HHS Public Access Author manuscript J Invest Dermatol. Author manuscript; available in PMC 2016 February 01.

Similar documents
Developing Molecularly Targeted Therapies for Basal Cell Carcinoma. Ivor Caro, MD, FAAD

Genomic Analysis of Smoothened Inhibitor Resistance in Basal Cell Carcinoma

TARGET THERAPY IN NON GLIOMA BRAIN TUMORS.

Nature Genetics: doi: /ng Supplementary Figure 1. Mutational signatures in BCC compared to melanoma.

Historically, cryosurgery has been the treatment of choice

John Strasswimmer MD, PhD

Molecular pathology of ameloblastoma: towards targeted therapy

Protein Domain-Centric Approach to Study Cancer Somatic Mutations from High-throughput Sequencing Studies

Cutaneous Adnexal Tumors

Basal Cell Carcinoma, Hedgehog Signaling, and Targeted Therapeutics: The Long and Winding Road

Nature Getetics: doi: /ng.3471

Supplementary Figure 1: Features of IGLL5 Mutations in CLL: a) Representative IGV screenshot of first

NCCN Non-Small Cell Lung Cancer V Meeting June 15, 2018

Evan J. Lipson, M.D.

AVENIO ctdna Analysis Kits The complete NGS liquid biopsy solution EMPOWER YOUR LAB

7.014 Problem Set 7 Solutions

DOES THE BRCAX GENE EXIST? FUTURE OUTLOOK

Molecular docking approaches in identification of High affinity inhibitors of Human SMO receptor

The Biology and Genetics of Cells and Organisms The Biology of Cancer

Supplementary Figure 1: Classification scheme for non-synonymous and nonsense germline MC1R variants. The common variants with previously established

Inhibiting the Hedgehog Pathway in Patients with the Basal-Cell Nevus Syndrome

Introduction. Acta Medica Mediterranea, 2018, 34: 1441 ABSTRACT

Supporting Information

Multistep nature of cancer development. Cancer genes

Supplementary Figure 1

Odomzo. Odomzo (sonidegib) Description

NGS ONCOPANELS: FDA S PERSPECTIVE

Mayo School of Continuous Professional Development. Scott W. Fosko, M.D. Chair, Department of Dermatology Mayo Clinic Florida Jacksonville, Florida

MEDICAL GENOMICS LABORATORY. Next-Gen Sequencing and Deletion/Duplication Analysis of NF1 Only (NF1-NG)

AVENIO family of NGS oncology assays ctdna and Tumor Tissue Analysis Kits

Src-INACTIVE / Src-INACTIVE

A Comprehensive Study of TP53 Mutations in Chronic Lymphocytic Leukemia: Analysis of 1,287 Diagnostic CLL Samples

Contents. Preface XV Acknowledgments XXI List of Abbreviations XXIII About the Companion Website XXIX

Cancer Research Coordinating Committee Abstracts for Awards Supported Through California Cancer Research Voluntary Tax Contributions

Supplementary Figure 1

w ª wy xvwz A ª vw xvw P ª w} xvw w Æ w Æ V w,x Æ w Æ w Æ y,z Æ { Æ y,z, w w w~ w wy}æ zy Æ wyw{ xæ wz w xywæ xx Æ wv Æ } w x w x w Æ w Æ wy} zy Æ wz

When bad things happen to good genes: mutation vs. selection

THE UMD TP53 MUTATION DATABASE UPDATES AND BENEFITS. Pr. Thierry Soussi

Introduction to Cancer Bioinformatics and cancer biology. Anthony Gitter Cancer Bioinformatics (BMI 826/CS 838) January 20, 2015

Rational targeting of very high-risk medulloblastoma of childhood - 1 st progress report

University of Pittsburgh Annual Progress Report: 2008 Formula Grant

HHS Public Access Author manuscript J Invest Dermatol. Author manuscript; available in PMC 2012 June 01.

Cancer develops after somatic mutations overcome the multiple

OncoPPi Portal A Cancer Protein Interaction Network to Inform Therapeutic Strategies

Review (V1) - The Hallmarks of Cancer

p53 cooperates with DNA methylation and a suicidal interferon response to maintain epigenetic silencing of repeats and noncoding RNAs

Osamu Tetsu, MD, PhD Associate Professor Department of Otolaryngology-Head and Neck Surgery School of Medicine, University of California, San

NGS IN ONCOLOGY: FDA S PERSPECTIVE

Chapter 9. Cells Grow and Reproduce

University of Groningen. Hidradenitis suppurativa Dickinson-Blok, Janine Louise

Cancer troublemakers: a tale of usual suspects and novel villains

Computer Science, Biology, and Biomedical Informatics (CoSBBI) Outline. Molecular Biology of Cancer AND. Goals/Expectations. David Boone 7/1/2015

DeSigN: connecting gene expression with therapeutics for drug repurposing and development. Bernard lee GIW 2016, Shanghai 8 October 2016

Functional validation of cancer susceptibility genes using gene editing

Histones modifications and variants

underlying metastasis and recurrence in HNSCC, we analyzed two groups of patients. The

The lymphoma-associated NPM-ALK oncogene elicits a p16ink4a/prb-dependent tumor-suppressive pathway. Blood Jun 16;117(24):

Supporting Information

Ch. 18 Regulation of Gene Expression

IDENTIFICATION OF BIOMARKERS FOR EARLY DIAGNOSIS OF BREAST CANCER"

Qué hemos aprendido hasta hoy? What have we learned so far?

MCB140: Second Midterm Spring 2010

Toll-like Receptors (TLRs): Biology, Pathology and Therapeutics

Predictive Assays in Radiation Therapy

Hedgehog pathway mutations in T-cell acute lymphoblastic leukemia

Introduction to Genetics

News. Stanford Cancer Institute News. Drug Development from Bench to Bedside A Precision Approach to Skin Cancer Y Y

Session 4 Rebecca Poulos

Prokaryotes and eukaryotes alter gene expression in response to their changing environment

Biology Developmental Biology Spring Quarter Midterm 1 Version A

Bio 1M: Evolutionary processes

10/15/2012. Biologic Subtypes of TNBC. Topics. Topics. Histopathology Molecular pathology Clinical relevance

Review (V1) - The Hallmarks of Cancer

SUPPLEMENTARY INFORMATION

Supplementary Information Titles Journal: Nature Medicine

SUPPLEMENTARY INFORMATION

supplementary information

Accepted Manuscript. Pancreatic Cancer Subtypes: Beyond Lumping and Splitting. Andrew J. Aguirre

"BRCAness," PARP and the Triple-Negative Phenotype

Supplemental Figure legends

Discuss the impact of targeted molecular skin cancer therapies on dermatological surgery

Module 3: Pathway and Drug Development

Studying The Role Of DNA Mismatch Repair In Brain Cancer Malignancy

Problem Set 8 Key 1 of 8

Basal cell carcinoma: PD-L1/PD-1 checkpoint expression and tumor regression after PD-1 blockade

7.012 Quiz 3 Answers

EPIGENETIC RE-EXPRESSION OF HIF-2α SUPPRESSES SOFT TISSUE SARCOMA GROWTH

Early Embryonic Development

AP VP DLP H&E. p-akt DLP

The vast majority of the two to three million

ARTICLE RESEARCH. Macmillan Publishers Limited. All rights reserved

Journal: Nature Medicine

Problem Set 5 KEY

The Complexity of Simple Genetics

Supplementary Materials for

Cancer. The fundamental defect is. unregulated cell division. Properties of Cancerous Cells. Causes of Cancer. Altered growth and proliferation

Joanna Hillman Michael Higgins Lab Oncology for Scientists I 10/29/2015

The Shh Receptor Boc Promotes Progression of Early Medulloblastoma to Advanced Tumors

Transcription:

Rolling the genetic dice: neutral and deleterious Smoothened mutations in drug-resistant basal cell carcinoma Scott X. Atwood 1, Kavita Y. Sarin 1, Jiang R. Li 1, Catherine Yao 1, Nicole M. Urman 1, Anne Lynn S. Chang 1, Jean Y. Tang 1, and Anthony E. Oro 1 1 Program in Epithelial Biology and Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA To the editor HHS Public Access Author manuscript Published in final edited form as: J Invest Dermatol. 2015 August ; 135(8): 2138 2141. doi:10.1038/jid.2015.115. Basal cell carcinomas (BCCs) are locally invasive epithelial tumors that are caused by activating mutations in the Hedgehog (HH) pathway, typically through the loss of the receptor Patched1 or by activating the G-protein coupled receptor Smoothened (SMO). Genomic analysis by our group and others have revealed that BCCs are typically diploid and carry a high frequency of non-silent single nucleotide variants (SNVs) compared to other cutaneous and non-cutaneous tumors (Alexandrov et al. 2013; Atwood et al. 2014; Atwood et al. 2015; Jayaraman et al. 2014). Given their high mutational load, how these variants confer selective tumor growth without deleterious effects remains poorly understood. We previously identified and functionally validated nine SMO mutations that drive the majority of drug resistance in BCC through two distinct mechanisms that maintain HH signaling in the presence of drug: induction of constitutive activity or disruption of ligand binding (Atwood et al. 2015). However, SMO mutations with unclear function are frequently found across many HH and non-hh dependent cancers with drug-resistant BCCs bearing the highest rate of recurrent mutations at 66% (Figure 1a). To determine how these additional SMO mutations promote tumor growth, we identified 28 mutations through our genomic analysis of 44 drug-resistant and 36 sporadic BCC that were either recurrent, found to overlap with the COSMIC database, or were regional-specific (ligand binding pocket or pivot region) and interrogated their ability to promote HH signaling (Figure 1b, c). We expressed wildtype human SMO (SMO-WT) or SMO mutants in Smo-null mouse embryonic fibroblasts (MEFs) to assess the ability of these variants to activate the HH pathway with or without ligand. SMO-W535L is a known constitutively active mutant that was present in many of our tumor samples and significantly increased basal HH activity in the absence of HH ligand as determined by mrna levels of the HH target gene Gli1 (Figure 2a). No other SMO variant induced constitutive activity, including SMO-WT and the known ligand binding pocket mutant SMO-D473G (Yauch et al. 2009), Users may view, print, copy, and download text and data-mine the content in such documents, for the purposes of academic research, subject always to the full Conditions of use:http://www.nature.com/authors/editorial_policies/license.html#terms Address Correspondence to: Anthony E. Oro, oro@stanford.edu, 269 Campus Drive, CCSR 2140, Stanford, CA 94305, T: (650) 723-7843 F: (650) 725-8762. Conflict of Interest: AO and AC are investigators in Genentech, Novartis, and Eli Lilly clinical trials. JT is a consultant to Genentech.

Atwood et al. Page 2 suggesting these variants could not confer tumor growth by themselves. This was surprising as several of the residues (A327P, T336I, V414A, and T534I) lie in the pivot regions of transmembrane helices 3, 5, and 7 that control SMO activation (Figure 1c) and correspond to residues 320-340, 410-415, and 530-540 from the SMO crystal structure (Atwood et al. 2015; Wang et al. 2013). Addition of HH ligand revealed a range of responses from the SMO variants to activate the pathway. No SMO mutation conferred a statistically significant increase in SMO activity with the majority of variants acting as passenger mutations (Figure 2b). However, 13 variants disrupted SMO activity by 50% or more with 7 of the variants effectively abolishing activity. How the tumor could withstand the loss of SMO activity remains unclear, although only one functional copy of SMO is necessary to transduce HH signal. To assess the ability of the SMO variants to confer drug resistance to vismodegib, the current FDA-approved SMO antagonist, we added both HH ligand and 100 nm vismodegib to the Smo-null MEFs and assayed for HH activation. SMO-D473G and SMO-W535L both resisted SMO inhibition and displayed robust activation as expected (Figure 2c). However, no other SMO variant conferred vismodegib resistance, suggesting these mutations could not confer drug resistance and that the resistance mechanism in these tumors would be independent of SMO. This is also surprising as seven of these variants (L221P, V386A, C390R, V404M, N476K, E481G, and P513L) lie in the ligand binding pocket of SMO where vismodegib sits to repress protein function (Figure 1c). Because the concentration of vismodegib in our screening assay was roughly 12-fold above the IC 50 and data from our previous studies demonstrated that even small changes in IC 50 appeared to provide a growth advantage (Atwood et al. 2015), we assessed vismodegib sensitivity of the SMO mutants at low drug concentrations near the IC 50 of 10 nm and 20 nm. Using this more sensitive assay, SMO-D473G and SMO W535L maintained Gli1 mrna levels as expected, however the other SMO mutants displayed a vismodegib response similar to SMO-WT (Figure 2d). Altogether, our results reveal a surprising frequency of neutral and inactivating SMO variants in our drug-resistant BCC tumor population that provides a broader view to our recently described set of variants that confer drug resistance (Atwood et al. 2015). Our data supports a model where tumors are permissive to genetic mutations, generating many genetically diverse clones that compete as a way to grow. This ability to roll the genetic dice allows many mutations in key genes like SMO that would have activating, neutral, or negative effects on the cell. However, a small percentage of clones fortunate enough to contain activating mutations would continue to divide and contribute to a larger fraction of the tumor mass. Interestingly, SMO loss-of-function mutations would have no adverse effect on tumor growth as only one normal SMO gene is necessary to confer HH pathway activation, essentially making loss-of-function alleles similar to neutral mutations. Our functional studies included many variants that are recurrent in other genomic databases and argue against recurrent alleles necessarily imparting functional relevance. Rather, asymmetric distribution of variations could reflect bias in genome-wide chromatin accessibility or DNA repair mechanisms. On a cellular level, this suggests that individual tumor cells can be genetically distinct from each other and harbor many mutations, even in drivers like SMO, and have no negative effects on the growth of the tumor. Although our

Atwood et al. Page 3 Acknowledgments References Abbreviations results focus on the SMO locus, similar strategies may be operative at other genetic loci and tumors with high SNV frequencies may generate drug-resistance at a higher rate. Moreover, as we expand our use of high-throughput sequencing of tumors for personalized medicine, our results present a cautionary tale to functionally validate any mutation before concluding their ability to exert oncogenic effects. The work was funded by the V Foundation Translational Award and NIAMS 5ARO54780, 2AR046786 (AEO), NIH Pathway to Independence Award 1K99CA176847 (SXA), the Damon Runyon Clinical Investigator Award (JT), the American Skin Association Clinical Scholar Award (AC), the Stanford Cancer Institute Grant and the Dermatology Foundation Career Development Award (KS). Alexandrov LB, Nik-Zainal S, Wedge DC, et al. Signatures of mutational processes in human cancer. Nature. 2013; 500:415 21. [PubMed: 23945592] Atwood SX, Sarin KY, Whitson RJ, et al. Smoothened variants explain the majority of drug resistance in basal cell carcinoma. Cancer Cell. 2015; 27:342 53. [PubMed: 25759020] Atwood SX, Whitson RJ, Oro AE. Advanced treatment for basal cell carcinomas. Cold Spring Harb Perspect Med. 2014; 4:a013581. [PubMed: 24985127] Jayaraman SS, Rayhan DJ, Hazany S, et al. Mutational landscape of Basal cell carcinomas by wholeexome sequencing. Journal of Investigative Dermatology. 2014; 134:213 20. [PubMed: 23774526] Kool M, Jones DTW, Jäger N, et al. Genome Sequencing of SHH Medulloblastoma Predicts Genotype-Related Response to Smoothened Inhibition. Cancer Cell. 2014; 25:393 405. [PubMed: 24651015] Lee CS, Bhaduri A, Mah A, et al. Recurrent point mutations in the kinetochore gene KNSTRN in cutaneous squamous cell carcinoma. Nat Genet. 2014; 46:1060 2. [PubMed: 25194279] Sweeney RT, McClary AC, Myers BR, et al. Identification of recurrent SMO and BRAF mutations in ameloblastomas. Nat Genet. 2014; 46:722 5. [PubMed: 24859340] Wang C, Wu H, Katritch V, et al. Structure of the human smoothened receptor bound to an antitumour agent. Nature. 2013; 497:338 43. [PubMed: 23636324] Yauch RL, Dijkgraaf GJP, Alicke B, et al. Smoothened mutation confers resistance to ahedgehog pathway inhibitor in medulloblastoma. Science. 2009; 326:572 4. [PubMed: 19726788] BCC HH SMO SNV basal cell carcinoma Hedgehog Smoothened single nucleotide variant

Atwood et al. Page 4 Figure 1. Mutational profile of SMO in advanced basal cell carcinoma (a) Frequency of SMO mutations in various cancers from the COSMIC database and current literature (Atwood et al. 2015; Kool et al. 2014; Lee et al. 2014; Sweeney et al. 2014). (b) Number of tumors bearing recurrent, COSMIC database, or regional-specific (ligand binding pocket or pivot region) mutations. (c) Schematic of SMO showing location of mutations.

Atwood et al. Page 5 Figure 2. Variation in the response of SMO mutations to Hedgehog ligand (a) SMO variants expressed in Smo-null MEFs under serum-starvation conditions demonstrating baseline HH pathway activity. Note that previously characterized SMO mutations conferring vismodegib resistance (Atwood et al. 2015) were excluded from this analysis. (b) SMO variant expressing cells treated with SHH-N CM reveal a range of SMO activity to promote HH target gene induction. (c) SMO variant expressing cells treated with SHH-N CM and 100 nm vismodegib or (d) with or without 10 or 20 nm vismodegib show no apparent resistance to drug.