10 results on '"Huei San Leong"'
Search Results
2. Development and Validation of the Gene Expression Predictor of High-grade Serous Ovarian Carcinoma Molecular SubTYPE (PrOTYPE)
- Author
-
Sven Mahner, Robertson Mackenzie, Aline Talhouk, Linda E. Kelemen, Gottfried E. Konecny, Jennifer Alsop, Rosalind Glasspool, Chiu-Chen Tseng, Joy Hendley, Dennis J. Slamon, Jennifer A. Doherty, Andrew Berchuck, Anna H. Wu, Anna M. Piskorz, Chen Wang, Cristina Rodríguez-Antona, D.G.H. de Silva, Valerie Rhenius, Peter A. Fasching, Stacey J. Winham, Gary L. Keeney, Teodora Goranova, Joshy George, Jan Lubinski, Michelle J. Henderson, Rex C. Bentley, Jenny Lester, Sabine Behrens, Joellen M. Schildkraut, Michael E. Carney, Timothy Budden, David G. Huntsman, Oleg Oszurek, Michael S. Anglesio, Jacek Gronwald, Ruby Yun-Ju Huang, Martin Köbel, Javier Benitez, Martin Widschwendter, Melissa C. Larson, Raghwa Sharma, Clara Bodelon, Usha Menon, Janusz Menkiszak, Blake Gilks, María Josefa Mosteiro García, Jesús García-Donas, Wafaa Elatre, Scott H. Kaufmann, Paul Haluska, Pamela J. Thompson, Boris Winterhoff, Susan J. Ramus, Louise A. Brinton, Simon A. Gayther, Mary Anne Rossing, Georgia Chenevix-Trench, Hugh Luk, Jolanta Lissowska, Marc T. Goodman, Billy Chen, Beth Y. Karlan, Naveena Singh, Sian Fereday, Mark E. Sherman, Ana Osorio, Lynne R. Wilkens, Maria P. Intermaggio, Brenda Y. Hernandez, Britton Trabert, Esther Herpel, Mercedes Jimenez-Linan, Janine Senz, Geyi Liu, Celeste Leigh Pearce, Samuel C Y Leong, Iain A. McNeish, Isabelle Ray-Coquard, Susana Banerjee, Malcolm C. Pike, Liz-Anne Lewsley, Helen Steed, Honglin Song, Samantha Hinsley, David D.L. Bowtell, James D. Brenton, Holly R. Harris, Tuan Zea Tan, Cezary Cybulski, Alicia Beeghly-Fadiel, A. Toloczko, Nikilyn Nevins, Robert S. Brown, Darren Ennis, Stephanie Chen, Euan A. Stronach, José Palacios, Sandra Orsulic, Anna deFazio, Geoff Macintyre, Kara L. Cushing-Haugen, Mila Volchek, Aleksandra Gentry-Maharaj, Jenny Chang-Claude, Ellen L. Goode, Paul D.P. Pharoah, Hanwei Sudderuddin, Stefan Kommoss, Derek S. Chiu, Huei San Leong, Peter Sinn, Catherine J. Kennedy, Chloe Karpinskyj, Alison Brand, Amy Lum, Veronica Chow, Nicolas Wentzensen, Tayyebeh M. Nazeran, Nadia Traficante, Dustin Johnson, Yoke-Eng Chiew, Casey S. Greene, Jennifer M Koziak, Renée T. Fortner, Imperial College Healthcare NHS Trust- BRC Funding, Cancer Research UK, Ovarian Cancer Action, Talhouk, Aline [0000-0001-7760-410X], George, Joshy [0000-0001-8510-8229], Wang, Chen [0000-0003-2638-3081], Tan, Tuan Zea [0000-0001-6624-1593], Behrens, Sabine [0000-0002-9714-104X], Bodelon, Clara [0000-0002-6578-2678], Brinton, Louise [0000-0003-3853-8562], Fortner, Renée T [0000-0002-1426-8505], García-Donas, Jesús [0000-0001-7731-3601], Gentry-Maharaj, Aleksandra [0000-0001-7270-9762], Glasspool, Rosalind [0000-0002-5000-1680], Greene, Casey S [0000-0001-8713-9213], Harris, Holly R [0000-0002-2572-6727], Kaufmann, Scott H [0000-0002-4900-7145], Kennedy, Catherine J [0000-0002-4465-5784], Köbel, Martin [0000-0002-6615-2037], Koziak, Jennifer M [0000-0001-5830-0397], Lissowska, Jolanta [0000-0003-2695-5799], McNeish, Iain A [0000-0002-9387-7586], Menkiszak, Janusz [0000-0001-8279-7196], Hinsley, Samantha [0000-0001-6903-4688], Pike, Malcolm C [0000-0003-4891-1199], Rodriguez-Antona, Cristina [0000-0001-8750-7338], Sinn, Peter [0000-0003-2836-6699], Trabert, Britton [0000-0002-1539-6090], Widschwendter, Martin [0000-0002-7778-8380], Winham, Stacey J [0000-0002-8492-9102], Brenton, James D [0000-0002-5738-6683], Brown, Robert [0000-0001-7960-5755], Chang-Claude, Jenny [0000-0001-8919-1971], deFazio, Anna [0000-0003-0057-4744], Fasching, Peter A [0000-0003-4885-8471], Kelemen, Linda E [0000-0003-4362-9784], Menon, Usha [0000-0003-3708-1732], Pharoah, Paul DP [0000-0001-8494-732X], Ramus, Susan J [0000-0003-0005-7798], Doherty, Jennifer A [0000-0002-1454-8187], Anglesio, Michael S [0000-0003-1639-5003], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Neoplasm, Residual ,Bevacizumab ,03 medical and health sciences ,Ovarian tumor ,Lymphocytes, Tumor-Infiltrating ,0302 clinical medicine ,Ovarian carcinoma ,Internal medicine ,medicine ,Humans ,1112 Oncology and Carcinogenesis ,Oncology & Carcinogenesis ,Stage (cooking) ,Aged ,Ovarian Neoplasms ,business.industry ,Cystadenoma, Serous ,Cancer ,Middle Aged ,Precision medicine ,Omics ,medicine.disease ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,Serous fluid ,030104 developmental biology ,030220 oncology & carcinogenesis ,Female ,Neoplasm Grading ,Transcriptome ,business ,Algorithms ,medicine.drug - Abstract
Purpose: Gene expression–based molecular subtypes of high-grade serous tubo-ovarian cancer (HGSOC), demonstrated across multiple studies, may provide improved stratification for molecularly targeted trials. However, evaluation of clinical utility has been hindered by nonstandardized methods, which are not applicable in a clinical setting. We sought to generate a clinical grade minimal gene set assay for classification of individual tumor specimens into HGSOC subtypes and confirm previously reported subtype-associated features. Experimental Design: Adopting two independent approaches, we derived and internally validated algorithms for subtype prediction using published gene expression data from 1,650 tumors. We applied resulting models to NanoString data on 3,829 HGSOCs from the Ovarian Tumor Tissue Analysis consortium. We further developed, confirmed, and validated a reduced, minimal gene set predictor, with methods suitable for a single-patient setting. Results: Gene expression data were used to derive the predictor of high-grade serous ovarian carcinoma molecular subtype (PrOTYPE) assay. We established a de facto standard as a consensus of two parallel approaches. PrOTYPE subtypes are significantly associated with age, stage, residual disease, tumor-infiltrating lymphocytes, and outcome. The locked-down clinical grade PrOTYPE test includes a model with 55 genes that predicted gene expression subtype with >95% accuracy that was maintained in all analytic and biological validations. Conclusions: We validated the PrOTYPE assay following the Institute of Medicine guidelines for the development of omics-based tests. This fully defined and locked-down clinical grade assay will enable trial design with molecular subtype stratification and allow for objective assessment of the predictive value of HGSOC molecular subtypes in precision medicine applications. See related commentary by McMullen et al., p. 5271
- Published
- 2020
3. SDH-deficient renal cell carcinoma associated with biallelic mutation in succinate dehydrogenase A: comprehensive genetic profiling and its relation to therapy response
- Author
-
Lisa Koe, Stephen B. Fox, Deme Karikios, Jeffrey D Plew, Huei San Leong, Andrew Fellowes, David Y.H. Choong, Huiling Xu, Christopher R McEvoy, and Owen W.J. Prall
- Subjects
0301 basic medicine ,Biallelic Mutation ,Cancer Research ,Somatic cell ,SDHA ,Context (language use) ,Case Report ,macromolecular substances ,Biology ,medicine.disease_cause ,urologic and male genital diseases ,lcsh:RC254-282 ,Germline ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Genetic testing ,medicine.diagnostic_test ,Cancer ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Carcinogenesis - Abstract
Succinate dehydrogenase (SDH)-deficient renal cell carcinoma (RCC) is a rare RCC subtype that is caused by biallelic mutation of one of the four subunits of the SDH complex (SDHA, B, C, and D) and results in inactivation of the SDH enzyme. Here we describe a case of genetically characterized SDH-deficient RCC caused by biallelic (germline plus somatic) SDHA mutations. SDHA pathogenic variants were detected using comprehensive genomic profiling and SDH absence was subsequently confirmed by immunohistochemistry. Very little is known regarding the genomic context of SDH-deficient RCC. Interestingly we found genomic amplifications commonly observed in RCC but there was an absence of additional variants in common cancer driver genes. Prior to genetic testing a PD-1 inhibitor treatment was administered. However, following the genetic results a succession of tyrosine kinase inhibitors were administered as targeted treatment options and we highlight how the genetic results provide a rationale for their effectiveness. We also describe how the genetic results benefited the patient by empowering him to adopt dietary and lifestyle changes in accordance with knowledge of the mechanisms of SDH-related tumorigenesis.
- Published
- 2018
4. CNspector: a web-based tool for visualisation and clinical diagnosis of copy number variation from next generation sequencing
- Author
-
Jason Li, Georgina L Ryland, Richard Lupat, Jason Ellul, Satwica Yerneni, Andrew Fellowes, Anthony T. Papenfuss, Stephen B. Fox, Ella R. Thompson, Michael Dickinson, Huei San Leong, Amit Kumar, Wasanthi De Silva, John F. Markham, and Piers Blombery
- Subjects
0301 basic medicine ,Source code ,Computer science ,Somatic cell ,lcsh:Medicine ,Web Browser ,medicine.disease_cause ,Genome ,Germline ,Exon ,chemistry.chemical_compound ,0302 clinical medicine ,Chromosome Duplication ,Gene duplication ,Copy-number variation ,lcsh:Science ,media_common ,Mutation ,Multidisciplinary ,medicine.diagnostic_test ,High-Throughput Nucleotide Sequencing ,Basal Cell Nevus Syndrome ,Exons ,Publisher Correction ,Chromosome Arm ,Chromosome Deletion ,DNA Copy Number Variations ,Sequence analysis ,media_common.quotation_subject ,Context (language use) ,Computational biology ,DNA sequencing ,03 medical and health sciences ,Chromosome (genetic algorithm) ,medicine ,Humans ,Web application ,Genetic testing ,Internet ,Genome, Human ,business.industry ,lcsh:R ,Chromosome ,Sequence Analysis, DNA ,030104 developmental biology ,chemistry ,Carcinoma, Basal Cell ,lcsh:Q ,Human genome ,business ,030217 neurology & neurosurgery ,DNA - Abstract
Next Generation Sequencing is now routinely used in the practice of diagnostic pathology to detect clinically relevant somatic and germline sequence variations in patient samples. However, clinical assessment of copy number variations (CNVs) and large-scale structural variations (SVs) is still challenging. While tools exist to estimate both, their results are typically presented separately in tables or static plots which can be difficult to read and are unable to show the context needed for clinical interpretation and reporting. We have addressed this problem with CNspector, a multi-scale interactive browser that shows CNVs in the context of other relevant genomic features to enable fast and effective clinical reporting. We illustrate the utility of CNspector at different genomic scales across a variety of sample types in a range of case studies. We show how CNspector can be used for diagnosis and reporting of exon-level deletions, focal gene-level amplifications, chromosome and chromosome arm level amplifications/deletions and in complex genomic rearrangements. CNspector is a web-based clinical variant browser tailored to the clinical application of next generation sequencing for CNV assessment. We have demonstrated the utility of this interactive software in typical applications across a range of tissue types and disease contexts encountered in the context of diagnostic pathology. CNspector is written in R and the source code is available for download under the GPL3 Licence from https://github.com/PapenfussLab/CNspector.
- Published
- 2019
5. Profound MEK inhibitor response in a cutaneous melanoma harboring a GOLGA4-RAF1 fusion
- Author
-
Judy Browning, Jayesh Desai, Stephen B. Fox, Dariush Etemadmoghadam, David Y.H. Choong, Chloe Khoo, Huiling Xu, Andrew Fellowes, Sophie Beck, Anthony Bell, Huei San Leong, Christopher R McEvoy, Richard W. Tothill, Owen W.J. Prall, Linda Mileshkin, David D.L. Bowtell, Kortnye Smith, Amir Iravani, Bindi M. Bates, David J Byrne, and Violeta Nastevski
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Male ,Skin Neoplasms ,Oncogene Proteins, Fusion ,medicine.medical_treatment ,MAP Kinase Kinase 1 ,Autoantigens ,Targeted therapy ,03 medical and health sciences ,0302 clinical medicine ,CDKN2A ,Fluorodeoxyglucose F18 ,Medicine ,Humans ,Neoplasm Metastasis ,Extracellular Signal-Regulated MAP Kinases ,Melanoma ,Protein Kinase Inhibitors ,Alleles ,Cyclin-Dependent Kinase Inhibitor p16 ,beta Catenin ,Aged ,business.industry ,MEK inhibitor ,Concise Communication ,Cancer ,General Medicine ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Proto-Oncogene Proteins c-raf ,030104 developmental biology ,Tumor progression ,030220 oncology & carcinogenesis ,Positron-Emission Tomography ,Cutaneous melanoma ,Cancer research ,business - Abstract
The serine/threonine kinases BRAF and CRAF are critical components of the MAPK signaling pathway that is activated in many cancer types. In approximately 1% of melanomas, BRAF or CRAF is activated through structural arrangements. We describe a metastatic melanoma with a GOLGA4-RAF1 fusion and pathogenic variants in catenin β 1 (CTNNB1) and cyclin-dependent kinase inhibitor 2A (CDKN2A). Anti–cytotoxic T-lymphocyte–associated protein 4/anti–programmed cell death 1 (anti-CTLA4/anti–PD-1) combination immunotherapy failed to control tumor progression. In the absence of other actionable variants, the patient was administered MEK inhibitor therapy on the basis of its potential action against RAF1 fusions. This resulted in a profound and clinically significant response. We demonstrated that GOLGA4-RAF1 expression was associated with ERK activation, elevated expression of the RAS/RAF downstream coeffector ETV5, and a high Ki67 index. These findings provide a rationale for the dramatic response to targeted therapy. This study shows that molecular characterization of treatment-resistant cancers can identify therapeutic targets and personalize therapy management, leading to improved patient outcomes.
- Published
- 2019
6. Immune response to RB1-regulated senescence limits radiation-induced osteosarcoma formation
- Author
-
Marieke L. Kuijjer, Jason Ellul, Dale W. Garsed, Maya Kansara, Huei San Leong, Sophie Popkiss, Carl R. Walkley, Rod Hicks, Puiyi Pang, Mark J. Smyth, Philip W. Hinds, Carleen Cullinane, Anne-Marie Cleton-Jansen, Nicole M. Haynes, Dan Mei Lin, and David Thomas
- Subjects
Senescence ,Neoplasms, Radiation-Induced ,Tumor suppressor gene ,Bone Neoplasms ,Retinoblastoma Protein ,Mice ,medicine ,Animals ,Humans ,Genes, Retinoblastoma ,Immunologic Surveillance ,Cellular Senescence ,Osteosarcoma ,Osteoblasts ,biology ,Interleukin-6 ,Retinoblastoma ,Calcium Radioisotopes ,Retinoblastoma protein ,General Medicine ,Prognosis ,medicine.disease ,Pediatric cancer ,eye diseases ,Neoplasm Proteins ,Mice, Inbred C57BL ,Immunosurveillance ,Phenotype ,Immunology ,Cancer research ,biology.protein ,Cytokines ,Intercellular Signaling Peptides and Proteins ,Natural Killer T-Cells ,RNA Interference ,Cell aging ,Neoplasm Transplantation ,Research Article - 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
7. Polycomb repressive complex 2 (PRC2) suppresses Eμ-myc lymphoma
- Author
-
Stanley Chun-Wei Lee, Craig D. Hyland, Stephen L. Nutt, Warren S. Alexander, Huei San Leong, Ian J. Majewski, Aaron T. L. Lun, Gordon K. Smyth, Marnie E. Blewitt, Belinda Phipson, Douglas J. Hilton, and Rhys S. Allan
- Subjects
Lymphoma, B-Cell ,Myeloid ,Immunology ,Mice, Transgenic ,macromolecular substances ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Biochemistry ,Proto-Oncogene Proteins c-myc ,Loss of heterozygosity ,Mice ,SUZ12 ,medicine ,Animals ,Enhancer of Zeste Homolog 2 Protein ,Lymphopoiesis ,Cells, Cultured ,B cell ,Polycomb Repressive Complex 1 ,B-Lymphocytes ,Gene knockdown ,biology ,business.industry ,EZH2 ,Polycomb Repressive Complex 2 ,Cell Biology ,Hematology ,Gene Expression Regulation, Neoplastic ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Cancer research ,biology.protein ,business ,PRC2 - Abstract
Deregulation of polycomb group complexes polycomb repressive complex 1 (PRC1) and 2 (PRC2) is associated with human cancers. Although inactivating mutations in PRC2-encoding genes EZH2, EED, and SUZ12 are present in T-cell acute lymphoblastic leukemia and in myeloid malignancies, gain-of-function mutations in EZH2 are frequently observed in B-cell lymphoma, implying disease-dependent effects of individual mutations. We show that, in contrast to PRC1, PRC2 is a tumor suppressor in Eµ-myc lymphomagenesis, because disease onset was accelerated by heterozygosity for Suz12 or by short hairpin RNA-mediated knockdown of Suz12 or Ezh2. Accelerated lymphomagenesis was associated with increased accumulation of B-lymphoid cells in the absence of effects on apoptosis or cell cycling. However, Suz12-deficient B-lymphoid progenitors exhibit enhanced serial clonogenicity. Thus, PRC2 normally restricts the self-renewal of B-lymphoid progenitors, the disruption of which contributes to lymphomagenesis. This finding provides new insight regarding the functional contribution of mutations in PRC2 in a range of leukemias.
- Published
- 2013
8. Epigenetic Regulator Smchd1 Functions as a Tumor Suppressor
- Author
-
Marnie E. Blewitt, Jason Corbin, Huei San Leong, Ian J. Majewski, Yifang Hu, Stanley Chun-Wei Lee, Kelan Chen, James M. Murphy, Warren S. Alexander, Gordon K. Smyth, Miha Pakusch, and Douglas J. Hilton
- Subjects
Male ,Genetically modified mouse ,Cancer Research ,Lymphoma, B-Cell ,Chromosomal Proteins, Non-Histone ,Transgene ,Down-Regulation ,Mice, Nude ,Mice, Transgenic ,Biology ,medicine.disease_cause ,Epigenesis, Genetic ,Gene Knockout Techniques ,Mice ,medicine ,Animals ,Humans ,Genes, Tumor Suppressor ,Fibroblasts ,medicine.disease ,Molecular biology ,Candidate Tumor Suppressor Gene ,Fusion protein ,Gene expression profiling ,Transplantation ,Leukemia ,Cell Transformation, Neoplastic ,Oncology ,Cancer research ,Carcinogenesis - Abstract
SMCHD1 is an epigenetic modifier of gene expression that is critical to maintain X chromosome inactivation. Here, we show in mouse that genetic inactivation of Smchd1 accelerates tumorigenesis in male mice. Loss of Smchd1 in transformed mouse embryonic fibroblasts increased tumor growth upon transplantation into immunodeficient nude mice. In addition, loss of Smchd1 in Eμ-Myc transgenic mice that undergo lymphomagenesis reduced disease latency by 50% relative to control animals. In premalignant Eμ-Myc transgenic mice deficient in Smchd1, there was an increase in the number of pre-B cells in the periphery, likely accounting for the accelerated disease in these animals. Global gene expression profiling suggested that Smchd1 normally represses genes activated by MLL chimeric fusion proteins in leukemia, implying that Smchd1 loss may work through the same pathways as overexpressed MLL fusion proteins do in leukemia and lymphoma. Notably, we found that SMCHD1 is underexpressed in many types of human hematopoietic malignancy. Together, our observations collectively highlight a hitherto uncharacterized role for SMCHD1 as a candidate tumor suppressor gene in hematopoietic cancers. Cancer Res; 73(5); 1591–9. ©2012 AACR.
- Published
- 2013
9. Whole-genome characterization of chemoresistant ovarian cancer
- Author
-
Catherine Kennedy, Peter Bailey, Michael Friedlander, Conrad Leonard, Euan A. Stronach, Jodie Leditschke, Prue A. Cowin, Darrin Taylor, David D.L. Bowtell, Chris Mitchell, Felicity Newell, Senel Idrisoglu, Ravikiran Vedururu, Kathryn Alsop, Ehsan Nourbakhsh, Patricia C. M. O’Brien, Nathan E. Hall, Collin Stewart, Ann-Marie Patch, Linda Mileshkin, Gisela Mir Arnau, Charlotte Wilhelm-Benartzi, Shivashankar H. Nagaraj, Nadia Traficante, Angelika N. Christ, Edward Curry, Qinying Xu, Stephen H. Kazakoff, Emma Markham, Kate Strachan, Timothy J. C. Bruxner, David Miller, Nick Waddell, Yoke Eng Chiew, Karin S. Kassahn, A. Jewell, Barsha Poudel, Ronny Drapkin, Ernst Lengyel, Oliver Holmes, George Au-Yeung, Joshy George, Kelly Quek, Richard W. Tothill, Orla McNally, John V. Pearson, J. Lynn Fink, Greg Young, Nicola Waddell, Elizabeth L. Christie, Jillian Hung, Michael C. J. Quinn, Ivon Harliwong, Jan Pyman, Jason Ellul, Walid J Azar, Katia Nones, Andrew Lonie, Sian Fereday, Craig Nourse, Stephen Cordner, Dariush Etemadmoghadam, Anna deFazio, Paul R. Harnett, Scott Wood, Maria A. Doyle, Michael C.J. Quinn, Robert S. Brown, Hani Gabra, Peter Wilson, Joy Hendley, Timothy P. Holloway, Sean M. Grimmond, Heather Thorne, Matthew J. Anderson, Mark Shackleton, Suzanne Manning, Anne Hamilton, Dale W. Garsed, Huei San Leong, Timothy Semple, and Paul Waring
- Subjects
DNA Mutational Analysis ,Genes, BRCA2 ,Genes, BRCA1 ,Drug resistance ,Biology ,Retinoblastoma Protein ,Germline ,Cohort Studies ,Germline mutation ,Cyclin E ,Genes, Neurofibromatosis 1 ,medicine ,PTEN ,Humans ,ATP Binding Cassette Transporter, Subfamily B, Member 1 ,Promoter Regions, Genetic ,Germ-Line Mutation ,Genetics ,Oncogene Proteins ,Ovarian Neoplasms ,Multidisciplinary ,Genome, Human ,PTEN Phosphohydrolase ,Combination chemotherapy ,DNA Methylation ,medicine.disease ,Cystadenocarcinoma, Serous ,DNA-Binding Proteins ,Serous fluid ,Drug Resistance, Neoplasm ,Mutagenesis ,DNA methylation ,Cancer research ,biology.protein ,Female ,Ovarian cancer - 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
- 2014
10. Retrovirus Mediated Malignant Transformation of Mouse Embryonic Fibroblasts
- Author
-
Huei San Leong and Marnie E. Blewitt
- Subjects
biology ,Chemistry ,Strategy and Management ,Mechanical Engineering ,Cell ,Metals and Alloys ,biology.organism_classification ,Embryonic stem cell ,Industrial and Manufacturing Engineering ,Malignant transformation ,Transplantation ,Small hairpin RNA ,Transformation (genetics) ,Retrovirus ,medicine.anatomical_structure ,Cancer research ,medicine ,HRAS - Abstract
[Abstract] Cellular transformation is a widely used method to artificially induce cells to form tumours in vivo. Here, we describe the methodology for malignant transformation of mouse embryonic fibroblasts (MEFs) for transplantation into immunodeficient nude mice, as used in Leong et al. (2013). The two-step process involves: 1) down-regulation of Trp53 expression using a short hairpin RNA (shRNA); and 2) overexpression of the oncogenic HRas V12 protein. Reduction of Trp53 expression leads to cell immortalisation, and the subsequent overexpression of oncogenic HRas V12 results in malignant transformation of a cell.
- Published
- 2013
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.