Try a new search

Format these results:

Searched for:

person:imielm01

in-biosketch:yes

Total Results:

110


SvABA: genome-wide detection of structural variants and indels by local assembly

Wala, Jeremiah A; Bandopadhayay, Pratiti; Greenwald, Noah F; O'Rourke, Ryan; Sharpe, Ted; Stewart, Chip; Schumacher, Steve; Li, Yilong; Weischenfeldt, Joachim; Yao, Xiaotong; Nusbaum, Chad; Campbell, Peter; Getz, Gad; Meyerson, Matthew; Zhang, Cheng-Zhong; Imielinski, Marcin; Beroukhim, Rameen
Structural variants (SVs), including small insertion and deletion variants (indels), are challenging to detect through standard alignment-based variant calling methods. Sequence assembly offers a powerful approach to identifying SVs, but is difficult to apply at scale genome-wide for SV detection due to its computational complexity and the difficulty of extracting SVs from assembly contigs. We describe SvABA, an efficient and accurate method for detecting SVs from short-read sequencing data using genome-wide local assembly with low memory and computing requirements. We evaluated SvABA's performance on the NA12878 human genome and in simulated and real cancer genomes. SvABA demonstrates superior sensitivity and specificity across a large spectrum of SVs and substantially improves detection performance for variants in the 20-300 bp range, compared with existing methods. SvABA also identifies complex somatic rearrangements with chains of short (<1000 bp) templated-sequence insertions copied from distant genomic regions. We applied SvABA to 344 cancer genomes from 11 cancer types and found that short templated-sequence insertions occur in ∼4% of all somatic rearrangements. Finally, we demonstrate that SvABA can identify sites of viral integration and cancer driver alterations containing medium-sized (50-300 bp) SVs.
PMCID:5880247
PMID: 29535149
ISSN: 1549-5469
CID: 4195292

Insertions and Deletions Target Lineage-Defining Genes in Human Cancers

Imielinski, Marcin; Guo, Guangwu; Meyerson, Matthew
Certain cell types function as factories, secreting large quantities of one or more proteins that are central to the physiology of the respective organ. Examples include surfactant proteins in lung alveoli, albumin in liver parenchyma, and lipase in the stomach lining. Whole-genome sequencing analysis of lung adenocarcinomas revealed noncoding somatic mutational hotspots near VMP1/MIR21 and indel hotspots in surfactant protein genes (SFTPA1, SFTPB, and SFTPC). Extrapolation to other solid cancers demonstrated highly recurrent and tumor-type-specific indel hotspots targeting the noncoding regions of highly expressed genes defining certain secretory cellular lineages: albumin (ALB) in liver carcinoma, gastric lipase (LIPF) in stomach carcinoma, and thyroglobulin (TG) in thyroid carcinoma. The sequence contexts of indels targeting lineage-defining genes were significantly enriched in the AATAATD DNA motif and specific chromatin contexts, including H3K27ac and H3K36me3. Our findings illuminate a prevalent and hitherto unrecognized mutational process linking cellular lineage and cancer.
PMCID:5564321
PMID: 28089356
ISSN: 1097-4172
CID: 5270172

Mapping the hallmarks of lung adenocarcinoma with massively parallel sequencing

Imielinski, Marcin; Berger, Alice H; Hammerman, Peter S; Hernandez, Bryan; Pugh, Trevor J; Hodis, Eran; Cho, Jeonghee; Suh, James; Capelletti, Marzia; Sivachenko, Andrey; Sougnez, Carrie; Auclair, Daniel; Lawrence, Michael S; Stojanov, Petar; Cibulskis, Kristian; Choi, Kyusam; de Waal, Luc; Sharifnia, Tanaz; Brooks, Angela; Greulich, Heidi; Banerji, Shantanu; Zander, Thomas; Seidel, Danila; Leenders, Frauke; Ansen, Sascha; Ludwig, Corinna; Engel-Riedel, Walburga; Stoelben, Erich; Wolf, Jurgen; Goparju, Chandra; Thompson, Kristin; Winckler, Wendy; Kwiatkowski, David; Johnson, Bruce E; Janne, Pasi A; Miller, Vincent A; Pao, William; Travis, William D; Pass, Harvey I; Gabriel, Stacey B; Lander, Eric S; Thomas, Roman K; Garraway, Levi A; Getz, Gad; Meyerson, Matthew
Lung adenocarcinoma, the most common subtype of non-small cell lung cancer, is responsible for more than 500,000 deaths per year worldwide. Here, we report exome and genome sequences of 183 lung adenocarcinoma tumor/normal DNA pairs. These analyses revealed a mean exonic somatic mutation rate of 12.0 events/megabase and identified the majority of genes previously reported as significantly mutated in lung adenocarcinoma. In addition, we identified statistically recurrent somatic mutations in the splicing factor gene U2AF1 and truncating mutations affecting RBM10 and ARID1A. Analysis of nucleotide context-specific mutation signatures grouped the sample set into distinct clusters that correlated with smoking history and alterations of reported lung adenocarcinoma genes. Whole-genome sequence analysis revealed frequent structural rearrangements, including in-frame exonic alterations within EGFR and SIK2 kinases. The candidate genes identified in this study are attractive targets for biological characterization and therapeutic targeting of lung adenocarcinoma.
PMCID:3557932
PMID: 22980975
ISSN: 0092-8674
CID: 753362

Genomic sequence context differs between germline and somatic structural variants allowing for their differentiation in tumor samples without paired normals

Chukwu, Wolu; Lee, Siyun; Crane, Alexander; Zhang, Shu; Mittra, Ipsa; Imielinski, Marcin; Beroukhim, Rameen; Dubois, Frank; Dalin, Simona
There is currently no method to distinguish between germline and somatic structural variants (SVs) in tumor samples that lack a matched normal sample. In this study, we analyzed several features of germline and somatic SVs from a cohort of 974 patients from The Cancer Genome Atlas (TCGA). We identified a total of 21 features that differed significantly between germline and somatic SVs. Several of the germline SV features were associated with each other, as were several of the somatic SV features. We also found that these associations differed between the germline and somatic classes, for example, we found that somatic inversions were more likely to be longer events than their germline counterparts. Using these features we trained a support vector machine (SVM) classifier on 555,849 TCGA SVs to computationally distinguish germline from somatic SVs in the absence of a matched normal. This classifier had an ROC curve AUC of 0.984 when tested on an independent test set of 277,925 TCGA SVs. In this dataset, we achieved a positive predictive value (PPV) of 0.81 for an SV called somatic by the classifier being truly somatic. We further tested the classifier on a separate set of 7,623 SVs from pediatric high-grade gliomas (pHGG). In this non-TCGA cohort, our classifier achieved a PPV of 0.828, showing robust performance across datasets.
PMCID:10723258
PMID: 38106141
CID: 5637392

The importance of escalating molecular diagnostics in patients with low-grade pediatric brain cancer

Al Assaad, Majd; Gundem, Gunes; Liechty, Benjamin; Sboner, Andrea; Medina, Juan; Papaemmanuil, Elli; Sternberg, Cora N; Marks, Asher; Souweidane, Mark M; Greenfield, Jeffrey P; Tran, Ivy; Snuderl, Matija; Elemento, Oliver; Imielinski, Marcin; Pisapia, David J; Mosquera, Juan Miguel
Pilocytic astrocytomas are the most common pediatric brain tumors, typically presenting as low-grade neoplasms. We report two cases of pilocytic astrocytoma with atypical tumor progression. Case 1 involves a 12-year-old boy with an unresectable suprasellar tumor, negative for BRAF rearrangement but harboring a BRAF p.V600E mutation. He experienced tumor size reduction and stable disease following dabrafenib treatment. Case 2 describes a 6-year-old boy with a thalamic tumor that underwent multiple resections, with no actionable driver detected using targeted next-generation sequencing (NGS). Whole-genome and RNAseq analysis identified an internal tandem duplication in FGFR1 and RAS pathway activation. Future management options include FGFR1 inhibitors. These cases demonstrate the importance of escalating molecular diagnostics for pediatric brain cancer, advocating for early reflexing to integrative whole-genome sequencing and transcriptomic profiling when targeted panels are uninformative. Identifying molecular drivers can significantly impact treatment decisions and improve patient outcomes.
PMID: 37652664
ISSN: 2373-2873
CID: 5607272

Molecular Evolution of Classic Hodgkin Lymphoma Revealed Through Whole-Genome Sequencing of Hodgkin and Reed Sternberg Cells

Maura, Francesco; Ziccheddu, Bachisio; Xiang, Jenny Z; Bhinder, Bhavneet; Rosiene, Joel; Abascal, Federico; Maclachlan, Kylee H; Eng, Kenneth Wha; Uppal, Manik; He, Feng; Zhang, Wei; Gao, Qi; Yellapantula, Venkata D; Trujillo-Alonso, Vicenta; Park, Sunita I; Oberley, Matthew J; Ruckdeschel, Elizabeth; Lim, Megan S; Wertheim, Gerald B; Barth, Matthew J; Horton, Terzah M; Derkach, Andriy; Kovach, Alexandra E; Forlenza, Christopher J; Zhang, Yanming; Landgren, Ola; Moskowitz, Craig H; Cesarman, Ethel; Imielinski, Marcin; Elemento, Olivier; Roshal, Mikhail; Giulino-Roth, Lisa
UNLABELLED:The rarity of malignant Hodgkin and Reed Sternberg (HRS) cells in classic Hodgkin lymphoma (cHL) limits the ability to study the genomics of cHL. To circumvent this, our group has previously optimized fluorescence-activated cell sorting to purify HRS cells. Using this approach, we now report the whole-genome sequencing landscape of HRS cells and reconstruct the chronology and likely etiology of pathogenic events leading to cHL. We identified alterations in driver genes not previously described in cHL, APOBEC mutational activity, and the presence of complex structural variants including chromothripsis. We found that high ploidy in cHL is often acquired through multiple, independent chromosomal gains events including whole-genome duplication. Evolutionary timing analyses revealed that structural variants enriched for RAG motifs, driver mutations in B2M, BCL7A, GNA13, and PTPN1, and the onset of AID-driven mutagenesis usually preceded large chromosomal gains. This study provides a temporal reconstruction of cHL pathogenesis. SIGNIFICANCE:Previous studies in cHL were limited to coding sequences and therefore not able to comprehensively decipher the tumor complexity. Here, leveraging cHL whole-genome characterization, we identify driver events and reconstruct the tumor evolution, finding that structural variants, driver mutations, and AID mutagenesis precede chromosomal gains. This article is highlighted in the In This Issue feature, p. 171.
PMID: 36723991
ISSN: 2643-3249
CID: 5637362

Genomic signature of Fanconi anaemia DNA repair pathway deficiency in cancer

Webster, Andrew L H; Sanders, Mathijs A; Patel, Krupa; Dietrich, Ralf; Noonan, Raymond J; Lach, Francis P; White, Ryan R; Goldfarb, Audrey; Hadi, Kevin; Edwards, Matthew M; Donovan, Frank X; Hoogenboezem, Remco M; Jung, Moonjung; Sridhar, Sunandini; Wiley, Tom F; Fedrigo, Olivier; Tian, Huasong; Rosiene, Joel; Heineman, Thomas; Kennedy, Jennifer A; Bean, Lorenzo; Rosti, Rasim O; Tryon, Rebecca; Gonzalez, Ashlyn-Maree; Rosenberg, Allana; Luo, Ji-Dung; Carroll, Thomas S; Shroff, Sanjana; Beaumont, Michael; Velleuer, Eunike; Rastatter, Jeff C; Wells, Susanne I; Surrallés, Jordi; Bagby, Grover; MacMillan, Margaret L; Wagner, John E; Cancio, Maria; Boulad, Farid; Scognamiglio, Theresa; Vaughan, Roger; Beaumont, Kristin G; Koren, Amnon; Imielinski, Marcin; Chandrasekharappa, Settara C; Auerbach, Arleen D; Singh, Bhuvanesh; Kutler, David I; Campbell, Peter J; Smogorzewska, Agata
Fanconi anaemia (FA), a model syndrome of genome instability, is caused by a deficiency in DNA interstrand crosslink repair resulting in chromosome breakage1-3. The FA repair pathway protects against endogenous and exogenous carcinogenic aldehydes4-7. Individuals with FA are hundreds to thousands fold more likely to develop head and neck (HNSCC), oesophageal and anogenital squamous cell carcinomas8 (SCCs). Molecular studies of SCCs from individuals with FA (FA SCCs) are limited, and it is unclear how FA SCCs relate to sporadic HNSCCs primarily driven by tobacco and alcohol exposure or infection with human papillomavirus9 (HPV). Here, by sequencing genomes and exomes of FA SCCs, we demonstrate that the primary genomic signature of FA repair deficiency is the presence of high numbers of structural variants. Structural variants are enriched for small deletions, unbalanced translocations and fold-back inversions, and are often connected, thereby forming complex rearrangements. They arise in the context of TP53 loss, but not in the context of HPV infection, and lead to somatic copy-number alterations of HNSCC driver genes. We further show that FA pathway deficiency may lead to epithelial-to-mesenchymal transition and enhanced keratinocyte-intrinsic inflammatory signalling, which would contribute to the aggressive nature of FA SCCs. We propose that the genomic instability in sporadic HPV-negative HNSCC may arise as a result of the FA repair pathway being overwhelmed by DNA interstrand crosslink damage caused by alcohol and tobacco-derived aldehydes, making FA SCC a powerful model to study tumorigenesis resulting from DNA-crosslinking damage.
PMID: 36450981
ISSN: 1476-4687
CID: 5459302

Whole-genome characterization of myoepithelial carcinomas of the soft tissue

Cyrta, Joanna; Rosiene, Joel; Bareja, Rohan; Kudman, Sarah; Al Zoughbi, Wael; Motanagh, Samaneh; Wilkes, David C; Eng, Kenneth; Zhang, Tuo; Sticca, Evan; Mathew, Susan; Rubin, Mark A; Sboner, Andrea; Elemento, Olivier; Rubin, Brian P; Imielinski, Marcin; Mosquera, Juan Miguel
Myoepithelial carcinomas (MECs) of soft tissue are rare and aggressive tumors affecting young adults and children, but their molecular landscape has not been comprehensively explored through genome sequencing. Here, we present the whole-exome sequencing (WES), whole-genome sequencing (WGS), and RNA sequencing findings of two MECs. Patients 1 and 2 (P1, P2), both male, were diagnosed at 27 and 37 yr of age, respectively, with shoulder (P1) and inguinal (P2) soft tissue tumors. Both patients developed metastatic disease, and P2 died of disease. P1 tumor showed a rhabdoid cytomorphology and a complete loss of INI1 (SMARCB1) expression, associated with a homozygous SMARCB1 deletion. The tumor from P2 showed a clear cell/small cell morphology, retained INI1 expression and strong S100 positivity. By WES and WGS, tumors from both patients displayed low tumor mutation burdens, and no targetable alterations in cancer genes were detected. P2's tumor harbored an EWSR1::KLF15 rearrangement, whereas the tumor from P1 showed a novel ASCC2::GGNBP2 fusion. WGS evidenced a complex genomic event involving mainly Chromosomes 17 and 22 in the tumor from P1, which was consistent with chromoplexy. These findings are consistent with previous reports of EWSR1 rearrangements (50% of cases) in MECs and provide a genetic basis for the loss of SMARCB1 protein expression observed through immunohistochemistry in 10% of 40% of MEC cases. The lack of additional driver mutations in these tumors supports the hypothesis that these alterations are the key molecular events in MEC evolution. Furthermore, the presence of complex structural variant patterns, invisible to WES, highlights the novel biological insights that can be gained through the application of WGS to rare cancers.
PMCID:9808553
PMID: 36577525
ISSN: 2373-2873
CID: 5459312

Recurrent somatic mutations as predictors of immunotherapy response

Gajic, Zoran Z; Deshpande, Aditya; Legut, Mateusz; Imieliński, Marcin; Sanjana, Neville E
Immune checkpoint blockade (ICB) has transformed the treatment of metastatic cancer but is hindered by variable response rates. A key unmet need is the identification of biomarkers that predict treatment response. To address this, we analyzed six whole exome sequencing cohorts with matched disease outcomes to identify genes and pathways predictive of ICB response. To increase detection power, we focus on genes and pathways that are significantly mutated following correction for epigenetic, replication timing, and sequence-based covariates. Using this technique, we identify several genes (BCLAF1, KRAS, BRAF, and TP53) and pathways (MAPK signaling, p53 associated, and immunomodulatory) as predictors of ICB response and develop the Cancer Immunotherapy Response CLassifiEr (CIRCLE). Compared to tumor mutational burden alone, CIRCLE led to superior prediction of ICB response with a 10.5% increase in sensitivity and a 11% increase in specificity. We envision that CIRCLE and more broadly the analysis of recurrently mutated cancer genes will pave the way for better prognostic tools for cancer immunotherapy.
PMCID:9270330
PMID: 35803911
ISSN: 2041-1723
CID: 5270412

SETD2 Haploinsufficiency Enhances Germinal Center-Associated AICDA Somatic Hypermutation to Drive B-cell Lymphomagenesis

Leung, Wilfred; Teater, Matt; Durmaz, Ceyda; Meydan, Cem; Chivu, Alexandra G; Chadburn, Amy; Rice, Edward J; Muley, Ashlesha; Camarillo, Jeannie M; Arivalagan, Jaison; Li, Ziyi; Flowers, Christopher R; Kelleher, Neil L; Danko, Charles G; Imielinski, Marcin; Dave, Sandeep S; Armstrong, Scott A; Mason, Christopher E; Melnick, Ari M
SETD2 is the sole histone methyltransferase responsible for H3K36me3, with roles in splicing, transcription initiation, and DNA damage response. Homozygous disruption of SETD2 yields a tumor suppressor effect in various cancers. However, SETD2 mutation is typically heterozygous in diffuse large B-cell lymphomas. Here we show that heterozygous Setd2 deficiency results in germinal center (GC) hyperplasia and increased competitive fitness, with reduced DNA damage checkpoint activity and apoptosis, resulting in accelerated lymphomagenesis. Impaired DNA damage sensing in Setd2-haploinsufficient germinal center B (GCB) and lymphoma cells associated with increased AICDA-induced somatic hypermutation, complex structural variants, and increased translocations including those activating MYC. DNA damage was selectively increased on the nontemplate strand, and H3K36me3 loss was associated with greater RNAPII processivity and mutational burden, suggesting that SETD2-mediated H3K36me3 is required for proper sensing of cytosine deamination. Hence, Setd2 haploinsufficiency delineates a novel GCB context-specific oncogenic pathway involving defective epigenetic surveillance of AICDA-mediated effects on transcribed genes.
PMCID:9262862
PMID: 35443279
ISSN: 2159-8290
CID: 5270382