Network


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

Hotspot


Dive into the research topics where Thomas B.K. Watkins is active.

Publication


Featured researches published by Thomas B.K. Watkins.


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.


The New England Journal of Medicine | 2017

Tracking the Evolution of Non–Small-Cell Lung Cancer

Mariam Jamal-Hanjani; Gareth A. Wilson; Nicholas McGranahan; Nicolai Juul Birkbak; Thomas B.K. Watkins; Selvaraju Veeriah; Seema Shafi; Diana Johnson; Richard Mitter; Rachel Rosenthal; Max Salm; Stuart Horswell; Mickael Escudero; Nik Matthews; Andrew Rowan; Tim Chambers; David Moore; Samra Turajlic; Hang Xu; Siow Ming Lee; Martin Forster; Tanya Ahmad; Crispin Hiley; Christopher Abbosh; Mary Falzon; Elaine Borg; Teresa Marafioti; David Lawrence; Martin Hayward; Shyam Kolvekar

BACKGROUND Among patients with non‐small‐cell lung cancer (NSCLC), data on intratumor heterogeneity and cancer genome evolution have been limited to small retrospective cohorts. We wanted to prospectively investigate intratumor heterogeneity in relation to clinical outcome and to determine the clonal nature of driver events and evolutionary processes in early‐stage NSCLC. METHODS In this prospective cohort study, we performed multiregion whole‐exome sequencing on 100 early‐stage NSCLC tumors that had been resected before systemic therapy. We sequenced and analyzed 327 tumor regions to define evolutionary histories, obtain a census of clonal and subclonal events, and assess the relationship between intratumor heterogeneity and recurrence‐free survival. RESULTS We observed widespread intratumor heterogeneity for both somatic copy‐number alterations and mutations. Driver mutations in EGFR, MET, BRAF, and TP53 were almost always clonal. However, heterogeneous driver alterations that occurred later in evolution were found in more than 75% of the tumors and were common in PIK3CA and NF1 and in genes that are involved in chromatin modification and DNA damage response and repair. Genome doubling and ongoing dynamic chromosomal instability were associated with intratumor heterogeneity and resulted in parallel evolution of driver somatic copy‐number alterations, including amplifications in CDK4, FOXA1, and BCL11A. Elevated copy‐number heterogeneity was associated with an increased risk of recurrence or death (hazard ratio, 4.9; P=4.4×10‐4), which remained significant in multivariate analysis. CONCLUSIONS Intratumor heterogeneity mediated through chromosome instability was associated with an increased risk of recurrence or death, a finding that supports the potential value of chromosome instability as a prognostic predictor. (Funded by Cancer Research UK and others; TRACERx ClinicalTrials.gov number, NCT01888601.)


Cancer Discovery | 2015

Tracking the genomic evolution of esophageal adenocarcinoma through neoadjuvant chemotherapy

Nirupa Murugaesu; Gareth A. Wilson; Nicolai Juul Birkbak; Thomas B.K. Watkins; Nicholas McGranahan; Sacheen Kumar; Nima Abbassi-Ghadi; Max Salm; Richard Mitter; Stuart Horswell; Andrew Rowan; Benjamin Phillimore; Jennifer Biggs; Sharmin Begum; Nik Matthews; Daniel Hochhauser; George B. Hanna; Charles Swanton

UNLABELLED Esophageal adenocarcinomas are associated with a dismal prognosis. Deciphering the evolutionary history of this disease may shed light on therapeutically tractable targets and reveal dynamic mutational processes during the disease course and following neoadjuvant chemotherapy (NAC). We exome sequenced 40 tumor regions from 8 patients with operable esophageal adenocarcinomas, before and after platinum-containing NAC. This revealed the evolutionary genomic landscape of esophageal adenocarcinomas with the presence of heterogeneous driver mutations, parallel evolution, early genome-doubling events, and an association between high intratumor heterogeneity and poor response to NAC. Multiregion sequencing demonstrated a significant reduction in thymine to guanine mutations within a CpTpT context when comparing early and late mutational processes and the presence of a platinum signature with enrichment of cytosine to adenine mutations within a CpC context following NAC. Esophageal adenocarcinomas are characterized by early chromosomal instability leading to amplifications containing targetable oncogenes persisting through chemotherapy, providing a rationale for future therapeutic approaches. SIGNIFICANCE This work illustrates dynamic mutational processes occurring during esophageal adenocarcinoma evolution and following selective pressures of platinum exposure, emphasizing the iatrogenic impact of therapy on cancer evolution. Identification of amplifications encoding targetable oncogenes maintained through NAC suggests the presence of stable vulnerabilities, unimpeded by cytotoxics, suitable for therapeutic intervention.


Nature | 2018

Chromosomal instability drives metastasis through a cytosolic DNA response

Samuel F. Bakhoum; Bryan Ngo; Ashley M. Laughney; Julie-Ann Cavallo; Charles J. Murphy; Peter Ly; Pragya Shah; Roshan K. Sriram; Thomas B.K. Watkins; Neil K. Taunk; Mercedes Duran; Chantal Pauli; Christine Shaw; Kalyani Chadalavada; Vinagolu K. Rajasekhar; Giulio Genovese; Subramanian Venkatesan; Nicolai Juul Birkbak; Nicholas McGranahan; Mark R. Lundquist; Quincey LaPlant; John H. Healey; Olivier Elemento; Christine H. Chung; Nancy Y. Lee; Marcin Imielenski; Gouri Nanjangud; Dana Pe’er; Don W. Cleveland; Simon N. Powell

Chromosomal instability is a hallmark of cancer that results from ongoing errors in chromosome segregation during mitosis. Although chromosomal instability is a major driver of tumour evolution, its role in metastasis has not been established. Here we show that chromosomal instability promotes metastasis by sustaining a tumour cell-autonomous response to cytosolic DNA. Errors in chromosome segregation create a preponderance of micronuclei whose rupture spills genomic DNA into the cytosol. This leads to the activation of the cGAS–STING (cyclic GMP-AMP synthase–stimulator of interferon genes) cytosolic DNA-sensing pathway and downstream noncanonical NF-κB signalling. Genetic suppression of chromosomal instability markedly delays metastasis even in highly aneuploid tumour models, whereas continuous chromosome segregation errors promote cellular invasion and metastasis in a STING-dependent manner. By subverting lethal epithelial responses to cytosolic DNA, chromosomally unstable tumour cells co-opt chronic activation of innate immune pathways to spread to distant organs.


Cell | 2018

Tracking Cancer Evolution Reveals Constrained Routes to Metastases: TRACERx Renal

Samra Turajlic; Hang Xu; Kevin Litchfield; Andrew Rowan; Tim Chambers; José I. López; David Nicol; Tim O’Brien; James Larkin; Stuart Horswell; Mark Stares; Lewis Au; Mariam Jamal-Hanjani; Ben Challacombe; Ashish Chandra; Steve Hazell; Claudia Eichler-Jonsson; Aspasia Soultati; Simon Chowdhury; Sarah Rudman; Joanna Lynch; Archana Fernando; Gordon Stamp; Emma Nye; Faiz Jabbar; Lavinia Spain; Sharanpreet Lall; Rosa Guarch; Mary Falzon; Ian Proctor

Summary Clear-cell renal cell carcinoma (ccRCC) exhibits a broad range of metastatic phenotypes that have not been systematically studied to date. Here, we analyzed 575 primary and 335 metastatic biopsies across 100 patients with metastatic ccRCC, including two cases sampledat post-mortem. Metastatic competence was afforded by chromosome complexity, and we identify 9p loss as a highly selected event driving metastasis and ccRCC-related mortality (p = 0.0014). Distinct patterns of metastatic dissemination were observed, including rapid progression to multiple tissue sites seeded by primary tumors of monoclonal structure. By contrast, we observed attenuated progression in cases characterized by high primary tumor heterogeneity, with metastatic competence acquired gradually and initial progression to solitary metastasis. Finally, we observed early divergence of primitive ancestral clones and protracted latency of up to two decades as a feature of pancreatic metastases.


Nature Genetics | 2017

Evolution and clinical impact of co-occurring genetic alterations in advanced-stage EGFR-mutant lung cancers

Collin M. Blakely; Thomas B.K. Watkins; Wei Wu; Beatrice Gini; Jacob J. Chabon; Caroline E. McCoach; Nicholas McGranahan; Gareth A. Wilson; Nicolai Juul Birkbak; Victor Olivas; Julia Rotow; Ashley Maynard; Victoria Wang; Matthew A. Gubens; Kimberly C. Banks; Richard B. Lanman; Aleah F. Caulin; John St. John; Anibal Cordero; Petros Giannikopoulos; Andrew Simmons; Philip C. Mack; David R. Gandara; Hatim Husain; Robert C. Doebele; Jonathan W. Riess; Maximilian Diehn; Charles Swanton; Trever G. Bivona

A widespread approach to modern cancer therapy is to identify a single oncogenic driver gene and target its mutant-protein product (for example, EGFR-inhibitor treatment in EGFR-mutant lung cancers). However, genetically driven resistance to targeted therapy limits patient survival. Through genomic analysis of 1,122 EGFR-mutant lung cancer cell-free DNA samples and whole-exome analysis of seven longitudinally collected tumor samples from a patient with EGFR-mutant lung cancer, we identified critical co-occurring oncogenic events present in most advanced-stage EGFR-mutant lung cancers. We defined new pathways limiting EGFR-inhibitor response, including WNT/β-catenin alterations and cell-cycle-gene (CDK4 and CDK6) mutations. Tumor genomic complexity increases with EGFR-inhibitor treatment, and co-occurring alterations in CTNNB1 and PIK3CA exhibit nonredundant functions that cooperatively promote tumor metastasis or limit EGFR-inhibitor response. This study calls for revisiting the prevailing single-gene driver-oncogene view and links clinical outcomes to co-occurring genetic alterations in patients with advanced-stage EGFR-mutant lung cancer.


Cell | 2018

Deterministic Evolutionary Trajectories Influence Primary Tumor Growth: TRACERx Renal

Samra Turajlic; Hang Xu; Kevin Litchfield; Andrew Rowan; Stuart Horswell; Tim Chambers; Tim O’Brien; José I. López; Thomas B.K. Watkins; David Nicol; Mark Stares; Ben Challacombe; Steve Hazell; Ashish Chandra; Thomas J. Mitchell; Lewis Au; Claudia Eichler-Jonsson; Faiz Jabbar; Aspasia Soultati; Simon Chowdhury; Sarah Rudman; Joanna Lynch; Archana Fernando; Gordon Stamp; Emma Nye; Aengus Stewart; Wei Xing; Jonathan C. Smith; Mickael Escudero; Adam Huffman

Summary The evolutionary features of clear-cell renal cell carcinoma (ccRCC) have not been systematically studied to date. We analyzed 1,206 primary tumor regions from 101 patients recruited into the multi-center prospective study, TRACERx Renal. We observe up to 30 driver events per tumor and show that subclonal diversification is associated with known prognostic parameters. By resolving the patterns of driver event ordering, co-occurrence, and mutual exclusivity at clone level, we show the deterministic nature of clonal evolution. ccRCC can be grouped into seven evolutionary subtypes, ranging from tumors characterized by early fixation of multiple mutational and copy number drivers and rapid metastases to highly branched tumors with >10 subclonal drivers and extensive parallel evolution associated with attenuated progression. We identify genetic diversity and chromosomal complexity as determinants of patient outcome. Our insights reconcile the variable clinical behavior of ccRCC and suggest evolutionary potential as a biomarker for both intervention and surveillance.


International Journal of Cancer | 2018

Expansion of airway basal epithelial cells from primary human non-small cell lung cancer tumors: 3T3+Y for NSCLC cell culture

Robert E. Hynds; Assma Ben Aissa; Kate H.C. Gowers; Thomas B.K. Watkins; Leticia Bosshard-Carter; Andrew Rowan; Selvaraju Veeriah; Gareth A. Wilson; Sergio A. Quezada; Charles Swanton; Sam M. Janes

Pre‐clinical non‐small cell lung cancer (NSCLC) models are poorly representative of the considerable inter‐ and intra‐tumor heterogeneity of the disease in patients. Primary cell‐based in vitro models of NSCLC are therefore desirable for novel therapy development and personalized cancer medicine. Methods have been described to generate rapidly proliferating epithelial cell cultures from multiple human epithelia using 3T3‐J2 feeder cell culture in the presence of Y‐27632, a RHO‐associated protein kinase (ROCK) inhibitor, in what are known as “conditional reprograming conditions” (CRC) or 3T3 + Y. In some cancer studies, variations of this methodology have allowed primary tumor cell expansion across a number of cancer types but other studies have demonstrated the preferential expansion of normal epithelial cells from tumors in such conditions. Here, we report our experience regarding the derivation of primary NSCLC cell cultures from 12 lung adenocarcinoma patients enrolled in the Tracking Cancer Evolution through Therapy (TRACERx) clinical study and discuss these in the context of improving the success rate for in vitro cultivation of cells from NSCLC tumors.


Cold Spring Harbor Perspectives in Medicine | 2018

Phylogenetic Quantification of Intratumor Heterogeneity

Thomas B.K. Watkins; Roland F. Schwarz

As sequencing efforts continue to reveal the extent of the intratumor heterogeneity (ITH) present in human cancers, the importance of evolutionary studies attempting to trace its etiology has increased. Sequencing multiple samples or tumor regions from the same patient has become affordable and is an effective way of tracing these evolutionary pathways, understanding selection, and detecting clonal expansions in ways impractical with single samples alone. In this article, we discuss and show the benefits of such multisample studies. We describe how multiple samples can guide tree inference through accurate phasing of germline variants and copy-number profiles. We show their relevance in detecting clonal expansions and deriving summary statistics quantifying the overall degree of ITH, and discuss how the relationship of metastatic clades might give us insight into the dominant mode of cancer progression. We further outline how multisample studies might help us better understand selective processes acting on cancer genomes and help to detect neutral evolution and mutator phenotypes.


Bioinformatics | 2018

Abstract LB-A03: Allele specific HLA loss and immune escape in lung cancer evolution

Nicholas McGranahan; Rachel Rosenthal; Crispin T. Hiley; Andrew Rowan; Thomas B.K. Watkins; Gareth A. Wilson; Nicolai Juul Birkbak; Selvaraju Veeriah; Peter Van Loo; Javier Herrero; Charles Swanton

Cancer cells adopt a variety of mechanisms to evade the immune system and avoid T-cell recognition. Disruption of human leukocyte antigen (HLA), which may lead to reduced neoantigen presentation, has been proposed as an immune escape strategy in many cancers, including lung cancer. Mutations in HLA class I genes are infrequent in early stage non-small cell lung cancers (NSCLC), suggesting alternative mechanisms of HLA disruption may be common. To date, the polymorphic nature of the locus has precluded copy number analysis and exploration of HLA loss. To investigate the prevalence and importance of HLA disruption, we present LOHHLA (Loss Of Heterozygosity in Human Leukocyte Antigen), a computational tool to determine HLA allele-specific copy number from sequencing data. Building upon previous work imputing HLA haplotypes from sequencing data (Shukla, 2015; Szolek, 2014) and utilizing previously published datasets (Jamal-Hanjani, 2017; Brastianos, 2015), we endeavored to address the prevalence and timing of HLA LOH in lung cancer and its impact on tumor evolution and neoantigen presentation. Using LOHHLA, we find HLA LOH occurs in 40% of NSCLCs and is associated with a high subclonal neoantigen burden, APOBEC mediated-mutagenesis, upregulation of cytolytic activity and PD-L1 positivity. The focal nature of these alterations, their subclonal frequencies, enrichment in metastatic sites, and occurrence as parallel events suggest that HLA LOH is an immune escape mechanism, selected later in NSCLC tumor evolution. Characterizing HLA LOH with LOHHLA refines neoantigen prediction and may have implications for immunotherapeutic approaches targeting neoantigens. Citation Format: Nicholas McGranahan, Rachel Rosenthal, Crispin T. Hiley, Andrew J. Rowan, Thomas B.K. Watkins, Gareth A. Wilson, Nicolai J. Birkbak, Selvaraju Veeriah, Peter Van Loo, Javier Herrero, Charles Swanton. Allele specific HLA loss and immune escape in lung cancer evolution [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2017 Oct 26-30; Philadelphia, PA. Philadelphia (PA): AACR; Mol Cancer Ther 2018;17(1 Suppl):Abstract nr LB-A03.

Collaboration


Dive into the Thomas B.K. Watkins's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew Rowan

London Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hang Xu

Francis Crick Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge