Network


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

Hotspot


Dive into the research topics where Catherine J. Wu is active.

Publication


Featured researches published by Catherine J. Wu.


Nature | 2013

Mutational heterogeneity in cancer and the search for new cancer-associated genes.

Michael S. Lawrence; Petar Stojanov; Paz Polak; Gregory V. Kryukov; Kristian Cibulskis; Andrey Sivachenko; Scott L. Carter; Chip Stewart; Craig H. Mermel; Steven A. Roberts; Adam Kiezun; Peter S. Hammerman; Aaron McKenna; Yotam Drier; Lihua Zou; Alex H. Ramos; Trevor J. Pugh; Nicolas Stransky; Elena Helman; Jaegil Kim; Carrie Sougnez; Lauren Ambrogio; Elizabeth Nickerson; Erica Shefler; Maria L. Cortes; Daniel Auclair; Gordon Saksena; Douglas Voet; Michael S. Noble; Daniel DiCara

Major international projects are underway that are aimed at creating a comprehensive catalogue of all the genes responsible for the initiation and progression of cancer. These studies involve the sequencing of matched tumour–normal samples followed by mathematical analysis to identify those genes in which mutations occur more frequently than expected by random chance. Here we describe a fundamental problem with cancer genome studies: as the sample size increases, the list of putatively significant genes produced by current analytical methods burgeons into the hundreds. The list includes many implausible genes (such as those encoding olfactory receptors and the muscle protein titin), suggesting extensive false-positive findings that overshadow true driver events. We show that this problem stems largely from mutational heterogeneity and provide a novel analytical methodology, MutSigCV, for resolving the problem. We apply MutSigCV to exome sequences from 3,083 tumour–normal pairs and discover extraordinary variation in mutation frequency and spectrum within cancer types, which sheds light on mutational processes and disease aetiology, and in mutation frequency across the genome, which is strongly correlated with DNA replication timing and also with transcriptional activity. By incorporating mutational heterogeneity into the analyses, MutSigCV is able to eliminate most of the apparent artefactual findings and enable the identification of genes truly associated with cancer.


Cell | 2013

Evolution and Impact of Subclonal Mutations in Chronic Lymphocytic Leukemia

Dan A. Landau; Scott L. Carter; Petar Stojanov; Aaron McKenna; Kristen E. Stevenson; Michael S. Lawrence; Carrie Sougnez; Chip Stewart; Andrey Sivachenko; Lili Wang; Youzhong Wan; Wandi Zhang; Sachet A. Shukla; Alexander R. Vartanov; Stacey M. Fernandes; Gordon Saksena; Kristian Cibulskis; Bethany Tesar; Stacey Gabriel; Nir Hacohen; Matthew Meyerson; Eric S. Lander; Donna Neuberg; Jennifer R. Brown; Gad Getz; Catherine J. Wu

Clonal evolution is a key feature of cancer progression and relapse. We studied intratumoral heterogeneity in 149 chronic lymphocytic leukemia (CLL) cases by integrating whole-exome sequence and copy number to measure the fraction of cancer cells harboring each somatic mutation. We identified driver mutations as predominantly clonal (e.g., MYD88, trisomy 12, and del(13q)) or subclonal (e.g., SF3B1 and TP53), corresponding to earlier and later events in CLL evolution. We sampled leukemia cells from 18 patients at two time points. Ten of twelve CLL cases treated with chemotherapy (but only one of six without treatment) underwent clonal evolution, predominantly involving subclones with driver mutations (e.g., SF3B1 and TP53) that expanded over time. Furthermore, presence of a subclonal driver mutation was an independent risk factor for rapid disease progression. Our study thus uncovers patterns of clonal evolution in CLL, providing insights into its stepwise transformation, and links the presence of subclones with adverse clinical outcomes.


The New England Journal of Medicine | 2011

SF3B1 and Other Novel Cancer Genes in Chronic Lymphocytic Leukemia

Lili Wang; Michael S. Lawrence; Youzhong Wan; Petar Stojanov; Carrie Sougnez; Kristen E. Stevenson; Lillian Werner; Andrey Sivachenko; David S. DeLuca; Li Zhang; Wandi Zhang; Alexander R. Vartanov; Stacey M. Fernandes; Natalie R. Goldstein; Eric G. Folco; Kristian Cibulskis; Bethany Tesar; Quinlan L. Sievers; Erica Shefler; Stacey B Gabriel; Nir Hacohen; Robin Reed; Matthew Meyerson; Todd R. Golub; Eric S. Lander; Donna Neuberg; Jennifer R. Brown; Gad Getz; Catherine J. Wu

BACKGROUND The somatic genetic basis of chronic lymphocytic leukemia, a common and clinically heterogeneous leukemia occurring in adults, remains poorly understood. METHODS We obtained DNA samples from leukemia cells in 91 patients with chronic lymphocytic leukemia and performed massively parallel sequencing of 88 whole exomes and whole genomes, together with sequencing of matched germline DNA, to characterize the spectrum of somatic mutations in this disease. RESULTS Nine genes that are mutated at significant frequencies were identified, including four with established roles in chronic lymphocytic leukemia (TP53 in 15% of patients, ATM in 9%, MYD88 in 10%, and NOTCH1 in 4%) and five with unestablished roles (SF3B1, ZMYM3, MAPK1, FBXW7, and DDX3X). SF3B1, which functions at the catalytic core of the spliceosome, was the second most frequently mutated gene (with mutations occurring in 15% of patients). SF3B1 mutations occurred primarily in tumors with deletions in chromosome 11q, which are associated with a poor prognosis in patients with chronic lymphocytic leukemia. We further discovered that tumor samples with mutations in SF3B1 had alterations in pre-messenger RNA (mRNA) splicing. CONCLUSIONS Our study defines the landscape of somatic mutations in chronic lymphocytic leukemia and highlights pre-mRNA splicing as a critical cellular process contributing to chronic lymphocytic leukemia.


Science | 2015

Genomic correlates of response to CTLA-4 blockade in metastatic melanoma

Eliezer M. Van Allen; Diana Miao; Bastian Schilling; Sachet A. Shukla; Christian U. Blank; Lisa Zimmer; Antje Sucker; Uwe Hillen; Marnix H. Geukes Foppen; Simone M. Goldinger; Jochen Utikal; Jessica C. Hassel; Benjamin Weide; Katharina C. Kaehler; Carmen Loquai; Peter Mohr; Ralf Gutzmer; Reinhard Dummer; Stacey Gabriel; Catherine J. Wu; Dirk Schadendorf; Levi A. Garraway

Is cancer immunotherapy a private affair? Immune checkpoint blockade, a relatively new cancer treatment, substantially extends the survival of a subset of patients. Previous work has shown that patients whose tumors harbor the largest number of mutations—and thus produce a large number of “neoantigens” recognized as foreign by the immune system—are most likely to benefit. Expanding on these earlier studies, Van Allen et al. studied over 100 patients with melanoma and found a similar correlation (see the Perspective by Gubin and Schreiber). There was no evidence, however, that specific neoantigen sequences were shared by patients who responded. Science, this issue p. 207, see also p. 158 Melanoma patients who respond to immunotherapy do not appear to share common tumor neoantigens. [Also see Perspective by Gubin and Schreiber ] Monoclonal antibodies directed against cytotoxic T lymphocyte–associated antigen-4 (CTLA-4), such as ipilimumab, yield considerable clinical benefit for patients with metastatic melanoma by inhibiting immune checkpoint activity, but clinical predictors of response to these therapies remain incompletely characterized. To investigate the roles of tumor-specific neoantigens and alterations in the tumor microenvironment in the response to ipilimumab, we analyzed whole exomes from pretreatment melanoma tumor biopsies and matching germline tissue samples from 110 patients. For 40 of these patients, we also obtained and analyzed transcriptome data from the pretreatment tumor samples. Overall mutational load, neoantigen load, and expression of cytolytic markers in the immune microenvironment were significantly associated with clinical benefit. However, no recurrent neoantigen peptide sequences predicted responder patient populations. Thus, detailed integrated molecular characterization of large patient cohorts may be needed to identify robust determinants of response and resistance to immune checkpoint inhibitors.


Science | 2016

Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade

Nicholas McGranahan; Andrew Furness; Rachel Rosenthal; Sofie Ramskov; Rikke Birgitte Lyngaa; Sunil Kumar Saini; Mariam Jamal-Hanjani; Gareth A. Wilson; Nicolai Juul Birkbak; Crispin Hiley; Thomas B.K. Watkins; Seema Shafi; Nirupa Murugaesu; Richard Mitter; Ayse U. Akarca; Joseph Linares; Teresa Marafioti; Jake Y. Henry; Eliezer M. Van Allen; Diana Miao; Bastian Schilling; Dirk Schadendorf; Levi A. Garraway; Vladimir Makarov; Naiyer A. Rizvi; Alexandra Snyder; Matthew D. Hellmann; Taha Merghoub; Jedd D. Wolchok; Sachet A. Shukla

The cellular ancestry of tumor antigens One contributing factor in antitumor immunity is the repertoire of neoantigens created by genetic mutations within tumor cells. Like the corresponding mutations, these neoantigens show intratumoral heterogeneity. Some are present in all tumor cells (clonal), and others are present in only a fraction of cells (subclonal). In a study of lung cancer and melanoma, McGranahan et al. found that a high burden of clonal tumor neoantigens correlated with improved patient survival, an increased presence of tumor-infiltrating lymphocytes, and a durable response to immunotherapy. Science, this issue p. 1463 Analysis of the cellular ancestry of tumor neoantigens can predict which are most likely to induce an immune response. As tumors grow, they acquire mutations, some of which create neoantigens that influence the response of patients to immune checkpoint inhibitors. We explored the impact of neoantigen intratumor heterogeneity (ITH) on antitumor immunity. Through integrated analysis of ITH and neoantigen burden, we demonstrate a relationship between clonal neoantigen burden and overall survival in primary lung adenocarcinomas. CD8+ tumor-infiltrating lymphocytes reactive to clonal neoantigens were identified in early-stage non–small cell lung cancer and expressed high levels of PD-1. Sensitivity to PD-1 and CTLA-4 blockade in patients with advanced NSCLC and melanoma was enhanced in tumors enriched for clonal neoantigens. T cells recognizing clonal neoantigens were detectable in patients with durable clinical benefit. Cytotoxic chemotherapy–induced subclonal neoantigens, contributing to an increased mutational load, were enriched in certain poor responders. These data suggest that neoantigen heterogeneity may influence immune surveillance and support therapeutic developments targeting clonal neoantigens.


Nature | 2015

Mutations driving CLL and their evolution in progression and relapse

Dan A. Landau; Eugen Tausch; Amaro Taylor-Weiner; Chip Stewart; Johannes G. Reiter; Jasmin Bahlo; Sandra Kluth; Ivana Bozic; Michael S. Lawrence; Sebastian Böttcher; Scott L. Carter; Kristian Cibulskis; Daniel Mertens; Carrie Sougnez; Mara Rosenberg; Julian Hess; Jennifer Edelmann; Sabrina Kless; Michael Kneba; Matthias Ritgen; Anna Maria Fink; Kirsten Fischer; Stacey Gabriel; Eric S. Lander; Martin A. Nowak; Hartmut Döhner; Michael Hallek; Donna Neuberg; Gad Getz; Stephan Stilgenbauer

Which genetic alterations drive tumorigenesis and how they evolve over the course of disease and therapy are central questions in cancer biology. Here we identify 44 recurrently mutated genes and 11 recurrent somatic copy number variations through whole-exome sequencing of 538 chronic lymphocytic leukaemia (CLL) and matched germline DNA samples, 278 of which were collected in a prospective clinical trial. These include previously unrecognized putative cancer drivers (RPS15, IKZF3), and collectively identify RNA processing and export, MYC activity, and MAPK signalling as central pathways involved in CLL. Clonality analysis of this large data set further enabled reconstruction of temporal relationships between driver events. Direct comparison between matched pre-treatment and relapse samples from 59 patients demonstrated highly frequent clonal evolution. Thus, large sequencing data sets of clinically informative samples enable the discovery of novel genes associated with cancer, the network of relationships between the driver events, and their impact on disease relapse and clinical outcome.


Nature | 2017

An immunogenic personal neoantigen vaccine for patients with melanoma

Patrick A. Ott; Zhuting Hu; Derin B. Keskin; Sachet A. Shukla; Jing Sun; David J. Bozym; Wandi Zhang; Adrienne M. Luoma; Anita Giobbie-Hurder; Lauren Peter; Christina Chen; Oriol Olive; Todd A. Carter; Shuqiang Li; David J. Lieb; Thomas Eisenhaure; Evisa Gjini; Jonathan Stevens; William J. Lane; Indu Javeri; Kaliappanadar Nellaiappan; Andres M. Salazar; Heather Daley; Michael S. Seaman; Elizabeth I. Buchbinder; Charles H. Yoon; Maegan Harden; Niall J. Lennon; Stacey Gabriel; Scott J. Rodig

Effective anti-tumour immunity in humans has been associated with the presence of T cells directed at cancer neoantigens, a class of HLA-bound peptides that arise from tumour-specific mutations. They are highly immunogenic because they are not present in normal tissues and hence bypass central thymic tolerance. Although neoantigens were long-envisioned as optimal targets for an anti-tumour immune response, their systematic discovery and evaluation only became feasible with the recent availability of massively parallel sequencing for detection of all coding mutations within tumours, and of machine learning approaches to reliably predict those mutated peptides with high-affinity binding of autologous human leukocyte antigen (HLA) molecules. We hypothesized that vaccination with neoantigens can both expand pre-existing neoantigen-specific T-cell populations and induce a broader repertoire of new T-cell specificities in cancer patients, tipping the intra-tumoural balance in favour of enhanced tumour control. Here we demonstrate the feasibility, safety, and immunogenicity of a vaccine that targets up to 20 predicted personal tumour neoantigens. Vaccine-induced polyfunctional CD4+ and CD8+ T cells targeted 58 (60%) and 15 (16%) of the 97 unique neoantigens used across patients, respectively. These T cells discriminated mutated from wild-type antigens, and in some cases directly recognized autologous tumour. Of six vaccinated patients, four had no recurrence at 25 months after vaccination, while two with recurrent disease were subsequently treated with anti-PD-1 (anti-programmed cell death-1) therapy and experienced complete tumour regression, with expansion of the repertoire of neoantigen-specific T cells. These data provide a strong rationale for further development of this approach, alone and in combination with checkpoint blockade or other immunotherapies.


Nature Genetics | 2016

Distinct patterns of somatic genome alterations in lung adenocarcinomas and squamous cell carcinomas

Joshua D. Campbell; Anton Alexandrov; Jaegil Kim; Jeremiah Wala; Alice H. Berger; Chandra Sekhar Pedamallu; Sachet A. Shukla; Guangwu Guo; Angela N. Brooks; Bradley A. Murray; Marcin Imielinski; Xin Hu; Shiyun Ling; Rehan Akbani; Mara Rosenberg; Carrie Cibulskis; Eric A. Collisson; David J. Kwiatkowski; Michael S. Lawrence; John N. Weinstein; Roel G.W. Verhaak; Catherine J. Wu; Peter S. Hammerman; Andrew D. Cherniack; Gad Getz; Maxim N. Artyomov; Robert D. Schreiber; Ramaswamy Govindan; Matthew Meyerson

To compare lung adenocarcinoma (ADC) and lung squamous cell carcinoma (SqCC) and to identify new drivers of lung carcinogenesis, we examined the exome sequences and copy number profiles of 660 lung ADC and 484 lung SqCC tumor–normal pairs. Recurrent alterations in lung SqCCs were more similar to those of other squamous carcinomas than to alterations in lung ADCs. New significantly mutated genes included PPP3CA, DOT1L, and FTSJD1 in lung ADC, RASA1 in lung SqCC, and KLF5, EP300, and CREBBP in both tumor types. New amplification peaks encompassed MIR21 in lung ADC, MIR205 in lung SqCC, and MAPK1 in both. Lung ADCs lacking receptor tyrosine kinase–Ras–Raf pathway alterations had mutations in SOS1, VAV1, RASA1, and ARHGAP35. Regarding neoantigens, 47% of the lung ADC and 53% of the lung SqCC tumors had at least five predicted neoepitopes. Although targeted therapies for lung ADC and SqCC are largely distinct, immunotherapies may aid in treatment for both subtypes.


Blood | 2014

Systematic identification of personal tumor-specific neoantigens in chronic lymphocytic leukemia.

Mohini Rajasagi; Sachet A. Shukla; Edward F. Fritsch; Derin B. Keskin; David S. DeLuca; Ellese M. Carmona; Wandi Zhang; Carrie Sougnez; Kristian Cibulskis; John Sidney; Kristen E. Stevenson; Jerome Ritz; Donna Neuberg; Vladimir Brusic; Stacey Gabriel; Eric S. Lander; Gad Getz; Nir Hacohen; Catherine J. Wu

Genome sequencing has revealed a large number of shared and personal somatic mutations across human cancers. In principle, any genetic alteration affecting a protein-coding region has the potential to generate mutated peptides that are presented by surface HLA class I proteins that might be recognized by cytotoxic T cells. To test this possibility, we implemented a streamlined approach for the prediction and validation of such neoantigens derived from individual tumors and presented by patient-specific HLA alleles. We applied our computational pipeline to 91 chronic lymphocytic leukemias (CLLs) that underwent whole-exome sequencing (WES). We predicted ∼22 mutated HLA-binding peptides per leukemia (derived from ∼16 missense mutations) and experimentally confirmed HLA binding for ∼55% of such peptides. Two CLL patients that achieved long-term remission following allogeneic hematopoietic stem cell transplantation were monitored for CD8(+) T-cell responses against predicted or confirmed HLA-binding peptides. Long-lived cytotoxic T-cell responses were detected against peptides generated from personal tumor mutations in ALMS1, C6ORF89, and FNDC3B presented on tumor cells. Finally, we applied our computational pipeline to WES data (N = 2488 samples) across 13 different cancer types and estimated dozens to thousands of predicted neoantigens per individual tumor, suggesting that neoantigens are frequent in most tumors.


Cell Reports | 2016

Genomic Correlates of Immune-Cell Infiltrates in Colorectal Carcinoma

Marios Giannakis; Xinmeng Jasmine Mu; Sachet A. Shukla; Zhi Rong Qian; Ofir Cohen; Reiko Nishihara; Samira Bahl; Yin Cao; Ali Amin-Mansour; Mai Yamauchi; Yasutaka Sukawa; Chip Stewart; Mara Rosenberg; Kosuke Mima; Kentaro Inamura; Katsuhiko Nosho; Jonathan A. Nowak; Michael S. Lawrence; Edward Giovannucci; Andrew T. Chan; Kimmie Ng; Jeffrey A. Meyerhardt; Eliezer M. Van Allen; Gad Getz; Stacey Gabriel; Eric S. Lander; Catherine J. Wu; Charles S. Fuchs; Shuji Ogino; Levi A. Garraway

Summary Large-scale genomic characterization of tumors from prospective cohort studies may yield new insights into cancer pathogenesis. We performed whole-exome sequencing of 619 incident colorectal cancers (CRCs) and integrated the results with tumor immunity, pathology, and survival data. We identified recurrently mutated genes in CRC, such as BCL9L, RBM10, CTCF, and KLF5, that were not previously appreciated in this disease. Furthermore, we investigated the genomic correlates of immune-cell infiltration and found that higher neoantigen load was positively associated with overall lymphocytic infiltration, tumor-infiltrating lymphocytes (TILs), memory T cells, and CRC-specific survival. The association with TILs was evident even within microsatellite-stable tumors. We also found positive selection of mutations in HLA genes and other components of the antigen-processing machinery in TIL-rich tumors. These results may inform immunotherapeutic approaches in CRC. More generally, this study demonstrates a framework for future integrative molecular epidemiology research in colorectal and other malignancies.

Collaboration


Dive into the Catherine J. Wu's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robert J. Soiffer

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Edwin P. Alyea

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge