scispace - formally typeset
Search or ask a question

Showing papers by "Ludmil B. Alexandrov published in 2018"


Journal ArticleDOI
28 Feb 2018-Nature
TL;DR: A pan-cancer study of somatic alterations, including single nucleotide variants, small insertions or deletions, structural variations, copy number alterations, gene fusions and internal tandem duplications in 1,699 paediatric leukaemias and solid tumours across six histotypes, provides a comprehensive genomic architecture for paediatric cancers.
Abstract: Analysis of molecular aberrations across multiple cancer types, known as pan-cancer analysis, identifies commonalities and differences in key biological processes that are dysregulated in cancer cells from diverse lineages. Pan-cancer analyses have been performed for adult but not paediatric cancers, which commonly occur in developing mesodermic rather than adult epithelial tissues. Here we present a pan-cancer study of somatic alterations, including single nucleotide variants, small insertions or deletions, structural variations, copy number alterations, gene fusions and internal tandem duplications in 1,699 paediatric leukaemias and solid tumours across six histotypes, with whole-genome, whole-exome and transcriptome sequencing data processed under a uniform analytical framework. We report 142 driver genes in paediatric cancers, of which only 45% match those found in adult pan-cancer studies; copy number alterations and structural variants constituted the majority (62%) of events. Eleven genome-wide mutational signatures were identified, including one attributed to ultraviolet-light exposure in eight aneuploid leukaemias. Transcription of the mutant allele was detectable for 34% of protein-coding mutations, and 20% exhibited allele-specific expression. These data provide a comprehensive genomic architecture for paediatric cancers and emphasize the need for paediatric cancer-specific development of precision therapies.

573 citations


Journal ArticleDOI
11 Apr 2018-Nature
TL;DR: The results indicate that colorectal cancer cells experience substantial increases in somatic mutation rate compared to normal coloreCTal cells, and that genetic diversification of each cancer is accompanied by pervasive, stable and inherited differences in the biological states of individual cancer cells.
Abstract: Every cancer originates from a single cell. During expansion of the neoplastic cell population, individual cells acquire genetic and phenotypic differences from each other. Here, to investigate the nature and extent of intra-tumour diversification, we characterized organoids derived from multiple single cells from three colorectal cancers as well as from adjacent normal intestinal crypts. Colorectal cancer cells showed extensive mutational diversification and carried several times more somatic mutations than normal colorectal cells. Most mutations were acquired during the final dominant clonal expansion of the cancer and resulted from mutational processes that are absent from normal colorectal cells. Intra-tumour diversification of DNA methylation and transcriptome states also occurred; these alterations were cell-autonomous, stable, and followed the phylogenetic tree of each cancer. There were marked differences in responses to anticancer drugs between even closely related cells of the same tumour. The results indicate that colorectal cancer cells experience substantial increases in somatic mutation rate compared to normal colorectal cells, and that genetic diversification of each cancer is accompanied by pervasive, stable and inherited differences in the biological states of individual cancer cells.

383 citations


Journal ArticleDOI
TL;DR: It is shown LCNECs represent a distinct transcriptional subgroup among lung cancers and comprise two molecular subgroups, type I (TP53 and STK11/KEAP1 alterations) and type II ( TP53 and RB1 inactivation).
Abstract: Pulmonary large-cell neuroendocrine carcinomas (LCNECs) have similarities with other lung cancers, but their precise relationship has remained unclear. Here we perform a comprehensive genomic (n = 60) and transcriptomic (n = 69) analysis of 75 LCNECs and identify two molecular subgroups: “type I LCNECs” with bi-allelic TP53 and STK11/KEAP1 alterations (37%), and “type II LCNECs” enriched for bi-allelic inactivation of TP53 and RB1 (42%). Despite sharing genomic alterations with adenocarcinomas and squamous cell carcinomas, no transcriptional relationship was found; instead LCNECs form distinct transcriptional subgroups with closest similarity to SCLC. While type I LCNECs and SCLCs exhibit a neuroendocrine profile with ASCL1high/DLL3high/NOTCHlow, type II LCNECs bear TP53 and RB1 alterations and differ from most SCLC tumors with reduced neuroendocrine markers, a pattern of ASCL1low/DLL3low/NOTCHhigh, and an upregulation of immune-related pathways. In conclusion, LCNECs comprise two molecularly defined subgroups, and distinguishing them from SCLC may allow stratified targeted treatment of high-grade neuroendocrine lung tumors. The molecular nature of large-cell neuroendocrine lung carcinomas (LCNEC) has remained unclear. Here, the authors show LCNECs represent a distinct transcriptional subgroup among lung cancers and comprise two molecular subgroups, type I (TP53 and STK11/KEAP1 alterations) and type II (TP53 and RB1 inactivation).

233 citations


Posted ContentDOI
15 May 2018-bioRxiv
TL;DR: The substantial dataset size compared to previous analyses enabled discovery of new signatures, separation of overlapping signatures and decomposition of signatures into components that may represent associated, but distinct, DNA damage, repair and or replication mechanisms.
Abstract: Somatic mutations in cancer genomes are caused by multiple mutational processes each of which generates a characteristic mutational signature. Using 84,729,690 somatic mutations from 4,645 whole cancer genome and 19,184 exome sequences encompassing most cancer types we characterised 49 single base substitution, 11 doublet base substitution, four clustered base substitution, and 17 small insertion and deletion mutational signatures. The substantial dataset size compared to previous analyses enabled discovery of new signatures, separation of overlapping signatures and decomposition of signatures into components that may represent associated, but distinct, DNA damage, repair and/or replication mechanisms. Estimation of the contribution of each signature to the mutational catalogues of individual cancer genomes revealed associations with exogenous and endogenous exposures and defective DNA maintenance processes. However, many signatures are of unknown cause. This analysis provides a systematic perspective on the repertoire of mutational processes contributing to the development of human cancer including a comprehensive reference set of mutational signatures in human cancer.

215 citations


Journal ArticleDOI
TL;DR: In this article, the authors combine whole-exome analyses from 40 primary squamous cell carcinoma (cSCC) cells, comprising 20 well-differentiated and 20 moderately/poorly differentiated, with clinical data from a longitudinal study of immunosuppressed and immunocompetent patients and integrate this analysis with independent gene expression studies.
Abstract: Cutaneous squamous cell carcinoma (cSCC) has a high tumour mutational burden (50 mutations per megabase DNA pair). Here, we combine whole-exome analyses from 40 primary cSCC tumours, comprising 20 well-differentiated and 20 moderately/poorly differentiated tumours, with accompanying clinical data from a longitudinal study of immunosuppressed and immunocompetent patients and integrate this analysis with independent gene expression studies. We identify commonly mutated genes, copy number changes and altered pathways and processes. Comparisons with tumour differentiation status suggest events which may drive disease progression. Mutational signature analysis reveals the presence of a novel signature (signature 32), whose incidence correlates with chronic exposure to the immunosuppressive drug azathioprine. Characterisation of a panel of 15 cSCC tumour-derived cell lines reveals that they accurately reflect the mutational signatures and genomic alterations of primary tumours and provide a valuable resource for the validation of tumour drivers and therapeutic targets.

190 citations


Journal ArticleDOI
David C. Wedge1, David C. Wedge2, Gunes Gundem3, Gunes Gundem1, Thomas J. Mitchell1, Dan J. Woodcock2, Inigo Martincorena1, Mohammed J. R. Ghori1, Jorge Zamora1, Adam Butler1, Hayley C. Whitaker4, Zsofia Kote-Jarai5, Ludmil B. Alexandrov1, Peter Van Loo6, Peter Van Loo1, Charles E. Massie7, Charles E. Massie8, Stefan C. Dentro2, Stefan C. Dentro6, Stefan C. Dentro1, Anne Y. Warren9, Clare Verrill2, Daniel M. Berney10, Nening Dennis11, Sue Merson5, Steve Hawkins8, William Howat9, Yong-Jie Lu10, Adam Lambert2, Jonathan Kay4, Barbara Kremeyer1, Katalin Karaszi2, Hayley J. Luxton4, Niedzica Camacho5, Niedzica Camacho3, Luke Marsden2, S. Edwards5, Lucy Matthews2, Valeria Bo, Daniel Leongamornlert1, Daniel Leongamornlert5, Stuart McLaren1, Anthony C. H. Ng12, Yongwei Yu13, Hongwei Zhang13, Tokhir Dadaev5, Sarah Thomas11, Douglas F. Easton7, Mahbubl Ahmed5, Elizabeth Bancroft5, Elizabeth Bancroft11, Cyril Fisher11, Naomi Livni11, David Nicol11, Simon Tavaré, Pelvender Gill2, Christopher Greenman14, Vincent Khoo11, Nicholas van As11, Pardeep Kumar11, Chris Ogden11, Declan Cahill11, Alan Thompson11, Erik Mayer11, Edward Rowe11, Tim Dudderidge11, Vincent Gnanapragasam7, Vincent Gnanapragasam9, Nimish Shah9, Keiran Raine1, David T. Jones1, Andrew Menzies1, Lucy Stebbings1, Jon W. Teague1, Steven Hazell11, Cathy Corbishley15, Johann S. de Bono5, Gerhardt Attard5, William B. Isaacs16, Tapio Visakorpi, Michael Fraser17, Paul C. Boutros18, Paul C. Boutros19, Robert G. Bristow17, Robert G. Bristow19, Paul Workman5, Chris Sander20, Freddie C. Hamdy2, Andrew Futreal1, Ultan McDermott1, Bissan Al-Lazikani5, Andy G. Lynch21, G. Steven Bova16, Christopher S. Foster22, Daniel Brewer23, Daniel Brewer14, Daniel Brewer5, David E. Neal7, David E. Neal8, Colin Cooper5, Colin Cooper14, Rosalind A. Eeles11, Rosalind A. Eeles5 
TL;DR: Joint analysis of new and previously published sequencing data for primary and metastatic prostate cancers identifies new candidate driver mutations and provides insights into disease progression and potential drug targets.
Abstract: Prostate cancer represents a substantial clinical challenge because it is difficult to predict outcome and advanced disease is often fatal. We sequenced the whole genomes of 112 primary and metastatic prostate cancer samples. From joint analysis of these cancers with those from previous studies (930 cancers in total), we found evidence for 22 previously unidentified putative driver genes harboring coding mutations, as well as evidence for NEAT1 and FOXA1 acting as drivers through noncoding mutations. Through the temporal dissection of aberrations, we identified driver mutations specifically associated with steps in the progression of prostate cancer, establishing, for example, loss of CHD1 and BRCA2 as early events in cancer development of ETS fusion-negative cancers. Computational chemogenomic (canSAR) analysis of prostate cancer mutations identified 11 targets of approved drugs, 7 targets of investigational drugs, and 62 targets of compounds that may be active and should be considered candidates for future clinical trials.

182 citations


Journal ArticleDOI
TL;DR: The genomic landscape at the SMM stage is very similar to MM, but trajectories of evolution can vary from patient to patient, and activation-induced cytidine deaminase plays a major role in shaping the mutational landscape of early subclinical phases.
Abstract: We analyzed whole genomes of unique paired samples from smoldering multiple myeloma (SMM) patients progressing to multiple myeloma (MM). We report that the genomic landscape, including mutational profile and structural rearrangements at the smoldering stage is very similar to MM. Paired sample analysis shows two different patterns of progression: a “static progression model”, where the subclonal architecture is retained as the disease progressed to MM suggesting that progression solely reflects the time needed to accumulate a sufficient disease burden; and a “spontaneous evolution model”, where a change in the subclonal composition is observed. We also observe that activation-induced cytidine deaminase plays a major role in shaping the mutational landscape of early subclinical phases, while progression is driven by APOBEC cytidine deaminases. These results provide a unique insight into myelomagenesis with potential implications for the definition of smoldering disease and timing of treatment initiation. Smoldering MM (SMM) is a premalignant stage of multiple myeloma (MM). Here the authors perform whole genome sequencing of unique paired samples of SMM progressing to MM, and show that the genomic landscape at the SMM stage is very similar to MM, but trajectories of evolution can vary from patient to patient.

144 citations


Journal ArticleDOI
31 Aug 2018-Science
TL;DR: The genomic events that give rise to EWSR1-ETS fusions in ES and chart their evolution from diagnosis to relapse are reconstructed to reveal complex DNA rearrangements to be a mutational process underpinning gene fusion in a large proportion of ES.
Abstract: INTRODUCTION Gene fusions are often disease-defining events in cancer. The mutational processes that give rise to fusions, their timing relative to initial diagnosis, and whether they change at relapse are largely unknown. Mutational processes leave distinct marks in the tumor genome, meaning that DNA sequencing can be used to reconstruct how fusions are generated. A prototypical fusion-driven tumor is Ewing sarcoma (ES), a bone cancer predominantly affecting children and young adults. ES is defined by fusions involving EWSR1 , a gene encoding an RNA binding protein, and genes encoding E26 transformation-specific (ETS) transcription factors such as FLI1 . We sought to reconstruct the genomic events that give rise to EWSR1-ETS fusions in ES and chart their evolution from diagnosis to relapse. RATIONALE We studied the processes underpinning gene fusions in ES using the whole-genome sequences of 124 primary tumors. We determined the timing of the emergence of EWSR1 fusions relative to other mutations. To measure ongoing mutation rates and evolutionary trajectories of ES, we studied the genomes of primary tumors, tumors at relapse, and metastatic tumors. RESULTS We found that EWSR1-ETS , the key ES fusion, arises in 42% of cases via complex, loop-like rearrangements called chromoplexy, rather than by simple reciprocal translocations. Similar loops forming canonical fusions were found in three other sarcoma types. Timing the emergence of loops revealed that they occur as bursts in early replicating DNA, as a primary event in ES development. Additional gene disruptions are generated concurrently with the fusions within the loops. Chromoplexy-generated EWSR1 fusions appear to be associated with an aggressive form of the disease and a higher chance of relapse. Numerous mutations present in every cell of the primary were absent at relapse, demonstrating that the primary and relapsed diseases evolved independently. This divergence occurs after formation of an ancestral clone harboring EWSR1 fusions. Importantly, we determined that divergence of the primary tumor and the future relapsed tumor occurs 1 to 2 years before initial diagnosis, as estimated from the number of cell division–associated mutations. CONCLUSION Our findings provide insights into the pathogenesis and natural history of human sarcomas. They reveal complex DNA rearrangements to be a mutational process underpinning gene fusions in a large proportion of ES. Similar observations in other fusion-defined sarcoma types indicate that this process operates more generally. Such complex rearrangements occur preferentially in early replicating and transcriptionally active genomic regions, as evidenced by the additional genes disrupted. EWSR1 fusions arising from chromoplexy correlated with worse clinical outcomes. Formation of the EWSR1 fusion genes is a primary event in the life history of ES. We found evidence of a latency period between this seeding event and diagnosis. This is in keeping with the often-indolent nature of symptoms before clinical disease presentation.

122 citations


Journal ArticleDOI
10 Dec 2018-PLOS ONE
TL;DR: It is shown that the D-Wave 2X can be effectively used as part of an unsupervised machine learning method that is able to learn facial features and accurately reproduce the set of facial images.
Abstract: D-Wave quantum annealers represent a novel computational architecture and have attracted significant interest. Much of this interest has focused on the quantum behavior of D-Wave machines, and there have been few practical algorithms that use the D-Wave. Machine learning has been identified as an area where quantum annealing may be useful. Here, we show that the D-Wave 2X can be effectively used as part of an unsupervised machine learning method. This method takes a matrix as input and produces two low-rank matrices as output-one containing latent features in the data and another matrix describing how the features can be combined to approximately reproduce the input matrix. Despite the limited number of bits in the D-Wave hardware, this method is capable of handling a large input matrix. The D-Wave only limits the rank of the two output matrices. We apply this method to learn the features from a set of facial images and compare the performance of the D-Wave to two classical tools. This method is able to learn facial features and accurately reproduce the set of facial images. The performance of the D-Wave shows some promise, but has some limitations. It outperforms the two classical codes in a benchmark when only a short amount of computational time is allowed (200-20,000 microseconds), but these results suggest heuristics that would likely outperform the D-Wave in this benchmark.

92 citations


Journal ArticleDOI
TL;DR: The findings reveal a cause for cancers arising at sites of persistent inflammation and identify potential therapeutic avenues to treat RDEB SCC and establish specific mutagenic mechanisms associated with chronic tissue damage.
Abstract: Recessive dystrophic epidermolysis bullosa (RDEB) is a rare inherited skin and mucous membrane fragility disorder complicated by early-onset, highly malignant cutaneous squamous cell carcinomas (SCCs). The molecular etiology of RDEB SCC, which arises at sites of sustained tissue damage, is unknown. We performed detailed molecular analysis using whole-exome, whole-genome, and RNA sequencing of 27 RDEB SCC tumors, including multiple tumors from the same patient and multiple regions from five individual tumors. We report that driver mutations were shared with spontaneous, ultraviolet (UV) light-induced cutaneous SCC (UV SCC) and head and neck SCC (HNSCC) and did not explain the early presentation or aggressive nature of RDEB SCC. Instead, endogenous mutation processes associated with apolipoprotein B mRNA-editing enzyme catalytic polypeptide-like (APOBEC) deaminases dominated RDEB SCC. APOBEC mutation signatures were enhanced throughout RDEB SCC tumor evolution, relative to spontaneous UV SCC and HNSCC mutation profiles. Sixty-seven percent of RDEB SCC driver mutations was found to emerge as a result of APOBEC and other endogenous mutational processes previously associated with age, potentially explaining a >1000-fold increased incidence and the early onset of these SCCs. Human papillomavirus-negative basal and mesenchymal subtypes of HNSCC harbored enhanced APOBEC mutational signatures and transcriptomes similar to those of RDEB SCC, suggesting that APOBEC deaminases drive other subtypes of SCC. Collectively, these data establish specific mutagenic mechanisms associated with chronic tissue damage. Our findings reveal a cause for cancers arising at sites of persistent inflammation and identify potential therapeutic avenues to treat RDEB SCC.

91 citations




Journal ArticleDOI
TL;DR: A cross-species comparative analysis between a large cohort of patients and four diverse mouse models focused on clinically and therapeutically relevant aspects of genomic and transcriptomic profiles is implemented and two of these models are proposed as valid for the study of different stages of human HCC.
Abstract: Cancer genomics has enabled the exhaustive molecular characterization of tumors and exposed hepatocellular carcinoma (HCC) as among the most complex cancers. This complexity is paralleled by dozens of mouse models that generate histologically similar tumors but have not been systematically validated at the molecular level. Accurate models of the molecular pathogenesis of HCC are essential for biomedical progress; therefore we compared genomic and transcriptomic profiles of four separate mouse models [MUP transgenic, TAK1-knockout, carcinogen-driven diethylnitrosamine (DEN), and Stelic Animal Model (STAM)] with those of 987 HCC patients with distinct etiologies. These four models differed substantially in their mutational load, mutational signatures, affected genes and pathways, and transcriptomes. STAM tumors were most molecularly similar to human HCC, with frequent mutations in Ctnnb1, similar pathway alterations, and high transcriptomic similarity to high-grade, proliferative human tumors with poor prognosis. In contrast, TAK1 tumors better reflected the mutational signature of human HCC and were transcriptionally similar to low-grade human tumors. DEN tumors were least similar to human disease and almost universally carried the Braf V637E mutation, which is rarely found in human HCC. Immune analysis revealed that strain-specific MHC-I genotype can influence the molecular makeup of murine tumors. Thus, different mouse models of HCC recapitulate distinct aspects of HCC biology, and their use should be adapted to specific questions based on the molecular features provided here.

Posted ContentDOI
20 Apr 2018-bioRxiv
TL;DR: Using whole genome bisulfite sequencing of breast cancers, it is comprehensively shown that loss of methylation in PMDs occurs in a large fraction of the genome and represents the prime source of variation in DNA methylation.
Abstract: Global loss of DNA methylation and CpG island (CGI) hypermethylation are regarded as key epigenomic aberrations in cancer. Global loss manifests itself in partially methylated domains (PMDs) which can extend up to megabases. However, the distribution of PMDs within and between tumor types, and their effects on key functional genomic elements including CGIs are poorly defined. Using whole genome bisulfite sequencing (WGBS) of breast cancers, we comprehensively show that loss of methylation in PMDs occurs in a large fraction of the genome and represents the prime source of variation in DNA methylation. PMDs are hypervariable in methylation level, size and distribution, and display elevated mutation rates. They impose intermediate DNA methylation levels incognizant of functional genomic elements including CGIs, underpinning a CGI methylator phenotype (CIMP). However, significant repression effects on cancer-genes are negligible as tumor suppressor genes are generally excluded from PMDs. The genomic distribution of PMDs reports tissue-of-origin of different cancers and may represent tissue-specific 9silent9 regions of the genome, which tolerate instability at the epigenetic, transcriptomic and genetic level.

Posted ContentDOI
30 Oct 2018-bioRxiv
TL;DR: It is inferred that metastasis occurs rapidly across multiple sites, constituting a model of metastatic spread the authors term clonal diaspora, which has implications for understanding metastatic progression, clinical staging and patient management.
Abstract: Continual evolution of cancer makes it challenging to predict clinical outcomes. Highly varied and unpredictable patient outcomes in esophageal adenocarcinoma (EAC) prompted us to question the pattern and timing of metastatic spread. Whole genome sequencing and phylogenetic analysis of 396 samples across 18 EAC cases demonstrated a stellate pattern on the phylogenetic trees in 90% cases. The age-dependent trinucleotide signature, which can serve as a molecular clock, was absent or reduced in the stellate branches beyond the trunk in most cases (p

Posted ContentDOI
27 Nov 2018-bioRxiv
TL;DR: In this paper, the authors investigated the stepwise process in which additional genomic alterations such as copy number alterations (SCNAs) or structural variants (SV) are acquired.
Abstract: Intratumor heterogeneity (ITH) and tumor evolution have been described for clear cell renal cell carcinomas (ccRCC), but only limited data are available for other kidney cancer subtypes. Moreover, previous ITH studies predominately focused on single nucleotide variants (SNVs); little is known of the stepwise process in which additional genomic alterations such as copy number alterations (SCNAs) or structural variants (SV) are acquired. We investigated ITH and clonal evolution of papillary renal cell carcinoma (pRCC) and rarer kidney cancer subtypes using whole-genome sequencing and multi-omics analyses in 124 samples from 29 subjects. We collected multiple samples from the center of the tumor to the periphery and matched metastatic lesions to capture changes occurring along the physical tumor expansion. We used phylogenetic analysis to order the impact of SCNAs, SNVs, and SVs along the evolutionary trajectory of these tumors. While the few mutations in cancer driver genes were clonal, pRCC ITH was lowest for SCNAs, intermediate for SNVs, and highest for SVs. The phylogenetic analysis confirmed a clonal expansion cascade along these genomic alteration types. Moreover, while SNVs and SCNAs were similar, SVs were >20 times more frequent in pRCC type 2 than pRCC type 1, suggesting a role for SVs in pRCC type 2 aggressive behavior. Unlike ccRCC or other cancer types, pRCCs tumorigenesis appears to begin from SCNAs and/or rare mutations in cancer driver genes. No effective treatment is available for this tumor. Our work highlights the need for tailored intervention against large-scale somatic alterations beyond SNVs.