Network


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

Hotspot


Dive into the research topics where Ian Majewski is active.

Publication


Featured researches published by Ian Majewski.


Nucleic Acids Research | 2011

ChIP-seq analysis reveals distinct H3K27me3 profiles that correlate with transcriptional activity

Matthew D. Young; Tracy A. Willson; Matthew J. Wakefield; Evelyn Trounson; Douglas J. Hilton; Marnie E. Blewitt; Alicia Oshlack; Ian Majewski

Transcriptional control is dependent on a vast network of epigenetic modifications. One epigenetic mark of particular interest is tri-methylation of lysine 27 on histone H3 (H3K27me3), which is catalysed and maintained by Polycomb Repressive Complex 2 (PRC2). Although this histone mark is studied widely, the precise relationship between its local pattern of enrichment and regulation of gene expression is currently unclear. We have used ChIP-seq to generate genome-wide maps of H3K27me3 enrichment, and have identified three enrichment profiles with distinct regulatory consequences. First, a broad domain of H3K27me3 enrichment across the body of genes corresponds to the canonical view of H3K27me3 as inhibitory to transcription. Second, a peak of enrichment around the transcription start site (TSS) is commonly associated with ‘bivalent’ genes, where H3K4me3 also marks the TSS. Finally and most surprisingly, we identified an enrichment profile with a peak in the promoter of genes that is associated with active transcription. Genes with each of these three profiles were found in different proportions in each of the cell types studied. The data analysis techniques developed here will be useful for the identification of common enrichment profiles for other histone modifications that have important consequences for transcriptional regulation.


International Journal of Cancer | 2014

Colorectal cancer intrinsic subtypes predict chemotherapy benefit, deficient mismatch repair and epithelial-to-mesenchymal transition.

Paul Roepman; Andreas Schlicker; Josep Tabernero; Ian Majewski; Sun Tian; Victor Moreno; Mireille Snel; Christine M. Chresta; Robert D. Rosenberg; Ulrich Nitsche; Teresa Macarulla; Gabriel Capellá; Ramon Salazar; George Orphanides; Lodewyk F. A. Wessels; René Bernards; Iris Simon

In most colorectal cancer (CRC) patients, outcome cannot be predicted because tumors with similar clinicopathological features can have differences in disease progression and treatment response. Therefore, a better understanding of the CRC biology is required to identify those patients who will benefit from chemotherapy and to find a more tailored therapy plan for other patients. Based on unsupervised classification of whole genome data from 188 stages I–IV CRC patients, a molecular classification was developed that consist of at least three major intrinsic subtypes (A‐, B‐ and C‐type). The subtypes were validated in 543 stages II and III patients and were associated with prognosis and benefit from chemotherapy. The heterogeneity of the intrinsic subtypes is largely based on three biological hallmarks of the tumor: epithelial‐to‐mesenchymal transition, deficiency in mismatch repair genes that result in high mutation frequency associated with microsatellite instability and cellular proliferation. A‐type tumors, observed in 22% of the patients, have the best prognosis, have frequent BRAF mutations and a deficient DNA mismatch repair system. C‐type patients (16%) have the worst outcome, a mesenchymal gene expression phenotype and show no benefit from adjuvant chemotherapy treatment. Both A‐type and B‐type tumors have a more proliferative and epithelial phenotype and B‐types benefit from adjuvant chemotherapy. B‐type tumors (62%) show a low overall mutation frequency consistent with the absence of DNA mismatch repair deficiency. Classification based on molecular subtypes made it possible to expand and improve CRC classification beyond standard molecular and immunohistochemical assessment and might help in the future to guide treatment in CRC patients.


Journal of Clinical Oncology | 2015

PIK3CA Mutations Are Associated With Decreased Benefit to Neoadjuvant Human Epidermal Growth Factor Receptor 2–Targeted Therapies in Breast Cancer

Ian Majewski; Paolo Nuciforo; Lorenza Mittempergher; Astrid Bosma; Holger Eidtmann; Eileen Holmes; Christos Sotiriou; Debora Fumagalli; Jose L. Jimenez; Claudia Aura; Ludmila Prudkin; Maria Carmen Díaz-Delgado; Lorena de la Peña; Sherene Loi; Catherine E. Ellis; Nikolaus Schultz; Evandro de Azambuja; Nadia Harbeck; Martine Piccart-Gebhart; René Bernards; José Baselga

PURPOSE We investigated whether mutations in the gene encoding the phosphatidylinositol 3-kinase (PI3K) catalytic subunit (PIK3CA) correlates with response to neoadjuvant human epidermal growth factor receptor 2 (HER2) -targeted therapies in patients with breast cancer. PATIENTS AND METHODS Baseline tissue biopsies were available from patients with HER2-positive early breast cancer who were enrolled onto the Neoadjuvant Lapatinib and/or Trastuzumab Treatment Optimization trial (NeoALTTO). Activating mutations in PIK3CA were identified using mass spectrometry-based genotyping. RESULTS PIK3CA mutations were identified in 23% of HER2-positive breast tumors, and these mutations were associated with poorer outcome in all of the treatment arms. Patients treated with a combination of trastuzumab and lapatinib who had wild-type PIK3CA obtained a total pathologic complete response (pCR) rate of 53.1%, which decreased to 28.6% in patients with tumors that carried PIK3CA activating mutations (P = .012). CONCLUSION Activating mutations in PIK3CA predicted poor pCR in patients with HER2-positive breast cancer treated with neoadjuvant therapies that target HER2. Consequently, the combination of anti-HER2 agents and PI3K inhibitors is being investigated.


Blood | 2010

Opposing roles of polycomb repressive complexes in hematopoietic stem and progenitor cells.

Ian Majewski; Matthew E. Ritchie; Belinda Phipson; Jason Corbin; Miha Pakusch; Anja Ebert; Meinrad Busslinger; Haruhiko Koseki; Yifang Hu; Gordon K. Smyth; Warren S. Alexander; Douglas J. Hilton; Marnie E. Blewitt

Polycomb group (PcG) proteins are transcriptional repressors with a central role in the establishment and maintenance of gene expression patterns during development. We have investigated the role of polycomb repressive complexes (PRCs) in hematopoietic stem cells (HSCs) and progenitor populations. We show that mice with loss of function mutations in PRC2 components display enhanced HSC/progenitor population activity, whereas mutations that disrupt PRC1 or pleiohomeotic repressive complex are associated with HSC/progenitor cell defects. Because the hierarchical model of PRC action would predict synergistic effects of PRC1 and PRC2 mutation, these opposing effects suggest this model does not hold true in HSC/progenitor cells. To investigate the molecular targets of each complex in HSC/progenitor cells, we measured genome-wide expression changes associated with PRC deficiency, and identified transcriptional networks that are differentially regulated by PRC1 and PRC2. These studies provide new insights into the mechanistic interplay between distinct PRCs and have important implications for approaching PcG proteins as therapeutic targets.


PLOS Biology | 2008

Polycomb Repressive Complex 2 (PRC2) Restricts Hematopoietic Stem Cell Activity

Ian Majewski; Marnie E. Blewitt; Carolyn A. de Graaf; Edward J. McManus; Melanie Bahlo; Adrienne A. Hilton; Craig D. Hyland; Gordon K. Smyth; Jason Corbin; Donald Metcalf; Warren S. Alexander; Douglas J. Hilton

Polycomb group proteins are transcriptional repressors that play a central role in the establishment and maintenance of gene expression patterns during development. Using mice with an N-ethyl-N-nitrosourea (ENU)-induced mutation in Suppressor of Zeste 12 (Suz12), a core component of Polycomb Repressive Complex 2 (PRC2), we show here that loss of Suz12 function enhances hematopoietic stem cell (HSC) activity. In addition to these effects on a wild-type genetic background, mutations in Suz12 are sufficient to ameliorate the stem cell defect and thrombocytopenia present in mice that lack the thrombopoietin receptor (c-Mpl). To investigate the molecular targets of the PRC2 complex in the HSC compartment, we examined changes in global patterns of gene expression in cells deficient in Suz12. We identified a distinct set of genes that are regulated by Suz12 in hematopoietic cells, including eight genes that appear to be highly responsive to PRC2 function within this compartment. These data suggest that PRC2 is required to maintain a specific gene expression pattern in hematopoiesis that is indispensable to normal stem cell function.


The Journal of Pathology | 2013

Identification of recurrent FGFR3 fusion genes in lung cancer through kinome‐centred RNA sequencing

Ian Majewski; Lorenza Mittempergher; Nadia M Davidson; Astrid Bosma; Stefan M. Willems; Hugo M. Horlings; Iris de Rink; Liliana Greger; Gerrit K.J. Hooijer; Dennis Peters; Petra M. Nederlof; Ingrid Hofland; Jeroen de Jong; Jelle Wesseling; Roelof Jc Kluin; Wim Brugman; Ron M. Kerkhoven; Frank Nieboer; Paul Roepman; Annegien Broeks; Thomas Muley; Jacek Jassem; Jacek Niklinski; Nico van Zandwijk; Alvis Brazma; Alicia Oshlack; Michel M. van den Heuvel; René Bernards

Oncogenic fusion genes that involve kinases have proven to be effective targets for therapy in a wide range of cancers. Unfortunately, the diagnostic approaches required to identify these events are struggling to keep pace with the diverse array of genetic alterations that occur in cancer. Diagnostic screening in solid tumours is particularly challenging, as many fusion genes occur with a low frequency. To overcome these limitations, we developed a capture enrichment strategy to enable high‐throughput transcript sequencing of the human kinome. This approach provides a global overview of kinase fusion events, irrespective of the identity of the fusion partner. To demonstrate the utility of this system, we profiled 100 non‐small cell lung cancers and identified numerous genetic alterations impacting fibroblast growth factor receptor 3 (FGFR3) in lung squamous cell carcinoma and a novel ALK fusion partner in lung adenocarcinoma.


Nature Medicine | 2011

Taming the dragon: genomic biomarkers to individualize the treatment of cancer

Ian Majewski; René Bernards

The gradual shift from cytotoxic drugs to highly selective, targeted therapeutic agents for cancer requires a parallel effort to characterize cancers at the molecular level to guide the choice of therapy for the individual patient. Here we review the genomic technologies that can be used to develop these drug response indicators, or biomarkers. We also discuss hurdles in their development and the implementation of biomarkers in clinical practice.


The Annals of Applied Statistics | 2016

Robust hyperparameter estimation protects against hypervariable genes and improves power to detect differential expression

Belinda Phipson; Stanley Chun-Wei Lee; Ian Majewski; Warren S. Alexander; Gordon K. Smyth

One of the most common analysis tasks in genomic research is to identify genes that are differentially expressed (DE) between experimental conditions. Empirical Bayes (EB) statistical tests using moderated genewise variances have been very effective for this purpose, especially when the number of biological replicate samples is small. The EB procedures can however be heavily influenced by a small number of genes with very large or very small variances. This article improves the differential expression tests by robustifying the hyperparameter estimation procedure. The robust procedure has the effect of decreasing the informativeness of the prior distribution for outlier genes while increasing its informativeness for other genes. This effect has the double benefit of reducing the chance that hypervariable genes will be spuriously identified as DE while increasing statistical power for the main body of genes. The robust EB algorithm is fast and numerically stable. The procedure allows exact small-sample null distributions for the test statistics and reduces exactly to the original EB procedure when no outlier genes are present. Simulations show that the robustified tests have similar performance to the original tests in the absence of outlier genes but have greater power and robustness when outliers are present. The article includes case studies for which the robust method correctly identifies and downweights genes associated with hidden covariates and detects more genes likely to be scientifically relevant to the experimental conditions. The new procedure is implemented in the limma software package freely available from the Bioconductor repository.


Blood | 2016

The BCL2 selective inhibitor venetoclax induces rapid onset apoptosis of CLL cells in patients via a TP53-independent mechanism

Mary Ann Anderson; Jing Deng; John F. Seymour; Constantine S. Tam; Su Young Kim; Joshua Fein; Lijian Yu; Jennifer R. Brown; David Westerman; Eric G. Si; Ian Majewski; David Segal; Sari H. Enschede; David C. S. Huang; Matthew S. Davids; Anthony Letai; Andrew W. Roberts

BCL2 blunts activation of the mitochondrial pathway to apoptosis, and high-level expression is required for chronic lymphocytic leukemia (CLL) survival. Venetoclax (ABT-199) is a small-molecule selective inhibitor of BCL2 currently in clinical trials for CLL and other malignancies. In conjunction with the phase 1 first-in-human clinical trial of venetoclax in patients with relapsed or refractory CLL (M12-175), we investigated the mechanism of action of venetoclax in vivo, explored whether in vitro sensitivity assays or BH3 profiling correlated with in vivo responses in patients, and determined whether loss of TP53 function affected responses in vitro and in vivo. In all samples tested, venetoclax induced death of CLL cells in vitro at concentrations achievable in vivo, with cell death evident within 4 hours. Apoptotic CLL cells were detected in vivo 6 or 24 hours after a single 20-mg or 50-mg dose in some patients. The extent of mitochondrial depolarization by a BIM BH3 peptide in vitro was correlated with percentage reduction of CLL in the blood and bone marrow in vivo, whereas the half lethal concentration derived from standard cytotoxicity assays was not. CLL cell death in vitro and the depth of clinical responses were independent of deletion of chromosome 17p, TP53 mutation, and TP53 function. These data provide direct evidence that venetoclax kills CLL cells in a TP53-independent fashion by inhibition of BCL2 in patients and support further assessment of BH3 profiling as a predictive biomarker for this drug.


The Journal of Pathology | 2013

An α‐E‐catenin (CTNNA1) mutation in hereditary diffuse gastric cancer

Ian Majewski; Irma Kluijt; Annemieke Cats; Thomas Scerri; Daphne de Jong; Roelof Jc Kluin; Samantha Hansford; Frans B. L. Hogervorst; Astrid Bosma; Ingrid Hofland; Marcel Winter; David Huntsman; Jos Jonkers; Melanie Bahlo; René Bernards

Diffuse gastric cancers typically present as late‐stage tumours and, as a result, the 5 year survival rate is poor. Some gastric cancers are hereditary and these tend to be of the diffuse type; 30–40% of hereditary diffuse gastric cancers (HDGCs) can be explained by defective germline alleles of E‐cadherin (CDH1), but for the remaining families the factors driving susceptibility remain unknown. We had access to a large HDGC pedigree with no obvious mutation in CDH1, and applied exome sequencing to identify new genes involved in gastric cancer. We identified a germline truncating allele of α‐E‐catenin (CTNNA1) that was present in two family members with invasive diffuse gastric cancer and four in which intramucosal signet ring cells were detected as part of endoscopic surveillance. The remaining CTNNA1 allele was silenced in the two diffuse gastric cancers from the family that were available for screening, and this was also true for signet ring cells identified in endoscopic biopsies. Since α‐E‐catenin functions in the same complex as E‐cadherin, our results call attention to the broader signalling network surrounding these proteins in HDGC. We also detected somatic mutations in one tumour and found substantial overlap with genes mutated in sporadic gastric cancer, including PIK3CA, ARID1A, MED12 and MED23.

Collaboration


Dive into the Ian Majewski's collaboration.

Top Co-Authors

Avatar

Warren S. Alexander

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

René Bernards

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Astrid Bosma

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Marnie E. Blewitt

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tesa Severson

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Christoffer Flensburg

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Gordon K. Smyth

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge