Sir Peter MacCallum Department of Oncology - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 10 of 19
  • Item
    Thumbnail Image
    The genomic and immune landscape of long-term survivors of high-grade serous ovarian cancer
    Garsed, DW ; Pandey, A ; Fereday, S ; Kennedy, CJ ; Takahashi, K ; Alsop, K ; Hamilton, PT ; Hendley, J ; Chiew, Y-E ; Traficante, N ; Provan, P ; Ariyaratne, D ; Au-Yeung, G ; Bateman, NW ; Bowes, L ; Brand, A ; Christie, EL ; Cunningham, JM ; Friedlander, M ; Grout, B ; Harnett, P ; Hung, J ; McCauley, B ; McNally, O ; Piskorz, AM ; Saner, FAM ; Vierkant, RA ; Wang, C ; Winham, SJ ; Pharoah, PDP ; Brenton, JD ; Conrads, TP ; Maxwell, GL ; Ramus, SJ ; Pearce, CL ; Pike, MC ; Nelson, BH ; Goode, EL ; DeFazio, A ; Bowtell, DDL (NATURE PORTFOLIO, 2022-12)
    Fewer than half of all patients with advanced-stage high-grade serous ovarian cancers (HGSCs) survive more than five years after diagnosis, but those who have an exceptionally long survival could provide insights into tumor biology and therapeutic approaches. We analyzed 60 patients with advanced-stage HGSC who survived more than 10 years after diagnosis using whole-genome sequencing, transcriptome and methylome profiling of their primary tumor samples, comparing this data to 66 short- or moderate-term survivors. Tumors of long-term survivors were more likely to have multiple alterations in genes associated with DNA repair and more frequent somatic variants resulting in an increased predicted neoantigen load. Patients clustered into survival groups based on genomic and immune cell signatures, including three subsets of patients with BRCA1 alterations with distinctly different outcomes. Specific combinations of germline and somatic gene alterations, tumor cell phenotypes and differential immune responses appear to contribute to long-term survival in HGSC.
  • Item
    Thumbnail Image
    Copy Number Variants Are Ovarian Cancer Risk Alleles at Known and Novel Risk Loci
    DeVries, AA ; Dennis, J ; Tyrer, JP ; Peng, P-C ; Coetzee, SG ; Reyes, AL ; Plummer, JT ; Davis, BD ; Chen, SS ; Dezem, FS ; Aben, KKH ; Anton-Culver, H ; Antonenkova, NN ; Beckmann, MW ; Beeghly-Fadiel, A ; Berchuck, A ; Bogdanova, N ; Bogdanova-Markov, N ; Brenton, JD ; Butzow, R ; Campbell, I ; Chang-Claude, J ; Chenevix-Trench, G ; Cook, LS ; DeFazio, A ; Doherty, JA ; Dork, T ; Eccles, DM ; Eliassen, AH ; Fasching, PA ; Fortner, RT ; Giles, GG ; Goode, EL ; Goodman, MT ; Gronwald, J ; Hakansson, N ; Hildebrandt, MAT ; Huff, C ; Huntsman, DG ; Jensen, A ; Kar, S ; Karlan, BY ; Khusnutdinova, EK ; Kiemeney, LA ; Kjaer, SK ; Kupryjanczyk, J ; Labrie, M ; Lambrechts, D ; Le, ND ; Lubinski, J ; May, T ; Menon, U ; Milne, RL ; Modugno, F ; Monteiro, AN ; Moysich, KB ; Odunsi, K ; Olsson, H ; Pearce, CL ; Pejovic, T ; Ramus, SJ ; Riboli, E ; Riggan, MJ ; Romieu, I ; Sandler, DP ; Schildkraut, JM ; Setiawan, VW ; Sieh, W ; Song, H ; Sutphen, R ; Terry, KL ; Thompson, PJ ; Titus, L ; Tworoger, SS ; Van Nieuwenhuysen, E ; Edwards, DV ; Webb, PM ; Wentzensen, N ; Whittemore, AS ; Wolk, A ; Wu, AH ; Ziogas, A ; Freedman, ML ; Lawrenson, K ; Pharoah, PDP ; Easton, DF ; Gayther, SA ; Jones, MR (OXFORD UNIV PRESS INC, 2022-11)
    BACKGROUND: Known risk alleles for epithelial ovarian cancer (EOC) account for approximately 40% of the heritability for EOC. Copy number variants (CNVs) have not been investigated as EOC risk alleles in a large population cohort. METHODS: Single nucleotide polymorphism array data from 13 071 EOC cases and 17 306 controls of White European ancestry were used to identify CNVs associated with EOC risk using a rare admixture maximum likelihood test for gene burden and a by-probe ratio test. We performed enrichment analysis of CNVs at known EOC risk loci and functional biofeatures in ovarian cancer-related cell types. RESULTS: We identified statistically significant risk associations with CNVs at known EOC risk genes; BRCA1 (PEOC = 1.60E-21; OREOC = 8.24), RAD51C (Phigh-grade serous ovarian cancer [HGSOC] = 5.5E-4; odds ratio [OR]HGSOC = 5.74 del), and BRCA2 (PHGSOC = 7.0E-4; ORHGSOC = 3.31 deletion). Four suggestive associations (P < .001) were identified for rare CNVs. Risk-associated CNVs were enriched (P < .05) at known EOC risk loci identified by genome-wide association study. Noncoding CNVs were enriched in active promoters and insulators in EOC-related cell types. CONCLUSIONS: CNVs in BRCA1 have been previously reported in smaller studies, but their observed frequency in this large population-based cohort, along with the CNVs observed at BRCA2 and RAD51C gene loci in EOC cases, suggests that these CNVs are potentially pathogenic and may contribute to the spectrum of disease-causing mutations in these genes. CNVs are likely to occur in a wider set of susceptibility regions, with potential implications for clinical genetic testing and disease prevention.
  • Item
    No Preview Available
    High Prediagnosis Inflammation-Related Risk Score Associated with Decreased Ovarian Cancer Survival
    Brieger, KK ; Phung, MT ; Mukherjee, B ; Bakulski, KM ; Anton-Culver, H ; Bandera, E ; Bowtell, DDL ; Cramer, DW ; DeFazio, A ; Doherty, JA ; Fereday, S ; Fortner, RT ; Gentry-Maharaj, A ; Goode, EL ; Goodman, MT ; Harris, HR ; Matsuo, K ; Menon, U ; Modugno, F ; Moysich, KB ; Qin, B ; Ramus, SJ ; Risch, HA ; Rossing, MA ; Schildkraut, JM ; Trabert, B ; Vierkant, RA ; Winham, SJ ; Wentzensen, N ; Wu, AH ; Ziogas, A ; Khoja, L ; Cho, KR ; McLean, K ; Richardson, J ; Grout, B ; Chase, A ; Deurloo, CM ; Odunsi, K ; Nelson, BH ; Brenton, JD ; Terry, KL ; Pharoah, PDP ; Berchuck, A ; Hanley, GE ; Webb, PM ; Pike, MC ; Pearce, CL (AMER ASSOC CANCER RESEARCH, 2022-02)
    BACKGROUND: There is suggestive evidence that inflammation is related to ovarian cancer survival. However, more research is needed to identify inflammation-related factors that are associated with ovarian cancer survival and to determine their combined effects. METHODS: This analysis used pooled data on 8,147 women with invasive epithelial ovarian cancer from the Ovarian Cancer Association Consortium. The prediagnosis inflammation-related exposures of interest included alcohol use; aspirin use; other nonsteroidal anti-inflammatory drug use; body mass index; environmental tobacco smoke exposure; history of pelvic inflammatory disease, polycystic ovarian syndrome, and endometriosis; menopausal hormone therapy use; physical inactivity; smoking status; and talc use. Using Cox proportional hazards models, the relationship between each exposure and survival was assessed in 50% of the data. A weighted inflammation-related risk score (IRRS) was developed, and its association with survival was assessed using Cox proportional hazards models in the remaining 50% of the data. RESULTS: There was a statistically significant trend of increasing risk of death per quartile of the IRRS [HR = 1.09; 95% confidence interval (CI), 1.03-1.14]. Women in the upper quartile of the IRRS had a 31% higher death rate compared with the lowest quartile (95% CI, 1.11-1.54). CONCLUSIONS: A higher prediagnosis IRRS was associated with an increased mortality risk after an ovarian cancer diagnosis. Further investigation is warranted to evaluate whether postdiagnosis exposures are also associated with survival. IMPACT: Given that pre- and postdiagnosis exposures are often correlated and many are modifiable, our study results can ultimately motivate the development of behavioral recommendations to enhance survival among patients with ovarian cancer.
  • Item
    No Preview Available
    Evolution of core archetypal phenotypes in progressive high grade serous ovarian cancer
    Nath, A ; Cosgrove, PA ; Mirsafian, H ; Christie, EL ; Pflieger, L ; Copeland, B ; Majumdar, S ; Cristea, MC ; Han, ES ; Lee, SJ ; Wang, EW ; Fereday, S ; Traficante, N ; Salgia, R ; Werner, T ; Cohen, AL ; Moos, P ; Chang, JT ; Bowtell, DDL ; Bild, AH (NATURE PORTFOLIO, 2021-05-24)
    The evolution of resistance in high-grade serous ovarian cancer (HGSOC) cells following chemotherapy is only partially understood. To understand the selection of factors driving heterogeneity before and through adaptation to treatment, we profile single-cell RNA-sequencing (scRNA-seq) transcriptomes of HGSOC tumors collected longitudinally during therapy. We analyze scRNA-seq data from two independent patient cohorts to reveal that HGSOC is driven by three archetypal phenotypes, defined as oncogenic states that describe the majority of the transcriptome variation. Using a multi-task learning approach to identify the biological tasks of each archetype, we identify metabolism and proliferation, cellular defense response, and DNA repair signaling as consistent cell states found across patients. Our analysis demonstrates a shift in favor of the metabolism and proliferation archetype versus cellular defense response archetype in cancer cells that received multiple lines of treatment. While archetypes are not consistently associated with specific whole-genome driver mutations, they are closely associated with subclonal populations at the single-cell level, indicating that subclones within a tumor often specialize in unique biological tasks. Our study reveals the core archetypes found in progressive HGSOC and shows consistent enrichment of subclones with the metabolism and proliferation archetype as resistance is acquired to multiple lines of therapy.
  • Item
    Thumbnail Image
    TRACEBACK: Testing of Historical Tubo-Ovarian Cancer Patients for Hereditary Risk Genes as a Cancer Prevention Strategy in Family Members
    Delahunty, R ; Nguyen, L ; Craig, S ; Creighton, B ; Ariyaratne, D ; Garsed, DW ; Christie, E ; Fereday, S ; Andrews, L ; Lewis, A ; Limb, S ; Pandey, A ; Hendley, J ; Traficante, N ; Carvajal, N ; Spurdle, AB ; Thompson, B ; Parsons, MT ; Beshay, V ; Volcheck, M ; Semple, T ; Lupat, R ; Doig, K ; Yu, J ; Chen, XQ ; Marsh, A ; Love, C ; Bilic, S ; Beilin, M ; Nichols, CB ; Greer, C ; Lee, YC ; Gerty, S ; Gill, L ; Newton, E ; Howard, J ; Williams, R ; Norris, C ; Stephens, AN ; Tutty, E ; Smyth, C ; O'Connell, S ; Jobling, T ; Stewart, CJR ; Tan, A ; Fox, SB ; Pachter, N ; Li, J ; Ellul, J ; Mir Arnau, G ; Young, M-A ; Gordon, L ; Forrest, L ; Harris, M ; Livingstone, K ; Hill, J ; Chenevix-Trench, G ; Cohen, PA ; Webb, PM ; Friedlander, M ; James, P ; Bowtell, D ; Alsop, K (LIPPINCOTT WILLIAMS & WILKINS, 2022-06-20)
    PURPOSE: Tubo-ovarian cancer (TOC) is a sentinel cancer for BRCA1 and BRCA2 pathogenic variants (PVs). Identification of a PV in the first member of a family at increased genetic risk (the proband) provides opportunities for cancer prevention in other at-risk family members. Although Australian testing rates are now high, PVs in patients with TOC whose diagnosis predated revised testing guidelines might have been missed. We assessed the feasibility of detecting PVs in this population to enable genetic risk reduction in relatives. PATIENTS AND METHODS: In this pilot study, deceased probands were ascertained from research cohort studies, identification by a relative, and gynecologic oncology clinics. DNA was extracted from archival tissue or stored blood for panel sequencing of 10 risk-associated genes. Testing of deceased probands ascertained through clinic records was performed with a consent waiver. RESULTS: We identified 85 PVs in 84 of 787 (11%) probands. Familial contacts of 39 of 60 (65%) deceased probands with an identified recipient (60 of 84; 71%) have received a written notification of results, with follow-up verbal contact made in 85% (33 of 39). A minority of families (n = 4) were already aware of the PV. For many (29 of 33; 88%), the genetic result provided new information and referral to a genetic service was accepted in most cases (66%; 19 of 29). Those who declined referral (4 of 29) were all male next of kin whose family member had died more than 10 years before. CONCLUSION: We overcame ethical and logistic challenges to demonstrate that retrospective genetic testing to identify PVs in previously untested deceased probands with TOC is feasible. Understanding reasons for a family member's decision to accept or decline a referral will be important for guiding future TRACEBACK projects.
  • Item
    Thumbnail Image
    Evolution of core archetypal phenotypes in progressive high grade serous ovarian cancer.
    Nath, A ; Cosgrove, P ; Copeland, B ; Mirsafian, H ; Christie, E ; Pflieger, L ; Majumdar, S ; Cristea, M ; Han, E ; Lee, S ; Wang, E ; Fereday, S ; Traficante, N ; Salgia, R ; Werner, T ; Cohen, A ; Moos, P ; Chang, J ; Bowtell, D ; Bild, A (AMER ASSOC CANCER RESEARCH, 2021-07-01)
    Abstract The evolution of resistance in high-grade serous ovarian cancer (HGSOC) cells following chemotherapy is only partially understood. To uncover phenotypic changes associated with chemotherapy resistance, we profiled single-cell RNA-sequencing (scRNA-seq) transcriptomes of HGSOC tumors collected longitudinally during patient treatment. Analysis of scRNA-seq data from two independent patient cohorts revealed that HGSOC is driven by three core archetypal phenotypes, defined as oncogenic tasks that describe the majority of the transcriptome variation. A multi-task learning approach to identify the biological tasks of each archetype identified metabolism and proliferation, cellular defense response, and DNA repair signaling. The metabolism and proliferation archetype evolved during treatment and was enriched in cancer cells from patients that received multiple-lines of treatment and had elevated tumor burden indicated by CA-125 levels. The emergence of archetypes was not consistently associated with specific whole-genome driver mutations. However, archetypes were closely associated with subclonal populations at the single-cell level, indicating that subclones within a tumor often specialize in unique biological tasks. Our study reveals the core archetypes found in progressive HGSOC and shows consistent enrichment of subclones with the metabolism archetype as resistance is acquired to multiple lines of therapy. Citation Format: Aritro Nath, Patrick Cosgrove, Benjamin Copeland, Hoda Mirsafian, Elizabeth Christie, Lance Pflieger, Sumana Majumdar, Mihaela Cristea, Ernest Han, Stephen Lee, Edward Wang, Sian Fereday, Nadia Traficante, Ravi Salgia, Theresa Werner, Adam Cohen, Phillip Moos, Jeffrey Chang, David Bowtell, Andrea Bild. Evolution of core archetypal phenotypes in progressive high grade serous ovarian cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 3141.
  • Item
    Thumbnail Image
    Therapeutic options for mucinous ovarian carcinoma
    Gorringe, KL ; Cheasley, D ; Wakefield, MJ ; Ryland, GL ; Allan, PE ; Alsop, K ; Amarasinghe, KC ; Ananda, S ; Bowtell, DDL ; Christie, M ; Chiew, Y-E ; Churchman, M ; DeFazio, A ; Fereday, S ; Gilks, CB ; Gourley, C ; Hadley, AM ; Hendley, J ; Hunter, SM ; Kaufmann, SH ; Kennedy, CJ ; Kobel, M ; Le Page, C ; Li, J ; Lupat, R ; McNally, OM ; McAlpine, JN ; Pyman, J ; Rowley, SM ; Salazar, C ; Saunders, H ; Semple, T ; Stephens, AN ; Thio, N ; Torres, MC ; Traficante, N ; Zethoven, M ; Antill, YC ; Campbell, IG ; Scott, CL (ACADEMIC PRESS INC ELSEVIER SCIENCE, 2020-03)
    OBJECTIVE: Mucinous ovarian carcinoma (MOC) is an uncommon ovarian cancer histotype that responds poorly to conventional chemotherapy regimens. Although long overall survival outcomes can occur with early detection and optimal surgical resection, recurrent and advanced disease are associated with extremely poor survival. There are no current guidelines specifically for the systemic management of recurrent MOC. We analyzed data from a large cohort of women with MOC to evaluate the potential for clinical utility from a range of systemic agents. METHODS: We analyzed gene copy number (n = 191) and DNA sequencing data (n = 184) from primary MOC to evaluate signatures of mismatch repair deficiency and homologous recombination deficiency, and other genetic events. Immunohistochemistry data were collated for ER, CK7, CK20, CDX2, HER2, PAX8 and p16 (n = 117-166). RESULTS: Molecular aberrations noted in MOC that suggest a match with current targeted therapies include amplification of ERBB2 (26.7%) and BRAF mutation (9%). Observed genetic events that suggest potential efficacy for agents currently in clinical trials include: KRAS/NRAS mutations (66%), TP53 missense mutation (49%), RNF43 mutation (11%), ARID1A mutation (10%), and PIK3CA/PTEN mutation (9%). Therapies exploiting homologous recombination deficiency (HRD) may not be effective in MOC, as only 1/191 had a high HRD score. Mismatch repair deficiency was similarly rare (1/184). CONCLUSIONS: Although genetically diverse, MOC has several potential therapeutic targets. Importantly, the lack of response to platinum-based therapy observed clinically corresponds to the lack of a genomic signature associated with HRD, and MOC are thus also unlikely to respond to PARP inhibition.
  • Item
    Thumbnail Image
    Prognostic gene expression signature for high-grade serous ovarian cancer
    Millstein, J ; Budden, T ; Goode, EL ; Anglesio, MS ; Talhouk, A ; Intermaggio, MP ; Leong, HS ; Chen, S ; Elatre, W ; Gilks, B ; Nazeran, T ; Volchek, M ; Bentley, RC ; Wang, C ; Chiu, DS ; Kommoss, S ; Leung, SCY ; Senz, J ; Lum, A ; Chow, V ; Sudderuddin, H ; Mackenzie, R ; George, J ; Fereday, S ; Hendley, J ; Traficante, N ; Steed, H ; Koziak, JM ; Kobel, M ; McNeish, IA ; Goranova, T ; Ennis, D ; Macintyre, G ; De Silva, DS ; Ramon y Cajal, T ; Garcia-Donas, J ; Hernando Polo, S ; Rodriguez, GC ; Cushing-Haugen, KL ; Harris, HR ; Greene, CS ; Zelaya, RA ; Behrens, S ; Fortner, RT ; Sinn, P ; Herpel, E ; Lester, J ; Lubinski, J ; Oszurek, O ; Toloczko, A ; Cybulski, C ; Menkiszak, J ; Pearce, CL ; Pike, MC ; Tseng, C ; Alsop, J ; Rhenius, V ; Song, H ; Jimenez-Linan, M ; Piskorz, AM ; Gentry-Maharaj, A ; Karpinskyj, C ; Widschwendter, M ; Singh, N ; Kennedy, CJ ; Sharma, R ; Harnett, PR ; Gao, B ; Johnatty, SE ; Sayer, R ; Boros, J ; Winham, SJ ; Keeney, GL ; Kaufmann, SH ; Larson, MC ; Luk, H ; Hernandez, BY ; Thompson, PJ ; Wilkens, LR ; Carney, ME ; Trabert, B ; Lissowska, J ; Brinton, L ; Sherman, ME ; Bodelon, C ; Hinsley, S ; Lewsley, LA ; Glasspool, R ; Banerjee, SN ; Stronach, EA ; Haluska, P ; Ray-Coquard, I ; Mahner, S ; Winterhoff, B ; Slamon, D ; Levine, DA ; Kelemen, LE ; Benitez, J ; Chang-Claude, J ; Gronwald, J ; Wu, AH ; Menon, U ; Goodman, MT ; Schildkraut, JM ; Wentzensen, N ; Brown, R ; Berchuck, A ; Chenevix-Trench, G ; DeFazio, A ; Gayther, SA ; Garcia, MJ ; Henderson, MJ ; Rossing, MA ; Beeghly-Fadiel, A ; Fasching, PA ; Orsulic, S ; Karlan, BY ; Konecny, GE ; Huntsman, DG ; Bowtell, DD ; Brenton, JD ; Doherty, JA ; Pharoah, PDP ; Ramus, SJ (ELSEVIER, 2020-09)
    BACKGROUND: Median overall survival (OS) for women with high-grade serous ovarian cancer (HGSOC) is ∼4 years, yet survival varies widely between patients. There are no well-established, gene expression signatures associated with prognosis. The aim of this study was to develop a robust prognostic signature for OS in patients with HGSOC. PATIENTS AND METHODS: Expression of 513 genes, selected from a meta-analysis of 1455 tumours and other candidates, was measured using NanoString technology from formalin-fixed paraffin-embedded tumour tissue collected from 3769 women with HGSOC from multiple studies. Elastic net regularization for survival analysis was applied to develop a prognostic model for 5-year OS, trained on 2702 tumours from 15 studies and evaluated on an independent set of 1067 tumours from six studies. RESULTS: Expression levels of 276 genes were associated with OS (false discovery rate < 0.05) in covariate-adjusted single-gene analyses. The top five genes were TAP1, ZFHX4, CXCL9, FBN1 and PTGER3 (P < 0.001). The best performing prognostic signature included 101 genes enriched in pathways with treatment implications. Each gain of one standard deviation in the gene expression score conferred a greater than twofold increase in risk of death [hazard ratio (HR) 2.35, 95% confidence interval (CI) 2.02-2.71; P < 0.001]. Median survival [HR (95% CI)] by gene expression score quintile was 9.5 (8.3 to -), 5.4 (4.6-7.0), 3.8 (3.3-4.6), 3.2 (2.9-3.7) and 2.3 (2.1-2.6) years. CONCLUSION: The OTTA-SPOT (Ovarian Tumor Tissue Analysis consortium - Stratified Prognosis of Ovarian Tumours) gene expression signature may improve risk stratification in clinical trials by identifying patients who are least likely to achieve 5-year survival. The identified novel genes associated with the outcome may also yield opportunities for the development of targeted therapeutic approaches.
  • Item
    Thumbnail Image
    Clinical and pathological associations of PTEN expression in ovarian cancer: a multicentre study from the Ovarian Tumour Tissue Analysis Consortium
    Martins, FC ; Couturier, D-L ; Paterson, A ; Karnezis, AN ; Christine, C ; Nazeran, TM ; Odunsi, A ; Gentry-Maharaj, A ; Vrvilo, A ; Hein, A ; Talhouk, A ; Osorio, A ; Hartkopf, AD ; Brooks-Wilson, A ; DeFazio, A ; Fischer, A ; Hartmann, A ; Hernandez, BY ; McCauley, BM ; Karpinskyj, C ; de Sousa, CB ; Hogdall, C ; Tiezzi, DG ; Herpel, E ; Taran, FA ; Modugno, F ; Keeney, G ; Nelson, G ; Steed, H ; Song, H ; Luk, H ; Benitez, J ; Alsop, J ; Koziak, JM ; Lester, J ; Rothstein, JH ; de Andrade, JM ; Lundvall, L ; Paz-Ares, L ; Robles-Diaz, L ; Wilkens, LR ; Garcia, MJ ; Intermaggio, MP ; Alcaraz, M-L ; Brett, MA ; Beckmann, MW ; Jimenez-Linan, M ; Anglesio, M ; Carney, ME ; Schneider, M ; Traficante, N ; Pejovic, N ; Singh, N ; Le, N ; Sinn, P ; Ghatage, P ; Erber, R ; Edwards, R ; Vierkant, R ; Ness, RB ; Leung, S ; Orsulic, S ; Brucker, SY ; Kaufmann, SH ; Fereday, S ; Gayther, S ; Winham, SJ ; Kommoss, S ; Pejovic, T ; Longacre, TA ; McGuire, V ; Rhenius, V ; Sieh, W ; Shvetsov, YB ; Whittemore, AS ; Staebler, A ; Karlan, BY ; Rodriguez-Antona, C ; Bowtell, DD ; Goode, EL ; Hogdall, E ; Candido dos Reis, FJ ; Gronwald, J ; Chang-Claude, J ; Moysich, KB ; Kelemen, LE ; Cook, LS ; Goodman, MT ; Fasching, PA ; Crawford, R ; Deen, S ; Menon, U ; Huntsman, DG ; Kobel, M ; Ramus, SJ ; Pharoah, PDP ; Brenton, JD (SPRINGERNATURE, 2020-09-01)
    BACKGROUND: PTEN loss is a putative driver in histotypes of ovarian cancer (high-grade serous (HGSOC), endometrioid (ENOC), clear cell (CCOC), mucinous (MOC), low-grade serous (LGSOC)). We aimed to characterise PTEN expression as a biomarker in epithelial ovarian cancer in a large population-based study. METHODS: Tumours from 5400 patients from a multicentre observational, prospective cohort study of the Ovarian Tumour Tissue Analysis Consortium were used to evaluate associations between immunohistochemical PTEN patterns and overall survival time, age, stage, grade, residual tumour, CD8+ tumour-infiltrating lymphocytes (TIL) counts, expression of oestrogen receptor (ER), progesterone receptor (PR) and androgen receptor (AR) by means of Cox proportional hazard models and generalised Cochran-Mantel-Haenszel tests. RESULTS: Downregulation of cytoplasmic PTEN expression was most frequent in ENOC (most frequently in younger patients; p value = 0.0001) and CCOC and was associated with longer overall survival in HGSOC (hazard ratio: 0.78, 95% CI: 0.65-0.94, p value = 0.022). PTEN expression was associated with ER, PR and AR expression (p values: 0.0008, 0.062 and 0.0002, respectively) in HGSOC and with lower CD8 counts in CCOC (p value < 0.0001). Heterogeneous expression of PTEN was more prevalent in advanced HGSOC (p value = 0.019) and associated with higher CD8 counts (p value = 0.0016). CONCLUSIONS: PTEN loss is a frequent driver in ovarian carcinoma associating distinctly with expression of hormonal receptors and CD8+ TIL counts in HGSOC and CCOC histotypes.
  • 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.