Network


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

Hotspot


Dive into the research topics where Guillaume Bergthold is active.

Publication


Featured researches published by Guillaume Bergthold.


Clinical Cancer Research | 2014

BET Bromodomain Inhibition of MYC-Amplified Medulloblastoma

Pratiti Bandopadhayay; Guillaume Bergthold; Brian Nguyen; Simone Schubert; Sharareh Gholamin; Yujie Tang; Sara Bolin; Steven E. Schumacher; Rhamy Zeid; Sabran Masoud; Furong Yu; Nujsaubnusi Vue; William J. Gibson; Brenton R. Paolella; Siddhartha Mitra; Samuel H. Cheshier; Jun Qi; Kun-Wei Liu; Robert J. Wechsler-Reya; William A. Weiss; Fredrik J. Swartling; Mark W. Kieran; James E. Bradner; Rameen Beroukhim; Yoon-Jae Cho

Purpose: MYC-amplified medulloblastomas are highly lethal tumors. Bromodomain and extraterminal (BET) bromodomain inhibition has recently been shown to suppress MYC-associated transcriptional activity in other cancers. The compound JQ1 inhibits BET bromodomain-containing proteins, including BRD4. Here, we investigate BET bromodomain targeting for the treatment of MYC-amplified medulloblastoma. Experimental Design: We evaluated the effects of genetic and pharmacologic inhibition of BET bromodomains on proliferation, cell cycle, and apoptosis in established and newly generated patient- and genetically engineered mouse model (GEMM)-derived medulloblastoma cell lines and xenografts that harbored amplifications of MYC or MYCN. We also assessed the effect of JQ1 on MYC expression and global MYC-associated transcriptional activity. We assessed the in vivo efficacy of JQ1 in orthotopic xenografts established in immunocompromised mice. Results: Treatment of MYC-amplified medulloblastoma cells with JQ1 decreased cell viability associated with arrest at G1 and apoptosis. We observed downregulation of MYC expression and confirmed the inhibition of MYC-associated transcriptional targets. The exogenous expression of MYC from a retroviral promoter reduced the effect of JQ1 on cell viability, suggesting that attenuated levels of MYC contribute to the functional effects of JQ1. JQ1 significantly prolonged the survival of orthotopic xenograft models of MYC-amplified medulloblastoma (P < 0.001). Xenografts harvested from mice after five doses of JQ1 had reduced the expression of MYC mRNA and a reduced proliferative index. Conclusion: JQ1 suppresses MYC expression and MYC-associated transcriptional activity in medulloblastomas, resulting in an overall decrease in medulloblastoma cell viability. These preclinical findings highlight the promise of BET bromodomain inhibitors as novel agents for MYC-amplified medulloblastoma. Clin Cancer Res; 20(4); 912–25. ©2013 AACR.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Genomic analysis of diffuse pediatric low-grade gliomas identifies recurrent oncogenic truncating rearrangements in the transcription factor MYBL1

Lori A. Ramkissoon; Peleg Horowitz; Justin M. Craig; Shakti Ramkissoon; Benjamin E. Rich; Steven E. Schumacher; Aaron McKenna; Michael S. Lawrence; Guillaume Bergthold; Priscilla K. Brastianos; Barbara Tabak; Matthew Ducar; Paul Van Hummelen; Laura E. MacConaill; Tina Pouissant-Young; Yoon-Jae Cho; Hala Taha; Madeha Mahmoud; Daniel C. Bowers; Linda R. Margraf; Uri Tabori; Cynthia Hawkins; Roger J. Packer; D. Ashley Hill; Scott L. Pomeroy; Charles G. Eberhart; Ian F. Dunn; Liliana Goumnerova; Gad Getz; Jennifer A. Chan

Pediatric low-grade gliomas (PLGGs) are among the most common solid tumors in children but, apart from BRAF kinase mutations or duplications in specific subclasses, few genetic driver events are known. Diffuse PLGGs comprise a set of uncommon subtypes that exhibit invasive growth and are therefore especially challenging clinically. We performed high-resolution copy-number analysis on 44 formalin-fixed, paraffin-embedded diffuse PLGGs to identify recurrent alterations. Diffuse PLGGs exhibited fewer such alterations than adult low-grade gliomas, but we identified several significantly recurrent events. The most significant event, 8q13.1 gain, was observed in 28% of diffuse astrocytoma grade IIs and resulted in partial duplication of the transcription factor MYBL1 with truncation of its C-terminal negative-regulatory domain. A similar recurrent deletion-truncation breakpoint was identified in two angiocentric gliomas in the related gene v-myb avian myeloblastosis viral oncogene homolog (MYB) on 6q23.3. Whole-genome sequencing of a MYBL1-rearranged diffuse astrocytoma grade II demonstrated MYBL1 tandem duplication and few other events. Truncated MYBL1 transcripts identified in this tumor induced anchorage-independent growth in 3T3 cells and tumor formation in nude mice. Truncated transcripts were also expressed in two additional tumors with MYBL1 partial duplication. Our results define clinically relevant molecular subclasses of diffuse PLGGs and highlight a potential role for the MYB family in the biology of low-grade gliomas.


Nature Communications | 2015

Predicting clinical response to anticancer drugs using an ex vivo platform that captures tumour heterogeneity

Biswanath Majumder; Ulaganathan Baraneedharan; Saravanan Thiyagarajan; Padhma Radhakrishnan; Harikrishna Narasimhan; Muthu Dhandapani; Nilesh Brijwani; Dency D. Pinto; Arun Prasath; Basavaraja Shanthappa; Allen Thayakumar; Rajagopalan Surendran; Govind K. Babu; Ashok M. Shenoy; Moni A. Kuriakose; Guillaume Bergthold; Peleg Horowitz; Massimo Loda; Rameen Beroukhim; Shivani Agarwal; Shiladitya Sengupta; Pradip K. Majumder

Predicting clinical response to anticancer drugs remains a major challenge in cancer treatment. Emerging reports indicate that the tumour microenvironment and heterogeneity can limit the predictive power of current biomarker-guided strategies for chemotherapy. Here we report the engineering of personalized tumour ecosystems that contextually conserve the tumour heterogeneity, and phenocopy the tumour microenvironment using tumour explants maintained in defined tumour grade-matched matrix support and autologous patient serum. The functional response of tumour ecosystems, engineered from 109 patients, to anticancer drugs, together with the corresponding clinical outcomes, is used to train a machine learning algorithm; the learned model is then applied to predict the clinical response in an independent validation group of 55 patients, where we achieve 100% sensitivity in predictions while keeping specificity in a desired high range. The tumour ecosystem and algorithm, together termed the CANScript technology, can emerge as a powerful platform for enabling personalized medicine.


Pediatric Blood & Cancer | 2014

Long‐term outcome of 4,040 children diagnosed with pediatric low‐grade gliomas: An analysis of the Surveillance Epidemiology and End Results (SEER) database

Pratiti Bandopadhayay; Guillaume Bergthold; Wendy B. London; Liliana Goumnerova; Andres Morales La Madrid; Karen J. Marcus; Dongjing Guo; Nicole J. Ullrich; Nathan Robison; Susan N. Chi; Rameen Beroukhim; Mark W. Kieran; Peter Manley

Children with pediatric low‐grade gliomas (PLGG) are known to have excellent 10‐year survival rates; however the outcomes of adult survivors of PLGG are unknown. We identified patients diagnosed with PLGG diagnosed between 1973 and 2008 through the Surveillance Epidemiology and End Results (SEER) database to examine outcomes of adult survivors of PLGG.


Nature Genetics | 2016

MYB-QKI rearrangements in angiocentric glioma drive tumorigenicity through a tripartite mechanism.

Pratiti Bandopadhayay; Lori A. Ramkissoon; Payal Jain; Guillaume Bergthold; Jeremiah Wala; Rhamy Zeid; Steven E. Schumacher; Laura M. Urbanski; Ryan O'Rourke; William J. Gibson; Kristine Pelton; Shakti Ramkissoon; Harry J. Han; Yuankun Zhu; Namrata Choudhari; Amanda Silva; Katie Boucher; Rosemary E. Henn; Yun Jee Kang; David Knoff; Brenton R. Paolella; Adrianne Gladden-Young; Pascale Varlet; Mélanie Pagès; Peleg Horowitz; Alexander J. Federation; Hayley Malkin; Adam Tracy; Sara Seepo; Matthew Ducar

Angiocentric gliomas are pediatric low-grade gliomas (PLGGs) without known recurrent genetic drivers. We performed genomic analysis of new and published data from 249 PLGGs, including 19 angiocentric gliomas. We identified MYB-QKI fusions as a specific and single candidate driver event in angiocentric gliomas. In vitro and in vivo functional studies show that MYB-QKI rearrangements promote tumorigenesis through three mechanisms: MYB activation by truncation, enhancer translocation driving aberrant MYB-QKI expression and hemizygous loss of the tumor suppressor QKI. To our knowledge, this represents the first example of a single driver rearrangement simultaneously transforming cells via three genetic and epigenetic mechanisms in a tumor.


Biochimica et Biophysica Acta | 2014

Pediatric low-grade gliomas: How modern biology reshapes the clinical field

Guillaume Bergthold; Pratiti Bandopadhayay; Wenya Linda Bi; Lori A. Ramkissoon; Charles D. Stiles; Rosalind A. Segal; Rameen Beroukhim; Keith L. Ligon; Jacques Grill; Mark W. Kieran

Low-grade gliomas represent the most frequent brain tumors arising during childhood. They are characterized by a broad and heterogeneous group of tumors that are currently classified by the WHO according to their morphological appearance. Here we review the clinical features of these tumors, current therapeutic strategies and the recent discovery of genomic alterations characteristic to these tumors. We further explore how these recent biological findings stand to transform the treatment for these tumors and impact the diagnostic criteria for pediatric low-grade gliomas.


Anti-Cancer Drugs | 2015

Inhibition of the NOTCH pathway using γ-secretase inhibitor RO4929097 has limited antitumor activity in established glial tumors.

Carmela Dantas-Barbosa; Guillaume Bergthold; Estelle Daudigeos-Dubus; Heike Blockus; John F. Boylan; Céline Ferreira; Stéphanie Puget; Michel Abely; Gilles Vassal; Jacques Grill; Birgit Geoerger

Notch signaling is altered in many cancers. Our previous findings in primary pediatric ependymoma support a role for NOTCH in glial oncogenesis. The present study evaluates the γ-secretase inhibitor RO4929097 in glial tumor models. The expression of Notch pathway genes was evaluated using real-time RT-PCR in 21 ependymoma and glioma models. NOTCH1 mutations were analyzed by DNA sequencing. RO4929097 activity was evaluated in vitro and in vivo, as a single agent and in combination, in glioma and ependymoma models. Notch pathway genes are overexpressed in ependymomas and gliomas along with FBXW7 downregulation. NOTCH1 mutations in the TAD domain were observed in 20% (2/10) of ependymoma primary cultures. Blocking the Notch pathway with the γ-secretase inhibitor RO4929097 reduced cell density and viability in ependymoma short-term cultures. When combined with chemotherapeutic agents, RO4929097 enhanced temozolomide effects in ependymoma short-term cultures and potentiated the cytotoxicity of etoposide, cisplatinum, and temozolomide in glioma cells. RO4929097, in combined treatment with mTOR inhibition, potentiated cytotoxicity in vitro, but did not enhance antitumor effects in vivo. In contrast, RO4929097 enhanced irradiation effects in glioma and ependymoma xenografts and showed tumor growth inhibition in advanced-stage IGRG121 glioblastoma xenografts. RO4929097-mediated effects were independent of NOTCH1 mutation status or expression levels, but associated with low IL-6 levels. In established glial tumor models, NOTCH inhibition had limited effects as a single agent, but enhanced efficacy when combined with DNA-interfering agents. These preclinical data need to be considered for further clinical development of NOTCH inhibitors in glial tumors.


Current Opinion in Oncology | 2011

Pediatric ependymomas: will molecular biology change patient management?

Jacques Grill; Guillaume Bergthold; Céline Ferreira

Purpose of review Ependymomas remain a therapeutic challenge in pediatric neuro-oncology. These tumors are chemoresistant and rather radioresistant and until recently little was known about their biology. Recent findings Histopathological grading of ependymomas according to the WHO classification is neither reproducible, nor correlated with outcome, especially in young children. Characterization of molecular abnormalities in ependymomas offers now a better understanding of their initiation and progression; different biological subtypes of tumors have been described and would need further validation. The identification of new prognostic biomarkers, such as tenascin-C overexpression or chromosome 1q gain, will considerably help patient stratification in future trials. Finally, the recent discovery of specific pathways involved in ependymomas oncogenesis, such as Notch-1or EPHB2 offers new perspectives for the development of targeted therapies. Summary A comprehensive biological work-out including CGHarray and immunohistochemistry for specific biomarkers should now be recommended for the current management of pediatric ependymoma, especially in young children if radiotherapy has to be omitted in the first line of treatment.


Cancer Research | 2016

Abstract 4372: MYB-QKI rearrangements in angiocentric glioma drive tumorigenicity through a tripartite mechanism

Lori A. Ramkissoon; Pratiti Bandopadhayay; Payal Jain; Guillaume Bergthold; Adam C. Resnick; Rameen Beroukhim; Keith L. Ligon

Diffuse pediatric low-grade gliomas (PLGGs) are among the most common solid tumors in children. While BRAF mutations and MYBL1 rearrangements have recently been identified as diagnostic and treatment related oncogenic drivers in pediatric gangliogliomas and diffuse astrocytomas, respectively, for the majority of diffuse PLGGs the oncogenic driver(s) remain unknown. Recent efforts to identify new oncogenic drivers lack sufficient power to determine the true frequency of alterations in genes or to associate specific alterations with rare histological subtypes, such as Angiocentric gliomas (AGs). To address this issue, we performed genomic analysis of new and published data from 249 PLGGs including 19 AGs. We identified MYB-QKI fusions as a specific, recurrent, single candidate driver event in AGs. Although MYB is expressed during early brain development in a subset of progenitor cells, it is not known to play a role in normal cortical brain where AGs frequently occur. In vitro functional studies show MYB-QKI rearrangements promote tumorigenesis in mouse neural stem cells (mNSCs) through three mechanisms: MYB activation by truncation, enhancer translocation driving aberrant MYB-QKI expression, and hemizygous loss of the tumor suppressor QKI. Expression of the MYB-QKI fusion increases mNSC proliferation and activates known MYB target genes including KIT and CDK6. H3K27ac enhancer profiling of AG tumors demonstrate active enhancer elements are translocated proximally to the MYB promoter while in vitro promoter assays confirmed that QKI enhancer sequences activate the MYB promoter. Together with functional studies demonstrating that MYB-QKI can also bind and activate the MYB promoter, these data support a positive auto-regulatory feed-back loop model in which active MYB-QKI is able to drive its own expression through enhanced MYB-promoter-activation. Furthermore, when injected orthotopically into immunodeficient mice, expression of truncated MYB or MYB-QKI is sufficient to induce tumor formation that recapitulate histologic and immunophenotypic characteristics of AGs. Analysis of AG specific MYB-QKI fusion represents the first example of how a single driver rearrangement simultaneously transforms cells via three genetic and epigenetic mechanisms in a cancer. Citation Format: Lori Ramkissoon, Pratiti Bandopadhayay, Payal Jain, Guillaume Bergthold, Adam Resnick, Rameen Beroukhim, Keith Ligon. MYB-QKI rearrangements in angiocentric glioma drive tumorigenicity through a tripartite mechanism. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 4372.


Cancer Cell | 2014

Genome sequencing of SHH medulloblastoma predicts genotype-related response to smoothened inhibition

Marcel Kool; David T. W. Jones; Natalie Jäger; Paul A. Northcott; Trevor J. Pugh; Volker Hovestadt; Rosario M. Piro; L. Adriana Esparza; Shirley L. Markant; Marc Remke; Till Milde; Franck Bourdeaut; Marina Ryzhova; Dominik Sturm; Elke Pfaff; Sebastian Stark; Sonja Hutter; Huriye Şeker-Cin; Pascal Johann; Sebastian Bender; Christin Schmidt; Tobias Rausch; David Shih; Jüri Reimand; Laura Sieber; Andrea Wittmann; Linda Linke; Hendrik Witt; Ursula Weber; Marc Zapatka

Collaboration


Dive into the Guillaume Bergthold's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jacques Grill

Centre national de la recherche scientifique

View shared research outputs
Researchain Logo
Decentralizing Knowledge