Network


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

Hotspot


Dive into the research topics where Kevin Litchfield is active.

Publication


Featured researches published by Kevin Litchfield.


Lancet Oncology | 2017

Insertion-and-deletion-derived tumour-specific neoantigens and the immunogenic phenotype: a pan-cancer analysis

Samra Turajlic; Kevin Litchfield; Hang Xu; Rachel Rosenthal; Nicholas McGranahan; James L. Reading; Yien Ning S Wong; Andrew Rowan; Nnennaya Kanu; Maise Al Bakir; Tim Chambers; Roberto Salgado; Peter Savas; Sherene Loi; Nicolai Juul Birkbak; Laurent Sansregret; Martin Gore; James Larkin; Sergio A. Quezada; Charles Swanton

BACKGROUND The focus of tumour-specific antigen analyses has been on single nucleotide variants (SNVs), with the contribution of small insertions and deletions (indels) less well characterised. We investigated whether the frameshift nature of indel mutations, which create novel open reading frames and a large quantity of mutagenic peptides highly distinct from self, might contribute to the immunogenic phenotype. METHODS We analysed whole-exome sequencing data from 5777 solid tumours, spanning 19 cancer types from The Cancer Genome Atlas. We compared the proportion and number of indels across the cohort, with a subset of results replicated in two independent datasets. We assessed in-silico tumour-specific neoantigen predictions by mutation type with pan-cancer analysis, together with RNAseq profiling in renal clear cell carcinoma cases (n=392), to compare immune gene expression across patient subgroups. Associations between indel burden and treatment response were assessed across four checkpoint inhibitor datasets. FINDINGS We observed renal cell carcinomas to have the highest proportion (0·12) and number of indel mutations across the pan-cancer cohort (p<2·2 × 10-16), more than double the median proportion of indel mutations in all other cancer types examined. Analysis of tumour-specific neoantigens showed that enrichment of indel mutations for high-affinity binders was three times that of non-synonymous SNV mutations. Furthermore, neoantigens derived from indel mutations were nine times enriched for mutant specific binding, as compared with non-synonymous SNV derived neoantigens. Immune gene expression analysis in the renal clear cell carcinoma cohort showed that the presence of mutant-specific neoantigens was associated with upregulation of antigen presentation genes, which correlated (r=0·78) with T-cell activation as measured by CD8-positive expression. Finally, analysis of checkpoint inhibitor response data revealed frameshift indel count to be significantly associated with checkpoint inhibitor response across three separate melanoma cohorts (p=4·7 × 10-4). INTERPRETATION Renal cell carcinomas have the highest pan-cancer proportion and number of indel mutations. Evidence suggests indels are a highly immunogenic mutational class, which can trigger an increased abundance of neoantigens and greater mutant-binding specificity. FUNDING Cancer Research UK, UK National Institute for Health Research (NIHR) at the Royal Marsden Hospital National Health Service Foundation Trust, Institute of Cancer Research and University College London Hospitals Biomedical Research Centres, the UK Medical Research Council, the Rosetrees Trust, Novo Nordisk Foundation, the Prostate Cancer Foundation, the Breast Cancer Research Foundation, the European Research Council.


Nature Communications | 2015

Whole-exome sequencing reveals the mutational spectrum of testicular germ cell tumours

Kevin Litchfield; Brenda Summersgill; Shawn Yost; Razvan Sultana; Karim Labreche; Darshna Dudakia; Anthony Renwick; Sheila Seal; Reem Al-Saadi; Peter Broderick; Nicholas C. Turner; Richard S. Houlston; Robert Huddart; Janet Shipley; Clare Turnbull

Testicular germ cell tumours (TGCTs) are the most common cancer in young men. Here we perform whole-exome sequencing (WES) of 42 TGCTs to comprehensively study the cancers mutational profile. The mutation rate is uniformly low in all of the tumours (mean 0.5 mutations per Mb) as compared with common cancers, consistent with the embryological origin of TGCT. In addition to expected copy number gain of chromosome 12p and mutation of KIT, we identify recurrent mutations in the tumour suppressor gene CDC27 (11.9%). Copy number analysis reveals recurring amplification of the spermatocyte development gene FSIP2 (15.3%) and a 0.4 Mb region at Xq28 (15.3%). Two treatment-refractory patients are shown to harbour XRCC2 mutations, a gene strongly implicated in defining cisplatin resistance. Our findings provide further insights into genes involved in the development and progression of TGCT.


Journal of Andrology | 2015

Common variants identified in genome-wide association studies of testicular germ cell tumour: an update, biological insights and clinical application

Kevin Litchfield; Janet Shipley; Clare Turnbull

Testicular germ cell tumour (TGCT) is the most common cause of cancer in young men (aged 15–45 years) in many populations. Multiple genome‐wide association studies (GWAS) of TGCT have now been conducted, yielding over 25 disease‐associated single‐nucleotide polymorphism (SNP)s at 19 independent loci. The genes at these loci have provided rich biological and genetic insight into possible mechanisms underlying testicular germ cell oncogenesis. In this review, we summarize these mechanisms which can be grouped into five distinct categories: KIT/KITLG signalling, other pathways of male germ cell development/differentiation, telomerase function, microtubule assembly and DNA damage repair. The TGCT risk markers identified through GWAS include individual SNPs carrying per allele odds ratios (OR) in excess of 2.5. These ORs are among the highest reported in GWAS of any cancer type, hence suggesting a potential clinical utility in risk determination. Here, we present analysis of such an approach, using polygenic risk scores to calculate the combined effect of all risk loci on overall TGCT risk and discuss how a potential screening strategy may fit within a broader clinical context.


Nature | 2016

Genomic evolution and chemoresistance in germ-cell tumours

Amaro Taylor-Weiner; Travis I. Zack; Elizabeth O’Donnell; Jennifer L. Guerriero; Brandon David Bernard; Anita Reddy; G. Celine Han; Saud H. Aldubayan; Ali Amin-Mansour; Steven E. Schumacher; Kevin Litchfield; Clare Turnbull; Stacey Gabriel; Rameen Beroukhim; Gad Getz; Scott L. Carter; Michelle S. Hirsch; Anthony Letai; Christopher Sweeney; Eliezer M. Van Allen

Germ-cell tumours (GCTs) are derived from germ cells and occur most frequently in the testes. GCTs are histologically heterogeneous and distinctly curable with chemotherapy. Gains of chromosome arm 12p and aneuploidy are nearly universal in GCTs, but specific somatic genomic features driving tumour initiation, chemosensitivity and progression are incompletely characterized. Here, using clinical whole-exome and transcriptome sequencing of precursor, primary (testicular and mediastinal) and chemoresistant metastatic human GCTs, we show that the primary somatic feature of GCTs is highly recurrent chromosome arm level amplifications and reciprocal deletions (reciprocal loss of heterozygosity), variations that are significantly enriched in GCTs compared to 19 other cancer types. These tumours also acquire KRAS mutations during the development from precursor to primary disease, and primary testicular GCTs (TGCTs) are uniformly wild type for TP53. In addition, by functional measurement of apoptotic signalling (BH3 profiling) of fresh tumour and adjacent tissue, we find that primary TGCTs have high mitochondrial priming that facilitates chemotherapy-induced apoptosis. Finally, by phylogenetic analysis of serial TGCTs that emerge with chemotherapy resistance, we show how TGCTs gain additional reciprocal loss of heterozygosity and that this is associated with loss of pluripotency markers (NANOG and POU5F1) in chemoresistant teratomas or transformed carcinomas. Our results demonstrate the distinct genomic features underlying the origins of this disease and associated with the chemosensitivity phenotype, as well as the rare progression to chemoresistance. These results identify the convergence of cancer genomics, mitochondrial priming and GCT evolution, and may provide insights into chemosensitivity and resistance in other cancers.


Nature Genetics | 2017

Identification of 19 new risk loci and potential regulatory mechanisms influencing susceptibility to testicular germ cell tumor

Kevin Litchfield; Max Levy; Giulia Orlando; Chey Loveday; Philip J. Law; Gabriele Migliorini; Amy Holroyd; Peter Broderick; Robert Karlsson; Trine B. Haugen; Wenche Kristiansen; Jérémie Nsengimana; Kerry Fenwick; Ioannis Assiotis; Zsofia Kote-Jarai; Alison M. Dunning; Kenneth Muir; Julian Peto; Rosalind Eeles; Douglas F. Easton; Darshna Dudakia; Nick Orr; Nora Pashayan; D. Timothy Bishop; Alison Reid; Robert Huddart; Janet Shipley; Tom Grotmol; Fredrik Wiklund; Richard S. Houlston

Genome-wide association studies (GWAS) have transformed understanding of susceptibility to testicular germ cell tumors (TGCTs), but much of the heritability remains unexplained. Here we report a new GWAS, a meta-analysis with previous GWAS and a replication series, totaling 7,319 TGCT cases and 23,082 controls. We identify 19 new TGCT risk loci, roughly doubling the number of known TGCT risk loci to 44. By performing in situ Hi-C in TGCT cells, we provide evidence for a network of physical interactions among all 44 TGCT risk SNPs and candidate causal genes. Our findings implicate widespread disruption of developmental transcriptional regulators as a basis of TGCT susceptibility, consistent with failed primordial germ cell differentiation as an initiating step in oncogenesis. Defective microtubule assembly and dysregulation of KIT–MAPK signaling also feature as recurrently disrupted pathways. Our findings support a polygenic model of risk and provide insight into the biological basis of TGCT.


Nature Reviews Urology | 2016

The genomic landscape of testicular germ cell tumours: from susceptibility to treatment

Kevin Litchfield; Max Levy; Robert Huddart; Janet Shipley; Clare Turnbull

The genomic landscape of testicular germ cell tumour (TGCT) can be summarized using four overarching hypotheses. Firstly, TGCT risk is dominated by inherited genetic factors, which determine nearly half of all disease risk and are highly polygenic in nature. Secondly KIT–KITLG signalling is currently the major pathway that is implicated in TGCT formation, both as a predisposition risk factor and a somatic driver event. Results from genome-wide association studies have also consistently suggested that other closely related pathways involved in male germ cell development and sex determination are associated with TGCT risk. Thirdly, the method of disease formation is unique, with tumours universally stemming from a noninvasive precursor lesion, probably of fetal origin, which lies dormant through childhood into adolescence and then eventually begins malignant growth in early adulthood. Formation of a 12p isochromosome, a hallmark of TGCT observed in nearly all tumours, is likely to be a key triggering event for malignant transformation. Finally, TGCT have been shown to have a distinctive somatic mutational profile, with a low rate of point mutations contrasted with frequent large-scale chromosomal gains. These four hypotheses by no means constitute a complete model that explains TGCT tumorigenesis, but advances in genomic technologies have enabled considerable progress in describing and understanding the disease. Further advancing our understanding of the genomic basis of TGCT offers a clear opportunity for clinical benefit in terms of preventing invasive cancer arising in young men, decreasing the burden of chemotherapy-related survivorship issues and reducing mortality in the minority of patients who have treatment-refractory disease.


Nature Genetics | 2017

Meta-analysis of five genome-wide association studies identifies multiple new loci associated with testicular germ cell tumor

Zhaoming Wang; Katherine A. McGlynn; Ewa Rajpert-De Meyts; D. Timothy Bishop; Charles C. Chung; Marlene Danner Dalgaard; Mark H. Greene; Ramneek Gupta; Tom Grotmol; Trine B. Haugen; Robert Karlsson; Kevin Litchfield; Nandita Mitra; Kasper Nielsen; Louise C. Pyle; Stephen M. Schwartz; Vesteinn Thorsson; Saran Vardhanabhuti; Fredrik Wiklund; Clare Turnbull; Stephen J. Chanock; Peter A. Kanetsky; Katherine L. Nathanson

The international Testicular Cancer Consortium (TECAC) combined five published genome-wide association studies of testicular germ cell tumor (TGCT; 3,558 cases and 13,970 controls) to identify new susceptibility loci. We conducted a fixed-effects meta-analysis, including, to our knowledge, the first analysis of the X chromosome. Eight new loci mapping to 2q14.2, 3q26.2, 4q35.2, 7q36.3, 10q26.13, 15q21.3, 15q22.31, and Xq28 achieved genome-wide significance (P < 5 × 10−8). Most loci harbor biologically plausible candidate genes. We refined previously reported associations at 9p24.3 and 19p12 by identifying one and three additional independent SNPs, respectively. In aggregate, the 39 independent markers identified to date explain 37% of father-to-son familial risk, 8% of which can be attributed to the 12 new signals reported here. Our findings substantially increase the number of known TGCT susceptibility alleles, move the field closer to a comprehensive understanding of the underlying genetic architecture of TGCT, and provide further clues to the etiology of TGCT.


Human Molecular Genetics | 2014

Pathway-based analysis of GWAs data identifies association of sex determination genes with susceptibility to testicular germ cell tumors

Roelof Koster; Nandita Mitra; Kurt D'Andrea; Saran Vardhanabhuti; Charles C. Chung; Zhaoming Wang; R. Loren Erickson; David J. Vaughn; Kevin Litchfield; Nazneen Rahman; Mark H. Greene; Katherine A. McGlynn; Clare Turnbull; Stephen J. Chanock; Katherine L. Nathanson; Peter A. Kanetsky

Genome-wide association (GWA) studies of testicular germ cell tumor (TGCT) have identified 18 susceptibility loci, some containing genes encoding proteins important in male germ cell development. Deletions of one of these genes, DMRT1, lead to male-to-female sex reversal and are associated with development of gonadoblastoma. To further explore genetic association with TGCT, we undertook a pathway-based analysis of SNP marker associations in the Penn GWAs (349 TGCT cases and 919 controls). We analyzed a custom-built sex determination gene set consisting of 32 genes using three different methods of pathway-based analysis. The sex determination gene set ranked highly compared with canonical gene sets, and it was associated with TGCT (FDRG = 2.28 × 10(-5), FDRM = 0.014 and FDRI = 0.008 for Gene Set Analysis-SNP (GSA-SNP), Meta-Analysis Gene Set Enrichment of Variant Associations (MAGENTA) and Improved Gene Set Enrichment Analysis for Genome-wide Association Study (i-GSEA4GWAS) analysis, respectively). The association remained after removal of DMRT1 from the gene set (FDRG = 0.0002, FDRM = 0.055 and FDRI = 0.009). Using data from the NCI GWA scan (582 TGCT cases and 1056 controls) and UK scan (986 TGCT cases and 4946 controls), we replicated these findings (NCI: FDRG = 0.006, FDRM = 0.014, FDRI = 0.033, and UK: FDRG = 1.04 × 10(-6), FDRM = 0.016, FDRI = 0.025). After removal of DMRT1 from the gene set, the sex determination gene set remains associated with TGCT in the NCI (FDRG = 0.039, FDRM = 0.050 and FDRI = 0.055) and UK scans (FDRG = 3.00 × 10(-5), FDRM = 0.056 and FDRI = 0.044). With the exception of DMRT1, genes in the sex determination gene set have not previously been identified as TGCT susceptibility loci in these GWA scans, demonstrating the complementary nature of a pathway-based approach for genome-wide analysis of TGCT.


Human Molecular Genetics | 2015

Multi-stage genome-wide association study identifies new susceptibility locus for testicular germ cell tumour on chromosome 3q25

Kevin Litchfield; Razvan Sultana; Anthony Renwick; Darshna Dudakia; Sheila Seal; Emma Ramsay; Silvana Powell; Anna Elliott; Margaret Warren-Perry; Rosalind Eeles; Julian Peto; Zsofia Kote-Jarai; Kenneth Muir; Jérémie Nsengimana; Uktcc; Michael R. Stratton; Douglas F. Easton; D. Timothy Bishop; Robert Huddart; Nazneen Rahman; Clare Turnbull

Recent genome-wide association studies (GWAS) and subsequent meta-analyses have identified over 25 SNPs at 18 loci, together accounting for >15% of the genetic susceptibility to testicular germ cell tumour (TGCT). To identify further common SNPs associated with TGCT, here we report a three-stage experiment, involving 4098 cases and 18 972 controls. Stage 1 comprised previously published GWAS analysis of 307 291 SNPs in 986 cases and 4946 controls. In Stage 2, we used previously published customised Illumina iSelect genotyping array (iCOGs) data across 694 SNPs in 1064 cases and 10 082 controls. Here, we report new genotyping of eight SNPs showing some evidence of association in combined analysis of Stage 1 and Stage 2 in an additional 2048 cases of TGCT and 3944 controls (Stage 3). Through fixed-effects meta-analysis across three stages, we identified a novel locus at 3q25.31 (rs1510272) demonstrating association with TGCT [per-allele odds ratio (OR) = 1.16, 95% confidence interval (CI) = 1.06-1.27; P = 1.2 × 10(-9)].


Blood | 2016

Germline mutations in shelterin complex genes are associated with familial chronic lymphocytic leukemia.

Helen E. Speedy; Ben Kinnersley; Daniel Chubb; Peter Broderick; Philip J. Law; Kevin Litchfield; Sandrine Jayne; Martin J. S. Dyer; Claire Dearden; George A. Follows; Daniel Catovsky; Richard S. Houlston

Chronic lymphocytic leukemia (CLL) can be familial; however, thus far no rare germ line disruptive alleles for CLL have been identified. We performed whole-exome sequencing of 66 CLL families, identifying 4 families where loss-of-function mutations in protection of telomeres 1 (POT1) co-segregated with CLL. The p.Tyr36Cys mutation is predicted to disrupt the interaction between POT1 and the telomeric overhang. The c.1164-1G>A splice-site, p.Gln358SerfsTer13 frameshift, and p.Gln376Arg missense mutations are likely to impact the interaction between POT1 and adrenocortical dysplasia homolog (ACD), which is a part of the telomere-capping shelterin complex. We also identified mutations in ACD (c.752-2A>C) and another shelterin component, telomeric repeat binding factor 2, interacting protein (p.Ala104Pro and p.Arg133Gln), in 3 CLL families. In a complementary analysis of 1083 cases and 5854 controls, the POT1 p.Gln376Arg variant, which has a global minor allele frequency of 0.0005, conferred a 3.61-fold increased risk of CLL (P = .009). This study further highlights telomere dysregulation as a key process in CLL development.

Collaboration


Dive into the Kevin Litchfield's collaboration.

Top Co-Authors

Avatar

Clare Turnbull

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Richard S. Houlston

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Robert Huddart

The Royal Marsden NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar

Peter Broderick

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Darshna Dudakia

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Alison Reid

The Royal Marsden NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar

Janet Shipley

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Max Levy

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge