Sir Peter MacCallum Department of Oncology - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 10 of 16
  • Item
    Thumbnail Image
    Chemotherapy weakly contributes to predicted neoantigen expression in ovarian cancer
    O'Donnell, T ; Christie, EL ; Ahuja, A ; Buros, J ; Aksoy, BA ; Bowtell, DDL ; Snyder, A ; Hammerbacher, J (BMC, 2018-01-22)
    BACKGROUND: Patients with highly mutated tumors, such as melanoma or smoking-related lung cancer, have higher rates of response to immune checkpoint blockade therapy, perhaps due to increased neoantigen expression. Many chemotherapies including platinum compounds are known to be mutagenic, but the impact of standard treatment protocols on mutational burden and resulting neoantigen expression in most human cancers is unknown. METHODS: We sought to quantify the effect of chemotherapy treatment on computationally predicted neoantigen expression for high grade serous ovarian carcinoma patients enrolled in the Australian Ovarian Cancer Study. In this series, 35 of 114 samples were collected after exposure to chemotherapy; 14 are matched with an untreated sample from the same patient. Our approach integrates whole genome and RNA sequencing of bulk tumor samples with class I MHC binding prediction and mutational signatures extracted from studies of chemotherapy-exposed Caenorhabditis elegans and Gallus gallus cells. We additionally investigated the relationship between neoantigens, tumor infiltrating immune cells estimated from RNA-seq with CIBERSORT, and patient survival. RESULTS: Greater neoantigen burden and CD8+ T cell infiltration in primary, pre-treatment samples were independently associated with improved survival. Relapse samples collected after chemotherapy harbored a median of 78% more expressed neoantigens than untreated primary samples, a figure that combines the effects of chemotherapy and other processes operative during relapse. The contribution from chemotherapy-associated signatures was small, accounting for a mean of 5% (range 0-16) of the expressed neoantigen burden in relapse samples. In both treated and untreated samples, most neoantigens were attributed to COSMIC Signature (3), associated with BRCA disruption, Signature (1), associated with a slow mutagenic process active in healthy tissue, and Signature (8), of unknown etiology. CONCLUSION: Relapsed ovarian cancers harbor more predicted neoantigens than primary tumors, but the increase is due to pre-existing mutational processes, not mutagenesis from chemotherapy.
  • Item
    Thumbnail Image
    Ovarian Carcinoma-Associated Mesenchymal Stem Cells Arise from Tissue-Specific Normal Stroma
    Coffman, LG ; Pearson, AT ; Frisbie, LG ; Freeman, Z ; Christie, E ; Bowtell, DD ; Buckanovich, RJ (WILEY, 2019-02)
    Carcinoma-associated mesenchymal stem cells (CA-MSCs) are critical stromal progenitor cells within the tumor microenvironment (TME). We previously demonstrated that CA-MSCs differentially express bone morphogenetic protein family members, promote tumor cell growth, increase cancer "stemness," and chemotherapy resistance. Here, we use RNA sequencing of normal omental MSCs and ovarian CA-MSCs to demonstrate global changes in CA-MSC gene expression. Using these expression profiles, we create a unique predictive algorithm to classify CA-MSCs. Our classifier accurately distinguishes normal omental, ovary, and bone marrow MSCs from ovarian cancer CA-MSCs. Suggesting broad applicability, the model correctly classifies pancreatic and endometrial cancer CA-MSCs and distinguishes cancer associated fibroblasts from CA-MSCs. Using this classifier, we definitively demonstrate ovarian CA-MSCs arise from tumor mediated reprograming of local tissue MSCs. Although cancer cells alone cannot induce a CA-MSC phenotype, the in vivo ovarian TME can reprogram omental or ovary MSCs to protumorigenic CA-MSCs (classifier score of >0.96). In vitro studies suggest that both tumor secreted factors and hypoxia are critical to induce the CA-MSC phenotype. Interestingly, although the breast cancer TME can reprogram bone marrow MSCs into CA-MSCs, the ovarian TME cannot, demonstrating for the first time that tumor mediated CA-MSC conversion is tissue and cancer type dependent. Together these findings (a) provide a critical tool to define CA-MSCs and (b) highlight cancer cell influence on distinct normal tissues providing powerful insights into the mechanisms underlying cancer specific metastatic niche formation. Stem Cells 2019;37:257-269.
  • Item
    Thumbnail Image
    Multiple ABCB1 transcriptional fusions in drug resistant high-grade serous ovarian and breast cancer
    Christie, EL ; Pattnaik, S ; Beach, J ; Copeland, A ; Rashoo, N ; Fereday, S ; Hendley, J ; Alsop, K ; Brady, SL ; Lamb, G ; Pandey, A ; deFazio, A ; Thorne, H ; Bild, A ; Bowtell, DDL (NATURE PUBLISHING GROUP, 2019-03-20)
    ABCB1 encodes Multidrug Resistance protein (MDR1), an ATP-binding cassette member involved in the cellular efflux of chemotherapeutic drugs. Here we report that ovarian and breast samples from chemotherapy treated patients are positive for multiple transcriptional fusions involving ABCB1, placing it under the control of a strong promoter while leaving its open reading frame intact. We identified 15 different transcriptional fusion partners involving ABCB1, as well as patients with multiple distinct fusion events. The partner gene selected depended on its structure, promoter strength, and chromosomal proximity to ABCB1. Fusion positivity was strongly associated with the number of lines of MDR1-substrate chemotherapy given. MDR1 inhibition in a fusion positive ovarian cancer cell line increased sensitivity to paclitaxel more than 50-fold. Convergent evolution of ABCB1 fusion is therefore frequent in chemotherapy resistant recurrent ovarian cancer. As most currently approved PARP inhibitors (PARPi) are MDR1 substrates, prior chemotherapy may precondition resistance to PARPi.
  • Item
    Thumbnail Image
    Genomic footprints of activated telomere maintenance mechanisms in cancer
    Sieverling, L ; Hong, C ; Koser, SD ; Ginsbach, P ; Kleinheinz, K ; Hutter, B ; Braun, DM ; Cortes-Ciriano, I ; Xi, R ; Kabbe, R ; Park, PJ ; Eils, R ; Schlesner, M ; Brors, B ; Rippe, K ; Jones, DTW ; Feuerbach, L (NATURE PORTFOLIO, 2020-02-05)
    Cancers require telomere maintenance mechanisms for unlimited replicative potential. They achieve this through TERT activation or alternative telomere lengthening associated with ATRX or DAXX loss. Here, as part of the ICGC/TCGA Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium, we dissect whole-genome sequencing data of over 2500 matched tumor-control samples from 36 different tumor types aggregated within the ICGC/TCGA Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium to characterize the genomic footprints of these mechanisms. While the telomere content of tumors with ATRX or DAXX mutations (ATRX/DAXXtrunc) is increased, tumors with TERT modifications show a moderate decrease of telomere content. One quarter of all tumor samples contain somatic integrations of telomeric sequences into non-telomeric DNA. This fraction is increased to 80% prevalence in ATRX/DAXXtrunc tumors, which carry an aberrant telomere variant repeat (TVR) distribution as another genomic marker. The latter feature includes enrichment or depletion of the previously undescribed singleton TVRs TTCGGG and TTTGGG, respectively. Our systematic analysis provides new insight into the recurrent genomic alterations associated with telomere maintenance mechanisms in cancer.
  • Item
    Thumbnail Image
    High-coverage whole-genome analysis of 1220 cancers reveals hundreds of genes deregulated by rearrangement-mediated cis-regulatory alterations
    Zhang, Y ; Chen, F ; Fonseca, NA ; He, Y ; Fujita, M ; Nakagawa, H ; Zhang, Z ; Brazma, A ; Creighton, CJ (NATURE PUBLISHING GROUP, 2020-02-05)
    The impact of somatic structural variants (SVs) on gene expression in cancer is largely unknown. Here, as part of the ICGC/TCGA Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium, which aggregated whole-genome sequencing data and RNA sequencing from a common set of 1220 cancer cases, we report hundreds of genes for which the presence within 100 kb of an SV breakpoint associates with altered expression. For the majority of these genes, expression increases rather than decreases with corresponding breakpoint events. Up-regulated cancer-associated genes impacted by this phenomenon include TERT, MDM2, CDK4, ERBB2, CD274, PDCD1LG2, and IGF2. TERT-associated breakpoints involve ~3% of cases, most frequently in liver biliary, melanoma, sarcoma, stomach, and kidney cancers. SVs associated with up-regulation of PD1 and PDL1 genes involve ~1% of non-amplified cases. For many genes, SVs are significantly associated with increased numbers or greater proximity of enhancer regulatory elements near the gene. DNA methylation near the promoter is often increased with nearby SV breakpoint, which may involve inactivation of repressor elements.
  • Item
    Thumbnail Image
    Comprehensive analysis of chromothripsis in 2,658 human cancers using whole-genome sequencing
    Cortes-Ciriano, I ; Lee, JJ-K ; Xi, R ; Jain, D ; Jung, YL ; Yang, L ; Gordenin, D ; Klimczak, LJ ; Zhang, C-Z ; Pellman, DS ; Park, PJ ; Akdemir, KC ; Alvarez, EG ; Baez-Ortega, A ; Beroukhim, R ; Boutros, PC ; Bowtell, DDL ; Brors, B ; Burns, KH ; Campbell, PJ ; Chan, K ; Chen, K ; Dueso-Barroso, A ; Dunford, AJ ; Edwards, PA ; Estivill, X ; Etemadmoghadam, D ; Feuerbach, L ; Fink, JL ; Frenkel-Morgenstern, M ; Garsed, DW ; Gerstein, M ; Gordenin, DA ; Haan, D ; Haber, JE ; Hess, JM ; Hutter, B ; Imielinski, M ; Jones, DTW ; Ju, YS ; Kazanov, MD ; Koh, Y ; Korbel, JO ; Kumar, K ; Lee, EA ; Li, Y ; Lynch, AG ; Macintyre, G ; Markowetz, F ; Martincorena, I ; Martinez-Fundichely, A ; Miyano, S ; Nakagawa, H ; Navarro, FCP ; Ossowski, S ; Pearson, J ; Puiggros, M ; Rippe, K ; Roberts, ND ; Roberts, SA ; Rodriguez-Martin, B ; Schumacher, SE ; Scully, R ; Shackleton, M ; Sidiropoulos, N ; Sieverling, L ; Stewart, C ; Torrents, D ; Tubio, JMC ; Villasante, I ; Waddell, N ; Wala, JA ; Weischenfeldt, J ; Yao, X ; Yoon, S-S ; Zamora, J ; Alsop, K ; Christie, EL ; Fereday, S ; Mileshkin, L ; Mitchell, C ; Thorne, H ; Traficante, N ; Cmero, M ; Cowin, PA ; Hamilton, A ; Mir Arnau, G ; Vedururu, R ; Grimmond, SM ; Hofmann, O ; Morrison, C ; Oien, KA ; Pairojkul, C ; Waring, PM ; van de Vijver, MJ ; Behren, A (Nature Research, 2020-03)
    Chromothripsis is a mutational phenomenon characterized by massive, clustered genomic rearrangements that occurs in cancer and other diseases. Recent studies in selected cancer types have suggested that chromothripsis may be more common than initially inferred from low-resolution copy-number data. Here, as part of the Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium of the International Cancer Genome Consortium (ICGC) and The Cancer Genome Atlas (TCGA), we analyze patterns of chromothripsis across 2,658 tumors from 38 cancer types using whole-genome sequencing data. We find that chromothripsis events are pervasive across cancers, with a frequency of more than 50% in several cancer types. Whereas canonical chromothripsis profiles display oscillations between two copy-number states, a considerable fraction of events involve multiple chromosomes and additional structural alterations. In addition to non-homologous end joining, we detect signatures of replication-associated processes and templated insertions. Chromothripsis contributes to oncogene amplification and to inactivation of genes such as mismatch-repair-related genes. These findings show that chromothripsis is a major process that drives genome evolution in human cancer.
  • Item
    Thumbnail Image
    Cancer LncRNA Census reveals evidence for deep functional conservation of long noncoding RNAs in tumorigenesis
    Carlevaro-Fita, J ; Lanzos, A ; Feuerbach, L ; Hong, C ; Mas-Ponte, D ; Pedersen, JS ; Johnson, R ; Abascal, F ; Amin, SB ; Bader, GD ; Barenboim, J ; Beroukhim, R ; Bertl, J ; Boroevich, KA ; Brunak, S ; Campbell, PJ ; Carlevaro-Fita, J ; Chakravarty, D ; Chan, CWY ; Chen, K ; Choi, JK ; Deu-Pons, J ; Dhingra, P ; Diamanti, K ; Feuerbach, L ; Fink, JL ; Fonseca, NA ; Frigola, J ; Gambacorti-Passerini, C ; Garsed, DW ; Gerstein, M ; Getz, G ; Gonzalez-Perez, A ; Guo, Q ; Gut, IG ; Haan, D ; Hamilton, MP ; Haradhvala, NJ ; Harmanci, AO ; Helmy, M ; Herrmann, C ; Hess, JM ; Hobolth, A ; Hodzic, E ; Hong, C ; Hornshoj, H ; Isaev, K ; Izarzugaza, JMG ; Johnson, TA ; Juul, M ; Juul, RI ; Kahles, A ; Kahraman, A ; Kellis, M ; Khurana, E ; Kim, J ; Kim, JK ; Kim, Y ; Komorowski, J ; Korbel, JO ; Kumar, S ; Lanzos, A ; Larsson, E ; Lawrence, MS ; Lee, D ; Lehmann, K-V ; Li, S ; Li, X ; Lin, Z ; Liu, EM ; Lochovsky, L ; Lou, S ; Madsen, T ; Marchal, K ; Martincorena, I ; Martinez-Fundichely, A ; Maruvka, YE ; McGillivray, PD ; Meyerson, W ; Muinos, F ; Mularoni, L ; Nakagawa, H ; Nielsen, MM ; Paczkowska, M ; Park, K ; Park, K ; Pedersen, JS ; Pich, O ; Pons, T ; Pulido-Tamayo, S ; Raphael, BJ ; Reimand, J ; Reyes-Salazar, I ; Reyna, MA ; Rheinbay, E ; Rubin, MA ; Rubio-Perez, C ; Sabarinathan, R ; Sahinalp, SC ; Saksena, G ; Salichos, L ; Sander, C ; Schumacher, SE ; Shackleton, M ; Shapira, O ; Shen, C ; Shrestha, R ; Shuai, S ; Sidiropoulos, N ; Sieverling, L ; Sinnott-Armstrong, N ; Stein, LD ; Stuart, JM ; Tamborero, D ; Tiao, G ; Tsunoda, T ; Umer, HM ; Uuskula-Reimand, L ; Valencia, A ; Vazquez, M ; Verbeke, LPC ; Wadelius, C ; Wadi, L ; Wang, J ; Warrell, J ; Waszak, SM ; Weischenfeldt, J ; Wheeler, DA ; Wu, G ; Yu, J ; Zhang, J ; Zhang, X ; Zhang, Y ; Zhao, Z ; Zou, L ; von Mering, C (NATURE PUBLISHING GROUP, 2020-02-05)
    Long non-coding RNAs (lncRNAs) are a growing focus of cancer genomics studies, creating the need for a resource of lncRNAs with validated cancer roles. Furthermore, it remains debated whether mutated lncRNAs can drive tumorigenesis, and whether such functions could be conserved during evolution. Here, as part of the ICGC/TCGA Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium, we introduce the Cancer LncRNA Census (CLC), a compilation of 122 GENCODE lncRNAs with causal roles in cancer phenotypes. In contrast to existing databases, CLC requires strong functional or genetic evidence. CLC genes are enriched amongst driver genes predicted from somatic mutations, and display characteristic genomic features. Strikingly, CLC genes are enriched for driver mutations from unbiased, genome-wide transposon-mutagenesis screens in mice. We identified 10 tumour-causing mutations in orthologues of 8 lncRNAs, including LINC-PINT and NEAT1, but not MALAT1. Thus CLC represents a dataset of high-confidence cancer lncRNAs. Mutagenesis maps are a novel means for identifying deeply-conserved roles of lncRNAs in tumorigenesis.
  • Item
    Thumbnail Image
    Integrative pathway enrichment analysis of multivariate omics data
    Paczkowska, M ; Barenboim, J ; Sintupisut, N ; Fox, NS ; Zhu, H ; Abd-Rabbo, D ; Mee, MW ; Boutros, PC ; Reimand, J (NATURE PUBLISHING GROUP, 2020-02-05)
    Multi-omics datasets represent distinct aspects of the central dogma of molecular biology. Such high-dimensional molecular profiles pose challenges to data interpretation and hypothesis generation. ActivePathways is an integrative method that discovers significantly enriched pathways across multiple datasets using statistical data fusion, rationalizes contributing evidence and highlights associated genes. As part of the ICGC/TCGA Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium, which aggregated whole genome sequencing data from 2658 cancers across 38 tumor types, we integrated genes with coding and non-coding mutations and revealed frequently mutated pathways and additional cancer genes with infrequent mutations. We also analyzed prognostic molecular pathways by integrating genomic and transcriptomic features of 1780 breast cancers and highlighted associations with immune response and anti-apoptotic signaling. Integration of ChIP-seq and RNA-seq data for master regulators of the Hippo pathway across normal human tissues identified processes of tissue regeneration and stem cell regulation. ActivePathways is a versatile method that improves systems-level understanding of cellular organization in health and disease through integration of multiple molecular datasets and pathway annotations.
  • Item
    Thumbnail Image
    Reconstructing evolutionary trajectories of mutation signature activities in cancer using TrackSig
    Rubanova, Y ; Shi, R ; Harrigan, CF ; Li, R ; Wintersinger, J ; Sahin, N ; Deshwar, A ; PCAWG Evolution and Heterogeneity Working Group, ; Morris, Q ; PCAWG Consortium, (Nature Research (part of Springer Nature), 2020-02-05)
    The type and genomic context of cancer mutations depend on their causes. These causes have been characterized using signatures that represent mutation types that co-occur in the same tumours. However, it remains unclear how mutation processes change during cancer evolution due to the lack of reliable methods to reconstruct evolutionary trajectories of mutational signature activity. Here, as part of the ICGC/TCGA Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium, which aggregated whole-genome sequencing data from 2658 cancers across 38 tumour types, we present TrackSig, a new method that reconstructs these trajectories using optimal, joint segmentation and deconvolution of mutation type and allele frequencies from a single tumour sample. In simulations, we find TrackSig has a 3-5% activity reconstruction error, and 12% false detection rate. It outperforms an aggressive baseline in situations with branching evolution, CNA gain, and neutral mutations. Applied to data from 2658 tumours and 38 cancer types, TrackSig permits pan-cancer insight into evolutionary changes in mutational processes.
  • Item
    Thumbnail Image
    Combined burden and functional impact tests for cancer driver discovery using DriverPower
    Shuai, S ; Gallinger, S ; Stein, L (NATURE PORTFOLIO, 2020-02-05)
    The discovery of driver mutations is one of the key motivations for cancer genome sequencing. Here, as part of the ICGC/TCGA Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium, which aggregated whole genome sequencing data from 2658 cancers across 38 tumour types, we describe DriverPower, a software package that uses mutational burden and functional impact evidence to identify driver mutations in coding and non-coding sites within cancer whole genomes. Using a total of 1373 genomic features derived from public sources, DriverPower's background mutation model explains up to 93% of the regional variance in the mutation rate across multiple tumour types. By incorporating functional impact scores, we are able to further increase the accuracy of driver discovery. Testing across a collection of 2583 cancer genomes from the PCAWG project, DriverPower identifies 217 coding and 95 non-coding driver candidates. Comparing to six published methods used by the PCAWG Drivers and Functional Interpretation Working Group, DriverPower has the highest F1 score for both coding and non-coding driver discovery. This demonstrates that DriverPower is an effective framework for computational driver discovery.