Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Stephen C. Mack is active.

Publication


Featured researches published by Stephen C. Mack.


Journal of Clinical Oncology | 2011

Medulloblastoma Comprises Four Distinct Molecular Variants

Paul A. Northcott; Andrey Korshunov; Hendrik Witt; Thomas Hielscher; Charles G. Eberhart; Stephen C. Mack; Eric Bouffet; Steven C. Clifford; Cynthia Hawkins; Pim J. French; James T. Rutka; Stefan Pfister; Michael D. Taylor

PURPOSE Recent genomic approaches have suggested the existence of multiple distinct subtypes of medulloblastoma. We studied a large cohort of medulloblastomas to determine how many subgroups of the disease exist, how they differ, and the extent of overlap between subgroups. METHODS We determined gene expression profiles and DNA copy number aberrations for 103 primary medulloblastomas. Bioinformatic tools were used for class discovery of medulloblastoma subgroups based on the most informative genes in the data set. Immunohistochemistry for subgroup-specific signature genes was used to determine subgroup affiliation for 294 nonoverlapping medulloblastomas on two independent tissue microarrays. RESULTS Multiple unsupervised analyses of transcriptional profiles identified the following four distinct, nonoverlapping molecular variants: WNT, SHH, group C, and group D. Supervised analysis of these four subgroups revealed significant subgroup-specific demographics, histology, metastatic status, and DNA copy number aberrations. Immunohistochemistry for DKK1 (WNT), SFRP1 (SHH), NPR3 (group C), and KCNA1 (group D) could reliably and uniquely classify formalin-fixed medulloblastomas in approximately 98% of patients. Group C patients (NPR3-positive tumors) exhibited a significantly diminished progression-free and overall survival irrespective of their metastatic status. CONCLUSION Our integrative genomics approach to a large cohort of medulloblastomas has identified four disparate subgroups with distinct demographics, clinical presentation, transcriptional profiles, genetic abnormalities, and clinical outcome. Medulloblastomas can be reliably assigned to subgroups through immunohistochemistry, thereby making medulloblastoma subclassification widely available. Future research on medulloblastoma and the development of clinical trials should take into consideration these four distinct types of medulloblastoma.


Nature Genetics | 2009

Multiple recurrent genetic events converge on control of histone lysine methylation in medulloblastoma

Paul A. Northcott; Yukiko Nakahara; Xiaochong Wu; Lars Feuk; David W. Ellison; Sid Croul; Stephen C. Mack; Paul N. Kongkham; John Peacock; Adrian Dubuc; Young Shin Ra; Karen Zilberberg; Jessica McLeod; Stephen W. Scherer; J. Sunil Rao; Charles G. Eberhart; Wiesia Grajkowska; Yancey Gillespie; Boleslaw Lach; Richard Grundy; Ian F. Pollack; Ronald L. Hamilton; Timothy Van Meter; Carlos Gilberto Carlotti; Frederick A. Boop; Darrell D. Bigner; Richard J. Gilbertson; James T. Rutka; Michael D. Taylor

We used high-resolution SNP genotyping to identify regions of genomic gain and loss in the genomes of 212 medulloblastomas, malignant pediatric brain tumors. We found focal amplifications of 15 known oncogenes and focal deletions of 20 known tumor suppressor genes (TSG), most not previously implicated in medulloblastoma. Notably, we identified previously unknown amplifications and homozygous deletions, including recurrent, mutually exclusive, highly focal genetic events in genes targeting histone lysine methylation, particularly that of histone 3, lysine 9 (H3K9). Post-translational modification of histone proteins is critical for regulation of gene expression, can participate in determination of stem cell fates and has been implicated in carcinogenesis. Consistent with our genetic data, restoration of expression of genes controlling H3K9 methylation greatly diminishes proliferation of medulloblastoma in vitro. Copy number aberrations of genes with critical roles in writing, reading, removing and blocking the state of histone lysine methylation, particularly at H3K9, suggest that defective control of the histone code contributes to the pathogenesis of medulloblastoma.


Genes & Development | 2015

Cancer stem cells in glioblastoma

Justin D. Lathia; Stephen C. Mack; Erin E. Mulkearns-Hubert; Claudia L.L. Valentim; Jeremy N. Rich

Tissues with defined cellular hierarchies in development and homeostasis give rise to tumors with cellular hierarchies, suggesting that tumors recapitulate specific tissues and mimic their origins. Glioblastoma (GBM) is the most prevalent and malignant primary brain tumor and contains self-renewing, tumorigenic cancer stem cells (CSCs) that contribute to tumor initiation and therapeutic resistance. As normal stem and progenitor cells participate in tissue development and repair, these developmental programs re-emerge in CSCs to support the development and progressive growth of tumors. Elucidation of the molecular mechanisms that govern CSCs has informed the development of novel targeted therapeutics for GBM and other brain cancers. CSCs are not self-autonomous units; rather, they function within an ecological system, both actively remodeling the microenvironment and receiving critical maintenance cues from their niches. To fulfill the future goal of developing novel therapies to collapse CSC dynamics, drawing parallels to other normal and pathological states that are highly interactive with their microenvironments and that use developmental signaling pathways will be beneficial.


Cancer Cell | 2011

Delineation of two clinically and molecularly distinct subgroups of posterior fossa ependymoma.

Hendrik Witt; Stephen C. Mack; Marina Ryzhova; Sebastian Bender; Martin Sill; Ruth Isserlin; Axel Benner; Thomas Hielscher; Till Milde; Marc Remke; David T. W. Jones; Paul A. Northcott; Livia Garzia; Kelsey C. Bertrand; Andrea Wittmann; Yuan Yao; Stephen S. Roberts; Luca Massimi; Tim Van Meter; William A. Weiss; Nalin Gupta; Wiesia Grajkowska; Boleslaw Lach; Yoon-Jae Cho; Andreas von Deimling; Andreas E. Kulozik; Olaf Witt; Gary D. Bader; Cynthia Hawkins; Uri Tabori

Despite the histological similarity of ependymomas from throughout the neuroaxis, the disease likely comprises multiple independent entities, each with a distinct molecular pathogenesis. Transcriptional profiling of two large independent cohorts of ependymoma reveals the existence of two demographically, transcriptionally, genetically, and clinically distinct groups of posterior fossa (PF) ependymomas. Group A patients are younger, have laterally located tumors with a balanced genome, and are much more likely to exhibit recurrence, metastasis at recurrence, and death compared with Group B patients. Identification and optimization of immunohistochemical (IHC) markers for PF ependymoma subgroups allowed validation of our findings on a third independent cohort, using a human ependymoma tissue microarray, and provides a tool for prospective prognostication and stratification of PF ependymoma patients.


Nature | 2010

Cross-species genomics matches driver mutations and cell compartments to model ependymoma

Robert A. Johnson; Karen Wright; Helen Poppleton; Kumarasamypet M. Mohankumar; David Finkelstein; Stanley Pounds; Vikki Rand; Sarah Leary; Elsie White; Christopher Eden; Twala L. Hogg; Paul A. Northcott; Stephen C. Mack; Geoffrey Neale; Yong Dong Wang; Beth Coyle; Jennifer M. Atkinson; Mariko DeWire; Tanya A. Kranenburg; Yancey Gillespie; Jeffrey C. Allen; Thomas E. Merchant; F.A. Boop; Robert A. Sanford; Amar Gajjar; David W. Ellison; Michael D. Taylor; Richard Grundy; Richard J. Gilbertson

Understanding the biology that underlies histologically similar but molecularly distinct subgroups of cancer has proven difficult because their defining genetic alterations are often numerous, and the cellular origins of most cancers remain unknown. We sought to decipher this heterogeneity by integrating matched genetic alterations and candidate cells of origin to generate accurate disease models. First, we identified subgroups of human ependymoma, a form of neural tumour that arises throughout the central nervous system (CNS). Subgroup-specific alterations included amplifications and homozygous deletions of genes not yet implicated in ependymoma. To select cellular compartments most likely to give rise to subgroups of ependymoma, we matched the transcriptomes of human tumours to those of mouse neural stem cells (NSCs), isolated from different regions of the CNS at different developmental stages, with an intact or deleted Ink4a/Arf locus (that encodes Cdkn2a and b). The transcriptome of human supratentorial ependymomas with amplified EPHB2 and deleted INK4A/ARF matched only that of embryonic cerebral Ink4a/Arf−/− NSCs. Notably, activation of Ephb2 signalling in these, but not other, NSCs generated the first mouse model of ependymoma, which is highly penetrant and accurately models the histology and transcriptome of one subgroup of human supratentorial tumour. Further, comparative analysis of matched mouse and human tumours revealed selective deregulation in the expression and copy number of genes that control synaptogenesis, pinpointing disruption of this pathway as a critical event in the production of this ependymoma subgroup. Our data demonstrate the power of cross-species genomics to meticulously match subgroup-specific driver mutations with cellular compartments to model and interrogate cancer subgroups.


Nature | 2014

Epigenomic alterations define lethal CIMP-positive ependymomas of infancy.

Stephen C. Mack; Hendrik Witt; Rosario M. Piro; Lei Gu; Scott Zuyderduyn; A. M. Stütz; Xiaosong Wang; Marco Gallo; Livia Garzia; Kory Zayne; Xiaoyang Zhang; Vijay Ramaswamy; Natalie Jäger; David T. W. Jones; Martin Sill; Trevor J. Pugh; M. Ryzhova; Khalida Wani; David Shih; Renee Head; Marc Remke; S. D. Bailey; Thomas Zichner; Claudia C. Faria; Mark Barszczyk; Sebastian Stark; Huriye Seker-Cin; Sonja Hutter; Pascal Johann; Sebastian Bender

Ependymomas are common childhood brain tumours that occur throughout the nervous system, but are most common in the paediatric hindbrain. Current standard therapy comprises surgery and radiation, but not cytotoxic chemotherapy as it does not further increase survival. Whole-genome and whole-exome sequencing of 47 hindbrain ependymomas reveals an extremely low mutation rate, and zero significant recurrent somatic single nucleotide variants. Although devoid of recurrent single nucleotide variants and focal copy number aberrations, poor-prognosis hindbrain ependymomas exhibit a CpG island methylator phenotype. Transcriptional silencing driven by CpG methylation converges exclusively on targets of the Polycomb repressive complex 2 which represses expression of differentiation genes through trimethylation of H3K27. CpG island methylator phenotype-positive hindbrain ependymomas are responsive to clinical drugs that target either DNA or H3K27 methylation both in vitro and in vivo. We conclude that epigenetic modifiers are the first rational therapeutic candidates for this deadly malignancy, which is epigenetically deregulated but genetically bland.


Journal of Clinical Oncology | 2014

Cytogenetic Prognostication Within Medulloblastoma Subgroups

David Shih; Paul A. Northcott; Marc Remke; Andrey Korshunov; Vijay Ramaswamy; Marcel Kool; Betty Luu; Yuan Yao; Xin Wang; Adrian Dubuc; Livia Garzia; John Peacock; Stephen C. Mack; Xiaochong Wu; Adi Rolider; A. Sorana Morrissy; Florence M.G. Cavalli; David T. W. Jones; Karel Zitterbart; Claudia C. Faria; Ulrich Schüller; Leos Kren; Toshihiro Kumabe; Teiji Tominaga; Young Shin Ra; Miklós Garami; Péter Hauser; Jennifer A. Chan; Shenandoah Robinson; László Bognár

PURPOSE Medulloblastoma comprises four distinct molecular subgroups: WNT, SHH, Group 3, and Group 4. Current medulloblastoma protocols stratify patients based on clinical features: patient age, metastatic stage, extent of resection, and histologic variant. Stark prognostic and genetic differences among the four subgroups suggest that subgroup-specific molecular biomarkers could improve patient prognostication. PATIENTS AND METHODS Molecular biomarkers were identified from a discovery set of 673 medulloblastomas from 43 cities around the world. Combined risk stratification models were designed based on clinical and cytogenetic biomarkers identified by multivariable Cox proportional hazards analyses. Identified biomarkers were tested using fluorescent in situ hybridization (FISH) on a nonoverlapping medulloblastoma tissue microarray (n = 453), with subsequent validation of the risk stratification models. RESULTS Subgroup information improves the predictive accuracy of a multivariable survival model compared with clinical biomarkers alone. Most previously published cytogenetic biomarkers are only prognostic within a single medulloblastoma subgroup. Profiling six FISH biomarkers (GLI2, MYC, chromosome 11 [chr11], chr14, 17p, and 17q) on formalin-fixed paraffin-embedded tissues, we can reliably and reproducibly identify very low-risk and very high-risk patients within SHH, Group 3, and Group 4 medulloblastomas. CONCLUSION Combining subgroup and cytogenetic biomarkers with established clinical biomarkers substantially improves patient prognostication, even in the context of heterogeneous clinical therapies. The prognostic significance of most molecular biomarkers is restricted to a specific subgroup. We have identified a small panel of cytogenetic biomarkers that reliably identifies very high-risk and very low-risk groups of patients, making it an excellent tool for selecting patients for therapy intensification and therapy de-escalation in future clinical trials.


Lancet Oncology | 2013

Methylation of the TERT promoter and risk stratification of childhood brain tumours: an integrative genomic and molecular study

Pedro Castelo-Branco; Sanaa Choufani; Stephen C. Mack; Denis Gallagher; Cindy Zhang; Tatiana Lipman; Nataliya Zhukova; Erin Walker; Dianna Martin; Diana Merino; Jonathan D. Wasserman; Cynthia Elizabeth; Noa Alon; Libo Zhang; Volker Hovestadt; Marcel Kool; David T. W. Jones; Gelareh Zadeh; Sidney Croul; Cynthia Hawkins; Johann Hitzler; Jean Cy Wang; Sylvain Baruchel; Peter Dirks; David Malkin; Stefan M. Pfister; Michael D. Taylor; Rosanna Weksberg; Uri Tabori

BACKGROUND Identification of robust biomarkers of malignancy and methods to establish disease progression is a major goal in paediatric neuro-oncology. We investigated whether methylation of the TERT promoter can be a biomarker for malignancy and patient outcome in paediatric brain tumours. METHODS For the discovery cohort, we used samples obtained from patients with paediatric brain tumours and individuals with normal brain tissues stored at the German Cancer Research Center (Heidelberg, Germany). We used methylation arrays for genome-wide assessment of DNA. For the validation cohort, we used samples obtained from several tissues for which full clinical and follow-up data were available from two hospitals in Toronto (ON, Canada). We did methylation analysis using quantitative Sequenom and pyrosequencing of an identified region of the TERT promoter. We assessed TERT expression by real-time PCR. To establish whether the biomarker could be used to assess and predict progression, we analysed methylation in paired samples of tumours that transformed from low to high grade and from localised to metastatic, and in choroid plexus tumours of different grades. Finally, we investigated overall survival in patients with posterior fossa ependymomas in which the identified region was hypermethylated or not. All individuals responsible for assays were masked to the outcome of the patients. FINDINGS Analysis of 280 samples in the discovery cohort identified one CpG site (cg11625005) in which 78 (99%) of 79 samples from normal brain tissues and low-grade tumours were not hypermethylated, but 145 (72%) of 201 samples from malignant tumours were hypermethylated (>15% methylated; p<0.0001). Analysis of 68 samples in the validation cohort identified a subset of five CpG sites (henceforth, upstream of the transcription start site [UTSS]) that was hypermethylated in all malignant paediatric brain tumours that expressed TERT but not in normal tissues that did not express TERT (p<0.0001). UTSS had a positive predictive value of 1.00 (95% CI 0.95-1.00) and a negative predictive value of 0.95 (0.87-0.99). In two paired samples of paediatric gliomas, UTSS methylation increased during transformation from low to high grade; it also increased in two paired samples that progressed from localised to metastatic disease. Two of eight atypical papillomas that had high UTSS methylation progressed to carcinomas, while the other six assessed did not progress or require additional treatment. 5-year overall survival was 51% (95% CI 31-71) for 25 patients with hypermethylated UTSS posterior fossa ependymomas and 95% (86-100) for 20 with non-hypermethylated tumours (p=0.0008). 5-year progression-free survival was 86% (68-100) for the 25 patients with non-hypermethylated UTSS tumours and 30% (10-50) for those with hypermethylated tumours (p=0.0008). INTERPRETATION Hypermethylation of the UTSS region in the TERT promoter is associated with TERT expression in cancers. In paediatric brain tumours, UTSS hypermethylation is associated with tumour progression and poor prognosis. This region is easy to amplify, and the assay to establish hypermethylation can be done on most tissues in most clinical laboratories. Therefore the UTSS region is a potentially accessible biomarker for various cancers. FUNDING The Canadian Institute of Health Research and the Terry Fox Foundation.


Cancer Research | 2016

A Three-Dimensional Organoid Culture System Derived from Human Glioblastomas Recapitulates the Hypoxic Gradients and Cancer Stem Cell Heterogeneity of Tumors Found In Vivo.

Christopher G. Hubert; Maricruz Rivera; Lisa C. Spangler; Qiulian Wu; Stephen C. Mack; Briana Prager; Marta Couce; Roger E. McLendon; Andrew E. Sloan; Jeremy N. Rich

Many cancers feature cellular hierarchies that are driven by tumor-initiating cancer stem cells (CSC) and rely on complex interactions with the tumor microenvironment. Standard cell culture conditions fail to recapitulate the original tumor architecture or microenvironmental gradients and are not designed to retain the cellular heterogeneity of parental tumors. Here, we describe a three-dimensional culture system that supports the long-term growth and expansion of tumor organoids derived directly from glioblastoma specimens, including patient-derived primary cultures, xenografts, genetically engineered glioma models, or patient samples. Organoids derived from multiple regions of patient tumors retain selective tumorigenic potential. Furthermore, organoids could be established directly from brain metastases not typically amenable to in vitro culture. Once formed, tumor organoids grew for months and displayed regional heterogeneity with a rapidly dividing outer region of SOX2(+), OLIG2(+), and TLX(+) cells surrounding a hypoxic core of primarily non-stem senescent cells and diffuse, quiescent CSCs. Notably, non-stem cells within organoids were sensitive to radiotherapy, whereas adjacent CSCs were radioresistant. Orthotopic transplantation of patient-derived organoids resulted in tumors displaying histologic features, including single-cell invasiveness, that were more representative of the parental tumor compared with those formed from patient-derived sphere cultures. In conclusion, we present a new ex vivo model in which phenotypically diverse stem and non-stem glioblastoma cell populations can be simultaneously cultured to explore new facets of microenvironmental influences and CSC biology. Cancer Res; 76(8); 2465-77. ©2016 AACR.


Childs Nervous System | 2009

The genetic and epigenetic basis of ependymoma.

Stephen C. Mack; Michael D. Taylor

IntroductionAlthough ependymoma is the third most common pediatric brain tumor, we know little about the genetic/epigenetic basis of its initiation, maintenance, or progression. This is due in part to the heterogeneity of the disease, as well as the small sample size of the cohorts analyzed in most studies.MethodsMany of the genetic aberrations identified to date are large genomic regions, making the differentiation between passenger and driver genes difficult. The finding of a balanced karyotype in a significant subset of pediatric posterior fossa ependymomas increases the difficulty of identifying targets for rationale therapy.ConclusionThe paucity of in vitro and in vivo model systems for ependymoma compound the difficulties outlined above. In this review, we discuss the published literature on ependymoma genetics and epigenetics and discuss possible future directions for the field.

Collaboration


Dive into the Stephen C. Mack's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jeremy N. Rich

University of California

View shared research outputs
Top Co-Authors

Avatar

Stefan M. Pfister

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xin Wang

University of Toronto

View shared research outputs
Top Co-Authors

Avatar

Andrey Korshunov

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge