50 results on '"Garsed DW"'
Search Results
2. Proteogenomic analysis of enriched HGSOC tumor epithelium identifies prognostic signatures and therapeutic vulnerabilities
- Author
-
Bateman, NW, Abulez, T, Soltis, AR, Mcpherson, A, Choi, S, Garsed, DW, Pandey, A, Tian, C, Hood, BL, Conrads, KA, Teng, P-N, Oliver, J, Gist, G, Mitchell, D, Litzi, TJ, Tarney, CM, Crothers, BA, Mhawech-Fauceglia, P, Dalgard, CL, Wilkerson, MD, Pierobon, M, Petricoin, EF, Yan, C, Meerzaman, D, Bodelon, C, Wentzensen, N, Lee, JSH, Huntsman, DG, Shah, S, Shriver, CD, Phippen, NT, Darcy, KM, Bowtell, DDL, Conrads, TP, Maxwell, GL, Bateman, NW, Abulez, T, Soltis, AR, Mcpherson, A, Choi, S, Garsed, DW, Pandey, A, Tian, C, Hood, BL, Conrads, KA, Teng, P-N, Oliver, J, Gist, G, Mitchell, D, Litzi, TJ, Tarney, CM, Crothers, BA, Mhawech-Fauceglia, P, Dalgard, CL, Wilkerson, MD, Pierobon, M, Petricoin, EF, Yan, C, Meerzaman, D, Bodelon, C, Wentzensen, N, Lee, JSH, Huntsman, DG, Shah, S, Shriver, CD, Phippen, NT, Darcy, KM, Bowtell, DDL, Conrads, TP, and Maxwell, GL
- Abstract
We performed a deep proteogenomic analysis of bulk tumor and laser microdissection enriched tumor cell populations from high-grade serous ovarian cancer (HGSOC) tissue specimens spanning a broad spectrum of purity. We identified patients with longer progression-free survival had increased immune-related signatures and validated proteins correlating with tumor-infiltrating lymphocytes in 65 tumors from an independent cohort of HGSOC patients, as well as with overall survival in an additional 126 HGSOC patient cohort. We identified that homologous recombination deficient (HRD) tumors are enriched in pathways associated with metabolism and oxidative phosphorylation that we validated in independent patient cohorts. We further identified that polycomb complex protein BMI-1 is elevated in HR proficient (HRP) tumors, that elevated BMI-1 correlates with poor overall survival in HRP but not HRD HGSOC patients, and that HRP HGSOC cells are uniquely sensitive to BMI-1 inhibition.
- Published
- 2024
3. The Prognostic Effect of Immune Cell Infiltration Depends on Molecular Subtype in Endometrioid Ovarian Carcinomas
- Author
-
Heinze, K, Cairns, ES, Thornton, S, Harris, B, Milne, K, Grube, M, Meyer, C, Karnezis, AN, Fereday, S, Garsed, DW, Leung, SCY, Chiu, DS, Moubarak, M, Harter, P, Heitz, F, Mcalpine, JN, Defazio, A, Bowtell, DDL, Goode, EL, Pike, M, Ramus, SJ, Pearce, CL, Staebler, A, Koebel, M, Kommoss, S, Talhouk, A, Nelson, BH, Anglesio, MS, Heinze, K, Cairns, ES, Thornton, S, Harris, B, Milne, K, Grube, M, Meyer, C, Karnezis, AN, Fereday, S, Garsed, DW, Leung, SCY, Chiu, DS, Moubarak, M, Harter, P, Heitz, F, Mcalpine, JN, Defazio, A, Bowtell, DDL, Goode, EL, Pike, M, Ramus, SJ, Pearce, CL, Staebler, A, Koebel, M, Kommoss, S, Talhouk, A, Nelson, BH, and Anglesio, MS
- Abstract
PURPOSE: Endometrioid ovarian carcinoma (ENOC) is the second most-common type of ovarian carcinoma, comprising 10%-20% of cases. Recently, the study of ENOC has benefitted from comparisons to endometrial carcinomas including defining ENOC with four prognostic molecular subtypes. Each subtype suggests differential mechanisms of progression, although tumor-initiating events remain elusive. There is evidence that the ovarian microenvironment may be critical to early lesion establishment and progression. However, while immune infiltrates have been well studied in high-grade serous ovarian carcinoma, studies in ENOC are limited. EXPERIMENTAL DESIGN: We report on 210 ENOC, with clinical follow-up and molecular subtype annotation. Using multiplex IHC and immunofluorescence, we examine the prevalence of T-cell lineage, B-cell lineage, macrophages, and populations with programmed cell death protein 1 or programmed death-ligand 1 across subtypes of ENOC. RESULTS: Immune cell infiltrates in tumor epithelium and stroma showed higher densities in ENOC subtypes with known high mutation burden (POLEmut and MMRd). While molecular subtypes were prognostically significant, immune infiltrates were not (overall survival P > 0.2). Analysis by molecular subtype revealed that immune cell density was prognostically significant in only the no specific molecular profile (NSMP) subtype, where immune infiltrates lacking B cells (TILB minus) had inferior outcome (disease-specific survival: HR, 4.0; 95% confidence interval, 1.1-14.7; P < 0.05). Similar to endometrial carcinomas, molecular subtype stratification was generally superior to immune response in predicting outcomes. CONCLUSIONS: Subtype stratification is critical for better understanding of ENOC, in particular the distribution and prognostic significance of immune cell infiltrates. The role of B cells in the immune response within NSMP tumors warrants further study.
- Published
- 2023
4. Multiomic analysis of homologous recombination-deficient end-stage high-grade serous ovarian cancer
- Author
-
Burdett, NL, Willis, MO, Alsop, K, Hunt, AL, Pandey, A, Hamilton, PT, Abulez, T, Liu, X, Hoang, T, Craig, S, Fereday, S, Hendley, J, Garsed, DW, Milne, K, Kalaria, S, Marshall, A, Hood, BL, Wilson, KN, Conrads, KA, Pishas, K, Ananda, S, Scott, CL, Antill, Y, McNally, O, Mileshkin, L, Hamilton, A, Au-Yeung, G, Devereux, L, Thorne, H, Bild, A, Bateman, NW, Maxwell, GL, Chang, JT, Conrads, TPP, Nelson, BH, Bowtell, DDL, Christie, ELL, Burdett, NL, Willis, MO, Alsop, K, Hunt, AL, Pandey, A, Hamilton, PT, Abulez, T, Liu, X, Hoang, T, Craig, S, Fereday, S, Hendley, J, Garsed, DW, Milne, K, Kalaria, S, Marshall, A, Hood, BL, Wilson, KN, Conrads, KA, Pishas, K, Ananda, S, Scott, CL, Antill, Y, McNally, O, Mileshkin, L, Hamilton, A, Au-Yeung, G, Devereux, L, Thorne, H, Bild, A, Bateman, NW, Maxwell, GL, Chang, JT, Conrads, TPP, Nelson, BH, Bowtell, DDL, and Christie, ELL
- Abstract
High-grade serous ovarian cancer (HGSC) is frequently characterized by homologous recombination (HR) DNA repair deficiency and, while most such tumors are sensitive to initial treatment, acquired resistance is common. We undertook a multiomics approach to interrogate molecular diversity in end-stage disease, using multiple autopsy samples collected from 15 women with HR-deficient HGSC. Patients had polyclonal disease, and several resistance mechanisms were identified within most patients, including reversion mutations and HR restoration by other means. We also observed frequent whole-genome duplication and global changes in immune composition with evidence of immune escape. This analysis highlights diverse evolutionary changes within HGSC that evade therapy and ultimately overwhelm individual patients.
- Published
- 2023
5. Methylation Signature Implicated in Immuno-Suppressive Activities in Tubo-Ovarian High-Grade Serous Carcinoma
- Author
-
Wang, C, Block, MS, Cunningham, JM, Sherman, ME, McCauley, BM, Armasu, SM, Vierkant, RA, Traficante, N, Talhouk, A, Doherty, JA, Pejovic, N, Kobel, M, Jorgensen, BD, Garsed, DW, Fereday, S, Ramus, SJ, Ariyaratne, D, Anglesio, MS, Widschwendter, M, Pejovic, T, Bosquet, JG, Bowtell, DD, Winham, SJ, Goode, EL, Wang, C, Block, MS, Cunningham, JM, Sherman, ME, McCauley, BM, Armasu, SM, Vierkant, RA, Traficante, N, Talhouk, A, Doherty, JA, Pejovic, N, Kobel, M, Jorgensen, BD, Garsed, DW, Fereday, S, Ramus, SJ, Ariyaratne, D, Anglesio, MS, Widschwendter, M, Pejovic, T, Bosquet, JG, Bowtell, DD, Winham, SJ, and Goode, EL
- Abstract
BACKGROUND: Better understanding of prognostic factors in tubo-ovarian high-grade serous carcinoma (HGSC) is critical, as diagnosis confers an aggressive disease course. Variation in tumor DNA methylation shows promise predicting outcome, yet prior studies were largely platform-specific and unable to evaluate multiple molecular features. METHODS: We analyzed genome-wide DNA methylation in 1,040 frozen HGSC, including 325 previously reported upon, seeking a multi-platform quantitative methylation signature that we evaluated in relation to clinical features, tumor characteristics, time to recurrence/death, extent of CD8+ tumor-infiltrating lymphocytes (TIL), gene expression molecular subtypes, and gene expression of the ATP-binding cassette transporter TAP1. RESULTS: Methylation signature was associated with shorter time to recurrence, independent of clinical factors (N = 715 new set, hazard ratio (HR), 1.65; 95% confidence interval (CI), 1.10-2.46; P = 0.015; N = 325 published set HR, 2.87; 95% CI, 2.17-3.81; P = 2.2 × 10-13) and remained prognostic after adjustment for gene expression molecular subtype and TAP1 expression (N = 599; HR, 2.22; 95% CI, 1.66-2.95; P = 4.1 × 10-8). Methylation signature was inversely related to CD8+ TIL levels (P = 2.4 × 10-7) and TAP1 expression (P = 0.0011) and was associated with gene expression molecular subtype (P = 5.9 × 10-4) in covariate-adjusted analysis. CONCLUSIONS: Multi-center analysis identified a novel quantitative tumor methylation signature of HGSC applicable to numerous commercially available platforms indicative of shorter time to recurrence/death, adjusting for other factors. Along with immune cell composition analysis, these results suggest a role for DNA methylation in the immunosuppressive microenvironment. IMPACT: This work aids in identification of targetable epigenome processes and stratification of patients for whom tailored treatment may be most beneficial.
- Published
- 2023
6. Concurrent RB1 loss and BRCA-deficiency predicts enhanced immunological response and long-term survival in tubo-ovarian high-grade serous carcinoma.
- Author
-
Saner, FAM, Takahashi, K, Budden, T, Pandey, A, Ariyaratne, D, Zwimpfer, TA, Meagher, NS, Fereday, S, Twomey, L, Pishas, KI, Hoang, T, Bolithon, A, Traficante, N, Alsop, K, Christie, EL, Kang, E-Y, Nelson, GS, Ghatage, P, Lee, C-H, Riggan, MJ, Alsop, J, Beckmann, MW, Boros, J, Brand, AH, Brooks-Wilson, A, Carney, ME, Coulson, P, Courtney-Brooks, M, Cushing-Haugen, KL, Cybulski, C, El-Bahrawy, MA, Elishaev, E, Erber, R, Gayther, SA, Gentry-Maharaj, A, Blake Gilks, C, Harnett, PR, Harris, HR, Hartmann, A, Hein, A, Hendley, J, AOCS Group, Hernandez, BY, Jakubowska, A, Jimenez-Linan, M, Jones, ME, Kaufmann, SH, Kennedy, CJ, Kluz, T, Koziak, JM, Kristjansdottir, B, Le, ND, Lener, M, Lester, J, Lubiński, J, Mateoiu, C, Orsulic, S, Ruebner, M, Schoemaker, MJ, Shah, M, Sharma, R, Sherman, ME, Shvetsov, YB, Singh, N, Rinda Soong, T, Steed, H, Sukumvanich, P, Talhouk, A, Taylor, SE, Vierkant, RA, Wang, C, Widschwendter, M, Wilkens, LR, Winham, SJ, Anglesio, MS, Berchuck, A, Brenton, JD, Campbell, I, Cook, LS, Doherty, JA, Fasching, PA, Fortner, RT, Goodman, MT, Gronwald, J, Huntsman, DG, Karlan, BY, Kelemen, LE, Menon, U, Modugno, F, Pharoah, PDP, Schildkraut, JM, Sundfeldt, K, Swerdlow, AJ, Goode, EL, DeFazio, A, Köbel, M, Ramus, SJ, Bowtell, DDL, Garsed, DW, Saner, FAM, Takahashi, K, Budden, T, Pandey, A, Ariyaratne, D, Zwimpfer, TA, Meagher, NS, Fereday, S, Twomey, L, Pishas, KI, Hoang, T, Bolithon, A, Traficante, N, Alsop, K, Christie, EL, Kang, E-Y, Nelson, GS, Ghatage, P, Lee, C-H, Riggan, MJ, Alsop, J, Beckmann, MW, Boros, J, Brand, AH, Brooks-Wilson, A, Carney, ME, Coulson, P, Courtney-Brooks, M, Cushing-Haugen, KL, Cybulski, C, El-Bahrawy, MA, Elishaev, E, Erber, R, Gayther, SA, Gentry-Maharaj, A, Blake Gilks, C, Harnett, PR, Harris, HR, Hartmann, A, Hein, A, Hendley, J, AOCS Group, Hernandez, BY, Jakubowska, A, Jimenez-Linan, M, Jones, ME, Kaufmann, SH, Kennedy, CJ, Kluz, T, Koziak, JM, Kristjansdottir, B, Le, ND, Lener, M, Lester, J, Lubiński, J, Mateoiu, C, Orsulic, S, Ruebner, M, Schoemaker, MJ, Shah, M, Sharma, R, Sherman, ME, Shvetsov, YB, Singh, N, Rinda Soong, T, Steed, H, Sukumvanich, P, Talhouk, A, Taylor, SE, Vierkant, RA, Wang, C, Widschwendter, M, Wilkens, LR, Winham, SJ, Anglesio, MS, Berchuck, A, Brenton, JD, Campbell, I, Cook, LS, Doherty, JA, Fasching, PA, Fortner, RT, Goodman, MT, Gronwald, J, Huntsman, DG, Karlan, BY, Kelemen, LE, Menon, U, Modugno, F, Pharoah, PDP, Schildkraut, JM, Sundfeldt, K, Swerdlow, AJ, Goode, EL, DeFazio, A, Köbel, M, Ramus, SJ, Bowtell, DDL, and Garsed, DW
- Abstract
BACKGROUND: Somatic loss of the tumour suppressor RB1 is a common event in tubo-ovarian high-grade serous carcinoma (HGSC), which frequently co-occurs with alterations in homologous recombination DNA repair genes including BRCA1 and BRCA2 (BRCA). We examined whether tumour expression of RB1 was associated with survival across ovarian cancer histotypes (HGSC, endometrioid (ENOC), clear cell (CCOC), mucinous (MOC), low-grade serous carcinoma (LGSC)), and how co-occurrence of germline BRCA pathogenic variants and RB1 loss influences long-term survival in a large series of HGSC. PATIENTS AND METHODS: RB1 protein expression patterns were classified by immunohistochemistry in epithelial ovarian carcinomas of 7436 patients from 20 studies participating in the Ovarian Tumor Tissue Analysis consortium and assessed for associations with overall survival (OS), accounting for patient age at diagnosis and FIGO stage. We examined RB1 expression and germline BRCA status in a subset of 1134 HGSC, and related genotype to survival, tumour infiltrating CD8+ lymphocyte counts and transcriptomic subtypes. Using CRISPR-Cas9, we deleted RB1 in HGSC cell lines with and without BRCA1 mutations to model co-loss with treatment response. We also performed genomic analyses on 126 primary HGSC to explore the molecular characteristics of concurrent homologous recombination deficiency and RB1 loss. RESULTS: RB1 protein loss was most frequent in HGSC (16.4%) and was highly correlated with RB1 mRNA expression. RB1 loss was associated with longer OS in HGSC (hazard ratio [HR] 0.74, 95% confidence interval [CI] 0.66-0.83, P = 6.8 ×10-7), but with poorer prognosis in ENOC (HR 2.17, 95% CI 1.17-4.03, P = 0.0140). Germline BRCA mutations and RB1 loss co-occurred in HGSC (P < 0.0001). Patients with both RB1 loss and germline BRCA mutations had a superior OS (HR 0.38, 95% CI 0.25-0.58, P = 5.2 ×10-6) compared to patients with either alteration alone, and their median OS was three times longer than non-carr
- Published
- 2023
7. Profiling the immune landscape in mucinous ovarian carcinoma
- Author
-
Meagher, NS, Hamilton, P, Milne, K, Thornton, S, Harris, B, Weir, A, Alsop, J, Bisinoto, C, Brenton, JD, Brooks-Wilsoni, A, Chiu, DS, Cushing-Haugen, KL, Fereday, S, Garsed, DW, Gayther, SA, Gentry-Maharaj, A, Gilks, B, Jimenez-Linan, M, Kennedy, CJ, Le, ND, Piskorz, AM, Riggan, MJ, Shah, M, Singh, N, Talhoukj, A, Widschwendter, M, Bowtell, DDL, dos Reis, FJC, Cook, LS, Fortner, RT, Garcia, MJ, Harris, HR, Huntsman, DG, Karnezis, AN, Kobel, M, Menon, U, Pharoah, PDP, Doherty, JA, Anglesioj, MS, Pike, MC, Pearce, CL, Friedlander, ML, DeFazio, A, Nelson, BH, Ramus, SJ, Meagher, NS, Hamilton, P, Milne, K, Thornton, S, Harris, B, Weir, A, Alsop, J, Bisinoto, C, Brenton, JD, Brooks-Wilsoni, A, Chiu, DS, Cushing-Haugen, KL, Fereday, S, Garsed, DW, Gayther, SA, Gentry-Maharaj, A, Gilks, B, Jimenez-Linan, M, Kennedy, CJ, Le, ND, Piskorz, AM, Riggan, MJ, Shah, M, Singh, N, Talhoukj, A, Widschwendter, M, Bowtell, DDL, dos Reis, FJC, Cook, LS, Fortner, RT, Garcia, MJ, Harris, HR, Huntsman, DG, Karnezis, AN, Kobel, M, Menon, U, Pharoah, PDP, Doherty, JA, Anglesioj, MS, Pike, MC, Pearce, CL, Friedlander, ML, DeFazio, A, Nelson, BH, and Ramus, SJ
- Abstract
OBJECTIVE: Mucinous ovarian carcinoma (MOC) is a rare histotype of ovarian cancer, with low response rates to standard chemotherapy, and very poor survival for patients diagnosed at advanced stage. There is a limited understanding of the MOC immune landscape, and consequently whether immune checkpoint inhibitors could be considered for a subset of patients. METHODS: We performed multicolor immunohistochemistry (IHC) and immunofluorescence (IF) on tissue microarrays in a cohort of 126 MOC patients. Cell densities were calculated in the epithelial and stromal components for tumor-associated macrophages (CD68+/PD-L1+, CD68+/PD-L1-), T cells (CD3+/CD8-, CD3+/CD8+), putative T-regulatory cells (Tregs, FOXP3+), B cells (CD20+/CD79A+), plasma cells (CD20-/CD79a+), and PD-L1+ and PD-1+ cells, and compared these values with clinical factors. Univariate and multivariable Cox Proportional Hazards assessed overall survival. Unsupervised k-means clustering identified patient subsets with common patterns of immune cell infiltration. RESULTS: Mean densities of PD1+ cells, PD-L1- macrophages, CD4+ and CD8+ T cells, and FOXP3+ Tregs were higher in the stroma compared to the epithelium. Tumors from advanced (Stage III/IV) MOC had greater epithelial infiltration of PD-L1- macrophages, and fewer PD-L1+ macrophages compared with Stage I/II cancers (p = 0.004 and p = 0.014 respectively). Patients with high epithelial density of FOXP3+ cells, CD8+/FOXP3+ cells, or PD-L1- macrophages, had poorer survival, and high epithelial CD79a + plasma cells conferred better survival, all upon univariate analysis only. Clustering showed that most MOC (86%) had an immune depleted (cold) phenotype, with only a small proportion (11/76,14%) considered immune inflamed (hot) based on T cell and PD-L1 infiltrates. CONCLUSION: In summary, MOCs are mostly immunogenically 'cold', suggesting they may have limited response to current immunotherapies.
- Published
- 2023
8. TRACEBACK: Testing of Historical Tubo-Ovarian Cancer Patients for Hereditary Risk Genes as a Cancer Prevention Strategy in Family Members
- Author
-
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, 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, and Alsop, K
- Abstract
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.
- Published
- 2022
9. Beating the odds: molecular characteristics of long-term survivors of ovarian cancer
- Author
-
Garsed, DW, Bowtell, DDL, Garsed, DW, and Bowtell, DDL
- Abstract
High-grade serous ovarian cancer, the most common form of the disease, is often fatal. This study investigated the genomic and immune characteristics of tumors from women who survived more than 10 years after their initial diagnosis, and compared them with short-term and moderate-term survivors.
- Published
- 2022
10. The genomic and immune landscape of long-term survivors of high-grade serous ovarian cancer
- Author
-
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, 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, and Bowtell, DDL
- Abstract
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.
- Published
- 2022
11. ESMO recommendations on predictive biomarker testing for homologous recombination deficiency and PARP inhibitor benefit in ovarian cancer
- Author
-
Miller, RE, Leary, A, Scott, CL, Serra, V, Lord, CJ, Bowtell, D, Chang, DK, Garsed, DW, Jonkers, J, Ledermann, JA, Nik-Zainal, S, Ray-Coquard, I, Shah, SP, Matias-Guiu, X, Swisher, EM, Yates, LR, Nik-Zainal Abidin, Serena [0000-0001-5054-1727], and Apollo - University of Cambridge Repository
- Subjects
Ovarian Neoplasms ,homologous recombination deficiency (HRD) ,genomic scar assays ,poly-ADP ribose inhibitors (PARPi) ,BRCA ,Humans ,Female ,Carcinoma, Ovarian Epithelial ,Poly(ADP-ribose) Polymerase Inhibitors ,Homologous Recombination ,Biomarkers - Abstract
BACKGROUND: Homologous recombination repair deficiency (HRD) is a frequent feature of high-grade serous ovarian, fallopian tube and peritoneal carcinoma (HGSC) and is associated with sensitivity to PARP inhibitor (PARPi) therapy. HRD testing provides an opportunity to optimise PARPi use in HGSC but methodologies are diverse and clinical application remains controversial. MATERIALS AND METHODS: To define best practice for HRD testing in HGSC the ESMO Translational Research and Precision Medicine Working Group launched a collaborative project that incorporated a systematic review approach. The main aims were to (i) define the term 'HRD test'; (ii) provide an overview of the biological rationale and the level of evidence supporting currently available HRD tests; (iii) provide recommendations on the clinical utility of HRD tests in clinical management of HGSC. RESULTS: A broad range of repair genes, genomic scars, mutational signatures and functional assays are associated with a history of HRD. Currently, the clinical validity of HRD tests in ovarian cancer is best assessed, not in terms of biological HRD status per se, but in terms of PARPi benefit. Clinical trials evidence supports the use of BRCA mutation testing and two commercially available assays that also incorporate genomic instability for identifying subgroups of HGSCs that derive different magnitudes of benefit from PARPi therapy, albeit with some variation by clinical scenario. These tests can be used to inform treatment selection and scheduling but their use is limited by a failure to consistently identify a subgroup of patients who derive no benefit from PARPis in most studies. Existing tests lack negative predictive value and inadequately address the complex and dynamic nature of the HRD phenotype. CONCLUSIONS: Currently available HRD tests are useful for predicting likely magnitude of benefit from PARPis but better biomarkers are urgently needed to better identify current homologous recombination proficiency status and stratify HGSC management.
- Published
- 2020
- Full Text
- View/download PDF
12. Comprehensive analysis of chromothripsis in 2,658 human cancers using whole-genome sequencing
- Author
-
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, 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, and Behren, A
- Abstract
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.
- Published
- 2020
13. Analyses of non-coding somatic drivers in 2,658 cancer whole genomes
- Author
-
Rheinbay, E, Nielsen, MM, Abascal, F, Wala, JA, Shapira, O, Tiao, G, Hornshoj, H, Hess, JM, Juul, RI, Lin, Z, Feuerbach, L, Sabarinathan, R, Madsen, T, Kim, J, Mularoni, L, Shuai, S, Lanzos, A, Herrmann, C, Maruvka, YE, Shen, C, Amin, SB, Bandopadhayay, P, Bertl, J, Boroevich, KA, Busanovich, J, Carlevaro-Fita, J, Chakravarty, D, Chan, CWY, Craft, D, Dhingra, P, Diamanti, K, Fonseca, NA, Gonzalez-Perez, A, Guo, Q, Hamilton, MP, Haradhvala, NJ, Hong, C, Isaev, K, Johnson, TA, Juul, M, Kahles, A, Kahraman, A, Kim, Y, Komorowski, J, Kumar, K, Kumar, S, Lee, D, Lehmann, K-V, Li, Y, Liu, EM, Lochovsky, L, Park, K, Pich, O, Roberts, ND, Saksena, G, Schumacher, SE, Sidiropoulos, N, Sieverling, L, Sinnott-Armstrong, N, Stewart, C, Tamborero, D, Tubio, JMC, Umer, HM, Uuskula-Reimand, L, Wadelius, C, Wadi, L, Yao, X, Zhang, C-Z, Zhang, J, Haber, JE, Hobolth, A, Imielinski, M, Kellis, M, Lawrence, MS, von Mering, C, Nakagawa, H, Raphael, BJ, Rubin, MA, Sander, C, Stein, LD, Stuart, JM, Tsunoda, T, Wheeler, DA, Johnson, R, Reimand, J, Gerstein, M, Khurana, E, Campbell, PJ, Lopez-Bigas, N, Weischenfeldt, J, Beroukhim, R, Martincorena, I, Pedersen, JS, Getz, G, Bader, GD, Barenboim, J, Brunak, S, Chen, K, Choi, JK, Deu-Pons, J, Fink, JL, Frigola, J, Gambacorti-Passerini, C, Garsed, DW, Gut, IG, Haan, D, Harmanci, AO, Helmy, M, Hodzic, E, Izarzugaza, JMG, Kim, JK, Korbel, JO, Larsson, E, Li, S, Li, X, Lou, S, Marchal, K, Martinez-Fundichely, A, McGillivray, PD, Meyerson, W, Muinos, F, Paczkowska, M, Pons, T, Pulido-Tamayo, S, Reyes-Salazar, I, Reyna, MA, Rubio-Perez, C, Sahinalp, SC, Salichos, L, Shackleton, M, Shrestha, R, Valencia, A, Vazquez, M, Verbeke, LPC, Wang, J, Warrell, J, Waszak, SM, Wu, G, Yu, J, Zhang, X, Zhang, Y, Zhao, Z, Zou, L, Akdemir, KC, Alvarez, EG, Baez-Ortega, A, Boutros, PC, Bowtell, DDL, Brors, B, Burns, KH, Chan, K, CortesCiriano, I, Dueso-Barroso, A, Dunford, AJ, Edwards, PA, Estivill, X, Etemadmoghadam, D, Frenkel-Morgenstern, M, Gordenin, DA, Hutter, B, Jones, DTW, Ju, YS, Kazanov, MD, Klimczak, LJ, Koh, Y, Lee, EA, Lee, JJ-K, Lynch, AG, Macintyre, G, Markowetz, F, Meyerson, M, Miyano, S, Navarro, FCP, Ossowski, S, Park, PJ, Pearson, J, Puiggros, M, Rippe, K, Roberts, SA, RodriguezMartin, B, Scully, R, Torrents, D, Villasante, I, Waddell, N, Yang, L, Yoon, S-S, Zamora, J, Rheinbay, E, Nielsen, MM, Abascal, F, Wala, JA, Shapira, O, Tiao, G, Hornshoj, H, Hess, JM, Juul, RI, Lin, Z, Feuerbach, L, Sabarinathan, R, Madsen, T, Kim, J, Mularoni, L, Shuai, S, Lanzos, A, Herrmann, C, Maruvka, YE, Shen, C, Amin, SB, Bandopadhayay, P, Bertl, J, Boroevich, KA, Busanovich, J, Carlevaro-Fita, J, Chakravarty, D, Chan, CWY, Craft, D, Dhingra, P, Diamanti, K, Fonseca, NA, Gonzalez-Perez, A, Guo, Q, Hamilton, MP, Haradhvala, NJ, Hong, C, Isaev, K, Johnson, TA, Juul, M, Kahles, A, Kahraman, A, Kim, Y, Komorowski, J, Kumar, K, Kumar, S, Lee, D, Lehmann, K-V, Li, Y, Liu, EM, Lochovsky, L, Park, K, Pich, O, Roberts, ND, Saksena, G, Schumacher, SE, Sidiropoulos, N, Sieverling, L, Sinnott-Armstrong, N, Stewart, C, Tamborero, D, Tubio, JMC, Umer, HM, Uuskula-Reimand, L, Wadelius, C, Wadi, L, Yao, X, Zhang, C-Z, Zhang, J, Haber, JE, Hobolth, A, Imielinski, M, Kellis, M, Lawrence, MS, von Mering, C, Nakagawa, H, Raphael, BJ, Rubin, MA, Sander, C, Stein, LD, Stuart, JM, Tsunoda, T, Wheeler, DA, Johnson, R, Reimand, J, Gerstein, M, Khurana, E, Campbell, PJ, Lopez-Bigas, N, Weischenfeldt, J, Beroukhim, R, Martincorena, I, Pedersen, JS, Getz, G, Bader, GD, Barenboim, J, Brunak, S, Chen, K, Choi, JK, Deu-Pons, J, Fink, JL, Frigola, J, Gambacorti-Passerini, C, Garsed, DW, Gut, IG, Haan, D, Harmanci, AO, Helmy, M, Hodzic, E, Izarzugaza, JMG, Kim, JK, Korbel, JO, Larsson, E, Li, S, Li, X, Lou, S, Marchal, K, Martinez-Fundichely, A, McGillivray, PD, Meyerson, W, Muinos, F, Paczkowska, M, Pons, T, Pulido-Tamayo, S, Reyes-Salazar, I, Reyna, MA, Rubio-Perez, C, Sahinalp, SC, Salichos, L, Shackleton, M, Shrestha, R, Valencia, A, Vazquez, M, Verbeke, LPC, Wang, J, Warrell, J, Waszak, SM, Wu, G, Yu, J, Zhang, X, Zhang, Y, Zhao, Z, Zou, L, Akdemir, KC, Alvarez, EG, Baez-Ortega, A, Boutros, PC, Bowtell, DDL, Brors, B, Burns, KH, Chan, K, CortesCiriano, I, Dueso-Barroso, A, Dunford, AJ, Edwards, PA, Estivill, X, Etemadmoghadam, D, Frenkel-Morgenstern, M, Gordenin, DA, Hutter, B, Jones, DTW, Ju, YS, Kazanov, MD, Klimczak, LJ, Koh, Y, Lee, EA, Lee, JJ-K, Lynch, AG, Macintyre, G, Markowetz, F, Meyerson, M, Miyano, S, Navarro, FCP, Ossowski, S, Park, PJ, Pearson, J, Puiggros, M, Rippe, K, Roberts, SA, RodriguezMartin, B, Scully, R, Torrents, D, Villasante, I, Waddell, N, Yang, L, Yoon, S-S, and Zamora, J
- Abstract
The discovery of drivers of cancer has traditionally focused on protein-coding genes1-4. Here we present analyses of driver point mutations and structural variants in non-coding regions across 2,658 genomes from the Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium5 of the International Cancer Genome Consortium (ICGC) and The Cancer Genome Atlas (TCGA). For point mutations, we developed a statistically rigorous strategy for combining significance levels from multiple methods of driver discovery that overcomes the limitations of individual methods. For structural variants, we present two methods of driver discovery, and identify regions that are significantly affected by recurrent breakpoints and recurrent somatic juxtapositions. Our analyses confirm previously reported drivers6,7, raise doubts about others and identify novel candidates, including point mutations in the 5' region of TP53, in the 3' untranslated regions of NFKBIZ and TOB1, focal deletions in BRD4 and rearrangements in the loci of AKR1C genes. We show that although point mutations and structural variants that drive cancer are less frequent in non-coding genes and regulatory sequences than in protein-coding genes, additional examples of these drivers will be found as more cancer genomes become available.
- Published
- 2020
14. Cancer LncRNA Census reveals evidence for deep functional conservation of long noncoding RNAs in tumorigenesis
- Author
-
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, Chakravarty, D, Chan, CWY, Chen, K, Choi, JK, Deu-Pons, J, Dhingra, P, Diamanti, K, 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, 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, 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, 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, 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, Chakravarty, D, Chan, CWY, Chen, K, Choi, JK, Deu-Pons, J, Dhingra, P, Diamanti, K, 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, 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, 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, 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, and von Mering, C
- Abstract
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.
- Published
- 2020
15. Disruption of chromatin folding domains by somatic genomic rearrangements in human cancer
- Author
-
Akdemir, KC, Le, VT, Chandran, S, Li, Y, Verhaak, RG, Beroukhim, R, Campbell, PJ, Chin, L, Dixon, JR, Futreal, PA, Alvarez, EG, Baez-Ortega, A, Boutros, PC, Bowtell, DDL, Brors, B, Burns, KH, Chan, K, Chen, K, Cortes-Ciriano, I, 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, Klimczak, LJ, Koh, Y, Korbel, JO, Kumar, K, Lee, EA, Lee, JJ-K, Lynch, AG, Macintyre, G, Markowetz, F, Martincorena, I, Martinez-Fundichely, A, Meyerson, M, Miyano, S, Nakagawa, H, Navarro, FCP, Ossowski, S, Park, PJ, Pearson, JV, 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, Yang, L, Yao, X, Yoon, S-S, Zamora, J, Zhang, C-Z, Akdemir, KC, Le, VT, Chandran, S, Li, Y, Verhaak, RG, Beroukhim, R, Campbell, PJ, Chin, L, Dixon, JR, Futreal, PA, Alvarez, EG, Baez-Ortega, A, Boutros, PC, Bowtell, DDL, Brors, B, Burns, KH, Chan, K, Chen, K, Cortes-Ciriano, I, 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, Klimczak, LJ, Koh, Y, Korbel, JO, Kumar, K, Lee, EA, Lee, JJ-K, Lynch, AG, Macintyre, G, Markowetz, F, Martincorena, I, Martinez-Fundichely, A, Meyerson, M, Miyano, S, Nakagawa, H, Navarro, FCP, Ossowski, S, Park, PJ, Pearson, JV, 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, Yang, L, Yao, X, Yoon, S-S, Zamora, J, and Zhang, C-Z
- Abstract
Chromatin is folded into successive layers to organize linear DNA. Genes within the same topologically associating domains (TADs) demonstrate similar expression and histone-modification profiles, and boundaries separating different domains have important roles in reinforcing the stability of these features. Indeed, domain disruptions in human cancers can lead to misregulation of gene expression. However, the frequency of domain disruptions in human cancers remains unclear. 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), which aggregated whole-genome sequencing data from 2,658 cancers across 38 tumor types, we analyzed 288,457 somatic structural variations (SVs) to understand the distributions and effects of SVs across TADs. Notably, SVs can lead to the fusion of discrete TADs, and complex rearrangements markedly change chromatin folding maps in the cancer genomes. Notably, only 14% of the boundary deletions resulted in a change in expression in nearby genes of more than twofold.
- Published
- 2020
16. The evolutionary history of 2,658 cancers
- Author
-
Gerstung, M, Jolly, C, Leshchiner, I, Dentro, SC, Gonzalez, S, Rosebrock, D, Mitchell, TJ, Rubanova, Y, Anur, P, Yu, K, Tarabichi, M, Deshwar, A, Wintersinger, J, Kleinheinz, K, Vazquez-Garcia, I, Haase, K, Jerman, L, Sengupta, S, Macintyre, G, Malikic, S, Donmez, N, Livitz, DG, Cmero, M, Demeulemeester, J, Schumacher, S, Fan, Y, Yao, X, Lee, J, Schlesner, M, Boutros, PC, Bowtell, DD, Zhu, H, Getz, G, Imielinski, M, Beroukhim, R, Sahinalp, SC, Ji, Y, Peifer, M, Markowetz, F, Mustonen, V, Yuan, K, Wang, W, Morris, QD, Spellman, PT, Wedge, DC, Van Loo, P, Deshwar, AG, Adams, DJ, Campbell, PJ, Cao, S, Christie, EL, Cun, Y, Dawson, KJ, Drews, RM, Eils, R, Fittall, M, Garsed, DW, Ha, G, Lee-Six, H, Martincorena, I, Oesper, L, Peto, M, Raphael, BJ, Salcedo, A, Shi, R, Shin, SJ, Spiro, O, Stein, LD, Vembu, S, Wheeler, DA, Yang, T-P, Gerstung, M, Jolly, C, Leshchiner, I, Dentro, SC, Gonzalez, S, Rosebrock, D, Mitchell, TJ, Rubanova, Y, Anur, P, Yu, K, Tarabichi, M, Deshwar, A, Wintersinger, J, Kleinheinz, K, Vazquez-Garcia, I, Haase, K, Jerman, L, Sengupta, S, Macintyre, G, Malikic, S, Donmez, N, Livitz, DG, Cmero, M, Demeulemeester, J, Schumacher, S, Fan, Y, Yao, X, Lee, J, Schlesner, M, Boutros, PC, Bowtell, DD, Zhu, H, Getz, G, Imielinski, M, Beroukhim, R, Sahinalp, SC, Ji, Y, Peifer, M, Markowetz, F, Mustonen, V, Yuan, K, Wang, W, Morris, QD, Spellman, PT, Wedge, DC, Van Loo, P, Deshwar, AG, Adams, DJ, Campbell, PJ, Cao, S, Christie, EL, Cun, Y, Dawson, KJ, Drews, RM, Eils, R, Fittall, M, Garsed, DW, Ha, G, Lee-Six, H, Martincorena, I, Oesper, L, Peto, M, Raphael, BJ, Salcedo, A, Shi, R, Shin, SJ, Spiro, O, Stein, LD, Vembu, S, Wheeler, DA, and Yang, T-P
- Abstract
Cancer develops through a process of somatic evolution1,2. Sequencing data from a single biopsy represent a snapshot of this process that can reveal the timing of specific genomic aberrations and the changing influence of mutational processes3. Here, by whole-genome sequencing analysis of 2,658 cancers 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)4, we reconstruct the life history and evolution of mutational processes and driver mutation sequences of 38 types of cancer. Early oncogenesis is characterized by mutations in a constrained set of driver genes, and specific copy number gains, such as trisomy 7 in glioblastoma and isochromosome 17q in medulloblastoma. The mutational spectrum changes significantly throughout tumour evolution in 40% of samples. A nearly fourfold diversification of driver genes and increased genomic instability are features of later stages. Copy number alterations often occur in mitotic crises, and lead to simultaneous gains of chromosomal segments. Timing analyses suggest that driver mutations often precede diagnosis by many years, if not decades. Together, these results determine the evolutionary trajectories of cancer, and highlight opportunities for early cancer detection.
- Published
- 2020
17. Patterns of somatic structural variation in human cancer genomes
- Author
-
Li, Y, Roberts, ND, Wala, JA, Shapira, O, Schumacher, SE, Kumar, K, Khurana, E, Waszak, S, Korbel, JO, Haber, JE, Imielinski, M, Weischenfeldt, J, Beroukhim, R, Campbell, PJ, Akdemir, KC, Alvarez, EG, Baez-Ortega, A, Boutros, PC, Bowtell, DDL, Brors, B, Burns, KH, Chan, K, Chen, K, Cortes-Ciriano, I, 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, Hess, JM, Hutter, B, Jones, DTW, Ju, YS, Kazanov, MD, Klimczak, LJ, Koh, Y, Lee, EA, Lee, JJ-K, Lynch, AG, Macintyre, G, Markowetz, F, Martincorena, I, Martinez-Fundichely, A, Meyerson, M, Miyano, S, Nakagawa, H, Navarro, FCP, Ossowski, S, Park, PJ, Pearson, J, Puiggros, M, Rippe, K, Roberts, SA, Rodriguez-Martin, B, Scully, R, Shackleton, M, Sidiropoulos, N, Sieverling, L, Stewart, C, Torrents, D, Tubio, JMC, Villasante, I, Waddell, N, Yang, L, Yao, X, Yoon, S-S, Zamora, J, Zhang, C-Z, Li, Y, Roberts, ND, Wala, JA, Shapira, O, Schumacher, SE, Kumar, K, Khurana, E, Waszak, S, Korbel, JO, Haber, JE, Imielinski, M, Weischenfeldt, J, Beroukhim, R, Campbell, PJ, Akdemir, KC, Alvarez, EG, Baez-Ortega, A, Boutros, PC, Bowtell, DDL, Brors, B, Burns, KH, Chan, K, Chen, K, Cortes-Ciriano, I, 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, Hess, JM, Hutter, B, Jones, DTW, Ju, YS, Kazanov, MD, Klimczak, LJ, Koh, Y, Lee, EA, Lee, JJ-K, Lynch, AG, Macintyre, G, Markowetz, F, Martincorena, I, Martinez-Fundichely, A, Meyerson, M, Miyano, S, Nakagawa, H, Navarro, FCP, Ossowski, S, Park, PJ, Pearson, J, Puiggros, M, Rippe, K, Roberts, SA, Rodriguez-Martin, B, Scully, R, Shackleton, M, Sidiropoulos, N, Sieverling, L, Stewart, C, Torrents, D, Tubio, JMC, Villasante, I, Waddell, N, Yang, L, Yao, X, Yoon, S-S, Zamora, J, and Zhang, C-Z
- Abstract
A key mutational process in cancer is structural variation, in which rearrangements delete, amplify or reorder genomic segments that range in size from kilobases to whole chromosomes1-7. Here we develop methods to group, classify and describe somatic structural variants, using data from the Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium of the International Cancer Genome Consortium (ICGC) and The Cancer Genome Atlas (TCGA), which aggregated whole-genome sequencing data from 2,658 cancers across 38 tumour types8. Sixteen signatures of structural variation emerged. Deletions have a multimodal size distribution, assort unevenly across tumour types and patients, are enriched in late-replicating regions and correlate with inversions. Tandem duplications also have a multimodal size distribution, but are enriched in early-replicating regions-as are unbalanced translocations. Replication-based mechanisms of rearrangement generate varied chromosomal structures with low-level copy-number gains and frequent inverted rearrangements. One prominent structure consists of 2-7 templates copied from distinct regions of the genome strung together within one locus. Such cycles of templated insertions correlate with tandem duplications, and-in liver cancer-frequently activate the telomerase gene TERT. A wide variety of rearrangement processes are active in cancer, which generate complex configurations of the genome upon which selection can act.
- Published
- 2020
18. Pan-cancer analysis of whole genomes identifies driver rearrangements promoted by LINE-1 retrotransposition
- Author
-
Rodriguez-Martin, B, Alvarez, EG, Baez-Ortega, A, Zamora, J, Supek, F, Demeulemeester, J, Santamarina, M, Ju, YS, Temes, J, Garcia-Souto, D, Detering, H, Li, Y, Rodriguez-Castro, J, Dueso-Barroso, A, Bruzos, AL, Dentro, SC, Blanco, MG, Contino, G, Ardeljan, D, Tojo, M, Roberts, ND, Zumalave, S, Edwards, PAW, Weischenfeldt, J, Puiggros, M, Chong, Z, Chen, K, Lee, EA, Wala, JA, Raine, K, Butler, A, Waszak, SM, Navarro, FCP, Schumacher, SE, Monlong, J, Maura, F, Bolli, N, Bourque, G, Gerstein, M, Park, PJ, Wedge, DC, Beroukhim, R, Torrents, D, Korbel, JO, Martincorena, I, Fitzgerald, RC, Van Loo, P, Kazazian, HH, Burns, KH, Campbell, PJ, Tubio, JMC, Akdemir, KC, Boutros, PC, Bowtell, DDL, Brors, B, Chan, K, Cortes-Ciriano, I, Dunford, AJ, Edwards, PA, Estivill, X, Etemadmoghadam, D, Feuerbach, L, Fink, JL, Frenkel-Morgenstern, M, Garsed, DW, Gordenin, DA, Haan, D, Haber, JE, Hess, JM, Hutter, B, Imielinski, M, Jones, DTW, Kazanov, MD, Klimczak, LJ, Koh, Y, Kumar, K, Lee, JJ-K, Lynch, AG, Macintyre, G, Markowetz, F, Martinez-Fundichely, A, Meyerson, M, Miyano, S, Nakagawa, H, Ossowski, S, Pearson, J, Rippe, K, Roberts, SA, Scully, R, Shackleton, M, Sidiropoulos, N, Sieverling, L, Stewart, C, Villasante, I, Waddell, N, Yang, L, Yao, X, Yoon, S-S, Zhang, C-Z, Rodriguez-Martin, B, Alvarez, EG, Baez-Ortega, A, Zamora, J, Supek, F, Demeulemeester, J, Santamarina, M, Ju, YS, Temes, J, Garcia-Souto, D, Detering, H, Li, Y, Rodriguez-Castro, J, Dueso-Barroso, A, Bruzos, AL, Dentro, SC, Blanco, MG, Contino, G, Ardeljan, D, Tojo, M, Roberts, ND, Zumalave, S, Edwards, PAW, Weischenfeldt, J, Puiggros, M, Chong, Z, Chen, K, Lee, EA, Wala, JA, Raine, K, Butler, A, Waszak, SM, Navarro, FCP, Schumacher, SE, Monlong, J, Maura, F, Bolli, N, Bourque, G, Gerstein, M, Park, PJ, Wedge, DC, Beroukhim, R, Torrents, D, Korbel, JO, Martincorena, I, Fitzgerald, RC, Van Loo, P, Kazazian, HH, Burns, KH, Campbell, PJ, Tubio, JMC, Akdemir, KC, Boutros, PC, Bowtell, DDL, Brors, B, Chan, K, Cortes-Ciriano, I, Dunford, AJ, Edwards, PA, Estivill, X, Etemadmoghadam, D, Feuerbach, L, Fink, JL, Frenkel-Morgenstern, M, Garsed, DW, Gordenin, DA, Haan, D, Haber, JE, Hess, JM, Hutter, B, Imielinski, M, Jones, DTW, Kazanov, MD, Klimczak, LJ, Koh, Y, Kumar, K, Lee, JJ-K, Lynch, AG, Macintyre, G, Markowetz, F, Martinez-Fundichely, A, Meyerson, M, Miyano, S, Nakagawa, H, Ossowski, S, Pearson, J, Rippe, K, Roberts, SA, Scully, R, Shackleton, M, Sidiropoulos, N, Sieverling, L, Stewart, C, Villasante, I, Waddell, N, Yang, L, Yao, X, Yoon, S-S, and Zhang, C-Z
- Abstract
About half of all cancers have somatic integrations of retrotransposons. Here, to characterize their role in oncogenesis, we analyzed the patterns and mechanisms of somatic retrotransposition in 2,954 cancer genomes from 38 histological cancer subtypes within the framework of the Pan-Cancer Analysis of Whole Genomes (PCAWG) project. We identified 19,166 somatically acquired retrotransposition events, which affected 35% of samples and spanned a range of event types. Long interspersed nuclear element (LINE-1; L1 hereafter) insertions emerged as the first most frequent type of somatic structural variation in esophageal adenocarcinoma, and the second most frequent in head-and-neck and colorectal cancers. Aberrant L1 integrations can delete megabase-scale regions of a chromosome, which sometimes leads to the removal of tumor-suppressor genes, and can induce complex translocations and large-scale duplications. Somatic retrotranspositions can also initiate breakage-fusion-bridge cycles, leading to high-level amplification of oncogenes. These observations illuminate a relevant role of L1 retrotransposition in remodeling the cancer genome, with potential implications for the development of human tumors.
- Published
- 2020
19. Going to extremes: determinants of extraordinary response and survival in patients with cancer
- Author
-
Saner, FAM, Herschtal, A, Nelson, BH, deFazio, A, Goode, EL, Ramus, SJ, Pandey, A, Beach, JA, Fereday, S, Berchuck, A, Lheureux, S, Pearce, CL, Pharoah, PD, Pike, MC, Garsed, DW, Bowtell, DDL, Saner, FAM, Herschtal, A, Nelson, BH, deFazio, A, Goode, EL, Ramus, SJ, Pandey, A, Beach, JA, Fereday, S, Berchuck, A, Lheureux, S, Pearce, CL, Pharoah, PD, Pike, MC, Garsed, DW, and Bowtell, DDL
- Abstract
Research into factors affecting treatment response or survival in patients with cancer frequently involves cohorts that span the most common range of clinical outcomes, as such patients are most readily available for study. However, attention has turned to highly unusual patients who have exceptionally favourable or atypically poor responses to treatment and/or overall survival, with the expectation that patients at the extremes may provide insights that could ultimately improve the outcome of individuals with more typical disease trajectories. While clinicians can often recount surprising patients whose clinical journey was very unusual, given known clinical characteristics and prognostic indicators, there is a lack of consensus among researchers on how best to define exceptional patients, and little has been proposed for the optimal design of studies to identify factors that dictate unusual outcome. In this Opinion article, we review different approaches to identifying exceptional patients with cancer and possible study designs to investigate extraordinary clinical outcomes. We discuss pitfalls with finding these rare patients, including challenges associated with accrual of patients across different treatment centres and time periods. We describe recent molecular and immunological factors that have been identified as contributing to unusual patient outcome and make recommendations for future studies on these intriguing patients.
- Published
- 2019
20. P147 Long-term survival in women with high-grade serous ovarian cancer: interplay between RB1 and BRCA1/2
- Author
-
Saner, FAM, primary, Garsed, DW, additional, Beach, JA, additional, Pandey, A, additional, Fereday, S, additional, Alsop, K, additional, Wouters, MCA, additional, Traficante, N, additional, Pearce, CL, additional, Pike, MC, additional, de Fazio, A, additional, Ramus, SJ, additional, Köbel, M, additional, Goode, EL, additional, Nelson, BH, additional, and Bowtell, DDL, additional
- Published
- 2019
- Full Text
- View/download PDF
21. Genomic Analysis Using Regularized Regression in High-Grade Serous Ovarian Cancer
- Author
-
Natanzon, Y, Earp, M, Cunningham, JM, Kalli, KR, Wang, C, Armasu, SM, Larson, MC, Bowtell, DDL, Garsed, DW, Fridley, BL, Winham, SJ, Goode, EL, Natanzon, Y, Earp, M, Cunningham, JM, Kalli, KR, Wang, C, Armasu, SM, Larson, MC, Bowtell, DDL, Garsed, DW, Fridley, BL, Winham, SJ, and Goode, EL
- Abstract
High-grade serous ovarian cancer (HGSOC) is a complex disease in which initiation and progression have been associated with copy number alterations, epigenetic processes, and, to a lesser extent, germline variation. We hypothesized that, when summarized at the gene level, tumor methylation and germline genetic variation, alone or in combination, influence tumor gene expression in HGSOC. We used Elastic Net (ENET) penalized regression method to evaluate these associations and adjust for somatic copy number in 3 independent data sets comprising tumors from more than 470 patients. Penalized regression models of germline variation, with or without methylation, did not reveal a role in HGSOC gene expression. However, we observed significant association between regional methylation and expression of 5 genes (WDPCP, KRT6C, BRCA2, EFCAB13, and ZNF283). CpGs retained in ENET model for BRCA2 and ZNF283 appeared enriched in several regulatory elements, suggesting that regularized regression may provide a novel utility for integrative genomic analysis.
- Published
- 2018
22. A newly characterized human well-differentiated liposarcoma cell line contains amplifications of the 12q12-21 and 10p11-14 regions
- Author
-
Pedeutour, F, Maire, G, Pierron, A, Thomas, DM, Garsed, DW, Bianchini, L, Duranton-Tanneur, V, Cortes-Maurel, A, Italiano, A, Squire, JA, Coindre, J-M, Pedeutour, F, Maire, G, Pierron, A, Thomas, DM, Garsed, DW, Bianchini, L, Duranton-Tanneur, V, Cortes-Maurel, A, Italiano, A, Squire, JA, and Coindre, J-M
- Abstract
While surgery is the usual treatment for localized well-differentiated and dedifferentiated liposarcomas (WDLPS/DDLPS), the therapeutic options for patients with advanced disease are limited. The classical antimitotic treatments are most often inefficient. The establishment of genetically characterized cell lines is therefore crucial for providing in vitro models for novel targeted therapies. We have used spectral karyotyping, fluorescence in situ hybridization with whole chromosome painting and locus-specific probes, and array-comparative genomic hybridization to identify the chromosomal and molecular alterations of a novel cell line established from a recurring sclerosing WDLPS. The karyotype was hypertriploid and showed multiple structural anomalies. All cells retained the presence of a giant marker chromosome that had been previously identified in the primary cell cultures. This giant chromosome contained high-level amplification of chromosomal regions 12q13-21 and lacked the alpha-satellite centromeric sequences associated with WDLPS/DDLPS. The 12q amplicon was large, containing 370 amplified genes. The DNA copy number ranged from 3 to 57. The highest levels of amplification were observed at 12q14.3 for GNS, WIF1, and HMGA2. We analyzed the mRNA expression status by real-time reverse transcription polymerase chain reaction for six genes from this amplicon: MDM2, HMGA2, CDK4, TSPAN31, WIF1, and YEATS4. mRNA overexpression was correlated with genomic amplification. A second amplicon originating from 10p11-14 was also present in the giant marker chromosome. The 10p amplicon contained 62 genes, including oncogenes such as MLLT10, previously described in chimeric fusion with MLL in leukemias, NEBL, and BMI1.
- Published
- 2012
23. Immunological and molecular features of the tumor microenvironment of long-term survivors of ovarian cancer.
- Author
-
Nelson BH, Hamilton PT, Phung MT, Milne K, Harris B, Thornton S, Stevens DL, Kalaria S, Singh K, Laumont CM, Moss E, Alimujiang A, Meagher NS, Bolithon A, Fereday S, Kennedy CJ, Hendley J, Ariyaratne D, Alsop K, Traficante N, Goode EL, Karnezis AN, Shen H, Richardson J, McKinnon Deurloo C, Chase A, Grout B, Doherty JA, Harris HR, Cushing-Haugen KL, Anglesio MS, Heinze K, Huntsman D, Talhouk A, Hanley GE, Alsop J, Jimenez-Linan M, Pharoah PD, Boros J, Brand AH, Harnett PR, Sharma R, Hecht JL, Sasamoto N, Terry KL, Karlan BY, Lester J, Carney ME, Goodman MT, Hernandez BY, Wilkens LR, Behrens S, Turzanski Fortner R, Fasching PA, Bisinotto C, Candido Dos Reis FJ, Ghatage P, Köbel M, Elishaev E, Modugno F, Cook LS, Le ND, Gentry-Maharaj A, Menon U, García MJ, Rodriguez-Antona C, Farrington KM, Kelemen LE, Kommoss S, Staebler A, Garsed DW, Brenton JD, Piskorz AM, Bowtell DD, DeFazio A, Ramus SJ, Pike MC, and Pearce CL
- Abstract
Background: Despite an overall poor prognosis, about 15% of patients with advanced-stage tubo-ovarian high-grade serous carcinoma (HGSC) survive ten or more years after standard treatment., Methods: We evaluated the tumor microenvironment of this exceptional, understudied group using a large international cohort enriched for long-term survivors (LTS; 10+ years; n = 374) compared to medium-term (MTS; 5-7.99 years; n = 433) and short-term survivors (STS; 2-4.99 years; n = 416). Primary tumor samples were immunostained and scored for intra-epithelial and intra-stromal densities of 10 immune-cell subsets (including T cells, B cells, plasma cells, myeloid cells, PD-1+ cells, and PD-L1+ cells) and epithelial content., Results: Positive associations with LTS compared to STS were seen for 9/10 immune-cell subsets. In particular, the combination of intra-epithelial CD8+ T cells and intra-stromal B cells showed near five-fold increased odds of LTS compared to STS. All of these associations were stronger in tumors with high epithelial content and/or the C4/Differentiated molecular subtype, despite immune-cell densities generally being higher in tumors with low epithelial content and/or the C2/Immunoreactive molecular subtype., Conclusions: The tumor microenvironment of HGSC long-term survivors is distinguished by the intersection of T and B cell co-infiltration, high epithelial content and C4/Differentiated molecular subtype, features which may inspire new approaches to immunotherapy., Funding: Ovarian Cancer Research Program (OCRP) of the Congressionally Directed Medical Research Program (CDMRP), U.S. Department of Defense (DOD); American Cancer Society; BC Cancer Foundation; Canada's Networks of Centres of Excellence; Canadian Cancer Society; Canadian Institutes of Health Research; Cancer Councils of New South Wales, Victoria, Queensland, South Australia and Tasmania, Cancer Foundation of Western Australia; Cancer Institute NSW; Cancer Research UK; Deutsche Forschungsgesellschaft; ELAN Funds of the University of Erlangen-Nuremberg; Fred C. and Katherine B. Andersen Foundation; Genome BC; German Cancer Research Center; German Federal Ministry of Education and Research, Programme of Clinical Biomedical Research; Instituto de Salud Carlos III; Mayo Foundation; Minnesota Ovarian Cancer Alliance; Ministerio de Economía y Competitividad; MRC; National Center for Advancing Translational Sciences; National Health and Medical Research Council of Australia (NHMRC); Ovarian Cancer Australia; Peter MacCallum Foundation; Sydney West Translational Cancer Research Centre; Terry Fox Research Institute; The Eve Appeal (The Oak Foundation); UK National Institute for Health Research Biomedical Research Centres at the University of Cambridge; University of Pittsburgh School of Medicine; U.S. National Cancer Institute of the National Institutes of Health; VGH & UBC Hospital Foundation; Victorian Cancer Agency.
- Published
- 2024
- Full Text
- View/download PDF
24. Predictive value of homologous recombination deficiency status for survival outcomes in primary tubo-ovarian high-grade serous carcinoma.
- Author
-
Zwimpfer TA, Ewald H, Bilir E, Jayawardana M, Appenzeller-Herzog C, Bizzarri N, Razumova Z, Kacperczyk-Bartnik J, Heinzelmann-Schwarz V, Friedlander M, Bowtell DD, and Garsed DW
- Subjects
- Humans, Female, Fallopian Tube Neoplasms genetics, Fallopian Tube Neoplasms mortality, Fallopian Tube Neoplasms pathology, Fallopian Tube Neoplasms drug therapy, Cystadenocarcinoma, Serous genetics, Cystadenocarcinoma, Serous mortality, Prognosis, Progression-Free Survival, Genes, BRCA2, Genes, BRCA1, Mutation, Ovarian Neoplasms genetics, Ovarian Neoplasms mortality
- Abstract
Objectives: This is a protocol for a Cochrane Review (prognosis). The objectives are as follows: To evaluate the predictive value of the prognostic factor HRD status, as determined by various clinically validated HRD assays at the time of staging laparotomy, compared to BRCA1/2 mutation status for progression-free survival and overall survival in patients with tubo-ovarian high-grade serous carcinoma treated in the first-line setting with a combination of surgery and platinum-based chemotherapy and/or maintenance with PARP inhibitors., (Copyright © 2024 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
25. Concurrent RB1 Loss and BRCA Deficiency Predicts Enhanced Immunologic Response and Long-term Survival in Tubo-ovarian High-grade Serous Carcinoma.
- Author
-
Saner FAM, Takahashi K, Budden T, Pandey A, Ariyaratne D, Zwimpfer TA, Meagher NS, Fereday S, Twomey L, Pishas KI, Hoang T, Bolithon A, Traficante N, Alsop K, Christie EL, Kang EY, Nelson GS, Ghatage P, Lee CH, Riggan MJ, Alsop J, Beckmann MW, Boros J, Brand AH, Brooks-Wilson A, Carney ME, Coulson P, Courtney-Brooks M, Cushing-Haugen KL, Cybulski C, El-Bahrawy MA, Elishaev E, Erber R, Gayther SA, Gentry-Maharaj A, Gilks CB, Harnett PR, Harris HR, Hartmann A, Hein A, Hendley J, Hernandez BY, Jakubowska A, Jimenez-Linan M, Jones ME, Kaufmann SH, Kennedy CJ, Kluz T, Koziak JM, Kristjansdottir B, Le ND, Lener M, Lester J, Lubiński J, Mateoiu C, Orsulic S, Ruebner M, Schoemaker MJ, Shah M, Sharma R, Sherman ME, Shvetsov YB, Soong TR, Steed H, Sukumvanich P, Talhouk A, Taylor SE, Vierkant RA, Wang C, Widschwendter M, Wilkens LR, Winham SJ, Anglesio MS, Berchuck A, Brenton JD, Campbell I, Cook LS, Doherty JA, Fasching PA, Fortner RT, Goodman MT, Gronwald J, Huntsman DG, Karlan BY, Kelemen LE, Menon U, Modugno F, Pharoah PDP, Schildkraut JM, Sundfeldt K, Swerdlow AJ, Goode EL, DeFazio A, Köbel M, Ramus SJ, Bowtell DDL, and Garsed DW
- Subjects
- Humans, Female, Prognosis, Ubiquitin-Protein Ligases genetics, Neoplasm Grading, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating metabolism, Middle Aged, Germ-Line Mutation, Gene Expression Regulation, Neoplastic, Aged, Biomarkers, Tumor genetics, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, Ovarian Neoplasms genetics, Ovarian Neoplasms mortality, Ovarian Neoplasms immunology, Ovarian Neoplasms pathology, BRCA2 Protein genetics, BRCA2 Protein deficiency, BRCA1 Protein genetics, BRCA1 Protein deficiency, Cystadenocarcinoma, Serous genetics, Cystadenocarcinoma, Serous pathology, Cystadenocarcinoma, Serous mortality, Cystadenocarcinoma, Serous immunology, Retinoblastoma Binding Proteins genetics
- Abstract
Purpose: The purpose of this study was to evaluate RB1 expression and survival across ovarian carcinoma histotypes and how co-occurrence of BRCA1 or BRCA2 (BRCA) alterations and RB1 loss influences survival in tubo-ovarian high-grade serous carcinoma (HGSC)., Experimental Design: RB1 protein expression was classified by immunohistochemistry in ovarian carcinomas of 7,436 patients from the Ovarian Tumor Tissue Analysis consortium. We examined RB1 expression and germline BRCA status in a subset of 1,134 HGSC, and related genotype to overall survival (OS), tumor-infiltrating CD8+ lymphocytes, and transcriptomic subtypes. Using CRISPR-Cas9, we deleted RB1 in HGSC cells with and without BRCA1 alterations to model co-loss with treatment response. We performed whole-genome and transcriptome data analyses on 126 patients with primary HGSC to characterize tumors with concurrent BRCA deficiency and RB1 loss., Results: RB1 loss was associated with longer OS in HGSC but with poorer prognosis in endometrioid ovarian carcinoma. Patients with HGSC harboring both RB1 loss and pathogenic germline BRCA variants had superior OS compared with patients with either alteration alone, and their median OS was three times longer than those without pathogenic BRCA variants and retained RB1 expression (9.3 vs. 3.1 years). Enhanced sensitivity to cisplatin and paclitaxel was seen in BRCA1-altered cells with RB1 knockout. Combined RB1 loss and BRCA deficiency correlated with transcriptional markers of enhanced IFN response, cell-cycle deregulation, and reduced epithelial-mesenchymal transition. CD8+ lymphocytes were most prevalent in BRCA-deficient HGSC with co-loss of RB1., Conclusions: Co-occurrence of RB1 loss and BRCA deficiency was associated with exceptionally long survival in patients with HGSC, potentially due to better treatment response and immune stimulation., (©2024 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
26. Homologous recombination proficient subtypes of high-grade serous ovarian cancer: treatment options for a poor prognosis group.
- Author
-
Stiegeler N, Garsed DW, Au-Yeung G, Bowtell DDL, Heinzelmann-Schwarz V, and Zwimpfer TA
- Abstract
Approximately 50% of tubo-ovarian high-grade serous carcinomas (HGSCs) have functional homologous recombination-mediated (HR) DNA repair, so-called HR-proficient tumors, which are often associated with primary platinum resistance (relapse within six months after completion of first-line therapy), minimal benefit from poly(ADP-ribose) polymerase (PARP) inhibitors, and shorter survival. HR-proficient tumors comprise multiple molecular subtypes including cases with CCNE1 amplification, AKT2 amplification or CDK12 alteration, and are often characterized as "cold" tumors with fewer infiltrating lymphocytes and decreased expression of PD-1/PD-L1. Several new treatment approaches aim to manipulate these negative prognostic features and render HR-proficient tumors more susceptible to treatment. Alterations in multiple different molecules and pathways in the DNA damage response are driving new drug development to target HR-proficient cancer cells, such as inhibitors of the CDK or P13K/AKT pathways, as well as ATR inhibitors. Treatment combinations with chemotherapy or PARP inhibitors and agents targeting DNA replication stress have shown promising preclinical and clinical results. New approaches in immunotherapy are also being explored, including vaccines or antibody drug conjugates. Many approaches are still in the early stages of development and further clinical trials will determine their clinical relevance. There is a need to include HR-proficient tumors in ovarian cancer trials and to analyze them in a more targeted manner to provide further evidence for their specific therapy, as this will be crucial in improving the overall prognosis of HGSC and ovarian cancer in general., Competing Interests: DB reports research support grants from AstraZeneca, Roche-Genentech and BeiGene paid to institution outside the submitted work; also personal consulting fees from Exo Therapeutics, that are outside the submitted work. GA-Y reports research support grants from AstraZeneca and Roche-Genentech paid to institution outside the submitted work. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 Stiegeler, Garsed, Au-Yeung, Bowtell, Heinzelmann-Schwarz and Zwimpfer.)
- Published
- 2024
- Full Text
- View/download PDF
27. Author Correction: Proteogenomic analysis of enriched HGSOC tumor epithelium identifies prognostic signatures and therapeutic vulnerabilities.
- Author
-
Bateman NW, Abulez T, Soltis AR, McPherson A, Choi S, Garsed DW, Pandey A, Tian C, Hood BL, Conrads KA, Teng PN, Oliver J, Gist G, Mitchell D, Litzi TJ, Tarney CM, Crothers BA, Mhawech-Fauceglia P, Dalgard CL, Wilkerson MD, Pierobon M, Petricoin EF, Yan C, Meerzaman D, Bodelon C, Wentzensen N, Lee JSH, Huntsman DG, Shah S, Shriver CD, Phippen NT, Darcy KM, Bowtell DDL, Conrads TP, and Maxwell GL
- Published
- 2024
- Full Text
- View/download PDF
28. Proteogenomic analysis of enriched HGSOC tumor epithelium identifies prognostic signatures and therapeutic vulnerabilities.
- Author
-
Bateman NW, Abulez T, Soltis AR, McPherson A, Choi S, Garsed DW, Pandey A, Tian C, Hood BL, Conrads KA, Teng PN, Oliver J, Gist G, Mitchell D, Litzi TJ, Tarney CM, Crothers BA, Mhawech-Fauceglia P, Dalgard CL, Wilkerson MD, Pierobon M, Petricoin EF, Yan C, Meerzaman D, Bodelon C, Wentzensen N, Lee JSH, Huntsman DG, Shah S, Shriver CD, Phippen NT, Darcy KM, Bowtell DDL, Conrads TP, and Maxwell GL
- Abstract
We performed a deep proteogenomic analysis of bulk tumor and laser microdissection enriched tumor cell populations from high-grade serous ovarian cancer (HGSOC) tissue specimens spanning a broad spectrum of purity. We identified patients with longer progression-free survival had increased immune-related signatures and validated proteins correlating with tumor-infiltrating lymphocytes in 65 tumors from an independent cohort of HGSOC patients, as well as with overall survival in an additional 126 HGSOC patient cohort. We identified that homologous recombination deficient (HRD) tumors are enriched in pathways associated with metabolism and oxidative phosphorylation that we validated in independent patient cohorts. We further identified that polycomb complex protein BMI-1 is elevated in HR proficient (HRP) tumors, that elevated BMI-1 correlates with poor overall survival in HRP but not HRD HGSOC patients, and that HRP HGSOC cells are uniquely sensitive to BMI-1 inhibition., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
29. The microtubule inhibitor eribulin demonstrates efficacy in platinum-resistant and refractory high-grade serous ovarian cancer patient-derived xenograft models.
- Author
-
Ho GY, Vandenberg CJ, Lim R, Christie EL, Garsed DW, Lieschke E, Nesic K, Kondrashova O, Ratnayake G, Radke M, Penington JS, Carmagnac A, Heong V, Kyran EL, Zhang F, Traficante N, Huang R, Dobrovic A, Swisher EM, McNally O, Kee D, Wakefield MJ, Papenfuss AT, Bowtell DDL, Barker HE, and Scott CL
- Abstract
Background: Despite initial response to platinum-based chemotherapy and PARP inhibitor therapy (PARPi), nearly all recurrent high-grade serous ovarian cancer (HGSC) will acquire lethal drug resistance; indeed, ~15% of individuals have de novo platinum-refractory disease., Objectives: To determine the potential of anti-microtubule agent (AMA) therapy (paclitaxel, vinorelbine and eribulin) in platinum-resistant or refractory (PRR) HGSC by assessing response in patient-derived xenograft (PDX) models of HGSC., Design and Methods: Of 13 PRR HGSC PDX, six were primary PRR, derived from chemotherapy-naïve samples (one was BRCA2 mutant) and seven were from samples obtained following chemotherapy treatment in the clinic (five were mutant for either BRCA1 or BRCA2 ( BRCA1/2) , four with prior PARPi exposure), recapitulating the population of individuals with aggressive treatment-resistant HGSC in the clinic. Molecular analyses and in vivo treatment studies were undertaken., Results: Seven out of thirteen PRR PDX (54%) were sensitive to treatment with the AMA, eribulin (time to progressive disease (PD) ⩾100 days from the start of treatment) and 11 out of 13 PDX (85%) derived significant benefit from eribulin [time to harvest (TTH) for each PDX with p < 0.002]. In 5 out of 10 platinum-refractory HGSC PDX (50%) and one out of three platinum-resistant PDX (33%), eribulin was more efficacious than was cisplatin, with longer time to PD and significantly extended TTH (each PDX p < 0.02). Furthermore, four of these models were extremely sensitive to all three AMA tested, maintaining response until the end of the experiment (120d post-treatment start). Despite harbouring secondary BRCA2 mutations, two BRCA2 -mutant PDX models derived from heavily pre-treated individuals were sensitive to AMA. PRR HGSC PDX models showing greater sensitivity to AMA had high proliferative indices and oncogene expression. Two PDX models, both with prior chemotherapy and/or PARPi exposure, were refractory to all AMA, one of which harboured the SLC25A40-ABCB1 fusion, known to upregulate drug efflux via MDR1., Conclusion: The efficacy observed for eribulin in PRR HGSC PDX was similar to that observed for paclitaxel, which transformed ovarian cancer clinical practice. Eribulin is therefore worthy of further consideration in clinical trials, particularly in ovarian carcinoma with early failure of carboplatin/paclitaxel chemotherapy., Competing Interests: Eisai Inc. provided drug support for this study. DDB declares Consultant for Exo Therapeutics. CLS declares Advisory Boards for AstraZeneca, Clovis Oncology, Roche, Eisai Inc., Sierra Oncology, Takeda, MSD, EpsilaBio and Grant/Research support from Clovis Oncology, Eisai Inc., Sierra Oncology, Roche, Beigene, AstraZeneca and Boehringer Ingelheim. EMS declares Scientific Advisory Board for Ideaya Biosciences and DSMB role for Novartis. Other authors declare no conflicts of interest., (© The Author(s), 2023.)
- Published
- 2023
- Full Text
- View/download PDF
30. Concurrent RB1 loss and BRCA -deficiency predicts enhanced immunological response and long-term survival in tubo-ovarian high-grade serous carcinoma.
- Author
-
Saner FAM, Takahashi K, Budden T, Pandey A, Ariyaratne D, Zwimpfer TA, Meagher NS, Fereday S, Twomey L, Pishas KI, Hoang T, Bolithon A, Traficante N, Alsop K, Christie EL, Kang EY, Nelson GS, Ghatage P, Lee CH, Riggan MJ, Alsop J, Beckmann MW, Boros J, Brand AH, Brooks-Wilson A, Carney ME, Coulson P, Courtney-Brooks M, Cushing-Haugen KL, Cybulski C, El-Bahrawy MA, Elishaev E, Erber R, Gayther SA, Gentry-Maharaj A, Blake Gilks C, Harnett PR, Harris HR, Hartmann A, Hein A, Hendley J, Hernandez BY, Jakubowska A, Jimenez-Linan M, Jones ME, Kaufmann SH, Kennedy CJ, Kluz T, Koziak JM, Kristjansdottir B, Le ND, Lener M, Lester J, Lubiński J, Mateoiu C, Orsulic S, Ruebner M, Schoemaker MJ, Shah M, Sharma R, Sherman ME, Shvetsov YB, Singh N, Rinda Soong T, Steed H, Sukumvanich P, Talhouk A, Taylor SE, Vierkant RA, Wang C, Widschwendter M, Wilkens LR, Winham SJ, Anglesio MS, Berchuck A, Brenton JD, Campbell I, Cook LS, Doherty JA, Fasching PA, Fortner RT, Goodman MT, Gronwald J, Huntsman DG, Karlan BY, Kelemen LE, Menon U, Modugno F, Pharoah PDP, Schildkraut JM, Sundfeldt K, Swerdlow AJ, Goode EL, DeFazio A, Köbel M, Ramus SJ, Bowtell DDL, and Garsed DW
- Abstract
Background: Somatic loss of the tumour suppressor RB1 is a common event in tubo-ovarian high-grade serous carcinoma (HGSC), which frequently co-occurs with alterations in homologous recombination DNA repair genes including BRCA1 and BRCA2 ( BRCA ). We examined whether tumour expression of RB1 was associated with survival across ovarian cancer histotypes (HGSC, endometrioid (ENOC), clear cell (CCOC), mucinous (MOC), low-grade serous carcinoma (LGSC)), and how co-occurrence of germline BRCA pathogenic variants and RB1 loss influences long-term survival in a large series of HGSC., Patients and Methods: RB1 protein expression patterns were classified by immunohistochemistry in epithelial ovarian carcinomas of 7436 patients from 20 studies participating in the Ovarian Tumor Tissue Analysis consortium and assessed for associations with overall survival (OS), accounting for patient age at diagnosis and FIGO stage. We examined RB1 expression and germline BRCA status in a subset of 1134 HGSC, and related genotype to survival, tumour infiltrating CD8+ lymphocyte counts and transcriptomic subtypes. Using CRISPR-Cas9, we deleted RB1 in HGSC cell lines with and without BRCA1 mutations to model co-loss with treatment response. We also performed genomic analyses on 126 primary HGSC to explore the molecular characteristics of concurrent homologous recombination deficiency and RB1 loss., Results: RB1 protein loss was most frequent in HGSC (16.4%) and was highly correlated with RB1 mRNA expression. RB1 loss was associated with longer OS in HGSC (hazard ratio [HR] 0.74, 95% confidence interval [CI] 0.66-0.83, P = 6.8 ×10
-7 ), but with poorer prognosis in ENOC (HR 2.17, 95% CI 1.17-4.03, P = 0.0140). Germline BRCA mutations and RB1 loss co-occurred in HGSC ( P < 0.0001). Patients with both RB1 loss and germline BRCA mutations had a superior OS (HR 0.38, 95% CI 0.25-0.58, P = 5.2 ×10-6 ) compared to patients with either alteration alone, and their median OS was three times longer than non-carriers whose tumours retained RB1 expression (9.3 years vs. 3.1 years). Enhanced sensitivity to cisplatin ( P < 0.01) and paclitaxel ( P < 0.05) was seen in BRCA1 mutated cell lines with RB1 knockout. Among 126 patients with whole-genome and transcriptome sequence data, combined RB1 loss and genomic evidence of homologous recombination deficiency was correlated with transcriptional markers of enhanced interferon response, cell cycle deregulation, and reduced epithelial-mesenchymal transition in primary HGSC. CD8+ lymphocytes were most prevalent in BRCA -deficient HGSC with co-loss of RB1 ., Conclusions: Co-occurrence of RB1 loss and BRCA mutation was associated with exceptionally long survival in patients with HGSC, potentially due to better treatment response and immune stimulation., Competing Interests: COMPETING INTERESTS DDLB is an Exo Therapeutics advisor and has received research grant funding from AstraZeneca, Genentech-Roche and BeiGene for unrelated work. SF, NT, KA, and ADeF received grant funding from AstraZeneca for unrelated work. AGM and UM report funded research collaborations for unrelated work with industry: Intelligent Lab on Fiber, RNA Guardian, Micronoma and Mercy BioAnalytics. UM had stock ownership (2011–2021) awarded by University College London (UCL) in Abcodia, which held the licence for the Risk of Ovarian Cancer Algorithm (ROCA). UM reports research collaboration contracts with Cambridge University and QIMR Berghofer Medical Research Institute. UM holds patent number EP10178345.4 for Breast Cancer Diagnostics. UM is a member of Tina’s Wish Scientific Advisory Board (USA) and Research Advisory Panel, Yorkshire Cancer Research (UK). The remaining authors declared no conflicts of interest.- Published
- 2023
- Full Text
- View/download PDF
31. The Prognostic Effect of Immune Cell Infiltration Depends on Molecular Subtype in Endometrioid Ovarian Carcinomas.
- Author
-
Heinze K, Cairns ES, Thornton S, Harris B, Milne K, Grube M, Meyer C, Karnezis AN, Fereday S, Garsed DW, Leung SCY, Chiu DS, Moubarak M, Harter P, Heitz F, McAlpine JN, DeFazio A, Bowtell DDL, Goode EL, Pike M, Ramus SJ, Pearce CL, Staebler A, Köbel M, Kommoss S, Talhouk A, Nelson BH, and Anglesio MS
- Subjects
- Female, Humans, Prognosis, Biomarkers, Tumor genetics, Carcinoma, Ovarian Epithelial, Tumor Microenvironment, Carcinoma, Endometrioid pathology, Ovarian Neoplasms pathology, Endometrial Neoplasms genetics, Endometrial Neoplasms pathology
- Abstract
Purpose: Endometrioid ovarian carcinoma (ENOC) is the second most-common type of ovarian carcinoma, comprising 10%-20% of cases. Recently, the study of ENOC has benefitted from comparisons to endometrial carcinomas including defining ENOC with four prognostic molecular subtypes. Each subtype suggests differential mechanisms of progression, although tumor-initiating events remain elusive. There is evidence that the ovarian microenvironment may be critical to early lesion establishment and progression. However, while immune infiltrates have been well studied in high-grade serous ovarian carcinoma, studies in ENOC are limited., Experimental Design: We report on 210 ENOC, with clinical follow-up and molecular subtype annotation. Using multiplex IHC and immunofluorescence, we examine the prevalence of T-cell lineage, B-cell lineage, macrophages, and populations with programmed cell death protein 1 or programmed death-ligand 1 across subtypes of ENOC., Results: Immune cell infiltrates in tumor epithelium and stroma showed higher densities in ENOC subtypes with known high mutation burden (POLEmut and MMRd). While molecular subtypes were prognostically significant, immune infiltrates were not (overall survival P > 0.2). Analysis by molecular subtype revealed that immune cell density was prognostically significant in only the no specific molecular profile (NSMP) subtype, where immune infiltrates lacking B cells (TILB minus) had inferior outcome (disease-specific survival: HR, 4.0; 95% confidence interval, 1.1-14.7; P < 0.05). Similar to endometrial carcinomas, molecular subtype stratification was generally superior to immune response in predicting outcomes., Conclusions: Subtype stratification is critical for better understanding of ENOC, in particular the distribution and prognostic significance of immune cell infiltrates. The role of B cells in the immune response within NSMP tumors warrants further study., (©2023 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
32. Methylation Signature Implicated in Immuno-Suppressive Activities in Tubo-Ovarian High-Grade Serous Carcinoma.
- Author
-
Wang C, Block MS, Cunningham JM, Sherman ME, McCauley BM, Armasu SM, Vierkant RA, Traficante N, Talhouk A, Ramus SJ, Pejovic N, Köbel M, Jorgensen BD, Garsed DW, Fereday S, Doherty JA, Ariyaratne D, Anglesio MS, Widschwendter M, Pejovic T, Bosquet JG, Bowtell DD, Winham SJ, and Goode EL
- Subjects
- Female, Humans, Prognosis, Proportional Hazards Models, DNA Methylation, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Tumor Microenvironment genetics, Ovarian Neoplasms pathology, Cystadenocarcinoma, Serous genetics, Cystadenocarcinoma, Serous pathology
- Abstract
Background: Better understanding of prognostic factors in tubo-ovarian high-grade serous carcinoma (HGSC) is critical, as diagnosis confers an aggressive disease course. Variation in tumor DNA methylation shows promise predicting outcome, yet prior studies were largely platform-specific and unable to evaluate multiple molecular features., Methods: We analyzed genome-wide DNA methylation in 1,040 frozen HGSC, including 325 previously reported upon, seeking a multi-platform quantitative methylation signature that we evaluated in relation to clinical features, tumor characteristics, time to recurrence/death, extent of CD8+ tumor-infiltrating lymphocytes (TIL), gene expression molecular subtypes, and gene expression of the ATP-binding cassette transporter TAP1., Results: Methylation signature was associated with shorter time to recurrence, independent of clinical factors (N = 715 new set, hazard ratio (HR), 1.65; 95% confidence interval (CI), 1.10-2.46; P = 0.015; N = 325 published set HR, 2.87; 95% CI, 2.17-3.81; P = 2.2 × 10-13) and remained prognostic after adjustment for gene expression molecular subtype and TAP1 expression (N = 599; HR, 2.22; 95% CI, 1.66-2.95; P = 4.1 × 10-8). Methylation signature was inversely related to CD8+ TIL levels (P = 2.4 × 10-7) and TAP1 expression (P = 0.0011) and was associated with gene expression molecular subtype (P = 5.9 × 10-4) in covariate-adjusted analysis., Conclusions: Multi-center analysis identified a novel quantitative tumor methylation signature of HGSC applicable to numerous commercially available platforms indicative of shorter time to recurrence/death, adjusting for other factors. Along with immune cell composition analysis, these results suggest a role for DNA methylation in the immunosuppressive microenvironment., Impact: This work aids in identification of targetable epigenome processes and stratification of patients for whom tailored treatment may be most beneficial., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
33. Multiomic analysis of homologous recombination-deficient end-stage high-grade serous ovarian cancer.
- Author
-
Burdett NL, Willis MO, Alsop K, Hunt AL, Pandey A, Hamilton PT, Abulez T, Liu X, Hoang T, Craig S, Fereday S, Hendley J, Garsed DW, Milne K, Kalaria S, Marshall A, Hood BL, Wilson KN, Conrads KA, Pishas KI, Ananda S, Scott CL, Antill Y, McNally O, Mileshkin L, Hamilton A, Au-Yeung G, Devereux L, Thorne H, Bild A, Bateman NW, Maxwell GL, Chang JT, Conrads TP, Nelson BH, Bowtell DDL, and Christie EL
- Subjects
- Humans, Female, Multiomics, Carcinoma, Ovarian Epithelial, Homologous Recombination genetics, Ovarian Neoplasms genetics, Cystadenocarcinoma, Serous genetics
- Abstract
High-grade serous ovarian cancer (HGSC) is frequently characterized by homologous recombination (HR) DNA repair deficiency and, while most such tumors are sensitive to initial treatment, acquired resistance is common. We undertook a multiomics approach to interrogate molecular diversity in end-stage disease, using multiple autopsy samples collected from 15 women with HR-deficient HGSC. Patients had polyclonal disease, and several resistance mechanisms were identified within most patients, including reversion mutations and HR restoration by other means. We also observed frequent whole-genome duplication and global changes in immune composition with evidence of immune escape. This analysis highlights diverse evolutionary changes within HGSC that evade therapy and ultimately overwhelm individual patients., (© 2023. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2023
- Full Text
- View/download PDF
34. Profiling the immune landscape in mucinous ovarian carcinoma.
- Author
-
Meagher NS, Hamilton P, Milne K, Thornton S, Harris B, Weir A, Alsop J, Bisinoto C, Brenton JD, Brooks-Wilson A, Chiu DS, Cushing-Haugen KL, Fereday S, Garsed DW, Gayther SA, Gentry-Maharaj A, Gilks B, Jimenez-Linan M, Kennedy CJ, Le ND, Piskorz AM, Riggan MJ, Shah M, Singh N, Talhouk A, Widschwendter M, Bowtell DDL, Candido Dos Reis FJ, Cook LS, Fortner RT, García MJ, Harris HR, Huntsman DG, Karnezis AN, Köbel M, Menon U, Pharoah PDP, Doherty JA, Anglesio MS, Pike MC, Pearce CL, Friedlander ML, DeFazio A, Nelson BH, and Ramus SJ
- Subjects
- Humans, Female, Carcinoma, Ovarian Epithelial pathology, CD8-Positive T-Lymphocytes, Forkhead Transcription Factors therapeutic use, Lymphocytes, Tumor-Infiltrating, Tumor Microenvironment, B7-H1 Antigen genetics, Ovarian Neoplasms drug therapy
- Abstract
Objective: Mucinous ovarian carcinoma (MOC) is a rare histotype of ovarian cancer, with low response rates to standard chemotherapy, and very poor survival for patients diagnosed at advanced stage. There is a limited understanding of the MOC immune landscape, and consequently whether immune checkpoint inhibitors could be considered for a subset of patients., Methods: We performed multicolor immunohistochemistry (IHC) and immunofluorescence (IF) on tissue microarrays in a cohort of 126 MOC patients. Cell densities were calculated in the epithelial and stromal components for tumor-associated macrophages (CD68+/PD-L1+, CD68+/PD-L1-), T cells (CD3+/CD8-, CD3+/CD8+), putative T-regulatory cells (Tregs, FOXP3+), B cells (CD20+/CD79A+), plasma cells (CD20-/CD79a+), and PD-L1+ and PD-1+ cells, and compared these values with clinical factors. Univariate and multivariable Cox Proportional Hazards assessed overall survival. Unsupervised k-means clustering identified patient subsets with common patterns of immune cell infiltration., Results: Mean densities of PD1+ cells, PD-L1- macrophages, CD4+ and CD8+ T cells, and FOXP3+ Tregs were higher in the stroma compared to the epithelium. Tumors from advanced (Stage III/IV) MOC had greater epithelial infiltration of PD-L1- macrophages, and fewer PD-L1+ macrophages compared with Stage I/II cancers (p = 0.004 and p = 0.014 respectively). Patients with high epithelial density of FOXP3+ cells, CD8+/FOXP3+ cells, or PD-L1- macrophages, had poorer survival, and high epithelial CD79a + plasma cells conferred better survival, all upon univariate analysis only. Clustering showed that most MOC (86%) had an immune depleted (cold) phenotype, with only a small proportion (11/76,14%) considered immune inflamed (hot) based on T cell and PD-L1 infiltrates., Conclusion: In summary, MOCs are mostly immunogenically 'cold', suggesting they may have limited response to current immunotherapies., Competing Interests: Declaration of Competing Interest NSM has received a travel grant from NanoString technologies, unrelated to this work. DDLB has received research support grants from Roche-Genentech, AstraZeneca, and BeiGene (paid to institution); and personal consulting fees from Exo Therapeutics, that are outside the submitted work. MF has participated in Advisory Boards/Consulting from Astra Zeneca, Novartis, GSK, Takeda, Lilly, MSD Eisei and received honoraria/speakers' fees from AstraZeneca, GSK, ACT-Genomics; Research funding to institution: from AstraZeneca, Novartis, Beigene, all unrelated to this work. ADeF has received funding from AstraZeneca, unrelated this work. NS has received honoraria for invited participation in advisory boards hosted by Astra-Zeneca-MSD and Glaxo SmithKline, unrelated to this work. UM had stock ownership in Abcodia until October 2021, and UCL had a license agreement (2011–2021) with Abcodia not related to this work. BHN is a co-founder of Innovakine Therapeutics Inc., unrelated to this work., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
35. The genomic and immune landscape of long-term survivors of high-grade serous ovarian cancer.
- Author
-
Garsed DW, Pandey A, Fereday S, Kennedy CJ, Takahashi K, Alsop K, Hamilton PT, Hendley J, Chiew YE, 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, and Bowtell DDL
- Subjects
- Female, Humans, Survivors, Genomics, Ovarian Neoplasms genetics
- Abstract
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., (© 2022. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2022
- Full Text
- View/download PDF
36. The Tumor Microenvironment of Clear-Cell Ovarian Cancer.
- Author
-
Devlin MJ, Miller R, Laforets F, Kotantaki P, Garsed DW, Kristeleit R, Bowtell DD, McDermott J, Maniati E, and Balkwill FR
- Subjects
- Female, Humans, Programmed Cell Death 1 Receptor, CD8-Positive T-Lymphocytes, Collagen, Tumor Microenvironment, Ovarian Neoplasms pathology
- Abstract
Some patients with advanced clear-cell ovarian cancer (CCOC) respond to immunotherapy; however, little is known about the tumor microenvironment (TME) of this relatively rare disease. Here, we describe a comprehensive quantitative and topographical analysis of biopsies from 45 patients, 9 with Federation Internationale des Gynaecologistes et Obstetristes (FIGO) stage I/II (early CCOC) and 36 with FIGO stage III/IV (advanced CCOC). We investigated 14 immune cell phenotype markers, PD-1 and ligands, and collagen structure and texture. We interrogated a microarray data set from a second cohort of 29 patients and compared the TMEs of ARID1A-wildtype (ARID1Awt) versus ARID1A-mutant (ARID1Amut) disease. We found significant variations in immune cell frequency and phenotype, checkpoint expression, and collagen matrix between the malignant cell area (MCA), leading edge (LE), and stroma. The MCA had the largest population of CD138+ plasma cells, the LE had more CD20+ B cells and T cells, whereas the stroma had more mast cells and αSMA+ fibroblasts. PD-L2 was expressed predominantly on malignant cells and was the dominant PD-1 ligand. Compared with early CCOC, advanced-stage disease had significantly more fibroblasts and a more complex collagen matrix, with microarray analysis indicating "TGFβ remodeling of the extracellular matrix" as the most significantly enriched pathway. Data showed significant differences in immune cell populations, collagen matrix, and cytokine expression between ARID1Awt and ARID1Amut CCOC, which may reflect different paths of tumorigenesis and the relationship to endometriosis. Increased infiltration of CD8+ T cells within the MCA and CD4+ T cells at the LE and stroma significantly associated with decreased overall survival., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
37. TRACEBACK: Testing of Historical Tubo-Ovarian Cancer Patients for Hereditary Risk Genes as a Cancer Prevention Strategy in Family Members.
- Author
-
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 MA, Gordon L, Forrest L, Harris M, Livingstone K, Hill J, Chenevix-Trench G, Cohen PA, Webb PM, Friedlander M, James P, Bowtell D, and Alsop K
- Subjects
- Australia, Carcinoma, Ovarian Epithelial genetics, Family, Female, Genetic Predisposition to Disease, Genetic Testing methods, Humans, Male, Pilot Projects, Retrospective Studies, Breast Neoplasms genetics, Ovarian Neoplasms genetics, Ovarian Neoplasms prevention & control
- Abstract
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., Competing Interests: Belinda CreightonEmployment: Cancer Specialists 1/84 Bridge Rd, Richmond VIC 3121 AustraliaConsulting or Advisory Role: Bristol Myers Squibb, Eisai Dinuka AriyaratneResearch Funding: AstraZeneca Elizabeth ChristieHonoraria: AstraZeneca Sian FeredayConsulting or Advisory Role: Geneseq Biosciences Pty LtdResearch Funding: AstraZeneca (Inst), AstraZeneca (Inst) Nadia TraficanteResearch Funding: AstraZeneca (Inst) Bryony ThompsonConsulting or Advisory Role: Genetic Technologies Kenneth DoigStock and Other Ownership Interests: CSL Limited Christopher LoveEmployment: Geneseq Biosciences Yeh Chen LeeConsulting or Advisory Role: GlaxoSmithKlineResearch Funding: AstraZeneca (Inst)Travel, Accommodations, Expenses: AstraZeneca Rachel WilliamsHonoraria: AstraZeneca Andrew N. StephensConsulting or Advisory Role: Invion Pty Ltd (Inst)Research Funding: Invion Pty Ltd (Inst) Adeline TanEmployment: Sonic HealthcareStock and Other Ownership Interests: Sonic Healthcare Stephen B. FoxConsulting or Advisory Role: Novartis (Inst), BMS (Inst), AstraZeneca (Inst), MSD (Inst), Pfizer (Inst), Roche (Inst)Research Funding: AstraZeneca (Inst), Amgen (Inst), Roche (Inst), BMS (Inst)Expert Testimony: AstraZeneca (Inst), MSD (Inst)Travel, Accommodations, Expenses: Ventana Medical Systems, AstraZeneca, MSD Mary-Anne YoungUncompensated Relationships: Illumina (Inst) Jane HillOther Relationship: Medicines Australia Paul A. CohenEmployment: St John of God HealthcareStock and Other Ownership Interests: Clinic IQHonoraria: AstraZenecaConsulting or Advisory Role: Clinic IQResearch Funding: ANZGOG, St John of God Foundation Penelope M. WebbResearch Funding: AstraZeneca (Inst) Michael FriedlanderHonoraria: AstraZeneca, MSD, Lilly, Takeda, Novartis, GlaxoSmithKlineConsulting or Advisory Role: AstraZeneca, MSD, AbbVie, Lilly, Takeda, Novartis, GlaxoSmithKlineSpeakers' Bureau: AstraZeneca, ACT GenomicsResearch Funding: BeiGene (Inst), AstraZeneca (Inst), Novartis (Inst)Travel, Accommodations, Expenses: AstraZeneca David BowtellHonoraria: AstraZenecaConsulting or Advisory Role: Exo TherapeuticsResearch Funding: Roche/Genentech, AstraZeneca, BeiGenePatents, Royalties, Other Intellectual Property: AstraZeneca Genentech Roche Kathryn AlsopResearch Funding: AstraZeneca (Inst)No other potential conflicts of interest were reported.
- Published
- 2022
- Full Text
- View/download PDF
38. ESMO recommendations on predictive biomarker testing for homologous recombination deficiency and PARP inhibitor benefit in ovarian cancer.
- Author
-
Miller RE, Leary A, Scott CL, Serra V, Lord CJ, Bowtell D, Chang DK, Garsed DW, Jonkers J, Ledermann JA, Nik-Zainal S, Ray-Coquard I, Shah SP, Matias-Guiu X, Swisher EM, and Yates LR
- Subjects
- Biomarkers, Carcinoma, Ovarian Epithelial, Female, Homologous Recombination, Humans, Ovarian Neoplasms drug therapy, Ovarian Neoplasms genetics, Poly(ADP-ribose) Polymerase Inhibitors therapeutic use
- Abstract
Background: Homologous recombination repair deficiency (HRD) is a frequent feature of high-grade serous ovarian, fallopian tube and peritoneal carcinoma (HGSC) and is associated with sensitivity to PARP inhibitor (PARPi) therapy. HRD testing provides an opportunity to optimise PARPi use in HGSC but methodologies are diverse and clinical application remains controversial., Materials and Methods: To define best practice for HRD testing in HGSC the ESMO Translational Research and Precision Medicine Working Group launched a collaborative project that incorporated a systematic review approach. The main aims were to (i) define the term 'HRD test'; (ii) provide an overview of the biological rationale and the level of evidence supporting currently available HRD tests; (iii) provide recommendations on the clinical utility of HRD tests in clinical management of HGSC., Results: A broad range of repair genes, genomic scars, mutational signatures and functional assays are associated with a history of HRD. Currently, the clinical validity of HRD tests in ovarian cancer is best assessed, not in terms of biological HRD status per se, but in terms of PARPi benefit. Clinical trials evidence supports the use of BRCA mutation testing and two commercially available assays that also incorporate genomic instability for identifying subgroups of HGSCs that derive different magnitudes of benefit from PARPi therapy, albeit with some variation by clinical scenario. These tests can be used to inform treatment selection and scheduling but their use is limited by a failure to consistently identify a subgroup of patients who derive no benefit from PARPis in most studies. Existing tests lack negative predictive value and inadequately address the complex and dynamic nature of the HRD phenotype., Conclusions: Currently available HRD tests are useful for predicting likely magnitude of benefit from PARPis but better biomarkers are urgently needed to better identify current homologous recombination proficiency status and stratify HGSC management., Competing Interests: Disclosures REM receives consultancy fees from Merck, TESARO-GSK and Clovis Oncology, speaker bureau from Roche and TESARO and travel grants from AstraZeneca and TESARO. LRY receives research funding from the Wellcome Trust (Clinical Career Development Fellowship 214584/Z/18/Z). AL receives consultancy fees from Ability, Astra Zeneca, Clovis, GamaMabs, GSK, MSD, Merck Serono, and TESARO, research funding from Merus, GamaMabs, Inivata, and Sanofi Investigator and fees for trials with Ability, Agenus, AstraZeneca, BMS, Boehringer. IRC is the principal investigator of PAOLA1 trial; receives honoraria from AstraZeneca, Clovis, TESARO and Pharma Mar; consulting/advisory board fees from AstraZeneca, Roche, Clovis, TESARO, Genmab, Pharma Mar, MSD and Pfizer; research funding from MSD; travel expenses from AstraZeneca, GSK and Roche. JAL received fees for Advisory Boards and Lectures for AstraZeneca; Clovis Oncology; GSK. Advisory Boards from MSD/Merck; Eisai; Artios; Amgen; Regeneron. Grants AstraZeneca; MSD/Merck. SPS is a founder and shareholder of Canexia Health Inc. VS developed EP3502700A1: methods based on the detection of rad51 foci in tumour cells and developed EP3502700A1: methods based on the detection of rad51 foci in tumour cells and receives research funding from AstraZeneca and Tesaro; received consultancy, SAB membership or honoraria payments from Abbvie. CJL receives research funding from AstraZeneca, Merck KGaA, and Artios; received consultancy, SAB membership or honoraria payments from Syncona, Sun Pharma, Gerson Lehrman Group, Merck KGaA, Vertex, AstraZeneca, Tango, 3rd Rock, Ono Pharma, and Artios. CJL has stock in Tango and OVIBIO and is also a named inventor on patents describing the use of DNA repair inhibitors and stands to gain from the development as part of the ICR ‘Rewards to Inventors’ scheme. JJ is an advisory board member of Artios Pharma. DKC received research funding from Cancer Research UK (C51058/A25407), Celgene and Astra Zeneca, and speaker bureau from Celgene. DB receives research grant funding from Astra Zeneca, Genentech/Roche, Beigene, and paid consultancy from Exo Therapeutics. SNZ holds multiple patents on mutational-signature-based clinical algorithms and is a current or past recipient of honoraria from Astra Zeneca, Artios Pharma and the Scottish Genomics Partnership. JJ is an advisory board member of Artios Pharma and receives research funding from Artios Pharma, Cancer Research UK, Dutch Cancer Society and Oncode Institute. All remaining authors have declared no conflicts of interest. CLS acts in an honorary advisory capacity for AstraZeneca, Clovis Oncology, Roche, Eisai Inc, Sierra Oncology, Takeda, MSD.; receives grant/research support from Clovis Oncology, Eisai Inc, Sierra Oncology, Roche, Beigene; travel support from AstraZeneca., (Copyright © 2020 European Society for Medical Oncology. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
39. Menopausal hormone therapy prior to the diagnosis of ovarian cancer is associated with improved survival.
- Author
-
Brieger KK, Peterson S, Lee AW, Mukherjee B, Bakulski KM, Alimujiang A, Anton-Culver H, Anglesio MS, Bandera EV, Berchuck A, Bowtell DDL, Chenevix-Trench G, Cho KR, Cramer DW, DeFazio A, Doherty JA, Fortner RT, Garsed DW, Gayther SA, Gentry-Maharaj A, Goode EL, Goodman MT, Harris HR, Høgdall E, Huntsman DG, Shen H, Jensen A, Johnatty SE, Jordan SJ, Kjaer SK, Kupryjanczyk J, Lambrechts D, McLean K, Menon U, Modugno F, Moysich K, Ness R, Ramus SJ, Richardson J, Risch H, Rossing MA, Trabert B, Wentzensen N, Ziogas A, Terry KL, Wu AH, Hanley GE, Pharoah P, Webb PM, Pike MC, and Pearce CL
- Subjects
- Aged, Female, Humans, Middle Aged, Neoplasm Staging, Neoplasm, Residual pathology, Ovarian Neoplasms pathology, Ovarian Neoplasms surgery, Postmenopause, Progression-Free Survival, Proportional Hazards Models, Survival Rate, Estrogen Replacement Therapy statistics & numerical data, Hormone Replacement Therapy statistics & numerical data, Ovarian Neoplasms mortality, Progestins administration & dosage
- Abstract
Purpose: Prior studies of menopausal hormone therapy (MHT) and ovarian cancer survival have been limited by lack of hormone regimen detail and insufficient sample sizes. To address these limitations, a comprehensive analysis of 6419 post-menopausal women with pathologically confirmed ovarian carcinoma was conducted to examine the association between MHT use prior to diagnosis and survival., Methods: Data from 15 studies in the Ovarian Cancer Association Consortium were included. MHT use was examined by type (estrogen-only (ET) or estrogen+progestin (EPT)), duration, and recency of use relative to diagnosis. Cox proportional hazards models were used to estimate the association between hormone therapy use and survival. Logistic regression and mediation analysis was used to explore the relationship between MHT use and residual disease following debulking surgery., Results: Use of ET or EPT for at least five years prior to diagnosis was associated with better ovarian cancer survival (hazard ratio, 0.80; 95% CI, 0.74 to 0.87). Among women with advanced stage, high-grade serous carcinoma, those who used MHT were less likely to have any macroscopic residual disease at the time of primary debulking surgery (p for trend <0.01 for duration of MHT use). Residual disease mediated some (17%) of the relationship between MHT and survival., Conclusions: Pre-diagnosis MHT use for 5+ years was a favorable prognostic factor for women with ovarian cancer. This large study is consistent with prior smaller studies, and further work is needed to understand the underlying mechanism., Competing Interests: Declaration of Competing Interest Usha Menon has stocks in Abcodia awarded to her by University College London., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
40. Survival Following Chemotherapy in Ovarian Clear Cell Carcinoma Is Not Associated with Pathological Misclassification of Tumor Histotype.
- Author
-
Takenaka M, Köbel M, Garsed DW, Fereday S, Pandey A, Etemadmoghadam D, Hendley J, Kawabata A, Noguchi D, Yanaihara N, Takahashi H, Kiyokawa T, Ikegami M, Takano H, Isonishi S, Ochiai K, Traficante N, Gadipally S, Semple T, Vassiliadis D, Amarasinghe K, Li J, Mir Arnau G, Okamoto A, Friedlander M, and Bowtell DDL
- Subjects
- Adenocarcinoma, Clear Cell drug therapy, Adenocarcinoma, Clear Cell etiology, Adult, Aged, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Biomarkers, Diagnostic Errors, Female, Humans, Immunohistochemistry, Middle Aged, Mutation, Neoplasm Grading, Neoplasm Staging, Ovarian Neoplasms drug therapy, Ovarian Neoplasms etiology, Prognosis, Treatment Outcome, Adenocarcinoma, Clear Cell mortality, Adenocarcinoma, Clear Cell pathology, Ovarian Neoplasms mortality, Ovarian Neoplasms pathology
- Abstract
Purpose: Although ovarian clear cell carcinomas (OCCC) are commonly resistant to platinum-based chemotherapy, good clinical outcomes are observed in a subset of patients. The explanation for this is unknown but may be due to misclassification of high-grade serous ovarian cancer (HGSOC) as OCCC or mixed histology., Experimental Design: To discover potential biomarkers of survival benefit following platinum-based chemotherapy, we ascertained a cohort of 68 Japanese and Australian patients in whom progression-free survival (PFS) and overall survival (OS) could be assessed. We performed IHC reclassification of tumors, and targeted sequencing and immunohistochemistry of known driver genes. Exome sequencing was performed in 10 patients who had either unusually long survival ( N = 5) or had a very short time to progression ( N = 5)., Results: The majority of mixed OCCC ( N = 6, 85.7%) and a small proportion of pure OCCC ( N = 3, 4.9%) were reclassified as likely HGSOC. However, the PFS and OS of patients with misclassified samples were similar to that of patients with pathologically validated OCCC. Absent HNF1B expression was significantly correlated with longer PFS and OS ( P = 0.0194 and 0.0395, respectively). Mutations in ARID1A, PIK3CA, PPP2R1A , and TP53 were frequent, but did not explain length of PFS and OS. An exploratory exome analysis of patients with favorable and unfavorable outcomes did not identify novel outcome-associated driver mutations., Conclusions: Survival benefit following chemotherapy in OCCC was not associated with pathological misclassification of tumor histotype. HNF1B loss may help identify the subset of patients with OCCC with a more favorable outcome., (©2019 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
41. Going to extremes: determinants of extraordinary response and survival in patients with cancer.
- Author
-
Saner FAM, Herschtal A, Nelson BH, deFazio A, Goode EL, Ramus SJ, Pandey A, Beach JA, Fereday S, Berchuck A, Lheureux S, Pearce CL, Pharoah PD, Pike MC, Garsed DW, and Bowtell DDL
- Subjects
- Humans, Lymphocytes, Tumor-Infiltrating immunology, Mutation, Neoplasms genetics, Neoplasms mortality, Survival Analysis, Treatment Outcome, Cancer Survivors, Neoplasms therapy
- Abstract
Research into factors affecting treatment response or survival in patients with cancer frequently involves cohorts that span the most common range of clinical outcomes, as such patients are most readily available for study. However, attention has turned to highly unusual patients who have exceptionally favourable or atypically poor responses to treatment and/or overall survival, with the expectation that patients at the extremes may provide insights that could ultimately improve the outcome of individuals with more typical disease trajectories. While clinicians can often recount surprising patients whose clinical journey was very unusual, given known clinical characteristics and prognostic indicators, there is a lack of consensus among researchers on how best to define exceptional patients, and little has been proposed for the optimal design of studies to identify factors that dictate unusual outcome. In this Opinion article, we review different approaches to identifying exceptional patients with cancer and possible study designs to investigate extraordinary clinical outcomes. We discuss pitfalls with finding these rare patients, including challenges associated with accrual of patients across different treatment centres and time periods. We describe recent molecular and immunological factors that have been identified as contributing to unusual patient outcome and make recommendations for future studies on these intriguing patients.
- Published
- 2019
- Full Text
- View/download PDF
42. Genomic Analysis Using Regularized Regression in High-Grade Serous Ovarian Cancer.
- Author
-
Natanzon Y, Earp M, Cunningham JM, Kalli KR, Wang C, Armasu SM, Larson MC, Bowtell DD, Garsed DW, Fridley BL, Winham SJ, and Goode EL
- Abstract
High-grade serous ovarian cancer (HGSOC) is a complex disease in which initiation and progression have been associated with copy number alterations, epigenetic processes, and, to a lesser extent, germline variation. We hypothesized that, when summarized at the gene level, tumor methylation and germline genetic variation, alone or in combination, influence tumor gene expression in HGSOC. We used Elastic Net (ENET) penalized regression method to evaluate these associations and adjust for somatic copy number in 3 independent data sets comprising tumors from more than 470 patients. Penalized regression models of germline variation, with or without methylation, did not reveal a role in HGSOC gene expression. However, we observed significant association between regional methylation and expression of 5 genes ( WDPCP, KRT6C, BRCA2, EFCAB13 , and ZNF283 ). CpGs retained in ENET model for BRCA2 and ZNF283 appeared enriched in several regulatory elements, suggesting that regularized regression may provide a novel utility for integrative genomic analysis., Competing Interests: Declaration of conflicting interests:The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
- Published
- 2018
- Full Text
- View/download PDF
43. Homologous Recombination DNA Repair Pathway Disruption and Retinoblastoma Protein Loss Are Associated with Exceptional Survival in High-Grade Serous Ovarian Cancer.
- Author
-
Garsed DW, Alsop K, Fereday S, Emmanuel C, Kennedy CJ, Etemadmoghadam D, Gao B, Gebski V, Garès V, Christie EL, Wouters MCA, Milne K, George J, Patch AM, Li J, Arnau GM, Semple T, Gadipally SR, Chiew YE, Hendley J, Mikeska T, Zapparoli GV, Amarasinghe K, Grimmond SM, Pearson JV, Waddell N, Hung J, Stewart CJR, Sharma R, Allan PE, Rambau PF, McNally O, Mileshkin L, Hamilton A, Ananda S, Grossi M, Cohen PA, Leung YC, Rome RM, Beale P, Blomfield P, Friedlander M, Brand A, Dobrovic A, Köbel M, Harnett P, Nelson BH, Bowtell DDL, and deFazio A
- Subjects
- Adult, Aged, Aged, 80 and over, Biomarkers, Tumor, Cystadenocarcinoma, Serous diagnosis, Female, Homologous Recombination, Humans, Middle Aged, Mutation, Neoplasm Grading, Neoplasm Staging, Ovarian Neoplasms diagnosis, Ovarian Neoplasms metabolism, Prognosis, Retinoblastoma Protein metabolism, Signal Transduction, Survival Analysis, Symptom Assessment, Cystadenocarcinoma, Serous genetics, Cystadenocarcinoma, Serous mortality, Ovarian Neoplasms genetics, Ovarian Neoplasms mortality, Recombinational DNA Repair, Retinoblastoma Protein genetics
- Abstract
Purpose: Women with epithelial ovarian cancer generally have a poor prognosis; however, a subset of patients has an unexpected dramatic and durable response to treatment. We sought to identify clinical, pathological, and molecular determinants of exceptional survival in women with high-grade serous cancer (HGSC), a disease associated with the majority of ovarian cancer deaths. Experimental Design: We evaluated the histories of 2,283 ovarian cancer patients and, after applying stringent clinical and pathological selection criteria, identified 96 with HGSC that represented significant outliers in terms of treatment response and overall survival. Patient samples were characterized immunohistochemically and by genome sequencing. Results: Different patterns of clinical response were seen: long progression-free survival (Long-PFS), multiple objective responses to chemotherapy (Multiple Responder), and/or greater than 10-year overall survival (Long-Term Survivors). Pathogenic germline and somatic mutations in genes involved in homologous recombination (HR) repair were enriched in all three groups relative to a population-based series. However, 29% of 10-year survivors lacked an identifiable HR pathway alteration, and tumors from these patients had increased Ki-67 staining. CD8
+ tumor-infiltrating lymphocytes were more commonly present in Long-Term Survivors. RB1 loss was associated with long progression-free and overall survival. HR deficiency and RB1 loss were correlated, and co-occurrence was significantly associated with prolonged survival. Conclusions: There was diversity in the clinical trajectory of exceptional survivors associated with multiple molecular determinants of exceptional outcome in HGSC patients. Concurrent HR deficiency and RB1 loss were associated with favorable outcomes, suggesting that co-occurrence of specific mutations might mediate durable responses in such patients. Clin Cancer Res; 24(3); 569-80. ©2017 AACR See related commentary by Peng and Mills, p. 508 ., (©2017 American Association for Cancer Research.)- Published
- 2018
- Full Text
- View/download PDF
44. EIF1AX and NRAS Mutations Co-occur and Cooperate in Low-Grade Serous Ovarian Carcinomas.
- Author
-
Etemadmoghadam D, Azar WJ, Lei Y, Moujaber T, Garsed DW, Kennedy CJ, Fereday S, Mitchell C, Chiew YE, Hendley J, Sharma R, Harnett PR, Li J, Christie EL, Patch AM, George J, Au-Yeung G, Mir Arnau G, Holloway TP, Semple T, Pearson JV, Waddell N, Grimmond SM, Köbel M, Rizos H, Lomakin IB, Bowtell DDL, and deFazio A
- Subjects
- Cell Line, Tumor, Cystadenocarcinoma, Serous pathology, Eukaryotic Initiation Factor-1 biosynthesis, Female, Gene Knockdown Techniques, Humans, Mutagenesis, Site-Directed, Neoplasm Grading, Neoplasm Staging, Ovarian Neoplasms pathology, Cystadenocarcinoma, Serous genetics, Eukaryotic Initiation Factor-1 genetics, GTP Phosphohydrolases genetics, Membrane Proteins genetics, Mutation, Ovarian Neoplasms genetics
- Abstract
Low-grade serous ovarian carcinomas (LGSC) are associated with a poor response to chemotherapy and are molecularly characterized by RAS pathway activation. Using exome and whole genome sequencing, we identified recurrent mutations in the protein translational regulator EIF1AX and in NF1, USP9X, KRAS, BRAF , and NRAS RAS pathway mutations were mutually exclusive; however, we found significant co-occurrence of mutations in NRAS and EIF1AX Missense EIF1AX mutations were clustered at the N-terminus of the protein in a region associated with its role in ensuring translational initiation fidelity. Coexpression of mutant NRAS and EIF1AX proteins promoted proliferation and clonogenic survival in LGSC cells, providing the first example of co-occurring, growth-promoting mutational events in ovarian cancer. Cancer Res; 77(16); 4268-78. ©2017 AACR ., (©2017 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
45. Corrigendum: Whole-genome characterization of chemoresistant ovarian cancer.
- Author
-
Patch AM, Christie EL, Etemadmoghadam D, Garsed DW, George J, Fereday S, Nones K, Cowin P, Alsop K, Bailey PJ, Kassahn KS, Newell F, Quinn MC, Kazakoff S, Quek K, Wilhelm-Benartzi C, Curry E, Leong HS, Hamilton A, Mileshkin L, Au-Yeung G, Kennedy C, Hung J, Chiew YE, Harnett P, Friedlander M, Quinn M, Pyman J, Cordner S, O'Brien P, Leditschke J, Young G, Strachan K, Waring P, Azar W, Mitchell C, Traficante N, Hendley J, Thorne H, Shackleton M, Miller DK, Arnau GM, Tothill RW, Holloway TP, Semple T, Harliwong I, Nourse C, Nourbakhsh E, Manning S, Idrisoglu S, Bruxner TJ, Christ AN, Poudel B, Holmes O, Anderson M, Leonard C, Lonie A, Hall N, Wood S, Taylor DF, Xu Q, Fink JL, Waddell N, Drapkin R, Stronach E, Gabra H, Brown R, Jewell A, Nagaraj SH, Markham E, Wilson PJ, Ellul J, McNally O, Doyle MA, Vedururu R, Stewart C, Lengyel E, Pearson JV, Waddell N, deFazio A, Grimmond SM, and Bowtell DD
- Published
- 2015
- Full Text
- View/download PDF
46. Whole-genome characterization of chemoresistant ovarian cancer.
- Author
-
Patch AM, Christie EL, Etemadmoghadam D, Garsed DW, George J, Fereday S, Nones K, Cowin P, Alsop K, Bailey PJ, Kassahn KS, Newell F, Quinn MC, Kazakoff S, Quek K, Wilhelm-Benartzi C, Curry E, Leong HS, Hamilton A, Mileshkin L, Au-Yeung G, Kennedy C, Hung J, Chiew YE, Harnett P, Friedlander M, Quinn M, Pyman J, Cordner S, O'Brien P, Leditschke J, Young G, Strachan K, Waring P, Azar W, Mitchell C, Traficante N, Hendley J, Thorne H, Shackleton M, Miller DK, Arnau GM, Tothill RW, Holloway TP, Semple T, Harliwong I, Nourse C, Nourbakhsh E, Manning S, Idrisoglu S, Bruxner TJ, Christ AN, Poudel B, Holmes O, Anderson M, Leonard C, Lonie A, Hall N, Wood S, Taylor DF, Xu Q, Fink JL, Waddell N, Drapkin R, Stronach E, Gabra H, Brown R, Jewell A, Nagaraj SH, Markham E, Wilson PJ, Ellul J, McNally O, Doyle MA, Vedururu R, Stewart C, Lengyel E, Pearson JV, Waddell N, deFazio A, Grimmond SM, and Bowtell DD
- Subjects
- ATP Binding Cassette Transporter, Subfamily B, Member 1 genetics, Cohort Studies, Cyclin E genetics, Cystadenocarcinoma, Serous drug therapy, Cystadenocarcinoma, Serous genetics, DNA Methylation, DNA Mutational Analysis, DNA-Binding Proteins genetics, Female, Genes, BRCA1, Genes, BRCA2, Genes, Neurofibromatosis 1, Germ-Line Mutation genetics, Humans, Mutagenesis genetics, Oncogene Proteins genetics, Ovarian Neoplasms drug therapy, PTEN Phosphohydrolase genetics, Promoter Regions, Genetic genetics, Retinoblastoma Protein genetics, Drug Resistance, Neoplasm drug effects, Drug Resistance, Neoplasm genetics, Genome, Human genetics, Ovarian Neoplasms genetics
- Abstract
Patients with high-grade serous ovarian cancer (HGSC) have experienced little improvement in overall survival, and standard treatment has not advanced beyond platinum-based combination chemotherapy, during the past 30 years. To understand the drivers of clinical phenotypes better, here we use whole-genome sequencing of tumour and germline DNA samples from 92 patients with primary refractory, resistant, sensitive and matched acquired resistant disease. We show that gene breakage commonly inactivates the tumour suppressors RB1, NF1, RAD51B and PTEN in HGSC, and contributes to acquired chemotherapy resistance. CCNE1 amplification was common in primary resistant and refractory disease. We observed several molecular events associated with acquired resistance, including multiple independent reversions of germline BRCA1 or BRCA2 mutations in individual patients, loss of BRCA1 promoter methylation, an alteration in molecular subtype, and recurrent promoter fusion associated with overexpression of the drug efflux pump MDR1.
- Published
- 2015
- Full Text
- View/download PDF
47. The architecture and evolution of cancer neochromosomes.
- Author
-
Garsed DW, Marshall OJ, Corbin VD, Hsu A, Di Stefano L, Schröder J, Li J, Feng ZP, Kim BW, Kowarsky M, Lansdell B, Brookwell R, Myklebost O, Meza-Zepeda L, Holloway AJ, Pedeutour F, Choo KH, Damore MA, Deans AJ, Papenfuss AT, and Thomas DM
- Subjects
- Aged, Carcinogenesis genetics, Cell Line, Tumor, Centromere genetics, Chromosome Aberrations, Female, Humans, Liposarcoma pathology, Models, Genetic, Mutagenesis, Oncogenes, Retroperitoneal Neoplasms pathology, Translocation, Genetic, Chromosomes, Human genetics, Liposarcoma genetics, Retroperitoneal Neoplasms genetics
- Abstract
We isolated and analyzed, at single-nucleotide resolution, cancer-associated neochromosomes from well- and/or dedifferentiated liposarcomas. Neochromosomes, which can exceed 600 Mb in size, initially arise as circular structures following chromothripsis involving chromosome 12. The core of the neochromosome is amplified, rearranged, and corroded through hundreds of breakage-fusion-bridge cycles. Under selective pressure, amplified oncogenes are overexpressed, while coamplified passenger genes may be silenced epigenetically. New material may be captured during punctuated chromothriptic events. Centromeric corrosion leads to crisis, which is resolved through neocentromere formation or native centromere capture. Finally, amplification terminates, and the neochromosome core is stabilized in linear form by telomere capture. This study investigates the dynamic mutational processes underlying the life history of a special form of cancer mutation., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
48. Immune response to RB1-regulated senescence limits radiation-induced osteosarcoma formation.
- Author
-
Kansara M, Leong HS, Lin DM, Popkiss S, Pang P, Garsed DW, Walkley CR, Cullinane C, Ellul J, Haynes NM, Hicks R, Kuijjer ML, Cleton-Jansen AM, Hinds PW, Smyth MJ, and Thomas DM
- Subjects
- Animals, Bone Neoplasms etiology, Bone Neoplasms genetics, Bone Neoplasms pathology, Calcium Radioisotopes toxicity, Cytokines physiology, Genes, Retinoblastoma, Humans, Immunologic Surveillance, Intercellular Signaling Peptides and Proteins physiology, Interleukin-6 deficiency, Interleukin-6 physiology, Mice, Mice, Inbred C57BL, Neoplasm Proteins antagonists & inhibitors, Neoplasm Proteins physiology, Neoplasm Transplantation immunology, Neoplasms, Radiation-Induced etiology, Neoplasms, Radiation-Induced genetics, Neoplasms, Radiation-Induced pathology, Osteoblasts pathology, Osteosarcoma etiology, Osteosarcoma genetics, Osteosarcoma pathology, Phenotype, Prognosis, RNA Interference, Retinoblastoma Protein antagonists & inhibitors, Bone Neoplasms immunology, Cellular Senescence physiology, Natural Killer T-Cells immunology, Neoplasms, Radiation-Induced immunology, Osteosarcoma immunology, Retinoblastoma Protein physiology
- Abstract
Ionizing radiation (IR) and germline mutations in the retinoblastoma tumor suppressor gene (RB1) are the strongest risk factors for developing osteosarcoma. Recapitulating the human predisposition, we found that Rb1+/- mice exhibited accelerated development of IR-induced osteosarcoma, with a latency of 39 weeks. Initial exposure of osteoblasts to carcinogenic doses of IR in vitro and in vivo induced RB1-dependent senescence and the expression of a panel of proteins known as senescence-associated secretory phenotype (SASP), dominated by IL-6. RB1 expression closely correlated with that of the SASP cassette in human osteosarcomas, and low expression of both RB1 and the SASP genes was associated with poor prognosis. In vivo, IL-6 was required for IR-induced senescence, which elicited NKT cell infiltration and a host inflammatory response. Mice lacking IL-6 or NKT cells had accelerated development of IR-induced osteosarcomas. These data elucidate an important link between senescence, which is a cell-autonomous tumor suppressor response, and the activation of host-dependent cancer immunosurveillance. Our findings indicate that overcoming the immune response to senescence is a rate-limiting step in the formation of IR-induced osteosarcoma.
- Published
- 2013
- Full Text
- View/download PDF
49. A newly characterized human well-differentiated liposarcoma cell line contains amplifications of the 12q12-21 and 10p11-14 regions.
- Author
-
Pedeutour F, Maire G, Pierron A, Thomas DM, Garsed DW, Bianchini L, Duranton-Tanneur V, Cortes-Maurel A, Italiano A, Squire JA, and Coindre JM
- Subjects
- Adult, Cell Differentiation, Comparative Genomic Hybridization, Gene Amplification, Humans, In Situ Hybridization, Fluorescence, Karyotyping, Liposarcoma pathology, Male, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Cell Line, Tumor, Chromosomes, Human, Pair 10 genetics, Chromosomes, Human, Pair 12 genetics, Liposarcoma genetics, Primary Cell Culture
- Abstract
While surgery is the usual treatment for localized well-differentiated and dedifferentiated liposarcomas (WDLPS/DDLPS), the therapeutic options for patients with advanced disease are limited. The classical antimitotic treatments are most often inefficient. The establishment of genetically characterized cell lines is therefore crucial for providing in vitro models for novel targeted therapies. We have used spectral karyotyping, fluorescence in situ hybridization with whole chromosome painting and locus-specific probes, and array-comparative genomic hybridization to identify the chromosomal and molecular alterations of a novel cell line established from a recurring sclerosing WDLPS. The karyotype was hypertriploid and showed multiple structural anomalies. All cells retained the presence of a giant marker chromosome that had been previously identified in the primary cell cultures. This giant chromosome contained high-level amplification of chromosomal regions 12q13-21 and lacked the alpha-satellite centromeric sequences associated with WDLPS/DDLPS. The 12q amplicon was large, containing 370 amplified genes. The DNA copy number ranged from 3 to 57. The highest levels of amplification were observed at 12q14.3 for GNS, WIF1, and HMGA2. We analyzed the mRNA expression status by real-time reverse transcription polymerase chain reaction for six genes from this amplicon: MDM2, HMGA2, CDK4, TSPAN31, WIF1, and YEATS4. mRNA overexpression was correlated with genomic amplification. A second amplicon originating from 10p11-14 was also present in the giant marker chromosome. The 10p amplicon contained 62 genes, including oncogenes such as MLLT10, previously described in chimeric fusion with MLL in leukemias, NEBL, and BMI1.
- Published
- 2012
- Full Text
- View/download PDF
50. Cancer-associated neochromosomes: a novel mechanism of oncogenesis.
- Author
-
Garsed DW, Holloway AJ, and Thomas DM
- Subjects
- Animals, Chromosome Aberrations, Chromosome Mapping, DNA Mutational Analysis, DNA, Neoplasm genetics, DNA, Satellite genetics, Gene Expression Regulation, Neoplastic, Humans, Liposarcoma genetics, Telomere ultrastructure, Cell Transformation, Neoplastic, Chromosomes ultrastructure, Neoplasms genetics
- Abstract
Malignant tumours are often characterised by significant rearrangement of the genome. This may be visible in the form of a deranged karyotype with both loss and gain of DNA sequences extending from chromosomal regions to whole chromosomes. In several tumour types, however, gross genomic derangements are minimal, and tumour cells contain one or more additional (supernumerary) chromosomes that may be unrecognisable in terms of a single origin. In this review we term such chromosomes cancer-associated neochromosomes (CaNCs). In the absence of other identified genomic abnormalities, and because the CaNC is a common feature of the cancer type, it is hypothesised that the genetic alterations required for cell transformation are contained within its structure. In this review, we discuss the potential impact of modern genomic technologies on our understanding of the nature and causes of CaNC formation, which is central to several cancer types, exemplified here by well-differentiated liposarcoma.
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.