kinase first day of the treatment. Treatment for all mice continued (MAPK) inhibition as a promising therapy that slows until tumors regressed or it was necessary to sacrifice the the growth of primary mouse sarcomas through effects mouse for animal welfare concerns (i.e., animal was mor- on tumor cell growth and the surrounding vasculature. ibund; tumor volume reached 2,000 mm3).

www.aacrjournals.org Mol Cancer Ther; 12(9) September 2013 1907

Downloaded from mct.aacrjournals.org on September 26, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst July 15, 2013; DOI: 10.1158/1535-7163.MCT-13-0189

Dodd et al.

Tumor growth analysis were transferred to polyvinylidene difluoride (PVDF) Criteria for defining treatment response include both by wet transfer. volumetric analysis and a delay in tumor growth kine- tics. A partial response (PR) is classified as a tumor that: Quantitative RT-PCR (i) shows a decrease in tumor volume from baseline RNA was isolated from tumors and cells by TRIzol. measurements and (ii) shows delayed growth kinetics cDNA synthesis was conducted with iScript (Bio-Rad). (defined as requiring more than 10 days to increase in qRT-PCR was conducted on an iQ5 instrument (Bio-Rad) DD volume 150%). For waterfall plot analyses, percentage of using the Ct method. Primer sequences can be found in maximal change is reported as the greatest percentage of Supplementary Table S2. loss in tumor volume from baseline for tumors respond- ing to treatment. Tumors that did not respond to treat- Results ment are shown as greatest percentage of gain in tumor Generating inducible NF1-deleted sarcomas in the volume from baseline over the course of the treatment. In mouse this presentation of the data, change in tumor volume is As NF1 mutations have been recently detected in a capped at more than 100%. variety of human soft-tissue sarcoma subtypes (3, 4), we sought to determine whether loss of NF1 and the tumor Statistical analysis suppressor Ink4a/Arf was sufficient to generate soft-tis- Graphs and statistics were conducted in GraphPad 4.0. sue sarcomas in a p53- and NF1-wild-type background. A nonparametric Student t test was conducted to deter- Furthermore, we wished to generate an inducible mouse mine differences between treatment groups. A P value of model of NF1-deleted sarcoma that could reflect the less than 0.05 was considered statistically significant. diverse spectrum of NF1-associated sarcomas found in patients. Therefore, we generated mice with conditional In vitro studies mutations in both NF1 and Ink4a/Arf(NF1flox/flox; Ink4a/ The mouse sarcoma cell lines 1863 and 3017 were Arfflox/flox). These mice were injected with Ad-Cre at one derived from primary sarcomas in NF1flox/flox;Ink4a/ of two sites: intramuscular injections to model tumors Arfflox/flox mice that developed after intramuscular Ad- arising in the skeletal muscle, and sciatic nerve injections Cre injection. These cell lines were authenticated by to model tumors arising in the nerve sheath. Mice were PCR genotyping. The cells were cultured in Dulbecco’s injected into either the hind limb muscle (intramuscular; modified Eagle medium (DMEM) þ 10% FBS. For col- n ¼ 10) or sciatic nerve (n ¼ 12) of the lower left leg. ony formation studies, cells were seeded at 500/well in Following injection, Cre recombinase can delete both a 6-well dish and treated with dimethyl sulfoxide alleles of NF1 and Ink4a/Arf (Fig. 1A). Mice developed (DMSO) or 50 nmol/L PD325901 (Sigma) for 3 to 7 tumors 3 to 12 months after Cre exposure at the site of days. The number of existing colonies was determined injection. We successfully derived two cell lines (1863 by crystal violet staining. Each experiment was carried and 3017) from sarcomas generated after intramuscular out in triplicate, and the experiment was repeated three injection of Ad-Cre. Using these sarcoma cell lines, we times. Data shown are representative of one indepen- have confirmed recombination of both alleles of NF1 and dent experiment. For colony formation studies with Ink4a/Arf by PCR (Fig. 1B), in addition to loss of both prior PD325901 treatment, cells were cultured overnight mRNA and protein for , p19, and NF1 (Supplementary in 50 nmol/L PD325901, followed by splitting the cells Fig. S1A–S1D). For clarity, we refer to sarcomas and cell into DMSO control or PD325901 for colony formation. lines that are deleted for NF1, p16, and p19 as "NF1- Experiments were analyzed as described previously. deleted." These cell lines maintained an intact p53-depen- Cell proliferation assays were conducted with cells dent DNA damage response, as shown by elevation of cultured in either DMSO or 50 nmol/L PD325901, with in response to doxorubicin treatment (Supplementary the number of cell doublings calculated daily. For Fig. S1E). We observed that the site of injection influences quantitative real-time PCR (qRT-PCR) analyses, cells tumor initiation because sciatic nerve–derived tumors were treated with 50 nmol/L PD325901 for 4 hours, developed more quickly than intramuscularly derived followed by RNA harvest into TRIzol. Data shown tumors (mean time to tumor, 4.1 vs. 6.2 months; P < represent the average of four independent experiments. 0.0001; Fig. 1C). Injection of Ad-Cre at both sites resulted in large, ovoid tumors that appear well vascularized Western blot analysis (Fig. 1D). 1863 cells were treated with 50 nmol/L PD325901 for Histopathologic analysis determined that these tumors either 1 or 4 hours before harvest. Cells were washed reflect the histologic properties and spectrum of disease once with cold PBS (Sigma) and lysed for 10 minutes seen in human NF1-associated sarcomas (Fig. 1E). Sciatic on ice with radioimmunoprecipitation assay (RIPA) nerve injections result in tumors with a fascicular pattern buffer (Sigma), supplemented with phosphatase inhi- of tightly packed spindle cells surrounding the nerve bitors (Sigma; P5726 and P0044). Protein concentration (Supplementary Fig. S2A). As seen in other mouse models was conducted with BCA Protein Concentration Assay of nerve sheath tumors (10, 22), these sarcomas show focal (Thermo Scientific). MiniProtean TGX gels (Bio-Rad) positivity for S100b, but do not show nuclear expression

1908 Mol Cancer Ther; 12(9) September 2013 Molecular Cancer Therapeutics

Downloaded from mct.aacrjournals.org on September 26, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst July 15, 2013; DOI: 10.1158/1535-7163.MCT-13-0189

A Novel Mouse Model of NF1-Deleted Soft-Tissue Sarcoma

Figure 1. Characterization of a B 1863 3017 mouse model of NF1-deleted soft- A Tail Cells Tail Cells tissue sarcoma. A, schematic of NF1 active NF1 flox NF1 and Ink4a/Arf deletion loxPNF1 loxP loxP Ink4a/Arf loxP Ink4a/Arf active NF1 Δ following injection of Ad-Cre. B, PCR showing recombination of Inject Cre both NF1 and Ink4a/Arf floxed NF1 lost Ink4a/Arf Δ loxP alleles in samples from paired tails Ink4a/Arf lost loxP Ink4a/Arf flox and NF1-deleted sarcoma cell lines, derived from tumors CD generated by intramuscular (IM) 100 injection of Ad-Cre. C, Kaplan– SN (n = 12) Meier curve of tumor development IM (n = 10) based on site of orthotopic injection. The average time for 50 P < 0.0001 tumor development is 4.1 months for mice injected in the sciatic nerve (SN) and 6.2 months for mice injected intramuscularly. D, survival Tumor-free photographs of NF1-deleted 0 SN IM 0 100 200 300 tumors generated from injections Days postinjection into the sciatic nerve (left) or muscle (right). E, histopathology of E intramuscular and sciatic nerve H&E S100 MyoD pERK pS6 tumors. Intramuscular tumors resemble high-grade myogenic sarcomas and stain focally for

MyoD1, but not S100. Sciatic nerve Muscle tumors resemble MPNSTs and stain focally for S100, but not for MyoD1. Both tumors are positive for pERK and pS6, indicating activity of the MAPK and mTOR pathways, respectively. H&E, Nerve hematoxylin and eosin.

for the myogenic-lineage marker MyoD1. In contrast, Importantly, these primary tumors develop from a intramuscular injections generate a spectrum of high- small population of initiating tumor cells within the grade spindle cell sarcomas, including rhabdomyosarco- native tumor stroma, facilitating the study of endogenous ma (n ¼ 5 of 10) and UPS (n ¼ 5 of 10), the most common immune cells, cellular growth, and native vasculature in a and aggressive form of soft tissue sarcoma in adults primary tumor context. Thus, we further characterized (Supplementary Fig. S2B). Both rhabdomyosarcoma and the similarities and differences between the sarcoma sub- UPS occur in patients with NF1 (6–8), and NF1 mutations types based on the site of Ad-Cre injection using histo- have been identified in these sarcoma subtypes. Similar to chemical analyses to examine mast cell infiltration, cell other mouse models of myogenic sarcoma and their proliferation, and vascularization. First, we examined human tumor subtypes (14, 28), the intramuscular tumors mast cells, granule-rich inflammatory cells that reside express the myogenic marker MyoD1, but do not stain within the connective tissue and are enriched in NF1- focally for S100b. NF1-associated sarcomas in patients deleted neurofibromas and MPNSTs in patients (31). have active RAS/MAPK/ERK and PI3K/AKT/mTOR Infiltrating mast cells are important stromal factors that signaling (29, 30). Therefore, we hypothesized that the contribute to tumor formation in mouse models of NF1- MEK target extracellular signal–regulated kinase (ERK) deleted neurofibromas (22). In sarcomas from NF1flox/flox; and the mTOR target S6 would be phosphorylated in these Ink4a/Arfflox/flox mice, toluidine blue staining shows sig- primary NF1-deleted mouse sarcomas. Immunohisto- nificant infiltration of mast cells into both the sciatic nerve chemical analysis for pERK and pS6 reveals that MAPK and intramuscular tumors. Mast cells were present at and mTOR pathways are active in both sciatic nerve similar levels in both orthotopic injection models (Fig. and intramuscular primary tumors from NF1flox/flox; 2A). Ki67 staining shows a high level of proliferation in Ink4a/Arfflox/flox mice (Fig. 1E). Taken together, these data both intramuscular and sciatic nerve tumors. Quantifica- þ suggest that this mouse model reflects the spectrum of tion of Ki67-positive (Ki67 ) nuclei shows that intramus- NF1-deleted sarcomas found in the human population, cular tumors have a higher proliferative index than sciatic and that distinct sarcoma subtypes can be generated from nerve tumors (P ¼ 0.04), although there is a wide range of the same mouse strain by Ad-Cre injection into different proliferative index between individual tumors within orthotopic sites. each subtype (Fig. 2B). In both tumor types, average

www.aacrjournals.org Mol Cancer Ther; 12(9) September 2013 1909

Downloaded from mct.aacrjournals.org on September 26, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst July 15, 2013; DOI: 10.1158/1535-7163.MCT-13-0189

Dodd et al.

A Mast cells 30 IM SN 20

10 Figure 2. Comparison of NF1- deleted sarcomas initiated by intramuscular (IM) or intrasciatic 0 Toluidine blue cells per HPF nerve (SN) injection of Ad-Cre. A, IM SN mast cells, visualized by toluidine blue, infiltrate both intramuscular B P < 0.05 and sciatic nerve tumors at similar Ki67+ cells levels. Quantification represents 50 IM SN the average number of cells per 20 field, with 10 fields counted 40 þ

nuclei per tumor. B, Ki67 proliferative + index is higher in intramuscular 30 tumors compared with sciatic nerve tumors (P < 0.05). 20 Quantification represents the percentage of Ki67þ cells 10 normalized to total nuclei per 40 field, with 6 fields counted per

Percentage of Ki67 0 tumor. C, microvessel density, IM SN visualized by CD31þ cells, is similar between intramuscular and sciatic nerve tumors. Quantification C + 800 CD31 cells represents the average number of fi fi IM SN cells per 20 eld, with 6 elds 600 counted per tumor. HPF, high- power field.

events 400 +

CD31 200

0 IM SN

þ CD31-positive (CD31 ) microvessel density is similar, NF1-deleted myogenic sarcomas would respond to although vessel distribution between individual tumors MAPK inhibition based on two lines of evidence: (i) the varies considerably (Fig. 2C). Taken together, these data role of NF1 as a negative regulator of Ras/MAPK/ERK suggest that while this mouse model consistently devel- signaling, and (ii) the elevation of pERK in myogenic sar- ops tumors with distinct histologic properties, there is comas generated from the NF1flox/flox; Ink4a/Arfflox/flox individual tumor-to-tumor heterogeneity in mice. To determine the effect of MEK inhibition on cell such as mast cell infiltration, proliferation, and vascular- proliferation in vitro, we generated cell lines (1863 and ity, with intramuscular tumors showing higher Ki67 3017) from NF1-deleted sarcomas that developed follow- levels. As human NF1-associated sarcomas also display ing intramuscular injection of Ad-Cre into NF1flox/flox; heterogeneity by histologic analysis (7), we hypothesize Ink4a/Arfflox/flox mice. We treated the cells with the MEK that the heterogeneity observed in this mouse model will inhibitor PD325901 and observed decreases in colony- be useful for testing therapies directed against NF1-delet- forming units and cell proliferation following exposure ed sarcoma in the preclinical setting. to the drug (Fig. 3A and Supplementary Fig. S3A). Similar results were seen when cells were treated with PD325901 Efficacy of MEK inhibition in NF1-deleted sarcomas before seeding for colony formation to account for cell– The identification of NF1 mutations in myogenic sar- cell interaction signals and growth factors that are usually coma (3, 4) provides a new foundation for preclinical present in subconfluent conditions and in the tumor studies examining targeted therapies for soft-tissue sar- microenvironment (Supplementary Fig. S3B). Western comas. To determine the use of this novel mouse model in blot analysis of 1863 cells treated with PD325901 for 4 examining therapeutic response, we hypothesized that hours showed a decrease in phosphorylation of ERK

1910 Mol Cancer Ther; 12(9) September 2013 Molecular Cancer Therapeutics

Downloaded from mct.aacrjournals.org on September 26, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst July 15, 2013; DOI: 10.1158/1535-7163.MCT-13-0189

A Novel Mouse Model of NF1-Deleted Soft-Tissue Sarcoma

ABP Figure 3. Activity of a MEK inhibitor 400 = 0.019 P = 0.0124 in NF1-deleted sarcomas. A, NF1- deleted cell lines (1863 and 3017) 300 Hours after PD325901 were exposed to either DMSO- 014 alone or the MEK inhibitor PD325901 (50 nmol/L) for 7 days in 200 p-ERK culture. The number of colonies formed after 7 days was Total ERK determined by crystal violet 100 staining. B, Western blot analysis Colonyforming units showing inhibition of pERK levels 0 following 4-hour exposure of 1863 DMSO PD325901 DMSO PD325901 cells to MEK inhibitor PD325901 1863 3017 (50 nmol/L). C, primary high-grade C D myogenic sarcomas generated 100 4 from intramuscular injection of Ad- PD325901 fl fl fl fl 80 Cre into NF1 ox/ ox; Ink4a/Arf ox/ ox Vehicle mice were treated with vehicle- 60 alone or PD325901. Exposure to 3 40 PD325901 slowed tumor growth in majority of tumors, as shown by 20 the fold change in tumor volume 2 0 following initiation of treatment. D, –20 a waterfall plot shows the absolute change in tumor volume for primary –40 NF1-deleted myogenic sarcomas 1 Fold change in volume –60 following vehicle or PD325901 treatment. For tumors that showed maximal change Percentage –80 a PR to treatment, the maximal 0 –100 percentage of loss in tumor volume 0 5 10 15 20 25 30 is reported. For tumors that did not Days of treatment Vehicle PD325901 respond to treatment, the maximal percentage of gain in E Vehicle Responder Nonresponder tumor volume is reported. E, immunohistochemistry of pERK and pS6 levels in primary myogenic sarcomas that received vehicle- pERK alone or PD325901. Tumors that responded to treatment show a decrease in pERK and pS6 levels compared with vehicle-alone tumors, whereas tumors that did not respond to treatment show a pS6 corresponding upregulation in both pERK and pS6 levels.

(Fig. 3B), with corresponding changes in phospho-S6 and ity of the mice, with a PR rate of 75% (Fig. 3C). Among the phospho-AKT levels (Supplementary Fig. S4). tumors that responded to treatment, the degree of To extend these studies to the primary mouse model of response varied, which has been observed in other pri- NF1-deleted myogenic sarcoma, we compared mice mary sarcoma models (20, 32). The range of treatment receiving PD325901 treatment (n ¼ 8) to mice receiving response for individual tumors during PD325901 expo- vehicle alone (n ¼ 5). Treatment began when tumors sure is similar to the individual tumor-to-tumor hetero- reached 200 to 300 mm3, with mice receiving treatment geneity observed in Ki67 and CD31 staining (Fig. 2). five times per week. Data are presented as both absolute Immunohistochemical analysis reveals that tumors tumor volume and in a waterfall plot that display maximal responding to treatment have lower levels of MAPK and percentage of change in volume for each tumor following mTOR signaling compared with vehicle-treated tumors, treatment (Fig. 3C and D). For the waterfall plots, tumors as determined by a decrease in pERK and pS6 levels (Fig. that partially responded to treatment are shown as great- 3E). In contrast, tumors that did not respond to treatment est percentage of loss in tumor volume. Tumors that did showed hyperelevated levels of both pERK and pS6, not respond to treatment are shown as greatest percentage suggesting that these tumors escaped from treatment by of gain in tumor volume during treatment. As predicted, alternative activation of these pathways. These results treatment with PD325901 delayed tumor growth in major- show that MEK inhibition slows growth of NF1-deleted

www.aacrjournals.org Mol Cancer Ther; 12(9) September 2013 1911

Downloaded from mct.aacrjournals.org on September 26, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst July 15, 2013; DOI: 10.1158/1535-7163.MCT-13-0189

Dodd et al.

rhabdomyosarcoma and UPS, as the histologies for each showing that the cell-cycle effects are cell autonomous of these high-grade sarcoma subtype were evenly distrib- (Fig. 4D and Supplementary Fig. S5). Taken together, uted between vehicle-treated, responders, and nonre- these data suggest that PD325901 inhibits NF1-deleted sponding tumors. sarcomas through cytostatic, but not cytotoxic, mechan- To determine the mechanism of tumor control pro- isms. Very recently, others have used PD325901 to treat duced by MEK inhibition, we examined the Ki67 index neurofibromatosis-associated diseases (34, 35), and treat- between vehicle and PD325901-treated tumors (Fig. 4A). ment of a primary neurofibroma model with PD325901 Tumors that responded to treatment show a decrease in resulted in decreased proliferation as measured by Ki67 proliferative index determined by Ki67 staining (P < 0.05). staining, but not an increase in apoptosis (36). However, it Of note, the two nonresponding tumors have Ki67 levels is important to note that both the cellular context of the similar to vehicle-treated tumors, despite exposure to the treated cancer and the specific target that is inhibited inhibitor. TUNEL staining shows that PD325901 treat- determine the effect of an agent to work in cytotoxic or ment did not significantly increase apoptosis, although cytostatic contexts. nonresponders paradoxically show a high number of þ TUNEL-positive (TUNEL ) cells (Fig. 4B). Tumors that MEK inhibition alters the tumor microenvironment responded to PD325901 treatment exhibit a significant The effect of targeted therapies on the surrounding decrease in levels of cyclin D1 mRNA (P < 0.05), support- tumor stroma is an important component of therapeutic ing previous reports that suggest inhibition of the cell efficacy. Recently, the MEK inhibitor PD325901 was cycle contributes to the decrease in tumor cell prolifera- shown to inhibit angiogenesis and VEGF signaling in tion observed with this agent (ref. 33; Fig. 4C). Similar xenografted tumor models of renal cell cancer (RCC) and downregulation of cyclin D1 mRNA is observed during in lung cancer (37, 38). Although these xenograft models vitro exposure of NF1-deleted sarcoma cells to PD325901, provide valuable information about tumor response, we

* Figure 4. NF1-deficient sarcomas A respond to MEK inhibition by 50 þ cytostatic mechanisms. A, Ki67 Ki67+ cells 40 proliferative index is lower in

nuclei tumors responding to PD325901 Vehicle PD325901 Nonresponder + 30 treatment than in vehicle-alone– treated tumors (, P < 0.05). Tumors 20 that did not respond to treatment (nonresponders, NR) show Ki67þ 10 levels similar to vehicle-alone– treated tumors. Quantification

Percentage of Ki67 Percentage þ 0 represents the percentage of Ki67 Vehicle PD325901 NR cells normalized to total nuclei per fi fi B 40 40 eld, with 6 elds counted per TUNEL+ cells tumor. B, TUNEL staining determined that the number of nuclei + 30 apoptotic nuclei is not statistically Vehicle PD325901 Nonresponder different between vehicle-alone 20 and PD325901-treated tumors. Tumors that did not respond to treatment (nonresponders) show 10 an upregulation in TUNELþ cells. Quantification represents the þ Percentage of TUNEL of Percentage percentage of TUNEL cells 0 Vehicle PD325901 NR normalized to total nuclei per 40 field, with 6 fields counted C D Cell lines per tumor. C, qRT-PCR shows downregulation of cyclin D1 2.0 Tumors * * mRNA in NF1-deleted myogenic 1.0 sarcomas responding to 1.5 PD325901 treatment (n ¼ 4) in comparison with vehicle-alone– treated tumors (n ¼ 4; , P < 0.05). 1.0 * 0.5 D, qRT-PCR shows downregulation of cyclin D1 mRNA 0.5 in sarcoma cell lines following a Cyclin D1 mRNA Cyclin D1 mRNA 4-hour treatment with 50 nmol/L P < 0.0 0.0 of PD325901 ( , 0.05). Data Vehicle PD325901 Vehicle PD325901Vehicle PD325901 represent an average of four 1863 3017 independent experiments.

1912 Mol Cancer Ther; 12(9) September 2013 Molecular Cancer Therapeutics

Downloaded from mct.aacrjournals.org on September 26, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst July 15, 2013; DOI: 10.1158/1535-7163.MCT-13-0189

A Novel Mouse Model of NF1-Deleted Soft-Tissue Sarcoma

sought to extend these findings to our NF1-deleted sar- mary tumors treated with PD325901 (Fig. 5D). Upon coma model to determine the effect of MEK inhibition on VEGF binding, the VEGFR-2 on endothe- the tumor stroma of primary immunocompetent tumors lial cells is phosphorylated, activating the VEGF angio- with native vasculature. First, we examined the effect of genesis signaling cascade (45). In vehicle-treated tumors, MEK inhibition on mast cell infiltration. Elevated levels of pVEGFR-2 foci are abundant and colocalize with endo- mast cells correlate with tumor aggressiveness in NF1- thelial cells, visualized by MECA-32 staining. In contrast, mutant neurofibromas (22). The role of mast cells follow- PD325901-treated tumors show marked decrease in ing targeted therapy has not been examined in a primary pVEGFR-2 foci, along with decay of endothelial cell archi- sarcoma model, and reports of their prognostic or sup- tecture. These data suggest that in addition to decreasing portive role in other cancer types are conflicting (39–42). In proliferation of tumor cells, MEK inhibition has important our NF1-deleted sarcomas, toluidine blue staining shows effects on the surrounding tumor microenvironment by that tumors treated with either vehicle alone or PD325901 blocking angiogenesis and decreasing tumor microvessel have similar levels of mast cell infiltration (Fig. 5A). Thus, density. Taken together, these preclinical data suggest mast cell levels were not altered by MEK inhibition in this that the MEK inhibitor PD325901 may be a useful therapy model. for patients with NF1-deficient sarcomas. In contrast, MEK inhibition results in a significant þ decrease in CD31 microvessel density (P ¼ 0.010; Fig. Discussion 5B). As similar effects on the endothelium have been We have described a novel genetically engineered observed with MEK inhibition in a primary model of inducible mouse model of NF1-deficient sarcomas. This neurofibroma (36), we further characterized the molecular primary mouse model can be used to study the tumor reprogramming responsible for vascular changes in our biology and therapeutic response of sarcomas with NF1 primary high-grade sarcomas. To determine the changes mutation. Using temporally and spatially controlled con- in several angiogenic markers that may correlate with the ditional gene deletion, we have determined that inactiva- decreased endothelial cell content in the tumors, we con- tion of NF1 and Ink4a/Arfin the tumor cells is sufficient to ducted qRT-PCR on vehicle- and PD325901-treated initiate both myogenic and MPNST-like sarcomas in mice tumors. Expression of the mRNA for the VEGFa isoforms with a wild-type stroma. High-grade myogenic sarcomas VEGF 120 and VEGF 165 was decreased in treated tumors, develop after intramuscular injection of Ad-Cre, and whereas message levels for the VEGF 188 isoform were MPNST-like sarcomas develop after sciatic nerve injec- not significantly altered (Fig. 5C). Levels of Angiopoietin 2 tion. Histopathologic analysis shows that the spectrum of (Angpt 2) mRNA were lower in treated tumors, but levels sarcomas that develop in this model are similar to sarco- of other endothelial cell markers were unaffected, includ- mas with NF1 deletion in the patient population. By ing Angiopoietin 1. In addition, mRNA levels of other initiating sarcomas through deletion of NF1 and Ink4a/ angiogenesis-related genes were unchanged, including Arf, these mice model the approximately 50% of NF1- Tie2, VegfR-1, VegfR-2, Pdgf-b, PdgfR, Neuropilin, Hif1a, deleted sarcomas with wild-type p53 and complement the and Hif2a (Supplementary Fig. S6). Angpt 2 can have existing models of p53-deleted NF1-mutant sarcomas for proangiogenic properties when combined with increased therapeutic studies (21). In this model, tumor growth is VEGFa expression (43). Therefore, decreased levels of delayed by the MEK inhibitor PD325901, which is accom- Angpt2, VEGF 120, and VEGF 165 mRNA suggest a panied by a decrease in tumor cell proliferation but does potential mechanism for the decreased vascularity seen not increase cellular apoptosis. Importantly, in this model with PD325901 exposure. To determine whether the tumors develop within their native microenvironment, change in angiogenic genes following PD325901 treat- facilitating the study of stromal contribution to therapeu- ment was primarily through action on the tumor cells or tic response, which may be more challenging in cell on tumor stroma, we treated NF1-deficient sarcoma cells transplant or xenograft studies. PD325901 treatment with PD325901. Following a 4-hour treatment with the decreases microvessel density, at least partially through inhibitor, both cell lines exhibited decreased levels of a decrease in expression of proangiogenic genes, such as VEGF 120 and VEGF 165 mRNA, similar to the primary VEGF 120, VEGF 165, and Angpt 2. Therefore, this study tumors (Fig. 5C). However, levels of Angpt 2 remain shows the ability of this system to model NF1-deleted unchanged in the PD325901-treated tumor cells. These sarcomas and shows its use in characterizing targeted data suggest that some of the decrease in VEGF mRNA therapies for NF1-deficient sarcomas. Several recent stud- levels following PD325901 treatment occurs in the sarco- ies have also reported encouraging results using MEK ma cells, but the changes in Angpt 2 levels observed in inhibition to treat other neurofibromin-deficient disorders intact tumors reflect changes in the surrounding tumor in mice (34–36). stroma, perhaps in response to altered VEGF levels in the To our knowledge, this is the first study to examine the tumor cells (44). biology of NF1-deleted high-grade myogenic sarcomas in To determine whether the decreased levels in VEGF genetically engineered mouse models, despite the pre- 120, VEGF 165, and Angpt 2 mRNA in the primary tumors valence of these tumors in the general population and correspond to decreased VEGFa signaling in vivo,we in patients with NF1. Histologic analysis shows that conducted immunofluoresence for pVEGFR-2 in the pri- neither tumor location nor sarcoma subtype influences

www.aacrjournals.org Mol Cancer Ther; 12(9) September 2013 1913

Downloaded from mct.aacrjournals.org on September 26, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst July 15, 2013; DOI: 10.1158/1535-7163.MCT-13-0189

Dodd et al.

A Mast cells 50 Vehicle PD325901 40

30

20

10

Toluidine blue cells per HPF blue Toluidine 0 Vehicle PD325901 NR B CD31+ cells P Vehicle PD325901 600 < 0.01

400

200 Number of vessels

0 C D Vehicle PD325901 NR 3

Tumors 1.5 Cell lines 2 1.0 * 1 * * 0.5 *

Relative mRNA Relative mRNA Relative * * * * 0 0.0 120 165 188 Angpt 1 Angpt 2 120 165 188 Angpt 1 Angpt 2 VEGF isoforms VEGF isoforms E Nuclei pVEGFR-2 MECA-32 Merge Vehicle PD325901

Figure 5. PD325901 alters vessel number and expression of angiogenic genes. A, mast cells, visualized by toluidine blue, infiltrate both vehicle- and PD325901- treated NF1-deleted sarcomas at similar levels. Quantification represents the average number of cells per 20 field, with 10 fields counted per tumor. þ B, microvessel density, visualized by CD31 cells, is significantly decreased in tumors responding to PD325901 treatment (P < 0.010). Quantification represents the average number of cells per 20 field, with 6 fields counted per tumor. C, qRT-PCR for angiogenic genes in primary NF1-deleted myogenic sarcomas treated with vehicle alone (black bars) or PD325901 (shaded bars). Data represent an average of four tumors per group. Levels of VEGF 120, VEGF 165, and Angpt2 mRNA were significantly decreased in tumors responding to PD325901 treatment. D, qRT-PCR for angiogenic genes in sarcoma cell lines (1863-black, 3017-red) following a 4-hour treatment with either vehicle alone (solid bars) or 50 nmol/L of PD325901 (shaded bars). Data represent an average of four independent experiments. Levels of VEGF isoforms [120 and 165 (1863 and 3017), and 188 (3017 only)], but not Angpt2 mRNA, were significantly decreased in cells exposed to PD325901. E, coimmunofluoresence for pVEGFR-2–positive endothelial cells in NF1-deleted myogenic sarcomas. In vehicle-treated tumors, pVEGFR-2 foci (green) are abundant and found in association with multiple endothelial cells (red; MECA-32). However, in PD325901-treated tumors, pVEGFR-2 foci are less abundant and the endothelial cell network is less extensive. NR, nonresponders. , P < 0.05.

the average level of mast cell infiltration or the extent of that they all have identical initiating mutations and rep- vascularization. However, these properties are fairly het- resent similar sarcoma subtypes. We observed a differ- erogeneous between individual tumors, despite the fact ence in Ki67 proliferative index between intramuscular

1914 Mol Cancer Ther; 12(9) September 2013 Molecular Cancer Therapeutics

Downloaded from mct.aacrjournals.org on September 26, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst July 15, 2013; DOI: 10.1158/1535-7163.MCT-13-0189

A Novel Mouse Model of NF1-Deleted Soft-Tissue Sarcoma

and sciatic nerve subtypes, although individual tumors ment in a primary tumor model. Our results support a display a broad range of proliferative capacity within each model in which PD325901 targets both the tumor cell group. This heterogeneity is a salient feature of primary itself and results in secondary effects on the tumor tumor systems (46, 47) that permit modeling of diverse vasculature. Following MEK inhibition, the sarcoma biology and may reflect differences in additional acquired cell decreases both cyclin D1 and VEGFa mRNA levels. somatic mutations and/or the influence of the complex Lower cyclin D1 mRNA levels decrease cell prolifera- microenvironment. tion, as shown by lower Ki67 levels. In parallel, lower Indeed, the heterogeneity observed across primary VEGFa mRNA levels prevent activation of pVEGFR-2 tumors make them excellent models for examining dif- on surrounding endothelial cells, ultimately lowering fering tumor responses to therapy, and this model facil- Angpt 2 mRNA levels in the vasculature. Together with itated examination of tumors that were resistant to decreased VEGFa, this results in decreased microvessel PD325901 treatment. Alternatively, these results can be density. This model is consistent with data from previ- viewed from the perspective that tumors may have suf- ous in vitro and xenograft studies using MEK inhibitors ficient phenotypic variation to account for the failure of in other tumors types (33, 37, 38). Alternatively, it is also treatment in nonresponders. Although this diversity of possible that PD325901 could have direct effects on the response could be viewed as a limitation, we view it as a tumor endothelial cells, which has been reported in vitro strength of the model. Although we are not able to deter- (50, 51). mine whether these tumors possessed a primary resis- In summary, we have developed a novel mouse model tance to MEK inhibition or acquired resistance rapidly of NF1-deleted sarcoma with wild-type p53 status. The following the initial treatment, examining tumors that did tumor spectrum observed after intramuscular and sciatic not respond to PD325901 provides potential mechanisms nerve injections of Ad-Cre is similar to the clinical spec- to circumvent MEK inhibition. First, the resistant tumors trum of NF1-deleted sarcoma. We have used this model to have hyperelevated Ki67 staining, suggesting they were assess the efficacy of PD325901 and to investigate its able to grow through the treatment with increased cellular mechanism of action in a primary tumor model. We proliferation—even when accompanied by increased cel- anticipate that this model will be useful as a preclinical lular apoptosis—resulting in a net gain in tumor growth. platform for other novel therapies and for further studies Alternatively, the increased apoptotic rate in nonrespond- to examine the interplay between the microenvironment ing tumors may be a reflection of a relatively low apoptotic and NF1-deficent sarcoma cells during therapeutic rate of the responding tumors at this time point assayed. It response. is conceivable that tumors that initially responded to therapies have elevated apoptosis during tumor response Disclosure of Potential Conflicts of Interest but have decreased apoptosis during tumor progression. No potential conflicts of interest were disclosed. Secondly, although all six tumors that responded to treat- ment showed low pERK and pS6 levels, the two tumors Authors' Contributions that did not respond to PD325901 treatment showed high Conception and design: R.D. Dodd, J. Barretina, D.G. Kirsch Development of methodology: R.D. Dodd, J.K. Mito, W.C. Eward levels of pERK and pS6, even above levels observed in Acquisition of data (provided animals, acquired and managed patients, vehicle-treated tumors. Thus, treatment may have been provided facilities, etc.): R.D. Dodd, J.K. Mito, W.C. Eward, M. Sachdeva, L. Dodd ineffective due to re-activation of these pathways. These Analysis and interpretation of data (e.g., statistical analysis, biostatis- results imply that blocking mTOR in addition to ERK tics, computational analysis): R.D. Dodd, J.K. Mito, R. Chitalia, L. Dodd, would be a promising therapeutic strategy for future D.G. Kirsch Writing, review, and/or revision of the manuscript: R.D. Dodd, J.K. Mito, studies. Indeed, others have shown that mTOR inhibition W.C. Eward, J. Barretina, L. Dodd, D.G. Kirsch is effective in slowing growth of NF1-deficient MPNSTs in Administrative, technical, or material support (i.e., reporting or orga- mouse models (21, 25, 48). However, we recognize that it nizing data, constructing databases): W.C. Eward, R. Chitalia, Y. Ma can be challenging to define therapeutic windows for combination treatment of molecularly targeted inhibitors Acknowledgments The authors thank Ron Dephino (MD Anderson Cancer Center, Hous- in clinical trials. For example, recent reports have shown ton, TX) for providing the Ink4a/Arf flox mice. The authors also thank that combination of the MEK inhibitor with Laura Jeffords, Rafaela Rodrigues, and Loretta Woodleif for assistance the AKT inhibitor MK-2206 results in increased toxicity with mouse work. when used at single-agent doses (49). Finally, the effect of Grant Support PD325901 on the tumor stroma of nonresponders was not This work was supported by a Children’s Tumor Foundation Young well defined, as changes in the levels of mast cells and Investigator Award (to R.D. Dodd), a Liddy Shriver Sarcoma Initiative microvessel density were varied. Although future studies Grant (to D.G. Kirsch and R.D. Dodd), and K02-AI093866 (to D.G. Kirsch). will be needed to fully explore the mechanisms of resis- The costs of publication of this article were defrayed in part by the tance discussed here, this study shows the use of this payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate mouse model to study the diversity of therapeutic this fact. responses found in NF1-deleted myogenic sarcoma. This study examined the effects of MEK inhibition on Received March 12, 2013; revised June 27, 2013; accepted July 2, 2013; the tumor cells and native surrounding microenviron- published OnlineFirst July 15, 2013.

www.aacrjournals.org Mol Cancer Ther; 12(9) September 2013 1915

Downloaded from mct.aacrjournals.org on September 26, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst July 15, 2013; DOI: 10.1158/1535-7163.MCT-13-0189

Dodd et al.

References 1. Borden EC, Baker LH, Bell RS, Bramwell V, Demetri GD, Eisenberg BL, 23. Abraham J, Nelon LD, Kubicek CB, Kilcoyne A, Hampton ST, Zarzabal et al. Soft tissue sarcomas of adults: state of the translational science. LA, et al. Preclinical testing of erlotinib in a transgenic alveolar rhab- Clin Cancer Res 2003;9:1941–56. domyosarcoma mouse model. Sarcoma 2011;2011:130484. 2. Taylor BS, Barretina J, Maki RG, Antonescu CR, Singer S, Ladanyi M. 24. Wu J, Dombi E, Jousma E, Scott Dunn R, Lindquist D, Schnell BM, et al. Advances in sarcoma genomics and new therapeutic targets. Nat Rev Preclincial testing of and RAD001 in the Nf(flox/flox);DhhCre Cancer 2011;11:541–57. mouse model of plexiform neurofibroma using magnetic resonance 3. Barretina J, Taylor BS, Banerji S, Ramos AH, Lagos-Quintana M, imaging. Pediatr Blood Cancer 2012;58:173–80. Decarolis PL, et al. Subtype-specific genomic alterations define new 25. Ghadimi MP, Lopez G, Torres KE, Belousov R, Young ED, Liu J, et al. targets for soft-tissue sarcoma therapy. Nat Genet 2010;42:715–21. Targeting the PI3K/mTOR axis, alone and in combination with autop- 4. Paulson V, Chandler G, Rakheja D, Galindo RL, Wilson K, Amatruda JF, hagy blockade, for the treatment of malignant peripheral nerve sheath et al. High-resolution array CGH identifies common mechanisms that tumors. Mol Cancer Ther 2012;11:1758–69. drive embryonal rhabdomyosarcoma pathogenesis. Genes Chromo- 26. Aguirre AJ, Bardeesy N, Sinha M, Lopez L, Tuveson DA, Horner J, et al. somes Cancer 2011;50:397–408. Activated Kras and Ink4a/Arf deficiency cooperate to produce meta- 5. Evans DG, Baser ME, McGaughran J, Sharif S, Howard E, Moran A. static pancreatic ductal adenocarcinoma. Genes Dev 2003;17: Malignant peripheral nerve sheath tumours in neurofibromatosis 1. J 3112–26. Med Genet 2002;39:311–4. 27. Brown AP, Carlson TC, Loi CM, Graziano MJ. Pharmacodynamic and 6. Ferrari A, Bisogno G, Macaluso A, Casanova M, D'Angelo P, Pierani P, toxicokinetic evaluation of the novel MEK inhibitor, PD0325901, in the et al. Soft-tissue sarcomas in children and adolescents with neurofi- rat following oral and intravenous administration. Cancer Chemother bromatosis type 1. Cancer 2007;109:1406–12. Pharmacol 2007;59:671–9. 7. Sorensen SA, Mulvihill JJ, Nielsen A. Long-term follow-up of von 28. Rubin BP, Nishijo K, Chen HI, Yi X, Schuetze DP, Pal R, et al. Evidence Recklinghausen neurofibromatosis. Survival and malignant neo- for an unanticipated relationship between undifferentiated pleomor- plasms. N Engl J Med 1986;314:1010–5. phic sarcoma and embryonal rhabdomyosarcoma. Cancer Cell 8. McKeen EA, Bodurtha J, Meadows AT, Douglass EC, Mulvihill JJ. 2011;19:177–91. Rhabdomyosarcoma complicating multiple neurofibromatosis. J 29. Lau N, Feldkamp MM, Roncari L, Loehr AH, Shannon P, Gutmann DH, Pediatr 1978;93:992–3. et al. Loss of neurofibromin is associated with activation of RAS/MAPK 9. Birindelli S, Perrone F, Oggionni M, Lavarino C, Pasini B, Vergani B, and PI3-K/AKT signaling in a neurofibromatosis 1 . J et al. Rb and TP53 pathway alterations in sporadic and NF1-related Neuropathol Exp Neurol 2000;59:759–67. malignant peripheral nerve sheath tumors. Lab Invest 2001;81:833–44. 30. Gregorian C, Nakashima J, Dry SM, Nghiemphu PL, Smith KB, Ao Y, 10. Wu J, Williams JP, Rizvi TA, Kordich JJ, Witte D, Meijer D, et al. et al. PTEN dosage is essential for neurofibroma development and Plexiform and dermal neurofibromas and pigmentation are caused by malignant transformation. Proc Natl Acad Sci U S A 2009;106: Nf1 loss in desert hedgehog-expressing cells. Cancer Cell 19479–84. 2008;13:105–16. 31. Donhuijsen K, Sastry M, Volker B, Leder LD. Mast cell frequency in soft 11. Zhu Y, Romero MI, Ghosh P, Ye Z, Charnay P, Rushing EJ, et al. tissue tumors. Relation to type and grade of malignancy. Pathol Res Ablation of NF1 function in induces abnormal development of Pract 1992;188:61–6. cerebral cortex and reactive gliosis in the brain. Genes Dev 2001; 32. De Raedt T, Walton Z, Yecies JL, Li D, Chen Y, Malone CF, et al. 15:859–76. Exploiting cancer cell vulnerabilities to develop a combination therapy 12. Joseph NM, Mosher JT, Buchstaller J, Snider P, McKeever PE, Lim M, for ras-driven tumors. Cancer Cell 2011;20:400–13. et al. The loss of Nf1 transiently promotes self-renewal but not 33. Halilovic E, She QB, Ye Q, Pagliarini R, Sellers WR, Solit DB, et al. tumorigenesis by neural crest stem cells. Cancer Cell 2008;13:129–40. PIK3CA mutation uncouples tumor growth and cyclin D1 regulation 13. King D, Yang G, Thompson MA, Hiebert SW. Loss of neurofibroma- from MEK/ERK and mutant KRAS signaling. Cancer Res 2010;70: tosis-1 and p19(ARF) cooperate to induce a multiple tumor . 6804–14. 2002;21:4978–82. 34. Staser K, Park SJ, Rhodes SD, Zeng Y, He YZ, Shew MA, et al. Normal 14. Kirsch DG, Dinulescu DM, Miller JB, Grimm J, Santiago PM, Young NP, hematopoiesis and neurofibromin-deficient myeloproliferative disease et al. A spatially and temporally restricted mouse model of soft tissue require Erk. J Clin Invest 2013;123:329–34. sarcoma. Nat Med 2007;13:992–7. 35. Chang T, Krisman K, Theobald EH, Xu J, Akutagawa J, Lauchle JO, 15. Cichowski K, Shih TS, Schmitt E, Santiago S, Reilly K, McLaughlin ME, et al. Sustained MEK inhibition abrogates myeloproliferative disease in et al. Mouse models of tumor development in neurofibromatosis type Nf1 mutant mice. J Clin Invest 2013;123:335–9. 1. Science 1999;286:2172–6. 36. Jessen WJ, Miller SJ, Jousma E, Wu J, Rizvi TA, Brundage ME, et al. 16. Vogel KS, Klesse LJ, Velasco-Miguel S, Meyers K, Rushing EJ, Parada MEK inhibition exhibits efficacy in human and mouse neurofibroma- LF. Mouse tumor model for neurofibromatosis type 1. Science tosis tumors. J Clin Invest 2013;123:340–7. 1999;286:2176–9. 37. TakahashiO,KomakiR,SmithPD,JurgensmeierJM,RyanA, 17. Nishijo K, Chen QR, Zhang L, McCleish AT, Rodriguez A, Cho MJ, et al. Bekele BN, et al. Combined MEK and VEGFR inhibition in orthotopic Credentialing a preclinical mouse model of alveolar rhabdomyosar- human lung cancer models results in enhanced inhibition of tumor coma. Cancer Res 2009;69:2902–11. angiogenesis, growth, and metastasis. Clin Cancer Res 2012;18: 18. Haldar M, Hancock JD, Coffin CM, Lessnick SL, Capecchi MR. A 1641–54. conditional mouse model of synovial sarcoma: insights into a myo- 38. Huang D, Ding Y, Luo WM, Bender S, Qian CN, Kort E, et al. Inhibition of genic origin. Cancer Cell 2007;11:375–88. MAPK kinase signaling pathways suppressed renal cell carcinoma 19. Dodd RD, Mito J, Kirsch DG. Animal models of soft-tissue sarcoma. Dis growth and angiogenesis in vivo. Cancer Res 2008;68:81–8. Model Mech 2010;3:557–66. 39. Tanaka T, Ishikawa H. Mast cells and inflammation-associated colo- 20. Kim S, Dodd RD, Mito JK, Ma Y, Kim Y, Riedel RF, et al. Efficacy of rectal carcinogenesis. Semin Immunopathol 2013;35:245–54. phosphatidylinositol-3 kinase (PI3K) inhibitors in a primary mouse 40. Xia Q, Wu XJ, Zhou Q, Jing Z, Hou JH, Pan ZZ, et al. No relationship model of undifferentiated pleomorphic sarcoma (UPS). Sarcoma between the distribution of mast cells and the survival of stage IIIB 2012;2012:680708. colon cancer patients. J Transl Med 2011;9:88. 21. Johannessen CM, Johnson BW, Williams SM, Chan AW, Reczek EE, 41. Chichlowski M, Westwood GS, Abraham SN, Hale LP. Role of mast Lynch RC, et al. TORC1 is essential for NF1-associated malignancies. cells in inflammatory bowel disease and inflammation-associated Curr Biol 2008;18:56–62. colorectal neoplasia in IL-10–deficient mice. PLoS ONE 2010;5: 22. Yang FC, Ingram DA, Chen S, Zhu Y, Yuan J, Li X, et al. Nf1-dependent e12220. tumors require a microenvironment containing Nf1þ/ and c-- 42. Gulubova M, Vlaykova T. Prognostic significance of mast cell dependent bone marrow. Cell 2008;135:437–48. number and microvascular density for the survival of patients

1916 Mol Cancer Ther; 12(9) September 2013 Molecular Cancer Therapeutics

Downloaded from mct.aacrjournals.org on September 26, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst July 15, 2013; DOI: 10.1158/1535-7163.MCT-13-0189

A Novel Mouse Model of NF1-Deleted Soft-Tissue Sarcoma

with primary colorectal cancer. J Gastroenterol Hepatol 2009;24: 48. Johansson G, Mahller YY, Collins MH, Kim MO, Nobukuni T, Perent- 1265–75. esis J, et al. Effective in vivo targeting of the mammalian target of 43. Lobov IB, Brooks PC, Lang RA. Angiopoietin-2 displays VEGF-depen- rapamycin pathway in malignant peripheral nerve sheath tumors. Mol dent modulation of capillary structure and endothelial cell survival in Cancer Ther 2008;7:1237–45. vivo. Proc Natl Acad Sci U S A 2002;99:11205–10. 49. Speranza RJK, Khin S, Weil MK, Do KT, Horneffer Y, Juwara L, et al. 44. Felcht M, Luck R, Schering A, Seidel P, Srivastava K, Hu J, et al. Pharmacodynamic biomarker-driven trial of MK-2206, and AKT inhib- Angiopoietin-2 differentially regulates angiogenesis through TIE2 and itor, with AZD6244 (selumetinib), a MEK inhibitor, in patients with integrin signaling. J Clin Invest 2012;122:1991–2005. advanced colorectal carcinoma [abstract]. J Clin Oncol 30, 2012 45. Carmeliet P, Jain RK. Molecular mechanisms and clinical applications (suppl; abstr 3529). of angiogenesis. Nature 2011;473:298–307. 50. Vart RJ, Nikitenko LL, Lagos D, Trotter MW, Cannon M, Bourboulia D, 46. Chen Z, Cheng K, Walton Z, Wang Y, Ebi H, Shimamura T, et al. A et al. Kaposi's sarcoma-associated herpesvirus-encoded interleukin- murine lung cancer co- identifies genetic modifiers of 6 and G-protein–coupled receptor regulate angiopoietin-2 expression therapeutic response. Nature 2012;483:613–7. in lymphatic endothelial cells. Cancer Res 2007;67:4042–51. 47. Singh M, Lima A, Molina R, Hamilton P, Clermont AC, Devasthali V, 51. Mavria G, Vercoulen Y, Yeo M, Paterson H, Karasarides M, Marais R, et al. Assessing therapeutic responses in Kras mutant cancers using et al. ERK–MAPK signaling opposes Rho-kinase to promote endothe- genetically engineered mouse models. Nat Biotechnol 2010;28: lial cell survival and sprouting during angiogenesis. Cancer Cell 2006; 585–93. 9:33–44.

www.aacrjournals.org Mol Cancer Ther; 12(9) September 2013 1917

Downloaded from mct.aacrjournals.org on September 26, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst July 15, 2013; DOI: 10.1158/1535-7163.MCT-13-0189

NF1 Deletion Generates Multiple Subtypes of Soft-Tissue Sarcoma That Respond to MEK Inhibition

Rebecca D. Dodd, Jeffrey K. Mito, William C. Eward, et al.

Mol Cancer Ther 2013;12:1906-1917. Published OnlineFirst July 15, 2013.

Updated version Access the most recent version of this article at: doi:10.1158/1535-7163.MCT-13-0189

Supplementary Access the most recent supplemental material at: Material http://mct.aacrjournals.org/content/suppl/2013/07/15/1535-7163.MCT-13-0189.DC1

Cited articles This article cites 50 articles, 16 of which you can access for free at: http://mct.aacrjournals.org/content/12/9/1906.full#ref-list-1

Citing articles This article has been cited by 9 HighWire-hosted articles. Access the articles at: http://mct.aacrjournals.org/content/12/9/1906.full#related-urls

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

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

Permissions To request permission to re-use all or part of this article, use this link http://mct.aacrjournals.org/content/12/9/1906. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from mct.aacrjournals.org on September 26, 2021. © 2013 American Association for Cancer Research.