11 results on '"Ole Gjoerup"'
Search Results
2. Comprehensive pan-cancer genomic landscape of KRAS altered cancers and real-world outcomes in solid tumors
- Author
-
Jessica K. Lee, Smruthy Sivakumar, Alexa B. Schrock, Russell Madison, David Fabrizio, Ole Gjoerup, Jeffrey S. Ross, Garrett M. Frampton, Pavel Napalkov, Meagan Montesion, Jennifer L. Schutzman, Xin Ye, Priti S. Hegde, Misako Nagasaka, Geoffrey R. Oxnard, Ethan S. Sokol, Sai-Hong Ignatius Ou, and Zhen Shi
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Recent clinical development of KRAS inhibitors has heightened interest in the genomic landscape of KRAS-altered cancers. We performed a pan-cancer analysis of KRAS-altered samples from 426,706 adult patients with solid or hematologic malignancies using comprehensive genomic profiling; additional analyses included 62,369 liquid biopsy and 7241 pediatric samples. 23% of adult pan-cancer samples had KRAS alterations; 88% were mutations, most commonly G12D/G12V/G12C/G13D/G12R, and prevalence was similar in liquid biopsies. Co-alteration landscapes were largely similar across KRAS mutations but distinct from KRAS wild-type, though differences were observed in some tumor types for tumor mutational burden, PD-L1 expression, microsatellite instability, and other mutational signatures. Prognosis of KRAS-mutant versus other genomic cohorts of lung, pancreatic, and colorectal cancer were assessed using a real-world clinicogenomic database. As specific KRAS inhibitors and combination therapeutic strategies are being developed, genomic profiling to understand co-alterations and other biomarkers that may modulate response to targeted or immunotherapies will be imperative.
- Published
- 2022
- Full Text
- View/download PDF
3. Clinical and analytical validation of FoundationOne®CDx, a comprehensive genomic profiling assay for solid tumors
- Author
-
Coren A. Milbury, James Creeden, Wai-Ki Yip, David L. Smith, Varun Pattani, Kristi Maxwell, Bethany Sawchyn, Ole Gjoerup, Wei Meng, Joel Skoletsky, Alvin D. Concepcion, Yanhua Tang, Xiaobo Bai, Ninad Dewal, Pei Ma, Shannon T. Bailey, James Thornton, Dean C. Pavlick, Garrett M. Frampton, Daniel Lieber, Jared White, Christine Burns, and Christine Vietz
- Subjects
Medicine ,Science - Abstract
FoundationOne®CDx (F1CDx) is a United States (US) Food and Drug Administration (FDA)-approved companion diagnostic test to identify patients who may benefit from treatment in accordance with the approved therapeutic product labeling for 28 drug therapies. F1CDx utilizes next-generation sequencing (NGS)-based comprehensive genomic profiling (CGP) technology to examine 324 cancer genes in solid tumors. F1CDx reports known and likely pathogenic short variants (SVs), copy number alterations (CNAs), and select rearrangements, as well as complex biomarkers including tumor mutational burden (TMB) and microsatellite instability (MSI), in addition to genomic loss of heterozygosity (gLOH) in ovarian cancer. CGP services can reduce the complexity of biomarker testing, enabling precision medicine to improve treatment decision-making and outcomes for cancer patients, but only if test results are reliable, accurate, and validated clinically and analytically to the highest standard available. The analyses presented herein demonstrate the extensive analytical and clinical validation supporting the F1CDx initial and subsequent FDA approvals to ensure high sensitivity, specificity, and reliability of the data reported. The analytical validation included several in-depth evaluations of F1CDx assay performance including limit of detection (LoD), limit of blank (LoB), precision, and orthogonal concordance for SVs (including base substitutions [SUBs] and insertions/deletions [INDELs]), CNAs (including amplifications and homozygous deletions), genomic rearrangements, and select complex biomarkers. The assay validation of >30,000 test results comprises a considerable and increasing body of evidence that supports the clinical utility of F1CDx to match patients with solid tumors to targeted therapies or immunotherapies based on their tumor’s genomic alterations and biomarkers. F1CDx meets the clinical needs of providers and patients to receive guideline-based biomarker testing, helping them keep pace with a rapidly evolving field of medicine.
- Published
- 2022
4. Clinical and analytical validation of FoundationOne®CDx, a comprehensive genomic profiling assay for solid tumors.
- Author
-
Coren A Milbury, James Creeden, Wai-Ki Yip, David L Smith, Varun Pattani, Kristi Maxwell, Bethany Sawchyn, Ole Gjoerup, Wei Meng, Joel Skoletsky, Alvin D Concepcion, Yanhua Tang, Xiaobo Bai, Ninad Dewal, Pei Ma, Shannon T Bailey, James Thornton, Dean C Pavlick, Garrett M Frampton, Daniel Lieber, Jared White, Christine Burns, and Christine Vietz
- Subjects
Medicine ,Science - Abstract
FoundationOne®CDx (F1CDx) is a United States (US) Food and Drug Administration (FDA)-approved companion diagnostic test to identify patients who may benefit from treatment in accordance with the approved therapeutic product labeling for 28 drug therapies. F1CDx utilizes next-generation sequencing (NGS)-based comprehensive genomic profiling (CGP) technology to examine 324 cancer genes in solid tumors. F1CDx reports known and likely pathogenic short variants (SVs), copy number alterations (CNAs), and select rearrangements, as well as complex biomarkers including tumor mutational burden (TMB) and microsatellite instability (MSI), in addition to genomic loss of heterozygosity (gLOH) in ovarian cancer. CGP services can reduce the complexity of biomarker testing, enabling precision medicine to improve treatment decision-making and outcomes for cancer patients, but only if test results are reliable, accurate, and validated clinically and analytically to the highest standard available. The analyses presented herein demonstrate the extensive analytical and clinical validation supporting the F1CDx initial and subsequent FDA approvals to ensure high sensitivity, specificity, and reliability of the data reported. The analytical validation included several in-depth evaluations of F1CDx assay performance including limit of detection (LoD), limit of blank (LoB), precision, and orthogonal concordance for SVs (including base substitutions [SUBs] and insertions/deletions [INDELs]), CNAs (including amplifications and homozygous deletions), genomic rearrangements, and select complex biomarkers. The assay validation of >30,000 test results comprises a considerable and increasing body of evidence that supports the clinical utility of F1CDx to match patients with solid tumors to targeted therapies or immunotherapies based on their tumor's genomic alterations and biomarkers. F1CDx meets the clinical needs of providers and patients to receive guideline-based biomarker testing, helping them keep pace with a rapidly evolving field of medicine.
- Published
- 2022
- Full Text
- View/download PDF
5. SMAD4 represses FOSL1 expression and pancreatic cancer metastatic colonization
- Author
-
Chao Dai, Jonathan P. Rennhack, Taylor E. Arnoff, Maneesha Thaker, Scott T. Younger, John G. Doench, August Yue Huang, Annan Yang, Andrew J. Aguirre, Belinda Wang, Evan Mun, Joyce T. O’Connell, Ying Huang, Katherine Labella, Jessica A. Talamas, Ji Li, Nina Ilic, Justin Hwang, Andrew L. Hong, Andrew O. Giacomelli, Ole Gjoerup, David E. Root, and William C. Hahn
- Subjects
PDAC ,metastasis ,colonization ,FOSL1 ,SMAD4 ,Biology (General) ,QH301-705.5 - Abstract
Summary: Metastasis is a complex and poorly understood process. In pancreatic cancer, loss of the transforming growth factor (TGF)-β/BMP effector SMAD4 is correlated with changes in altered histopathological transitions, metastatic disease, and poor prognosis. In this study, we use isogenic cancer cell lines to identify SMAD4 regulated genes that contribute to the development of metastatic colonization. We perform an in vivo screen identifying FOSL1 as both a SMAD4 target and sufficient to drive colonization to the lung. The targeting of these genes early in treatment may provide a therapeutic benefit.
- Published
- 2021
- Full Text
- View/download PDF
6. STRIPAK directs PP2A activity toward MAP4K4 to promote oncogenic transformation of human cells
- Author
-
Jong Wook Kim, Christian Berrios, Miju Kim, Amy E Schade, Guillaume Adelmant, Huwate Yeerna, Emily Damato, Amanda Balboni Iniguez, Laurence Florens, Michael P Washburn, Kim Stegmaier, Nathanael S Gray, Pablo Tamayo, Ole Gjoerup, Jarrod A Marto, James DeCaprio, and William C Hahn
- Subjects
PP2A ,STRIPAK ,small t ,transformation ,MAP4K4 ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Alterations involving serine-threonine phosphatase PP2A subunits occur in a range of human cancers, and partial loss of PP2A function contributes to cell transformation. Displacement of regulatory B subunits by the SV40 Small T antigen (ST) or mutation/deletion of PP2A subunits alters the abundance and types of PP2A complexes in cells, leading to transformation. Here, we show that ST not only displaces common PP2A B subunits but also promotes A-C subunit interactions with alternative B subunits (B’’’, striatins) that are components of the Striatin-interacting phosphatase and kinase (STRIPAK) complex. We found that STRN4, a member of STRIPAK, is associated with ST and is required for ST-PP2A-induced cell transformation. ST recruitment of STRIPAK facilitates PP2A-mediated dephosphorylation of MAP4K4 and induces cell transformation through the activation of the Hippo pathway effector YAP1. These observations identify an unanticipated role of MAP4K4 in transformation and show that the STRIPAK complex regulates PP2A specificity and activity.
- Published
- 2020
- Full Text
- View/download PDF
7. CREB5 Promotes Resistance to Androgen-Receptor Antagonists and Androgen Deprivation in Prostate Cancer
- Author
-
Justin H. Hwang, Ji-Heui Seo, Michael L. Beshiri, Stephanie Wankowicz, David Liu, Alexander Cheung, Ji Li, Xintao Qiu, Andrew L. Hong, Ginevra Botta, Lior Golumb, Camden Richter, Jonathan So, Gabriel J. Sandoval, Andrew O. Giacomelli, Seav Huong Ly, Celine Han, Chao Dai, Hubert Pakula, Anjali Sheahan, Federica Piccioni, Ole Gjoerup, Massimo Loda, Adam G. Sowalsky, Leigh Ellis, Henry Long, David E. Root, Kathleen Kelly, Eliezer M. Van Allen, Matthew L. Freedman, Atish D. Choudhury, and William C. Hahn
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Androgen-receptor (AR) inhibitors, including enzalutamide, are used for treatment of all metastatic castration-resistant prostate cancers (mCRPCs). However, some patients develop resistance or never respond. We find that the transcription factor CREB5 confers enzalutamide resistance in an open reading frame (ORF) expression screen and in tumor xenografts. CREB5 overexpression is essential for an enzalutamide-resistant patient-derived organoid. In AR-expressing prostate cancer cells, CREB5 interactions enhance AR activity at a subset of promoters and enhancers upon enzalutamide treatment, including MYC and genes involved in the cell cycle. In mCRPC, we found recurrent amplification and overexpression of CREB5. Our observations identify CREB5 as one mechanism that drives resistance to AR antagonists in prostate cancers. : Advanced prostate cancers develop resistance to androgen receptor (AR)-targeting therapies. Hwang et al. show that resistant prostate cancers overexpress or amplify CREB5, mediating resistance to AR inhibition. CREB5 suppression reduces the viability of therapy-resistant patient-derived models, suggesting that CREB5 is a target in prostate cancer patients with limited treatment options. Keywords: metastatic castration-resistant prostate cancer, androgen deprivation therapy, therapy resistance, CREB5, androgen receptor, ORF screen
- Published
- 2019
- Full Text
- View/download PDF
8. Renal medullary carcinomas depend upon SMARCB1 loss and are sensitive to proteasome inhibition
- Author
-
Andrew L Hong, Yuen-Yi Tseng, Jeremiah A Wala, Won-Jun Kim, Bryan D Kynnap, Mihir B Doshi, Guillaume Kugener, Gabriel J Sandoval, Thomas P Howard, Ji Li, Xiaoping Yang, Michelle Tillgren, Mahmhoud Ghandi, Abeer Sayeed, Rebecca Deasy, Abigail Ward, Brian McSteen, Katherine M Labella, Paula Keskula, Adam Tracy, Cora Connor, Catherine M Clinton, Alanna J Church, Brian D Crompton, Katherine A Janeway, Barbara Van Hare, David Sandak, Ole Gjoerup, Pratiti Bandopadhayay, Paul A Clemons, Stuart L Schreiber, David E Root, Prafulla C Gokhale, Susan N Chi, Elizabeth A Mullen, Charles WM Roberts, Cigall Kadoch, Rameen Beroukhim, Keith L Ligon, Jesse S Boehm, and William C Hahn
- Subjects
renal medullary carcinoma ,SMARCB1 ,MLN2238 ,ubiquitin-proteasome system ,cell cycle ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Renal medullary carcinoma (RMC) is a rare and deadly kidney cancer in patients of African descent with sickle cell trait. We have developed faithful patient-derived RMC models and using whole-genome sequencing, we identified loss-of-function intronic fusion events in one SMARCB1 allele with concurrent loss of the other allele. Biochemical and functional characterization of these models revealed that RMC requires the loss of SMARCB1 for survival. Through integration of RNAi and CRISPR-Cas9 loss-of-function genetic screens and a small-molecule screen, we found that the ubiquitin-proteasome system (UPS) was essential in RMC. Inhibition of the UPS caused a G2/M arrest due to constitutive accumulation of cyclin B1. These observations extend across cancers that harbor SMARCB1 loss, which also require expression of the E2 ubiquitin-conjugating enzyme, UBE2C. Our studies identify a synthetic lethal relationship between SMARCB1-deficient cancers and reliance on the UPS which provides the foundation for a mechanism-informed clinical trial with proteasome inhibitors.
- Published
- 2019
- Full Text
- View/download PDF
9. An alternative splicing switch in FLNB promotes the mesenchymal cell state in human breast cancer
- Author
-
Ji Li, Peter S Choi, Christine L Chaffer, Katherine Labella, Justin H Hwang, Andrew O Giacomelli, Jong Wook Kim, Nina Ilic, John G Doench, Seav Huong Ly, Chao Dai, Kimberly Hagel, Andrew L Hong, Ole Gjoerup, Shom Goel, Jennifer Y Ge, David E Root, Jean J Zhao, Angela N Brooks, Robert A Weinberg, and William C Hahn
- Subjects
alternative splicing ,QKI ,RBFOX1 ,FLNB ,EMT ,basal-like breast cancer ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Alternative splicing of mRNA precursors represents a key gene expression regulatory step and permits the generation of distinct protein products with diverse functions. In a genome-scale expression screen for inducers of the epithelial-to-mesenchymal transition (EMT), we found a striking enrichment of RNA-binding proteins. We validated that QKI and RBFOX1 were necessary and sufficient to induce an intermediate mesenchymal cell state and increased tumorigenicity. Using RNA-seq and eCLIP analysis, we found that QKI and RBFOX1 coordinately regulated the splicing and function of the actin-binding protein FLNB, which plays a causal role in the regulation of EMT. Specifically, the skipping of FLNB exon 30 induced EMT by releasing the FOXC1 transcription factor. Moreover, skipping of FLNB exon 30 is strongly associated with EMT gene signatures in basal-like breast cancer patient samples. These observations identify a specific dysregulation of splicing, which regulates tumor cell plasticity and is frequently observed in human cancer.
- Published
- 2018
- Full Text
- View/download PDF
10. Viral interference with DNA repair by targeting of the single-stranded DNA binding protein RPA.
- Author
-
Pubali Banerjee, Rowena DeJesus, Ole Gjoerup, and Brian S Schaffhausen
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Correct repair of damaged DNA is critical for genomic integrity. Deficiencies in DNA repair are linked with human cancer. Here we report a novel mechanism by which a virus manipulates DNA damage responses. Infection with murine polyomavirus sensitizes cells to DNA damage by UV and etoposide. Polyomavirus large T antigen (LT) alone is sufficient to sensitize cells 100 fold to UV and other kinds of DNA damage. This results in activated stress responses and apoptosis. Genetic analysis shows that LT sensitizes via the binding of its origin-binding domain (OBD) to the single-stranded DNA binding protein replication protein A (RPA). Overexpression of RPA protects cells expressing OBD from damage, and knockdown of RPA mimics the LT phenotype. LT prevents recruitment of RPA to nuclear foci after DNA damage. This leads to failure to recruit repair proteins such as Rad51 or Rad9, explaining why LT prevents repair of double strand DNA breaks by homologous recombination. A targeted intervention directed at RPA based on this viral mechanism could be useful in circumventing the resistance of cancer cells to therapy.
- Published
- 2013
- Full Text
- View/download PDF
11. Cellular and viral factors regulating Merkel cell polyomavirus replication.
- Author
-
Huichen Feng, Hyun Jin Kwun, Xi Liu, Ole Gjoerup, Donna B Stolz, Yuan Chang, and Patrick S Moore
- Subjects
Medicine ,Science - Abstract
Merkel cell polyomavirus (MCV), a previously unrecognized component of the human viral skin flora, was discovered as a mutated and clonally-integrated virus inserted into Merkel cell carcinoma (MCC) genomes. We reconstructed a replicating MCV clone (MCV-HF), and then mutated viral sites required for replication or interaction with cellular proteins to examine replication efficiency and viral gene expression. Three days after MCV-HF transfection into 293 cells, although replication is not robust, encapsidated viral DNA and protein can be readily isolated by density gradient centrifugation and typical ∼40 nm diameter polyomavirus virions are identified by electron microscopy. The virus has an orderly gene expression cascade during replication in which large T (LT) and 57kT proteins are first expressed by day 2, followed by expression of small T (sT) and VP1 proteins. VP1 and sT proteins are not detected, and spliced 57kT is markedly diminished, in the replication-defective virus suggesting that early gene splicing and late gene transcription may be dependent on viral DNA replication. MCV replication and encapsidation is increased by overexpression of MCV sT, consistent with sT being a limiting factor during virus replication. Mutation of the MCV LT vacuolar sorting protein hVam6p (Vps39) binding site also enhances MCV replication while exogenous hVam6p overexpression reduces MCV virion production by >90%. Although MCV-HF generates encapsidated wild-type MCV virions, we did not find conditions for persistent transmission to recipient cell lines suggesting that MCV has a highly restricted tropism. These studies identify and highlight the role of polyomavirus DNA replication in viral gene expression and show that viral sT and cellular hVam6p are important factors regulating MCV replication. MCV-HF is a molecular clone that can be readily manipulated to investigate factors affecting MCV replication.
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.