Skip to main content
  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

AACR logo

  • Register
  • Log in
  • My Cart
Advertisement

Main menu

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Computer Resources
      • Highly Cited Collection
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Early Career Award
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citations
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

User menu

  • Register
  • Log in
  • My Cart

Search

  • Advanced search
Cancer Research
Cancer Research
  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Computer Resources
      • Highly Cited Collection
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Early Career Award
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citations
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

Tumor and Stem Cell Biology

RAS/MAPK Activation Drives Resistance to Smo Inhibition, Metastasis, and Tumor Evolution in Shh Pathway–Dependent Tumors

Xuesong Zhao, Tatyana Ponomaryov, Kimberly J. Ornell, Pengcheng Zhou, Sukriti K. Dabral, Ekaterina Pak, Wei Li, Scott X. Atwood, Ramon J. Whitson, Anne Lynn S. Chang, Jiang Li, Anthony E. Oro, Jennifer A. Chan, Joseph F. Kelleher and Rosalind A. Segal
Xuesong Zhao
1Cancer Biology and Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
2Neurobiology, Harvard Medical School, Boston, Massachusetts.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Tatyana Ponomaryov
1Cancer Biology and Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
2Neurobiology, Harvard Medical School, Boston, Massachusetts.
3University of Birmingham, Centre for Cardiovascular Sciences, College of Medical and Dental Sciences, Edgbaston, Birmingham, United Kingdom.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Kimberly J. Ornell
1Cancer Biology and Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
2Neurobiology, Harvard Medical School, Boston, Massachusetts.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Pengcheng Zhou
1Cancer Biology and Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
2Neurobiology, Harvard Medical School, Boston, Massachusetts.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Sukriti K. Dabral
1Cancer Biology and Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
2Neurobiology, Harvard Medical School, Boston, Massachusetts.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Ekaterina Pak
1Cancer Biology and Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
2Neurobiology, Harvard Medical School, Boston, Massachusetts.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Wei Li
4Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard School of Public Health, Boston, Massachusetts.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Scott X. Atwood
5Program in Epithelial Biology, Stanford University School of Medicine, Stanford, California.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Ramon J. Whitson
5Program in Epithelial Biology, Stanford University School of Medicine, Stanford, California.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Anne Lynn S. Chang
5Program in Epithelial Biology, Stanford University School of Medicine, Stanford, California.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jiang Li
5Program in Epithelial Biology, Stanford University School of Medicine, Stanford, California.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Anthony E. Oro
5Program in Epithelial Biology, Stanford University School of Medicine, Stanford, California.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jennifer A. Chan
6Pathology and Laboratory Medicine, University of Calgary, Calgary, Alberta, Canada.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Joseph F. Kelleher
7Novartis Institutes for Biomedical Research, Cambridge, Massachusetts.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Rosalind A. Segal
1Cancer Biology and Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
2Neurobiology, Harvard Medical School, Boston, Massachusetts.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: rosalind_segal@dfci.harvard.edu
DOI: 10.1158/0008-5472.CAN-14-2999-T Published September 2015
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Aberrant Shh signaling promotes tumor growth in diverse cancers. The importance of Shh signaling is particularly evident in medulloblastoma and basal cell carcinoma (BCC), where inhibitors targeting the Shh pathway component Smoothened (Smo) show great therapeutic promise. However, the emergence of drug resistance limits long-term efficacy, and the mechanisms of resistance remain poorly understood. Using new medulloblastoma models, we identify two distinct paradigms of resistance to Smo inhibition. Sufu mutations lead to maintenance of the Shh pathway in the presence of Smo inhibitors. Alternatively activation of the RAS–MAPK pathway circumvents Shh pathway dependency, drives tumor growth, and enhances metastatic behavior. Strikingly, in BCC patients treated with Smo inhibitor, squamous cell cancers with RAS/MAPK activation emerged from the antecedent BCC tumors. Together, these findings reveal a critical role of the RAS–MAPK pathway in drug resistance and tumor evolution of Shh pathway–dependent tumors. Cancer Res; 75(17); 3623–35. ©2015 AACR.

Introduction

Sonic Hedgehog (Shh) signaling plays a critical role in growth and patterning during development, and aberrant activation of Shh signaling is implicated in several cancers (1). Germline mutations that activate Shh signaling predispose to basal cell carcinoma (BCC) and medulloblastoma in humans and mice, whereas somatic mutations in Shh components are frequently observed in such tumors (2, 3).

Signaling is initiated when Shh, a secreted protein, binds its receptor, Patched (Ptch). In the absence of Shh, Ptch suppresses activity of Smoothened (Smo). When Shh binds Ptch, Smo initiates a signaling cascade, which inactivates the tumor-suppressor Suppressor of Fused (Sufu), and activates Gli transcription factors. Gli target genes include Gli1, Ptch, Nmyc, and Cyclin D1 (Ccnd1). Small-molecule antagonists of Smo, including Vismodegib (GDC-0449; Roche), Sonidegib (LDE225; Novartis), and XL-139 (BMS/Exelixis), provide promising targeted therapy for BCC and medulloblastoma (1, 4–7), and are in clinical trials or in use for these indications.

Despite initial success of Smo inhibitors, long-term efficacy is limited by pre-existing or acquired drug resistance (8, 9). Studies of other cancers indicate that both cell-autonomous mutations and microenvironment-derived factors contribute to therapeutic resistance (10). Amplification of Gli2 and point mutations in Smo that prevent drug binding have been reported to cause resistance in preclinical and clinical studies (4, 5, 11). Increased activation of PI3K, aPKC-ι/λ, or cell-cycle components may also contribute to resistance (5, 12, 13). Additional mechanisms of resistance are likely to arise in clinical practice, and must be understood to develop more effective therapeutic strategies for Shh-dependent tumors.

To date, the absence of reliable in vitro systems for growing and maintaining Shh-dependent tumors has been a major impediment for studying these cancers (14). Here, we report an approach for generating stable medulloblastoma cell lines that are tumorigenic and retain key characteristics of Shh-subtype medulloblastoma. Using these models, we identify two paradigms of resistance to Smo inhibitors. Loss of Sufu reactivates the Shh pathway downstream of Smo and thereby causes acquired therapeutic resistance. In a second scenario, activation of the RAS–MAPK pathway overrides oncogenic addiction to Shh signaling and enables proliferation of resistant tumors with enhanced metastatic behavior. In human cancers, MAPK pathway activation is increased in metastatic medulloblastoma tumor cells. Strikingly, the MAPK pathway also becomes activated after vismodegib treatment as Shh-dependent basal cell cancer transitions to squamous cell cancer resistant to Smo inhibitors. Together, these results indicate that reactivation of the Shh pathway or interactions between Shh and MAPK pathways can alter tumor behavior and therapeutic responses. Therefore, future treatments must consider these distinct mechanisms of tumor evolution.

Materials and Methods

Detailed description is given in Supplemental Materials.

Animals

All experimental procedures were done in accordance with the NIH guidelines and approved by the Dana-Farber Cancer Institutional Animal Care and Use Committee. Ptch+/− mice (The Jackson Laboratory; ref. 2). nu/nu mice (Charles River Laboratories).

Human studies

All human subjects work was reviewed by the Institutional Review Board Committees of Brigham and Women's Hospital and Dana-Farber Cancer Institute, University of Calgary, and Stanford University for appropriate use, that informed consent was obtained from all subjects when required, and appropriate waiver of consent requirements was obtained for minimal risk studies.

SMB cell culture

Shh-subtype medulloblastoma (SMB) cells were cultured as neurospheres in DMEM/F12 media (2% B27, 1% Pen/Strep). SMB(GF) cells were generated by culturing parental SMB cells for ≥3 weeks with the above media supplemented with EGF, bFGF (20 ng/mL each), 0.2% heparin.

Cell survival assays

SMB cells in 96-well plates (3 × 104 cells/well) were incubated for 72 hours in LDE225, vismodegib, LEQ506 or ATO, or for 120 hours in BKM120, BEZ235, PD325901, or CI-1040. Viability was measured using CellTiter 96 Aqueous One Solution (Promega), and calculated as the percentage of control (DMSO treated).

Gene copy number analysis

Genomic DNA was extracted with the DNeasy Blood and Tissue Kit (Qiagen). Genomic copy number for Sufu was determined by qPCR with custom-designed primers using 5 ng of genomic DNA/reaction. Copy number was calculated as described in Supplemental Information.

Immunohistochemistry, immunocytochemistry, and immunoblotting

Human medulloblastoma and matched metastases were stained with hematoxylin and eosin (H&E), or with anti-pERK1/2 (Cell Signaling Technology; 1:400), visualized using the Envision Plus Detection Kit (DAKO). Human skin tumors were immunostained with: anti-Keratin14(ab7800; Abcam); anti-Gli1 (C-18; Santa Cruz Biotechnology); anti-pERK (#9101; Cell Signaling Technology). Immunoreactivity was visualized with Alexa-Fluor secondary antibodies and confocal microscopy (Leica SP8). Staining antibodies: Ki67 (Leica Microsystems; 1:400), Nestin (Abcam; 1:400), Tuj1 (Covance; 1:400), GFP (Aves Labs; 1:1,000), and Zic (made in house, 1:400; ref. 15). Immunoblot antibodies: pAKT (S473), AKT, pERK1/2 (T202/Y204), ERK1/2, pS6, S6, pan-Ras, Gli1, Sufu, p53, cleaved caspase-3, Nmyc, Flag tag (Cell Signaling Technology; 1:1,000), actin (Sigma; 1:10,000), HA-tag (Millipore; 1:1,000), Gli2 (Aviva; 1:1,000), c-MYC (Santa Cruz Biotechnology; 1:1,000), V5-tag (Invitrogen; 1:1,000).

Transplantation and in vivo treatment

A total of 5 × 106 cells in 100 μL were injected s.c. in flank of nu/nu mice (6–8 weeks old). Tumor volumes (V = 0.5 × A × B2) were measured twice/week. When tumors reached 150 mm3, animals were randomly grouped for treatment with vehicle or LDE225 (diphosphate salt in 0.5% methylcellulose, 0.5% Tween 80, at 80 mg/kg by oral gavage once daily). Mice with tumors ≥2,000 mm3 were euthanized. For orthotopic tumors, 1 × 106 cells in 2 μL were injected into cerebella of nu/nu mice (6–8 weeks old). Animals were sacrificed when symptomatic.

Skin tumor sequencing

Sequencing of clinical samples was performed under Institutional Review Board-approval at Stanford University. Medically qualified patients 18 years or older with advanced BCCs were enrolled and informed consent was obtained for tumor sequencing (protocol #18325). Tissue samples were stored in RNALater at −20°C (Ambion). DNA was isolated using the DNeasy Blood & Tissue Kit. Capture libraries were constructed from 2 μg DNA from BCC and normal skin using the Agilent SureSelect XT Human All Exon V4 Kit. Enriched exome libraries were multiplexed and sequenced on Illumina HiSeq 2500 platform to generate 100-bp paired-end reads. Sequencing reads were aligned to human reference genome sequence (hg19) using Burrows-Wheeler Aligner. SAM to BAM conversion and marking of PCR duplicates were performed using Picard tools (version 1.86), followed by local realignment around indels and base quality score recalibration using the Genome Analysis Toolkit (GATK; v2.3.9). Mean target coverage was 114X over coding regions. Somatic SNVs and indels were called using GATK. Variants were annotated for standard quality metrics and for presence in dbSNP138.

Accession numbers

The microarray data accession number is GSE69359.

Results

SMB cell lines maintain features of Shh-subtype medulloblastoma

Studies of Shh-dependent tumors have been limited by the lack of appropriate tumor cell lines. To establish new Shh pathway–dependent tumor cell lines, we propagated tumor cells from spontaneous medulloblastoma of Ptch+/− mice as neurospheres (2). To retain Shh pathway dependency, we omitted EGF and bFGF from the media as these growth factors (GF) can promote differentiation of granule cell precursors (GCP), cells of origin for Shh-subtype medulloblastoma (16, 17). Several tumor samples tested (7 of 35, or 20%) grew for many passages and were designated SMB lines. The three most stable lines, SMB21, SMB55, and SMB56, were used in this study.

SMB lines are tumorigenic and retain key characteristics of in vivo Shh-subtype medulloblastoma. SMB cells are small (∼5 μm in diameter) and exhibit a morphology similar to cerebellar GCPs. These tumor-derived cells express medulloblastoma markers, including stem cell/progenitor marker Nestin, neuronal marker Tuj1, cerebellar marker Zic1, cell proliferation marker Ki67, and Math1(Atoh1), a hallmark of Shh-subtype medulloblastoma (Fig. 1A and B). Importantly, SMB cells exhibit constitutively activated Shh signaling, as Shh signaling components and target genes Gli1, Gli2, Boc, Ccnd1, Nmyc, and SFRP1, are highly expressed in SMB cells and in primary medulloblastoma from Ptch+/− mice (Fig. 1B). To compare SMB cells with human medulloblastoma, we performed gene-expression analysis of SMB cells, in vivo primary medulloblastoma, and cerebellum of P6 and adult mice. We compared these profiles with signature profiles of human medulloblastoma (WNT, SHH, group C, group D; ref. 18). Similarity to each subtype for each sample is defined by the “signature score,” which quantitatively measures similarity of gene-expression patterns to predefined signature genes. Subtype signature score indicates that SMB cells and in vivo primary medulloblastoma both closely resembled Shh-subtype medulloblastoma (Fig. 1C). We used a second algorithm, agreement of differential expression (AGDEX; ref. 19), to compare SMB cells with human medulloblastoma. AGDEX analysis also indicates that both Ptch+/− primary medulloblastoma and SMB cells exhibit the highest agreement with human SHH subtype (Supplementary Fig. S1F). Notably, activated Shh signaling in SMB cells is exquisitely sensitive to Smo inhibitors, as demonstrated by reduced Gli1 mRNA and protein following treatment with Smo inhibitor LDE225 (Fig. 1D and E). Importantly, proliferation and survival of SMB cells also depend on active Shh signaling as demonstrated in cell survival assays with three different Smo inhibitors LDE225, vismodegib, and LEQ506 (20). These inhibitors reduce cell number in all SMB lines, and increase apoptosis as assessed by activated caspase-3 (Fig. 1E and F; Supplementary Fig. S1A).

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

SMB cells maintain key features of Shh-subtype medulloblastoma. A, immunostaining of SMB cells with anti-Nestin, Ki67, Tuj1 Zic1; DAPI, blue; scale bar, 20 μm. B, microarray analysis reveals Shh-subtype signature in SMB lines and medulloblastoma from Ptch+/− mice, compared with adult and P6 cerebellum. C, expression of SMB cells, in vivo primary medulloblastoma, P6, and adult cerebellum, analyzed using signature profiles of human medulloblastomas (WNT, SHH, group C, group D). Similarity to each subtype for each sample defined by “signature score” and normalized by normal mouse samples. D, quantitative RT-PCR analysis of Gli1 in SMB cells treated with DMSO or 1 μmol/L LDE225 for 24 hours; mean ± SD; n = 3; ***, P < 0.0001, Student t test. E, dose-dependent inhibition of Shh signaling by LDE225 (0, 5, 50, 500 nmol/L, 48 hours). Shh signaling assessed by immunoblot for Gli1; apoptosis assessed by cleaved caspase-3. F, survival analysis of SMB cells treated with indicated LDE225 concentrations (72 hours; mean ± SEM; n = 6).

SMB cells, even after more than 20 passages in culture, initiate tumors in vivo when transplanted into nude mice, either subcutaneously or as orthotopic xenografts in the brain (Supplementary Fig. S1B). Transplanted SMB cells exhibit typical Shh-subtype histology (Supplementary Fig. S1C–S1E). Recent sequencing studies of large cohorts of medulloblastoma patients revealed that p53 is among the most frequently mutated genes in Shh-subtype medulloblastoma (10%–20%; refs. 21, 22). To evaluate p53 status in our SMB cells, we first sequenced coding exons of p53. Y233C and C138R point mutations were detected in SMB21 and SMB55 cells, respectively, but no mutations were detected in SMB56 cells. However, other alterations can impinge on p53 activity. All three SMB lines showed elevated levels of p53 protein compared with wild-type murine neural progenitor cells, and p53 expression in SMB cells did not change in response to gamma irradiation (Supplementary Fig. S1G), indicating that the p53 signaling axis is dysregulated in all SMB lines. Together, these data indicate that our culturing protocol can establish Shh-subtype cell lines with dysregulated p53, and these lines can be used to study Shh-subtype medulloblastoma in vitro and in vivo and provide a platform for rapid, large-scale functional study and drug screening.

SMB cell lines provide a model to study drug resistance

A major concern with Smo inhibitors or other targeted therapies is the emergence of drug resistance. To determine whether SMB cell lines can help address this challenge, we asked whether the effects of known resistance mechanisms, such as Smo mutants (D477G, L225R, and S391N) and Gli2 overexpression, can be recapitulated in SMB cells. To achieve stable expression of exogenous genes, we adapted the piggyBac transposon system to integrate exogenous genes into the SMB genome (Supplementary Fig. S2A). SMB21 cells stably expressing GFP, Smo(WT), Smo(D477G), Smo(L225R), Smo(S391N), Gli2, or Gli2ΔN, constitutively active Gli2 lacking the amino-terminal repressor domain (23), were tested for sensitivity to LDE225 in a cell survival assay. Although cells expressing GFP or Smo(WT) remain sensitive to Smo inhibitors LDE225, LEQ506, or vismodegib, cells expressing Smo mutants, Gli2, or Gli2ΔN are resistant to Smo inhibitors (Fig. 2A; Supplementary Fig. S2B–S2D). Consistent with previous studies, cells expressing Smo mutants or Gli2ΔN exhibit constitutively activated Shh signaling even in the presence of LDE225, as demonstrated by Gli1 expression (Fig. 2B).

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

SMB cells provide a model to study resistance to Smo inhibition. A, survival analysis of SMB21 cells expressing GFP (control), Smo mutants, or Gli2ΔN treated with indicated LDE225 concentrations (72 hours; mean ± SEM; n = 4). B, Shh signaling was analyzed by immunoblot for Gli1 in SMB21 cells expressing GFP, HA-tagged wild-type or mutant Smo, treated with DMSO or 1 μmol/L LDE225 for 24 hours. C, SMB21 or SMB21 cells expressing Smo(WT) retain LDE225 responsiveness in vivo; SMB21 cells expressing Smo(D477G) or Gli2ΔN initiate resistant tumors; tumor volume over time; mean ± SEM; n = 5.

To test resistance of engineered SMB cells in vivo, SMB21 parental, Smo(WT), Smo(D477G), and Gli2ΔN cells were subcutaneously transplanted into nude mice. Although SMB21 parental and Smo(WT)-expressing cells form tumors responsive to LDE225 in vivo, cells expressing Smo(D477G) or Gli2ΔN generate tumors resistant to LDE225 (Fig. 2C). Together, these results indicate that mutations that confer clinical resistance to LDE225 are effective in SMB cells, demonstrating that these lines provide attractive systems for studying drug resistance.

Identification of novel routes for circumventing Smo inhibition

To identify novel routes through which medulloblastoma cells can escape Smo inhibition, we used SMB cells to test three groups of candidates. The first group includes key Shh pathway components downstream of Smo: Sufu, Gli1, Gli3, and Nmyc. We used the piggyBac transposon system to overexpress Gli1, Gli3, or Nmyc, or used shRNA to knockdown Sufu. Unlike Gli2, expression of Gli1, Gli3, or Nmyc in SMB cells cannot bypass Smo inhibition, reactivate Shh signaling, or promote proliferation and survival in cells treated with LDE225 (Supplementary Fig. S2D and S2F). In contrast, shRNA knockdown of Sufu reactivates Shh signaling and confers robust resistance to LDE225 in SMB cells (Fig. 3A and B). Consistent with the key role of Sufu, several of the resistant tumors that arose spontaneously from subcutaneously implanted SMB cells following treatment with LDE225 showed drastic reduction of Sufu protein levels compared with sensitive tumors not exposed to LDE225 (Fig. 3C), and many exhibited genomic loss of Sufu (Fig. 3D). To identify possible treatment for tumors that are resistant due to Sufu loss, we tested arsenic trioxide (ATO), an FDA-approved drug shown to antagonize Gli action (24). ATO inhibits proliferation of SMB cells and Sufu knockdown cells with an effective dose similar to previous studies (Fig. 3E; ref. 24). Together, these data indicate that Gli inhibitors can treat intrinsic Shh pathway activation.

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

Loss of Sufu confers resistance to Smo inhibition. A, relative survival for SMB21, SMB55, SMB56 with shRNA knockdown of Sufu treated with indicated LDE225 concentrations (72 hours; mean ± SEM, n = 3). B, shRNA knockdown of Sufu causes constitutive activation of Shh signaling; DMSO or 1 μmol/L LDE225 for 24 hours; immunoblot for Gli1. C, subcutaneously grafted SMB21 cells initially responded to LDE225, but developed resistance after 40 days. Sufu immunoblot of vehicle (n = 4)- and LDE225 (n = 5)-treated tumors showed drastically reduced Sufu protein in resistant tumors (#1, 3, 4, 5). D, genomic copy number of Sufu in sensitive and resistant tumors determined by quantitative PCR. E, relative survival for SMB21 and Sufu knockdown cells treated with ATO (72 hours; mean ± SEM, n = 3).

In addition to Shh pathway–specific components, we tested molecules that are key drivers of WNT and group 3 (MYC) medulloblastoma subtypes (25). Neither expression of wild-type nor constitutively activated form of CTNNB1, CTNNB1(S33Y), nor overexpression of stabilized MYC, MYC(T58A), confer resistance to Smo inhibitors (Supplementary Fig. S2F–S2I). Thus, oncogenic mutations critical for other medulloblastoma subtypes cannot alter subtype identity, and thereby confer resistance. Recent studies indicate that WNT and Shh-subtype medulloblastoma have distinct cellular origins, as Shh-subtype medulloblastoma originate from GCPs and WNT subtype from dorsal brainstem cells (26). Thus, the cellular context may explain why signaling pathways driving other medulloblastoma subtypes fail to confer Smo-inhibitor resistance.

The third group of candidates tested encompassed genes involved in RTK/RAS signaling, as RTK/RAS signaling is implicated in development of normal cerebellar GCPs and medulloblastoma (27, 28). Expression of HRAS(G12V) or BRAF(V600E), but not PI3KCA(H1047R) or AKT(Myristoylated), induced resistance to LDE225 in SMB cells (Fig. 4A–C; Supplementary Fig. S3A and S3C). As expected, both HRAS(G12V) and BRAF(V600E) activate the MAPK pathway in SMB cells, as demonstrated by increased phosphorylation of ERK (Fig. 4A′; Supplementary Fig. S3B and S3D). HRAS(G12V) and BRAF(V600E) also cause resistance to other Smo inhibitors LEQ506 and vismodegib, indicating that this effect is generalizable for Smo antagonists (Supplementary Fig. S3E and S3F). SMB(HRAS) cells subcutaneously transplanted in nude mice were resistant to treatment with Smo inhibitors (Fig. 4D). Furthermore, MAPK activation is greater in SMB tumors that spontaneously develop resistance to Smo inhibitors following treatment with LDE225 than in vehicle treated, sensitive tumors (Fig. 4E). Taken together, these data indicate that activation of RAS/MAPK provides a novel way for cells to evade Smo inhibition.

Figure 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 4.

RAS/MAPK signaling confers resistance to Smo inhibitor. A–C, relative survival for SMB21 cells expressing candidate genes treated with LDE225 (72 hours). HRAS(G12V), BRAF(V600E), not PIK3CA(H1047R) or myristoylated AKT, confer resistance; mean ± SEM; n > 5. A′, elevated phospho-Erk in SMB21 cells expressing HRAS(G12V) or BRAF(V600E). B′ and C′, elevated phospho-AKT in SMB21 cells expressing PIK3CA(H1047R) or AKT(Myr). D, SMB21(HRAS) cells initiate resistant tumors in vivo; tumor volume over time; mean ± SEM; n = 5. (Experiments were performed concurrently with Fig. 2C, the same SMB21 control shown here and Fig. 2C.) E, phospho-Erk in sensitive and resistant tumors from engrafted SMB21 cells mean ± SEM; *, P < 0.05, unpaired Student t test.

Surprisingly, HRAS(G12V) does not confer resistance by reactivating Shh signaling downstream of Smo (Fig. 5A). Instead, HRAS(G12V) suppresses Shh signaling in SMB cells, as expression of multiple Shh pathway targets and components are downregulated in SMB(HRAS) cells (Fig. 5A–C; Supplementary Fig. S4A–S4D). Notably, Math1(Atoh1), a hallmark of Shh-subtype medulloblastoma that is not a direct target of SHH signaling, is decreased in SMB(HRAS) cells. These results suggest that HRAS renders SMB cells independent of Shh-signaling for growth, and thereby causes resistance.

Figure 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 5.

FGF/RAS/RAF signaling suppresses Shh-signaling. A–C, Shh-pathway targets and components decrease in SMB(HRAS) cells, by immunoblot, qRT-PCR, and microarrays; mean ± SD; n = 3; ***, P < 0.0005, Student t test. D, GFs induce Shh-signaling independence in SMB cells. SMB cells cultured with or without GF for 2 weeks, then treated with LDE225 (72 hours; relative survival; mean ± SEM; n = 3 ∼ 8). E, GF treatment activates AKT and MAPK signaling. F and G, GF treatment suppresses Shh-pathway targets and components; qRT-PCR and microarray analysis of SMB and SMB(GF) cells; means ± SD; n = 3; *, P < 0.05, Student t test.

RAS is normally regulated by upstream GFs and receptor tyrosine kinases, and so receptor activation might mimic the effects of HRAS in SMB cells. During normal cerebellar GCP development, GF, such as bFGF, antagonize Shh pathway activity and promote differentiation (29, 30). When SMB cells were exposed to GFs that are common components of stem cell media (bFGF and EGF; 20 ng/mL) both PI3K–AKT and RAS–RAF-MAPK pathways were activated, Shh signaling was suppressed, and SMB cells became resistant to LDE225 (Fig. 5D–G). We tested the GFs individually, and found that bFGF, not EGF, suppresses Shh signaling and causes resistance (Supplementary Fig. S4E and S4F). Together, these results demonstrate that sustained activation of FGF/RAS/RAF signaling enables Shh-subtype medulloblastoma to grow in a Shh pathway–independent manner.

FGF/RAS-mediated resistance to Smo inhibitors is reversible

To investigate whether FGF/RAS/MAPK signaling is required for SMB cells to both develop and maintain resistance to Smo inhibitors, we removed oncogenic HRAS from SMB(HRAS) cells using lentiviral delivered Flp to cleave FRT sites within the transposon (Supplementary Fig. S5A and S5B). Removal of RAS decreases Erk phosphorylation and restores Shh signaling activity and susceptibility to Smo inhibitors (Fig. 6A–C). Similarly, cells resistant to Smo inhibitors due to prolonged bFGF treatment regained Shh signaling activity and susceptibility to Smo inhibitors when bFGF was removed from media for prolonged time periods (Fig. 6D and E). Together, these data indicate that prolonged activation of FGF/RAS/MAPK signaling both initiates and maintains Shh-signaling independence and resistance to Smo inhibitors.

Figure 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 6.

FGF/RAS-dependent resistance is reversible. A, removal of HRAS(G12V) restores LDE225 sensitivity. SMB(HRAS) cells were infected with lentivirus expressing Flp or tdTomato (control), then assayed by relative survival at indicated LDE225 concentrations (mean ± SEM; n = 3). B and C, Shh signaling assayed by immunoblot and qRT-PCR; mean ± SD; n = 3, **, P < 0.01; ***, P < 0.001, one-way ANOVA, Bonferroni correction. D and E, SMB21(GF) and SMB55(GF) cells were cultured in media without GF for 3 weeks. D, relative survival was then analyzed in indicated LDE225 concentrations (72 hours); mean ± SEM; n > 9. E, qRT-PCR analysis of Atoh1, Gli1; mean ± SD, n = 6, ***, P < 0.001, one-way ANOVA with Bonferroni. F, relative survival for SMB21 and SMB21(HRAS) cells treated with MEK inhibitor PD325901 (120 hours; mean ± SEM; n = 4). G, MEK inhibitor PD325901 (0, 10, 100, 1,000 nmol/L for 24 hours) reduce phospho-Erk analyzed by immunoblot.

To identify therapeutic approaches for treating SMB cells resistant to Smo inhibitors, we tested PI3K and MEK pathway inhibitors. Although HRAS cells showed similar sensitivity as SMB parental cells to PI3K inhibitors BEZ235 and BKM120, they were much more sensitive to MEK inhibitors CI-1040 and PD325901. Thus, MEK inhibitors provide therapeutic treatment for resistant tumors driven by activation of FGF/RAS/RAF signaling (Fig. 6F and G; Supplementary Figs. S5C–S5E, S6).

RAS/MAPK activation alters characteristics of Shh pathway–dependent tumors

Morphologically, SMB(HRAS) cells exhibit a dramatically different appearance from SMB parental cells. SMB parental cells are small, with little cytoplasmic material, whereas SMB(HRAS) cells appear larger with extended cellular processes (Supplementary Fig. S7). Immunohistochemical characterization revealed that SMB(HRAS) cells are proliferative, as indicated by Ki67, and poorly differentiated, as indicated by Nestin, a stem cell/progenitor marker (Fig. 7A).

Figure 7.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 7.

RAS activation alters characteristics of Shh pathway–dependent tumors. A, SMB and SMB(HRAS) cells stained with anti-Nestin, Ki67; DAPI, blue; scale bar, 50 μm. B, cell invasion of SMB(HRAS) or SMB cells assessed using Matrigel-coated Transwell apparatus. Invasion measured at 18 hours and normalized to SMB cells; mean ± SD; n = 3; *, P < 0.05, Student t test. C and D, H&E-stained lung sections from animals engrafted with SMB (C) or SMB(HRAS) cells (D); arrow, metastasis. Metastases detected in 2 of 9 HRAS mice; 0 of 8 SMB parental mice; scale bar, 50 μm. E and F, phospho-ERK in primary and metastatic samples from human Shh-subtype medulloblastoma. Phospho-ERK high in metastatic, frontal lobe tumor (F), but not in matched primary cerebellar tumor (E); scale bar, 50 μm. G, pretreatment BCC with characteristic H&E, keratin14, and Gli1 with an absence of phospho-ERK immunoreactivity. SCC tumor that developed at the same location after vismodegib displayed a spindle-like morphology characteristic of SCC and lacked keratin14 and Gli1 immunostaining, but was positive for phospho-ERK; scale bar, 100 μm. H, sequencing suggests shared lineage for posttreatment-resistant SCC and pretreatment BCC. BCC and SCC share most genetic variations (n = 1248; ratio = 0.91); SCC-normal share (n = 84; ratio = 0.06); SCC unique (n = 43; ratio = 0.03).

The striking morphologic differences in SMB(HRAS) cells suggests that they may be more motile than parental cells. Indeed these cells were more invasive when tested in a Matrigel invasion assay (Fig. 7B). Interestingly, when SMB(HRAS) cells were s.c. injected in nude mice, they initiated resistant tumors, and also generated lung metastases in 2 of 9 mice. Metastases, were never found in 8 mice injected with SMB cells (Fig. 7C and D). Together, these data indicate that activation of RAS/MAPK increases tumor invasiveness.

Clinical observations from a rare set of matched primary and metastatic lesions from an individual patient provide additional evidence that RAS activation promotes metastasis in medulloblastoma, as the primary lesion exhibits low level of MAPK activation, whereas the metastatic frontal lobe lesion exhibits robust MAPK activation (Fig. 7E and F). In human primary Shh-subtype medulloblastoma, most areas are predominantly negative for MAPK activation (Supplementary Fig. S8, Table S2); however, in one tumor, cancer cells in the perivascular niche were strikingly positive for ERK phosphorylation (Supplementary Fig. S8D) whereas in a desmoplastic/nodular medulloblastoma, ERK phosphorylation was elevated in perinodular regions (Supplementary Fig. S8B). Both tumors exhibited regions with high FGF immunostaining (Supplementary Fig. S8B and S8D). Cells with MAPK activation within human Shh-subtype medulloblastomas may generate resistant tumors when challenged by Smo inhibitors.

Vismodegib treatment of BCC engenders RAS/MAPK-dependent tumors

Smo inhibitor vismodegib is approved for clinical use for patients with advanced BCC, but is not yet approved for medulloblastoma. Among BCC patients, approximately 21% that initially respond subsequently develop tumors resistant to this inhibitor (31). In some cases, posttreatment resistant tumors exhibit characteristics of squamous cell carcinoma (SCC; refs. 32–36). We analyzed three patients who developed SCC at the site of the antecedent BCC tumor following treatment with vismodegib. Posttreatment resistant tumors display low level of Gli1 and high level of phospho-ERK, suggesting upregulated RAS/MAPK and downregulated Shh signaling (Fig. 7G). To determine whether SCCs that develop following vismodegib are derived from the antecedent BCC in the same location, we analyzed patient-matched normal tissue or blood, and pre- and post-relapse tumor samples by exome sequencing with germline and dbSNP variants removed. Tumors we assayed initially responded to vismodegib before acquiring resistance to the inhibitor. Strikingly, 91% of genetic variations (n = 1248) in the SCC sample were shared between pretreatment BCC and posttreatment SCC, whereas only 3% and 6% of somatic genetic variations (n = 43 and n = 84) were unique to the SCC or shared with patient-matched normal sample, respectively, suggesting that the SCC arose from the BCC (Fig. 7H). These results suggest that tumors can evolve from Shh pathway–dependent BCC to the RAS–MAPK pathway–dependent SCCs, and thereby develop resistance to Smo inhibitors.

Discussion

The studies presented here introduce a set of Shh pathway–dependent medulloblastoma cell lines (SMB) and identify two distinct mechanisms of therapeutic resistance to Smo inhibitors. Loss of Sufu drives resistance to Smo inhibition by activating downstream Shh signaling. Alternatively, activation of RAS/MAPK signaling, either due to new mutations or to microenvironmental factors, constitutes a novel mechanism of resistance that circumvents Shh pathway dependence in a growing tumor.

SMB cells as a model for Shh-subtype medulloblastoma

Cell lines that faithfully model the cancer from which they are derived provide important tools for studying disease mechanism and discovering novel therapies. Investigation of medulloblastoma biology has been limited by the lack of stable lines that are tumorigenic and Shh pathway dependent. Most established medulloblastoma lines, are adherent cell cultures maintained in serum-containing media, and do not depend on the Shh pathway for growth and survival (6, 14). Current protocols can only culture freshly isolated tumor cells for a short time before key characteristics of Shh-subtype medulloblastoma are lost. Here, we present Shh pathway–dependent medulloblastoma lines (SMB) that can be used as effective and faithful in vitro models to study Shh-subtype medulloblastoma.

The protocol that enabled development of SMB lines is a modified version of neural stem cell culture methods. Key aspects include growing cells as nonadherent spheres and eliminating serum, EGF and bFGF from media. Generation of tumor spheres is commonly used to enrich for cancer stem-like cells (37). Indeed high-grade gliomas can be perpetuated as neurospheres by maintaining cells in neural stem cell media with EGF and bFGF (37, 38). However, these conditions do not maintain tumorigenicity and Shh pathway dependency of medulloblastoma cells (39), as FGF signaling has an antagonistic effect on Shh signaling (29, 30, 40). Instead, medulloblastoma neurospheres from Ptch+/− mice cultured without exogenous EGF or bFGF generate Shh-subtype medulloblastoma cell lines (SMB) that are tumorigenic, maintain markers of Shh-subtype medulloblastoma and remain dependent on Shh pathway activity even after many passages in vitro.

A previous study isolated rare lines from medulloblastomas of Ptch+/−;p53−/− mice (41). We observe distinct modes of p53 signaling dysregulation in each SMB line. Because all three lines are derived from Ptch+/−;p53+/+ mice, mutations or inactivation of p53 signaling may have developed during primary medulloblastoma tumorigenesis in each individual animal. In human medulloblastoma, the importance of p53 has become increasingly apparent. Among human medulloblastoma, p53 mutations were detected in 13% to 21% of Shh-subtype MBs (21, 22). We conclude that SMB cells offer a faithful model for investigating the Shh pathway in medulloblastoma, and facilitate high-throughput drug testing and large-scale functional screens. In the future, a similar approach might enable generation of human Shh pathway–dependent medulloblastoma lines.

Mechanisms of resistance to Smo inhibition

Several Smo inhibitors show promise in preclinical and clinical studies. Vismodegib was the first drug of this class approved by the FDA to treat BCCs. In one study, 6 of 28 BCC patients treated with vismodegib developed resistance to Smo inhibitors (31). Here, we demonstrate that Sufu mutation can occur after treatment with Smo inhibitors, and cause secondary resistance by activating Shh signaling downstream of Smo. As loss of Sufu generates medulloblastomas that never respond to Smo inhibition (21, 42), this finding provides proof of principle that clinically relevant resistance mechanisms can be studied in SMB cells.

Data that overexpression of Gli1 or Nmyc does not confer resistance to Smo inhibition may seem surprising. However, Gli2 is the primary activator of the Shh signaling pathway in GCP development and medulloblastoma, whereas Gli1 is not essential (43, 44). In preclinical and clinical settings, amplification and constitutive activation of Gli2 generates Shh-subtype medulloblastoma resistant to Smo inhibition (5, 12, 21). In a recent study of human Shh-subtype medulloblastoma (n = 133), 10 cases of Gli2 amplification were identified, but no cases of Gli1 amplification were seen (21). Therefore, our results with overexpression of Gli transcription factors are consistent with clinical observations. In contrast, Nmyc amplification is reported in Shh-subtype medulloblastoma that do not respond to Smo inhibition (21). Although we cannot exclude the possibility that higher expression achieved by other means might confer resistance, our results suggest that other genetic changes may be needed in conjunction with Nmyc for tumors to grow in the presence of Smo inhibitors.

An important finding of this study is identification of a Shh pathway–independent mechanism of resistance. RAS-mediated resistance involves shifting oncogenic addiction to a second oncogenic pathway. De novo mutations or a microenvironment with abundant GF can stimulate RAS/MAPK signaling, eliminate Shh pathway dependency, and cause resistance in medulloblastoma. Indeed, in human medulloblastoma, components of the RTK–RAS–MAPK pathway are often overexpressed (27), and epigenetic inactivation of RAS association domain family 1A (RASSF1A) tumor-suppressor gene is frequently observed (45, 46). Although de novo RAS/RAF mutations have not been detected in primary human medulloblastoma, such mutations might be favored following treatment with Smo inhibitors (28), as mutations that confer resistance are often only detected following treatment with targeted therapies (11, 47). Strikingly in our studies, xenografts with spontaneous resistance to Smo inhibitors display activation of ERK signaling in vivo. Thus, our data indicate that GF stimulation, genetic or epigenetic changes affecting the RAS–RAF–MEK pathway should be assessed in patients that develop resistance to Smo inhibitors.

In addition to intrinsic mutations in tumor cells, tumor microenvironment may alter drug efficacy (48). Our study suggests that microenvironments with abundant FGF could provide protective niches for cells exposed to Smo inhibitors. We show that in human medulloblastoma, ERK activation occurs in locations adjacent to blood vessels or in perinodular spaces. Recent work suggested that stromal production of placental GF (PlGF) in human medulloblastoma promotes cancer cell survival by activating the MAPK cascade (49). Thus, paracrine PlGF-mediated RAS/MAPK signaling could also attenuate efficacy of Smo inhibitors.

Cross-talk between Shh and FGFR/RAS signaling has been widely recognized during organogenesis in multiple tissues (50, 51). Depending on biologic context, interactions between FGF/RAS and Shh pathways can be synergistic or antagonistic. In cerebellar GCPs and Ptch+/− medulloblastoma cells, acute bFGF treatment suppresses Shh signaling and proliferation, and concomitantly promotes cell differentiation (29, 30). Similarly, oncogenic RAS can block Shh signaling in NIH3T3 cells and pancreatic cancer models (52). Here, we again observe an antagonistic relationship between Shh and FGF/RAS signaling in SMB cells. Importantly, however, this process does not promote terminal differentiation; instead tumor cells remain proliferative and tumorigenic.

RAS/MAPK signaling in metastasis and tumor evolution

Strikingly, RAS/MAPK activation alters multiple characteristics of Shh-dependent tumors. Morphologic and transcriptional profiles of SMB(HRAS) cells differ from SMB cells; although SMB cells are small with classic medulloblastoma histology, SMB(HRAS) cells display an extended morphology, are more invasive in vitro and more metastatic in vivo. Comparison of Shh-subtype primary medulloblastoma and matched metastatic lesions from the same person, reveal high level of phosphorylated ERK1/2 in metastases. Consistent with our findings, ectopic expression of Eras (embryonic stem cell–expressed Ras), which is structurally similar to RAS oncoprotein (53), increases leptomeningeal metastases in models of Shh-subtype medulloblastoma, and these metastatic cells differ genetically and epigenetically from primary tumor cells (54, 55). Together, these data indicate that RAS activation results in resistance to Smo inhibitors and alters tumor characteristics.

Clinical studies of resistant BCC also suggest a role of RAS/MAPK in tumor evolution. Several studies have reported occurrences of SCCs during treatment of BCC with vismodegib (32–36). Sequencing of matched pre- and posttreatment skin tumor samples supports the hypothesis that BCC tumors activate RTK/RAS/MAPK signaling and generate SCCs under selective pressure by Smo inhibition (56). Thus, our findings indicate a novel scenario of resistance by which Shh pathway–dependent tumor cells evolve and escape Shh signaling dependence. Future studies are required to assess the prevalence of this resistance mechanism in patients.

Disclosure of Potential Conflicts of Interest

A.L.S. Chang reports receiving other commercial research support and is a consultant/advisory board member for Genentech. A.E. Oro reports receiving a commercial research grant from Novartis and Genentech. J.F. Kelleher has ownership interest (including patents) in Novartis. No potential conflicts of interest were disclosed by the other authors.

Authors' Contributions

Conception and design: X. Zhao, T. Ponomaryov, K.J. Ornell, J.F. Kelleher, R.A. Segal

Development of methodology: X. Zhao, T. Ponomaryov, K.J. Ornell, E. Pak

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): X. Zhao, T. Ponomaryov, K.J. Ornell, P. Zhou, S.K. Dabral, S.X. Atwood, R.J. Whitson, A.L.S. Chang, A.E. Oro, J.A. Chan, R.A. Segal

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): X. Zhao, T. Ponomaryov, K.J. Ornell, P. Zhou, E. Pak, W. Li, S.X. Atwood, R.J. Whitson, A.L.S. Chang, J. Li, A.E. Oro, J.A. Chan

Writing, review, and/or revision of the manuscript: X. Zhao, T. Ponomaryov, K.J. Ornell, P. Zhou, E. Pak, W. Li, S.X. Atwood, R.J. Whitson, A.L.S. Chang, A.E. Oro, J.A. Chan, J.F. Kelleher, R.A. Segal

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): X. Zhao, K.J. Ornell, E. Pak

Study supervision: X. Zhao, R.A. Segal

Grant Support

This study was supported by grants from the NIH (CA142536 to RAS, ARO4786, and 5ARO54780 to A.E. Oro; HG4069 supports W. Li), Novartis Pharmaceuticals, the Emerald Foundation, Alex's Lemonade Stand Foundation, Autism Speaks Foundation, and the American Brain Tumor Association.

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.

Footnotes

  • Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/).

  • Received October 20, 2014.
  • Revision received June 2, 2015.
  • Accepted June 18, 2015.
  • ©2015 American Association for Cancer Research.

References

  1. 1.↵
    1. Rubin LL,
    2. de Sauvage FJ
    . Targeting the Hedgehog pathway in cancer. Nat Rev Drug Discov 2006;5:1026–33.
    OpenUrlCrossRefPubMed
  2. 2.↵
    1. Goodrich LV,
    2. Milenkovic L,
    3. Higgins KM,
    4. Scott MP
    . Altered neural cell fates and medulloblastoma in mouse patched mutants. Science 1997;277:1109–13.
    OpenUrlAbstract/FREE Full Text
  3. 3.↵
    1. Hahn H,
    2. Wicking C,
    3. Zaphiropoulous PG,
    4. Gailani MR,
    5. Shanley S,
    6. Chidambaram A,
    7. et al.
    Mutations of the human homolog of Drosophila patched in the nevoid basal cell carcinoma syndrome. Cell 1996;85:841–51.
    OpenUrlCrossRefPubMed
  4. 4.↵
    1. Rudin CM,
    2. Hann CL,
    3. Laterra J,
    4. Yauch RL,
    5. Callahan CA,
    6. Fu L,
    7. et al.
    Treatment of medulloblastoma with hedgehog pathway inhibitor GDC-0449. N Engl J Med 2009;361:1173–8.
    OpenUrlCrossRefPubMed
  5. 5.↵
    1. Buonamici S,
    2. Williams J,
    3. Morrissey M,
    4. Wang A,
    5. Guo R,
    6. Vattay A,
    7. et al.
    Interfering with resistance to smoothened antagonists by inhibition of the PI3K pathway in medulloblastoma. Sci Transl Med 2010;2:51ra70.
    OpenUrlAbstract/FREE Full Text
  6. 6.↵
    1. Romer JT,
    2. Kimura H,
    3. Magdaleno S,
    4. Sasai K,
    5. Fuller C,
    6. Baines H,
    7. et al.
    Suppression of the Shh pathway using a small-molecule inhibitor eliminates medulloblastoma in Ptc1(+/−)p53(−/−) mice. Cancer Cell 2004;6:229–40.
    OpenUrlCrossRefPubMed
  7. 7.↵
    1. Lee MJ,
    2. Hatton BA,
    3. Villavicencio EH,
    4. Khanna PC,
    5. Friedman SD,
    6. Ditzler S,
    7. et al.
    Hedgehog pathway inhibitor saridegib (IPI-926) increases lifespan in a mouse medulloblastoma model. Proc Natl Acad Sci U S A 2012;109:7859–64.
    OpenUrlAbstract/FREE Full Text
  8. 8.↵
    1. Metcalfe C,
    2. de Sauvage FJ
    . Hedgehog fights back: mechanisms of acquired resistance against Smoothened antagonists. Cancer Res 2011;71:5057–61.
    OpenUrlAbstract/FREE Full Text
  9. 9.↵
    1. Gajjar A,
    2. Stewart CF,
    3. Ellison DW,
    4. Kaste S,
    5. Kun LE,
    6. Packer RJ,
    7. et al.
    Phase I study of vismodegib in children with recurrent or refractory medulloblastoma: a pediatric brain tumor consortium study. Clin Cancer Res 2013;19:6305–12.
    OpenUrlAbstract/FREE Full Text
  10. 10.↵
    1. Bucheit AD,
    2. Davies MA
    . Emerging insights into resistance to BRAF inhibitors in melanoma. Biochem Pharmacol 2014;87:381–9.
    OpenUrlCrossRefPubMed
  11. 11.↵
    1. Yauch RL,
    2. Dijkgraaf GJ,
    3. Alicke B,
    4. Januario T,
    5. Ahn CP,
    6. Holcomb T,
    7. et al.
    Smoothened mutation confers resistance to a Hedgehog pathway inhibitor in medulloblastoma. Science 2009;326:572–4.
    OpenUrlAbstract/FREE Full Text
  12. 12.↵
    1. Dijkgraaf GJ,
    2. Alicke B,
    3. Weinmann L,
    4. Januario T,
    5. West K,
    6. Modrusan Z,
    7. et al.
    Small molecule inhibition of GDC-0449 refractory smoothened mutants and downstream mechanisms of drug resistance. Cancer Res 2011;71:435–44.
    OpenUrlAbstract/FREE Full Text
  13. 13.↵
    1. Atwood SX,
    2. Li M,
    3. Lee A,
    4. Tang JY,
    5. Oro AE
    . GLI activation by atypical protein kinase C iota/lambda regulates the growth of basal cell carcinomas. Nature 2013;494:484–8.
    OpenUrlCrossRefPubMed
  14. 14.↵
    1. Sasai K,
    2. Romer JT,
    3. Lee Y,
    4. Finkelstein D,
    5. Fuller C,
    6. McKinnon PJ,
    7. et al.
    Shh pathway activity is down-regulated in cultured medulloblastoma cells: implications for preclinical studies. Cancer Res 2006;66:4215–22.
    OpenUrlAbstract/FREE Full Text
  15. 15.↵
    1. Borghesani PR,
    2. Peyrin JM,
    3. Klein R,
    4. Rubin J,
    5. Carter AR,
    6. Schwartz PM,
    7. et al.
    BDNF stimulates migration of cerebellar granule cells. Development 2002;129:1435–42.
    OpenUrlPubMed
  16. 16.↵
    1. Schuller U,
    2. Heine VM,
    3. Mao J,
    4. Kho AT,
    5. Dillon AK,
    6. Han YG,
    7. et al.
    Acquisition of granule neuron precursor identity is a critical determinant of progenitor cell competence to form Shh-induced medulloblastoma. Cancer Cell 2008;14:123–34.
    OpenUrlCrossRefPubMed
  17. 17.↵
    1. Yang ZJ,
    2. Ellis T,
    3. Markant SL,
    4. Read TA,
    5. Kessler JD,
    6. Bourboulas M,
    7. et al.
    Medulloblastoma can be initiated by deletion of Patched in lineage-restricted progenitors or stem cells. Cancer Cell 2008;14:135–45.
    OpenUrlCrossRefPubMed
  18. 18.↵
    1. Northcott PA,
    2. Korshunov A,
    3. Witt H,
    4. Hielscher T,
    5. Eberhart CG,
    6. Mack S,
    7. et al.
    Medulloblastoma comprises four distinct molecular variants. J Clin Oncol 2011;29:1408–14.
    OpenUrlAbstract/FREE Full Text
  19. 19.↵
    1. Poschl J,
    2. Stark S,
    3. Neumann P,
    4. Grobner S,
    5. Kawauchi D,
    6. Jones DT,
    7. et al.
    Genomic and transcriptomic analyses match medulloblastoma mouse models to their human counterparts. Acta Neuropathol 2014;128:123–36.
    OpenUrlCrossRefPubMed
  20. 20.↵
    1. Peukert S,
    2. He F,
    3. Dai M,
    4. Zhang R,
    5. Sun Y,
    6. Miller-Moslin K,
    7. et al.
    Discovery of NVP-LEQ506, a second-generation inhibitor of smoothened. ChemMedChem 2013;8:1261–5.
    OpenUrlCrossRefPubMed
  21. 21.↵
    1. Kool M,
    2. Jones DT,
    3. Jager N,
    4. Northcott PA,
    5. Pugh TJ,
    6. Hovestadt V,
    7. et al.
    Genome sequencing of SHH medulloblastoma predicts genotype-related response to smoothened inhibition. Cancer Cell 2014;25:393–405.
    OpenUrlCrossRefPubMed
  22. 22.↵
    1. Zhukova N,
    2. Ramaswamy V,
    3. Remke M,
    4. Pfaff E,
    5. Shih DJ,
    6. Martin DC,
    7. et al.
    Subgroup-specific prognostic implications of TP53 mutation in medulloblastoma. J Clin Oncol 2013;31:2927–35.
    OpenUrlAbstract/FREE Full Text
  23. 23.↵
    1. Pasca di Magliano M,
    2. Sekine S,
    3. Ermilov A,
    4. Ferris J,
    5. Dlugosz AA,
    6. Hebrok M
    . Hedgehog/Ras interactions regulate early stages of pancreatic cancer. Genes Dev 2006;20:3161–73.
    OpenUrlAbstract/FREE Full Text
  24. 24.↵
    1. Kim J,
    2. Aftab BT,
    3. Tang JY,
    4. Kim D,
    5. Lee AH,
    6. Rezaee M,
    7. et al.
    Itraconazole and arsenic trioxide inhibit Hedgehog pathway activation and tumor growth associated with acquired resistance to smoothened antagonists. Cancer Cell 2013;23:23–34.
    OpenUrlCrossRefPubMed
  25. 25.↵
    1. Taylor MD,
    2. Northcott PA,
    3. Korshunov A,
    4. Remke M,
    5. Cho YJ,
    6. Clifford SC,
    7. et al.
    Molecular subgroups of medulloblastoma: the current consensus. Acta Neuropathol 2012;123:465–72.
    OpenUrlCrossRefPubMed
  26. 26.↵
    1. Gibson P,
    2. Tong Y,
    3. Robinson G,
    4. Thompson MC,
    5. Currle DS,
    6. Eden C,
    7. et al.
    Subtypes of medulloblastoma have distinct developmental origins. Nature 2010;468:1095–9.
    OpenUrlCrossRefPubMed
  27. 27.↵
    1. MacDonald TJ,
    2. Brown KM,
    3. LaFleur B,
    4. Peterson K,
    5. Lawlor C,
    6. Chen Y,
    7. et al.
    Expression profiling of medulloblastoma: PDGFRA and the RAS/MAPK pathway as therapeutic targets for metastatic disease. Nat Genet 2001;29:143–52.
    OpenUrlCrossRefPubMed
  28. 28.↵
    1. Gilbertson RJ,
    2. Langdon JA,
    3. Hollander A,
    4. Hernan R,
    5. Hogg TL,
    6. Gajjar A,
    7. et al.
    Mutational analysis of PDGFR-RAS/MAPK pathway activation in childhood medulloblastoma. Eur J Cancer 2006;42:646–9.
    OpenUrlCrossRefPubMed
  29. 29.↵
    1. Fogarty MP,
    2. Emmenegger BA,
    3. Grasfeder LL,
    4. Oliver TG,
    5. Wechsler-Reya RJ
    . Fibroblast growth factor blocks Sonic hedgehog signaling in neuronal precursors and tumor cells. Proc Natl Acad Sci U S A 2007;104:2973–8.
    OpenUrlAbstract/FREE Full Text
  30. 30.↵
    1. Emmenegger BA,
    2. Hwang EI,
    3. Moore C,
    4. Markant SL,
    5. Brun SN,
    6. Dutton JW,
    7. et al.
    Distinct roles for fibroblast growth factor signaling in cerebellar development and medulloblastoma. Oncogene 2013;32:4181–8.
    OpenUrlCrossRefPubMed
  31. 31.↵
    1. Chang AL,
    2. Oro AE
    . Initial assessment of tumor regrowth after vismodegib in advanced Basal cell carcinoma. Arch Dermatol 2012;148:1324–5.
    OpenUrlCrossRefPubMed
  32. 32.↵
    1. Aasi S,
    2. Silkiss R,
    3. Tang JY,
    4. Wysong A,
    5. Liu A,
    6. Epstein E,
    7. et al.
    New onset of keratoacanthomas after vismodegib treatment for locally advanced basal cell carcinomas: a report of 2 cases. JAMA Dermatol 2013;149:242–3.
    OpenUrlCrossRefPubMed
  33. 33.↵
    1. Gill HS,
    2. Moscato EE,
    3. Chang AL,
    4. Soon S,
    5. Silkiss RZ
    . Vismodegib for periocular and orbital basal cell carcinoma. JAMA Ophthalmol 2013;131:1591–4.
    OpenUrlCrossRefPubMed
  34. 34.↵
    1. Iarrobino A,
    2. Messina JL,
    3. Kudchadkar R,
    4. Sondak VK
    . Emergence of a squamous cell carcinoma phenotype following treatment of metastatic basal cell carcinoma with vismodegib. J Am Acad Dermatol 2013;69:e33–4.
    OpenUrlCrossRefPubMed
  35. 35.↵
    1. Zhu GA,
    2. Sundram U,
    3. Chang AL
    . Two different scenarios of squamous cell carcinoma within advanced Basal cell carcinomas: cases illustrating the importance of serial biopsy during vismodegib usage. JAMA Dermatol 2014;150:970–3.
    OpenUrlCrossRefPubMed
  36. 36.↵
    1. Saintes C,
    2. Saint-Jean M,
    3. Brocard A,
    4. Peuvrel L,
    5. Renaut JJ,
    6. Khammari A,
    7. et al.
    Development of squamous cell carcinoma into basal cell carcinoma under treatment with Vismodegib. J Eur Acad Dermatol Venereol 2015;29:1006–9.
    OpenUrlCrossRefPubMed
  37. 37.↵
    1. Singh SK,
    2. Clarke ID,
    3. Hide T,
    4. Dirks PB
    . Cancer stem cells in nervous system tumors. Oncogene 2004;23:7267–73.
    OpenUrlCrossRefPubMed
  38. 38.↵
    1. Lee J,
    2. Kotliarova S,
    3. Kotliarov Y,
    4. Li A,
    5. Su Q,
    6. Donin NM,
    7. et al.
    Tumor stem cells derived from glioblastomas cultured in bFGF and EGF more closely mirror the phenotype and genotype of primary tumors than do serum-cultured cell lines. Cancer Cell 2006;9:391–403.
    OpenUrlCrossRefPubMed
  39. 39.↵
    1. Huang X,
    2. Ketova T,
    3. Litingtung Y,
    4. Chiang C
    . Isolation, enrichment, and maintenance of medulloblastoma stem cells. J Vis Exp 2010;1:2086.
    OpenUrl
  40. 40.↵
    1. Duplan SM,
    2. Theoret Y,
    3. Kenigsberg RL
    . Antitumor activity of fibroblast growth factors (FGFs) for medulloblastoma may correlate with FGF receptor expression and tumor variant. Clin Cancer Res 2002;8:246–57.
    OpenUrlAbstract/FREE Full Text
  41. 41.↵
    1. Ward RJ,
    2. Lee L,
    3. Graham K,
    4. Satkunendran T,
    5. Yoshikawa K,
    6. Ling E,
    7. et al.
    Multipotent CD15+ cancer stem cells in patched-1-deficient mouse medulloblastoma. Cancer Res 2009;69:4682–90.
    OpenUrlAbstract/FREE Full Text
  42. 42.↵
    1. Taylor MD,
    2. Liu L,
    3. Raffel C,
    4. Hui CC,
    5. Mainprize TG,
    6. Zhang X,
    7. et al.
    Mutations in SUFU predispose to medulloblastoma. Nat Genet 2002;31:306–10.
    OpenUrlCrossRefPubMed
  43. 43.↵
    1. Weiner HL,
    2. Bakst R,
    3. Hurlbert MS,
    4. Ruggiero J,
    5. Ahn E,
    6. Lee WS,
    7. et al.
    Induction of medulloblastomas in mice by sonic hedgehog, independent of Gli1. Cancer Res 2002;62:6385–9.
    OpenUrlAbstract/FREE Full Text
  44. 44.↵
    1. Bai CB,
    2. Auerbach W,
    3. Lee JS,
    4. Stephen D,
    5. Joyner AL
    . Gli2, but not Gli1, is required for initial Shh signaling and ectopic activation of the Shh pathway. Development 2002;129:4753–61.
    OpenUrlPubMed
  45. 45.↵
    1. Lusher ME,
    2. Lindsey JC,
    3. Latif F,
    4. Pearson AD,
    5. Ellison DW,
    6. Clifford SC
    . Biallelic epigenetic inactivation of the RASSF1A tumor suppressor gene in medulloblastoma development. Cancer Res 2002;62:5906–11.
    OpenUrlAbstract/FREE Full Text
  46. 46.↵
    1. Lindsey JC,
    2. Lusher ME,
    3. Anderton JA,
    4. Bailey S,
    5. Gilbertson RJ,
    6. Pearson AD,
    7. et al.
    Identification of tumour-specific epigenetic events in medulloblastoma development by hypermethylation profiling. Carcinogenesis 2004;25:661–8.
    OpenUrlAbstract/FREE Full Text
  47. 47.↵
    1. Engelman JA,
    2. Settleman J
    . Acquired resistance to tyrosine kinase inhibitors during cancer therapy. Curr Opin Genet Dev 2008;18:73–9.
    OpenUrlCrossRefPubMed
  48. 48.↵
    1. Sebens S,
    2. Schafer H
    . The tumor stroma as mediator of drug resistance—a potential target to improve cancer therapy? Curr Pharm Biotechnol 2012;13:2259–72.
    OpenUrlCrossRefPubMed
  49. 49.↵
    1. Snuderl M,
    2. Batista A,
    3. Kirkpatrick ND,
    4. Ruiz de Almodovar C,
    5. Riedemann L,
    6. Walsh EC,
    7. et al.
    Targeting placental growth factor/neuropilin 1 pathway inhibits growth and spread of medulloblastoma. Cell 2013;152:1065–76.
    OpenUrlCrossRefPubMed
  50. 50.↵
    1. Benazet JD,
    2. Zeller R
    . Vertebrate limb development: moving from classical morphogen gradients to an integrated 4-dimensional patterning system. Cold Spring Harb Perspect Biol 2009;1:a001339.
    OpenUrlAbstract/FREE Full Text
  51. 51.↵
    1. Bertrand N,
    2. Dahmane N
    . Sonic hedgehog signaling in forebrain development and its interactions with pathways that modify its effects. Trends Cell Biol 2006;16:597–605.
    OpenUrlCrossRefPubMed
  52. 52.↵
    1. Lauth M,
    2. Bergstrom A,
    3. Shimokawa T,
    4. Tostar U,
    5. Jin Q,
    6. Fendrich V,
    7. et al.
    DYRK1B-dependent autocrine-to-paracrine shift of Hedgehog signaling by mutant RAS. Nat Struct Mol Biol 2010;17:718–25.
    OpenUrlCrossRefPubMed
  53. 53.↵
    1. Takahashi K,
    2. Mitsui K,
    3. Yamanaka S
    . Role of ERas in promoting tumour-like properties in mouse embryonic stem cells. Nature 2003;423:541–5.
    OpenUrlCrossRefPubMed
  54. 54.↵
    1. Wu X,
    2. Northcott PA,
    3. Dubuc A,
    4. Dupuy AJ,
    5. Shih DJ,
    6. Witt H,
    7. et al.
    Clonal selection drives genetic divergence of metastatic medulloblastoma. Nature 2012;482:529–33.
    OpenUrlCrossRefPubMed
  55. 55.↵
    1. Mumert M,
    2. Dubuc A,
    3. Wu X,
    4. Northcott PA,
    5. Chin SS,
    6. Pedone CA,
    7. et al.
    Functional genomics identifies drivers of medulloblastoma dissemination. Cancer Res 2012;72:4944–53.
    OpenUrlAbstract/FREE Full Text
  56. 56.↵
    1. Ratushny V,
    2. Gober MD,
    3. Hick R,
    4. Ridky TW,
    5. Seykora JT
    . From keratinocyte to cancer: the pathogenesis and modeling of cutaneous squamous cell carcinoma. J Clin Invest 2012;122:464–72.
    OpenUrlCrossRefPubMed
PreviousNext
Back to top
Cancer Research: 75 (17)
September 2015
Volume 75, Issue 17
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover

Sign up for alerts

View this article with LENS

Open full page PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Cancer Research article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
RAS/MAPK Activation Drives Resistance to Smo Inhibition, Metastasis, and Tumor Evolution in Shh Pathway–Dependent Tumors
(Your Name) has forwarded a page to you from Cancer Research
(Your Name) thought you would be interested in this article in Cancer Research.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
RAS/MAPK Activation Drives Resistance to Smo Inhibition, Metastasis, and Tumor Evolution in Shh Pathway–Dependent Tumors
Xuesong Zhao, Tatyana Ponomaryov, Kimberly J. Ornell, Pengcheng Zhou, Sukriti K. Dabral, Ekaterina Pak, Wei Li, Scott X. Atwood, Ramon J. Whitson, Anne Lynn S. Chang, Jiang Li, Anthony E. Oro, Jennifer A. Chan, Joseph F. Kelleher and Rosalind A. Segal
Cancer Res September 1 2015 (75) (17) 3623-3635; DOI: 10.1158/0008-5472.CAN-14-2999-T

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
RAS/MAPK Activation Drives Resistance to Smo Inhibition, Metastasis, and Tumor Evolution in Shh Pathway–Dependent Tumors
Xuesong Zhao, Tatyana Ponomaryov, Kimberly J. Ornell, Pengcheng Zhou, Sukriti K. Dabral, Ekaterina Pak, Wei Li, Scott X. Atwood, Ramon J. Whitson, Anne Lynn S. Chang, Jiang Li, Anthony E. Oro, Jennifer A. Chan, Joseph F. Kelleher and Rosalind A. Segal
Cancer Res September 1 2015 (75) (17) 3623-3635; DOI: 10.1158/0008-5472.CAN-14-2999-T
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Introduction
    • Materials and Methods
    • Results
    • Discussion
    • Disclosure of Potential Conflicts of Interest
    • Authors' Contributions
    • Grant Support
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

  • mtDNA-Oncogene Cross-Talk
  • Alternative NFκB Regulates ALDH+ Ovarian Cancer Cells
  • R-Spondin1/LGR5 Activates TGFβ in Colon Cancer Metastasis
Show more Tumor and Stem Cell Biology
  • Home
  • Alerts
  • Feedback
  • Privacy Policy
Facebook  Twitter  LinkedIn  YouTube  RSS

Articles

  • Online First
  • Current Issue
  • Past Issues
  • Meeting Abstracts

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians

About Cancer Research

  • About the Journal
  • Editorial Board
  • Permissions
  • Submit a Manuscript
AACR logo

Copyright © 2021 by the American Association for Cancer Research.

Cancer Research Online ISSN: 1538-7445
Cancer Research Print ISSN: 0008-5472
Journal of Cancer Research ISSN: 0099-7013
American Journal of Cancer ISSN: 0099-7374

Advertisement