Network


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

Hotspot


Dive into the research topics where Nicholas McGranahan is active.

Publication


Featured researches published by Nicholas McGranahan.


Nature Biotechnology | 2016

Large-scale detection of antigen-specific T cells using peptide-MHC-I multimers labeled with DNA barcodes

Amalie Kai Bentzen; Andrea Marion Marquard; Rikke Birgitte Lyngaa; Sunil Kumar Saini; Sofie Ramskov; Marco Donia; Lina Such; Andrew Furness; Nicholas McGranahan; Rachel Rosenthal; Per thor Straten; Zoltan Szallasi; Inge Marie Svane; Charles Swanton; Sergio A. Quezada; Soren Nyboe Jakobsen; Aron Charles Eklund; Sine Reker Hadrup

Identification of the peptides recognized by individual T cells is important for understanding and treating immune-related diseases. Current cytometry-based approaches are limited to the simultaneous screening of 10–100 distinct T-cell specificities in one sample. Here we use peptide–major histocompatibility complex (MHC) multimers labeled with individual DNA barcodes to screen >1,000 peptide specificities in a single sample, and detect low-frequency CD8 T cells specific for virus- or cancer-restricted antigens. When analyzing T-cell recognition of shared melanoma antigens before and after adoptive cell therapy in melanoma patients, we observe a greater number of melanoma-specific T-cell populations compared with cytometry-based approaches. Furthermore, we detect neoepitope-specific T cells in tumor-infiltrating lymphocytes and peripheral blood from patients with non-small cell lung cancer. Barcode-labeled pMHC multimers enable the combination of functional T-cell analysis with large-scale epitope recognition profiling for the characterization of T-cell recognition in various diseases, including in small clinical samples.


Genome Biology | 2016

DNA replication stress mediates APOBEC3 family mutagenesis in breast cancer

Nnennaya Kanu; Maria Cerone; Gerald Goh; Lykourgos Panagiotis Zalmas; Jirina Bartkova; Michelle Dietzen; Nicholas McGranahan; Rebecca Rogers; Emily K. Law; Irina Gromova; Maik Kschischo; Michael I. Walton; Olivia W. Rossanese; Jiri Bartek; Reuben S. Harris; Subramanian Venkatesan; Charles Swanton

BackgroundThe APOBEC3 family of cytidine deaminases mutate the cancer genome in a range of cancer types. Although many studies have documented the downstream effects of APOBEC3 activity through next-generation sequencing, less is known about their upstream regulation. In this study, we sought to identify a molecular basis for APOBEC3 expression and activation.ResultsHER2 amplification and PTEN loss promote DNA replication stress and APOBEC3B activity in vitro and correlate with APOBEC3 mutagenesis in vivo. HER2-enriched breast carcinomas display evidence of elevated levels of replication stress-associated DNA damage in vivo. Chemical and cytotoxic induction of replication stress, through aphidicolin, gemcitabine, camptothecin or hydroxyurea exposure, activates transcription of APOBEC3B via an ATR/Chk1-dependent pathway in vitro. APOBEC3B activation can be attenuated through repression of oncogenic signalling, small molecule inhibition of receptor tyrosine kinase signalling and alleviation of replication stress through nucleoside supplementation.ConclusionThese data link oncogene, loss of tumour suppressor gene and drug-induced replication stress with APOBEC3B activity, providing new insights into how cytidine deaminase-induced mutagenesis might be activated in tumourigenesis and limited therapeutically.


Annals of Oncology | 2016

Consensus on precision medicine for metastatic cancers: a report from the MAP conference

Charles Swanton; Jean-Charles Soria; Alberto Bardelli; Andrew V. Biankin; Carlos Caldas; Sarat Chandarlapaty; L de Koning; Ch Dive; J Feunteun; S-Y Leung; Richard Marais; Elaine R. Mardis; Nicholas McGranahan; Gary Middleton; Sergio A. Quezada; Jordi Rodon; Nitzan Rosenfeld; Christos Sotiriou; Fabrice Andre

Recent advances in biotechnologies have led to the development of multiplex genomic and proteomic analyses for clinical use. Nevertheless, guidelines are currently lacking to determine which molecular assays should be implemented in metastatic cancers. The first MAP conference was dedicated to exploring the use of genomics to better select therapies in the treatment of metastatic cancers. Sixteen consensus items were covered. There was a consensus that new technologies like next-generation sequencing of tumors and ddPCR on circulating free DNA have convincing analytical validity. Further work needs to be undertaken to establish the clinical utility of liquid biopsies and the added clinical value of expanding from individual gene tests into large gene panels. Experts agreed that standardized bioinformatics methods for biological interpretation of genomic data are needed and that precision medicine trials should be stratified based on the level of evidence available for the genomic alterations identified.


Biochimica et Biophysica Acta | 2015

Inferring mutational timing and reconstructing tumour evolutionary histories

Samra Turajlic; Nicholas McGranahan; Charles Swanton

Cancer evolution can be considered within a Darwinian framework. Both micro and macro-evolutionary theories can be applied to understand tumour progression and treatment failure. Owing to cancers complexity and heterogeneity the rules of tumour evolution, such as the role of selection, remain incompletely understood. The timing of mutational events during tumour evolution presents diagnostic, prognostic and therapeutic opportunities. Here we review the current sampling and computational approaches for inferring mutational timing and the evidence from next generation sequencing-informed data on mutational timing across all tumour types. We discuss how this knowledge can be used to illuminate the genes and pathways that drive cancer initiation and relapse; and to support drug development and clinical trial design.


Cancer Discovery | 2017

APC/C Dysfunction Limits Excessive Cancer Chromosomal Instability

Laurent Sansregret; James O. Patterson; Sally M. Dewhurst; Carlos López-García; André Koch; Nicholas McGranahan; William C. H. Chao; David J. Barry; Andrew Rowan; Rachael Instrell; Stuart Horswell; Michael Way; Michael Howell; Martin R. Singleton; René H. Medema; Paul Nurse; Mark Petronczki; Charles Swanton

Intercellular heterogeneity, exacerbated by chromosomal instability (CIN), fosters tumor heterogeneity and drug resistance. However, extreme CIN correlates with improved cancer outcome, suggesting that karyotypic diversity required to adapt to selection pressures might be balanced in tumors against the risk of excessive instability. Here, we used a functional genomics screen, genome editing, and pharmacologic approaches to identify CIN-survival factors in diploid cells. We find partial anaphase-promoting complex/cyclosome (APC/C) dysfunction lengthens mitosis, suppresses pharmacologically induced chromosome segregation errors, and reduces naturally occurring lagging chromosomes in cancer cell lines or following tetraploidization. APC/C impairment caused adaptation to MPS1 inhibitors, revealing a likely resistance mechanism to therapies targeting the spindle assembly checkpoint. Finally, CRISPR-mediated introduction of cancer somatic mutations in the APC/C subunit cancer driver gene CDC27 reduces chromosome segregation errors, whereas reversal of an APC/C subunit nonsense mutation increases CIN. Subtle variations in mitotic duration, determined by APC/C activity, influence the extent of CIN, allowing cancer cells to dynamically optimize fitness during tumor evolution.nnnSIGNIFICANCEnWe report a mechanism whereby cancers balance the evolutionary advantages associated with CIN against the fitness costs caused by excessive genome instability, providing insight into the consequence of CDC27 APC/C subunit driver mutations in cancer. Lengthening of mitosis through APC/C modulation may be a common mechanism of resistance to cancer therapeutics that increase chromosome segregation errors. Cancer Discov; 7(2); 218-33. ©2017 AACR.See related commentary by Burkard and Weaver, p. 134This article is highlighted in the In This Issue feature, p. 115.


Annals of Oncology | 2016

Cyclin D mediates tolerance of genome-doubling in cancers with functional p53.

A Crockford; Lp Zalmas; E. Grönroos; Sm Dewhurst; Nicholas McGranahan; Me Cuomo; Encheva; Ap Snijders; J Begum; S Purewal; J Cerveira; H Patel; Mj Renshaw; Charles Swanton

Abstract Background Aneuploidy and chromosomal instability (CIN) are common features of human malignancy that fuel genetic heterogeneity. Although tolerance to tetraploidization, an intermediate state that further exacerbates CIN, is frequently mediated by TP53 dysfunction, we find that some genome-doubled tumours retain wild-type TP53. We sought to understand how tetraploid cells with a functional p53/p21-axis tolerate genome-doubling events. Methods We performed quantitative proteomics in a diploid/tetraploid pair within a system of multiple independently derived TP53 wild-type tetraploid clones arising spontaneously from a diploid progenitor. We characterized adapted and acute tetraploidization in a variety of flow cytometry and biochemical assays and tested our findings against human tumours through bioinformatics analysis of the TCGA dataset. Results Cyclin D1 was found to be specifically overexpressed in early but not late passage tetraploid clones, and this overexpression was sufficient to promote tolerance to spontaneous and pharmacologically induced tetraploidy. We provide evidence that this role extends to D-type cyclins and their overexpression confers specific proliferative advantage to tetraploid cells. We demonstrate that tetraploid clones exhibit elevated levels of functional p53 and p21 but override the p53/p21 checkpoint by elevated expression of cyclin D1, via a stoichiometry-dependent and CDK activity-independent mechanism. Tetraploid cells do not exhibit increased sensitivity to abemaciclib, suggesting that cyclin D-overexpressing tumours might not be specifically amenable to treatment with CDK4/6 inhibitors. Conclusions Our study suggests that D-type cyclin overexpression is an acute event, permissive for rapid adaptation to a genome-doubled state in TP53 wild-type tumours and that its overexpression is dispensable in later stages of tumour progression.


Nature Communications | 2017

Oncogenic PIK3CA induces centrosome amplification and tolerance to genome doubling.

Inma M. Berenjeno; Roberto Piñeiro; Sandra D. Castillo; Wayne Pearce; Nicholas McGranahan; Sally M. Dewhurst; Valerie Meniel; Nicolai Juul Birkbak; Evelyn Lau; Laurent Sansregret; Daniele Morelli; Nnennaya Kanu; Shankar Srinivas; Mariona Graupera; Victoria Parker; Karen G. Montgomery; Larissa S. Moniz; Cheryl L. Scudamore; Wayne A. Phillips; Robert K. Semple; Alan Richard Clarke; Charles Swanton; Bart Vanhaesebroeck

Mutations in PIK3CA are very frequent in cancer and lead to sustained PI3K pathway activation. The impact of acute expression of mutant PIK3CA during early stages of malignancy is unknown. Using a mouse model to activate the Pik3caH1047R hotspot mutation in the heterozygous state from its endogenous locus, we here report that mutant Pik3ca induces centrosome amplification in cultured cells (through a pathway involving AKT, ROCK and CDK2/Cyclin E-nucleophosmin) and in mouse tissues, and increased in vitro cellular tolerance to spontaneous genome doubling. We also present evidence that the majority of PIK3CAH1047R mutations in the TCGA breast cancer cohort precede genome doubling. These previously unappreciated roles of PIK3CA mutation show that PI3K signalling can contribute to the generation of irreversible genomic changes in cancer. While this can limit the impact of PI3K-targeted therapies, these findings also open the opportunity for therapeutic approaches aimed at limiting tumour heterogeneity and evolution.Activated PI3K causes cancer, but the role of active PI3K mutations in early stages of malignancy are unclear. Here, the authors show in a mouse model that active PI3K induces centrosome amplification via AKT, ROCK, CDK2/Cyclin E and nucleophosmin, and increased tolerance of genome doubling.


Annals of Oncology | 2018

Perspective: APOBEC mutagenesis in drug resistance and immune escape in HIV and cancer evolution

Subramanian Venkatesan; Rachel Rosenthal; Nnennaya Kanu; Nicholas McGranahan; Jiri Bartek; Sergio A. Quezada; J. Hare; Reuben S. Harris; Charles Swanton

Abstract The apolipoprotein B mRNA-editing enzyme, catalytic polypeptide-like (APOBEC) mutational signature has only recently been detected in a multitude of cancers through next-generation sequencing. In contrast, APOBEC has been a focus of virology research for over a decade. Many lessons learnt regarding APOBEC within virology are likely to be applicable to cancer. In this review, we explore the parallels between the role of APOBEC enzymes in HIV and cancer evolution. We discuss data supporting the role of APOBEC mutagenesis in creating HIV genome heterogeneity, drug resistance, and immune escape variants. We hypothesize similar functions of APOBEC will also hold true in cancer.


Annals of Oncology | 2017

Intratumoural evolutionary landscape of high-risk prostate cancer: The PROGENY study of genomic and immune parameters

Mark Linch; Gerald Goh; Crispin Hiley; Yaalini Shanmugabavan; Nicholas McGranahan; Andrew Rowan; Yien Ning Sophia Wong; H. King; Andrew J.S. Furness; Alex Freeman; J. Linares; A. Akarca; Javier Herrero; Rachel Rosenthal; N. Harder; G. Schmidt; Gareth A. Wilson; Nicolai Juul Birkbak; Richard Mitter; Stefan Dentro; Paul Cathcart; Manit Arya; E Johnston; R. Scott; M. Hung; Mark Emberton; Gerhardt Attard; Zoltan Szallasi; Shonit Punwani; Sergio A. Quezada

Abstract Background Intratumoural heterogeneity (ITH) is well recognised in prostate cancer (PC), but its role in high-risk disease is uncertain. A prospective, single-arm, translational study using targeted multiregion prostate biopsies was carried out to study genomic and T-cell ITH in clinically high-risk PC aiming to identify drivers and potential therapeutic strategies. Patients and methods Forty-nine men with elevated prostate-specific antigen and multiparametric-magnetic resonance imaging detected PC underwent image-guided multiregion transperineal biopsy. Seventy-nine tumour regions from 25 patients with PC underwent sequencing, analysis of mutations, copy number and neoepitopes combined with tumour infiltrating T-cell subset quantification. Results We demonstrated extensive somatic nucleotide variation and somatic copy number alteration heterogeneity in high-risk PC. Overall, the mutational burden was low (0.93/Megabase), but two patients had hypermutation, with loss of mismatch repair (MMR) proteins, MSH2 and MSH6. Somatic copy number alteration burden was higher in patients with metastatic hormone-naive PC (mHNPC) than in those with high-risk localised PC (hrlPC), independent of Gleason grade. Mutations were rarely ubiquitous and mutational frequencies were similar for mHNPC and hrlPC patients. Enrichment of focal 3q26.2 and 3q21.3, regions containing putative metastasis drivers, was seen in mHNPC patients. We found evidence of parallel evolution with three separate clones containing activating mutations of β-catenin in a single patient. We demonstrated extensive intratumoural and intertumoural T-cell heterogeneity and high inflammatory infiltrate in the MMR-deficient (MMRD) patients and the patient with parallel evolution of β-catenin. Analysis of all patients with activating Wnt/β-catenin mutations demonstrated a low CD8+/FOXP3+u2009ratio, a potential surrogate marker of immune evasion. Conclusions The PROGENY (PROstate cancer GENomic heterogeneitY) study provides a diagnostic platform suitable for studying tumour ITH. Genetic aberrations in clinically high-risk PC are associated with altered patterns of immune infiltrate in tumours. Activating mutations of Wnt/β-catenin signalling pathway or MMRD could be considered as potential biomarkers for immunomodulation therapies. Clinical Trials.gov Identifier NCT02022371


bioRxiv | 2017

Evolution and clinical impact of genetic epistasis within 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; Philip C. Mack; David R. Gandara; Hatim Husain; Robert C. Doebele; Jonathan W. Riess; Maximilian Diehn; Charles Swanton; Trever G. Bivona

The current understanding of tumorigenesis is largely centered on a monogenic driver oncogene model. This paradigm is incompatible with the prevailing clinical experience in most solid malignancies: monotherapy with a drug directed against an individual oncogenic driver typically results in incomplete clinical responses and eventual tumor progression1-7. By profiling the somatic genetic alterations present in over 2,000 cases of lung cancer, the leading cause of cancer mortality worldwide8,9, we show that combinations of functional genetic alterations, i.e. genetic collectives dominate the landscape of advanced-stage disease. We highlight this polygenic landscape and evolution of advanced-stage non-small cell lung cancer (NSCLC) through the spatial-temporal genomic profiling of 7 distinct tumor biopsy specimens and 6 plasma specimens obtained from an EGFR-mutant NSCLC patient at (1) initial diagnosis of early-stage disease, (2) metastatic progression, (3) sequential treatment and resistance to 2 EGFR inhibitors, (4) death. The comprehensive genomic analysis of this case, coupled with circulating free (cf) tumor DNA profiling of additional advanced-stage EGFR-mutant NSCLC clinical cohorts with associated treatment responses uncovered features of evolutionary selection for multiple concurrent gene alterations: including the presence of EGFR inhibitor-sensitive (EGFRL858R;EGFRexon19del) or inhibitor-resistant (EGFRT790M;EGFRC797S) forms of oncogenic EGFR along with cell cycle gene alterations (e.g. in CDK4/6, CCNE1, RB1) and activating alterations in WNT/β-catenin and PI3K pathway genes, which our data suggest can cooperatively impart non-redundant functions to limit EGFR targeted therapy response and/or promote tumor progression. Moreover, evidence of an unanticipated parallel evolution of both EGFRT790M and two distinct forms of oncogenic PIK3CA was observed. Our study provides a large-scale clinical and genetic dataset of advanced-stage EGFR-mutant NSCLC, a rationale for specific polytherapy strategies such as EGFR and CDK4/6 inhibitor co-treatment to potentially enhance clinical outcomes, and prompts a re-evaluation of the prevailing paradigm of monogenic-based molecular stratification for targeted therapy. Instead, our findings highlight an alternative model of genetic collectives that operate through epistasis to drive lung cancer progression and therapy resistance.

Collaboration


Dive into the Nicholas McGranahan's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew Rowan

Francis Crick Institute

View shared research outputs
Top Co-Authors

Avatar

Nicolai Juul Birkbak

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Reuben S. Harris

Howard Hughes Medical Institute

View shared research outputs
Top Co-Authors

Avatar

Zoltan Szallasi

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Maik Kschischo

Münster University of Applied Sciences

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge