1
|
Chen JF, Guo SJ, He B, Zheng W, Jiang WJ, Yuan Z, Xiang Y, Peng C, Xiong W, Shi JY. Advances of dual inhibitors based on ALK for the treatment of cancer. Bioorg Chem 2025; 159:108417. [PMID: 40168884 DOI: 10.1016/j.bioorg.2025.108417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/17/2025] [Accepted: 03/25/2025] [Indexed: 04/03/2025]
Abstract
Anaplastic lymphoma kinase (ALK), which encodes a highly conserved receptor tyrosine kinase (RTK), is important for the development and progression of many tumors, especially non-small cell lung cancer (NSCLC). Currently, third-generation ALK inhibitors are used to treat ALK-mutant NSCLC, but the rapid emergence of resistance during treatment greatly limits their efficacy in clinic. In comparison to single-target inhibitors, ALK dual inhibitors offer the benefits of reducing the emergence of drug resistance, improving treatment efficacy, and optimizing pharmacokinetic features due to the synergistic function of ALK and other associated targets involved in tumor progression. Therefore, we outline the development of ALK dual inhibitors, highlight their design approaches and structure-activity relationship (SAR), and offer insights into new challenges and potential future directions in this area.
Collapse
Affiliation(s)
- Jin-Feng Chen
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731. China; Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Shu-Jin Guo
- Department of Health Management Center, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Bin He
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Wei Zheng
- Department of Integrative Medicine, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Wen-Jie Jiang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Zhuo Yuan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yu Xiang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Cheng Peng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; State Key Laboratory of Southwestern Chinese Medicine Resources, School of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Wei Xiong
- Department of urology, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Chengdu 610072, China.
| | - Jian-You Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China.
| |
Collapse
|
2
|
Jiang A, Li H, Li D, Chen H, Zhao L, Zhang Y, Li Y, Rong R, Li B, Xiao S. A Novel ACVR2A::RAF1 Fusion in Spindle Cell Sarcoma. Genes Chromosomes Cancer 2025; 64:e70033. [PMID: 39950347 DOI: 10.1002/gcc.70033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 02/02/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Kinase-rearranged spindle cell sarcomas are characterized by unique molecular features. The advent of next-generation sequencing (NGS) has enabled the detection of a multitude of kinase fusions, thereby contributing to the accurate categorization of these tumors. CASE PRESENTATION A 37-year-old woman experienced the fourth recurrence of a cranial base tumor 25 years following the initial surgery and radiation therapy. Histological analysis disclosed spindle-shaped and oval tumor cells, along with a high number of mitotic figures. Immunohistochemistry showed a null immunophenotype, negative for pan-TRK, S-100, CD34, pan-CK, GFAP, and Olig2. Molecular analysis of the tumor tissue identified a novel ACVR2A::RAF1 fusion, comprising the first four exons of ACVR2A and the last nine exons of RAF1. The resulting fusion protein retains the extracellular and transmembrane domains of ACVR2A, while its kinase domain is replaced by the kinase domain of RAF1. This hybrid protein likely contributes to tumorigenesis by activating RAF1 signaling in response to ACVR2A ligands from the TGF-β superfamily. TREATMENT AND OUTCOME The patient was treated with the MEK1 inhibitor Trametinib, 2 mg per time and once a day. One month later, MRI showed significant tumor shrinkage and pain relief. CONCLUSION The ACVR2A::RAF1 fusion represents a novel genomic profile in RAF1-rearranged spindle cell sarcoma, offering a rational basis for targeted therapy. This case highlights the importance of molecular diagnostics in identifying actionable targets and guiding treatment, potentially leading to significant clinical benefits.
Collapse
Affiliation(s)
- Anfeng Jiang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Huan Li
- Advanced Molecular Pathology Institute of Soochow University and SANO, Suzhou, China
- Suzhou Sano Precision Medicine Ltd, Suzhou, China
| | - Dongbing Li
- Advanced Molecular Pathology Institute of Soochow University and SANO, Suzhou, China
- Suzhou Sano Precision Medicine Ltd, Suzhou, China
| | - Huafei Chen
- Advanced Molecular Pathology Institute of Soochow University and SANO, Suzhou, China
- Suzhou Sano Precision Medicine Ltd, Suzhou, China
| | - Lina Zhao
- Advanced Molecular Pathology Institute of Soochow University and SANO, Suzhou, China
- Suzhou Sano Precision Medicine Ltd, Suzhou, China
| | - Ying Zhang
- Advanced Molecular Pathology Institute of Soochow University and SANO, Suzhou, China
- Suzhou Sano Precision Medicine Ltd, Suzhou, China
| | - Yangyang Li
- Advanced Molecular Pathology Institute of Soochow University and SANO, Suzhou, China
- Suzhou Sano Precision Medicine Ltd, Suzhou, China
| | - Rong Rong
- Department of Biological Sciences, Xi An Jiaotong-Liverpool University, Suzhou, China
| | - Bin Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Sheng Xiao
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
3
|
Rusidzé M, Poumeaud F, Akiki B, Valentin T, Ferron G, Ducassou A, Pissaloux D, Evrard S, Brousset P, Le Guellec S, Rochaix P. EWSR1::CREM rearranged intra-abdominal malignant epithelioid neoplasm: two new cases of an emerging entity with clinicopathological characteristics and histological pitfalls. Virchows Arch 2025:10.1007/s00428-025-04034-4. [PMID: 39888444 DOI: 10.1007/s00428-025-04034-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 01/14/2025] [Accepted: 01/15/2025] [Indexed: 02/01/2025]
Abstract
The EWSR1::CREM rearranged intra-abdominal malignant epithelioid neoplasm is an emerging tumor, with only a few publications describing it to date. Here, we report two new cases of this highly aggressive tumor, primarily involving the peritoneal surface. The tumors presented as a widespread diffuse peritoneal lesion associated with a 4-cm pelvic mass in a 28-year-old woman (Case 1) and as a 10-cm intra-abdominal mass infiltrating the stomach with multiple hepatic metastases in a 53-year-old woman (Case 2). The tumors shared predominant epithelioid morphology with minimal nuclear polymorphism. One of them additionally harbored spindle and rhabdoid cell populations. Both tumors displayed immunoreactivity for pan-cytokeratins, EMA, and CD99, and variable positivity for MUC4, progesterone and estrogen receptors, pan-NTRK, and synaptophysin. This misleading histology and immunophenotype give rise to a wide spectrum of differential diagnoses and highlight the crucial role of RNA sequencing in diagnostic accuracy and thus in appropriate therapeutic approaches.
Collapse
Affiliation(s)
- Mariam Rusidzé
- Department of Pathology, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse, 1 Av. Irène Joliot-Curie, 31100, Toulouse, France.
| | - François Poumeaud
- Department of Medical Oncology, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse, 1 Av. Irène Joliot-Curie, 31100, Toulouse, France
| | - Béatrice Akiki
- Department of Pathology, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse, 1 Av. Irène Joliot-Curie, 31100, Toulouse, France
| | - Thibaud Valentin
- Department of Medical Oncology, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse, 1 Av. Irène Joliot-Curie, 31100, Toulouse, France
| | - Gwenaël Ferron
- Department of Surgical Oncology, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse, 1 Av. Irène Joliot-Curie, 31100, Toulouse, France
| | - Anne Ducassou
- Department of Radiotherapy, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse, 1 Av. Irène Joliot-Curie, 31100, Toulouse, France
| | - Daniel Pissaloux
- Department of Biopathology, Unicancer, Léon Bérard Center, 28 Promenade Léa Et Napoléon Bullukian, 69008, Lyon, France
| | - Solène Evrard
- Department of Pathology, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse, 1 Av. Irène Joliot-Curie, 31100, Toulouse, France
| | - Pierre Brousset
- Department of Pathology, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse, 1 Av. Irène Joliot-Curie, 31100, Toulouse, France
| | - Sophie Le Guellec
- Medipath, Les Feuillants, 116 Route d'Espagne, Helios 3 BatD, 31100, Toulouse, France
| | - Philippe Rochaix
- Department of Pathology, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse, 1 Av. Irène Joliot-Curie, 31100, Toulouse, France
| |
Collapse
|
4
|
Cornish AJ, Gruber AJ, Kinnersley B, Chubb D, Frangou A, Caravagna G, Noyvert B, Lakatos E, Wood HM, Thorn S, Culliford R, Arnedo-Pac C, Househam J, Cross W, Sud A, Law P, Leathlobhair MN, Hawari A, Woolley C, Sherwood K, Feeley N, Gül G, Fernandez-Tajes J, Zapata L, Alexandrov LB, Murugaesu N, Sosinsky A, Mitchell J, Lopez-Bigas N, Quirke P, Church DN, Tomlinson IPM, Sottoriva A, Graham TA, Wedge DC, Houlston RS. The genomic landscape of 2,023 colorectal cancers. Nature 2024; 633:127-136. [PMID: 39112709 PMCID: PMC11374690 DOI: 10.1038/s41586-024-07747-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 06/24/2024] [Indexed: 08/17/2024]
Abstract
Colorectal carcinoma (CRC) is a common cause of mortality1, but a comprehensive description of its genomic landscape is lacking2-9. Here we perform whole-genome sequencing of 2,023 CRC samples from participants in the UK 100,000 Genomes Project, thereby providing a highly detailed somatic mutational landscape of this cancer. Integrated analyses identify more than 250 putative CRC driver genes, many not previously implicated in CRC or other cancers, including several recurrent changes outside the coding genome. We extend the molecular pathways involved in CRC development, define four new common subgroups of microsatellite-stable CRC based on genomic features and show that these groups have independent prognostic associations. We also characterize several rare molecular CRC subgroups, some with potential clinical relevance, including cancers with both microsatellite and chromosomal instability. We demonstrate a spectrum of mutational profiles across the colorectum, which reflect aetiological differences. These include the role of Escherichia colipks+ colibactin in rectal cancers10 and the importance of the SBS93 signature11-13, which suggests that diet or smoking is a risk factor. Immune-escape driver mutations14 are near-ubiquitous in hypermutant tumours and occur in about half of microsatellite-stable CRCs, often in the form of HLA copy number changes. Many driver mutations are actionable, including those associated with rare subgroups (for example, BRCA1 and IDH1), highlighting the role of whole-genome sequencing in optimizing patient care.
Collapse
Affiliation(s)
- Alex J Cornish
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - Andreas J Gruber
- Department of Biology, University of Konstanz, Konstanz, Germany
- Manchester Cancer Research Centre, Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Ben Kinnersley
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
- University College London Cancer Institute, London, UK
| | - Daniel Chubb
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - Anna Frangou
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany
| | - Giulio Caravagna
- Department of Mathematics and Geosciences, University of Trieste, Trieste, Italy
- Centre for Evolution and Cancer, Institute of Cancer Research, London, UK
| | - Boris Noyvert
- Cancer Research UK Centre and Centre for Computational Biology, Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Eszter Lakatos
- Centre for Evolution and Cancer, Institute of Cancer Research, London, UK
- Department of Mathematical Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Henry M Wood
- Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Steve Thorn
- Department of Oncology, University of Oxford, Oxford, UK
| | - Richard Culliford
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - Claudia Arnedo-Pac
- Institute for Research in Biomedicine Barcelona, The Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Jacob Househam
- Centre for Evolution and Cancer, Institute of Cancer Research, London, UK
| | - William Cross
- Centre for Evolution and Cancer, Institute of Cancer Research, London, UK
- Research Department of Pathology, University College London, UCL Cancer Institute, London, UK
| | - Amit Sud
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - Philip Law
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | | | - Aliah Hawari
- Manchester Cancer Research Centre, Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Connor Woolley
- Department of Oncology, University of Oxford, Oxford, UK
| | - Kitty Sherwood
- Department of Oncology, University of Oxford, Oxford, UK
- Edinburgh Cancer Research, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Nathalie Feeley
- Department of Oncology, University of Oxford, Oxford, UK
- Edinburgh Cancer Research, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Güler Gül
- Edinburgh Cancer Research, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | | | - Luis Zapata
- Centre for Evolution and Cancer, Institute of Cancer Research, London, UK
| | - Ludmil B Alexandrov
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA, USA
- Department of Bioengineering, UC San Diego, La Jolla, CA, USA
- Moores Cancer Center, UC San Diego, La Jolla, CA, USA
| | - Nirupa Murugaesu
- Genomics England, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Alona Sosinsky
- Genomics England, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Jonathan Mitchell
- Genomics England, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Nuria Lopez-Bigas
- Institute for Research in Biomedicine Barcelona, The Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Philip Quirke
- Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - David N Church
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford NIHR Comprehensive Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | | | - Andrea Sottoriva
- Centre for Evolution and Cancer, Institute of Cancer Research, London, UK
- Computational Biology Research Centre, Human Technopole, Milan, Italy
| | - Trevor A Graham
- Centre for Evolution and Cancer, Institute of Cancer Research, London, UK
| | - David C Wedge
- Manchester Cancer Research Centre, Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Richard S Houlston
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| |
Collapse
|
5
|
Renna FJ, Gonzalez CD, Vaccaro MI. Decoding the Versatile Landscape of Autophagic Protein VMP1 in Cancer: A Comprehensive Review across Tissue Types and Regulatory Mechanisms. Int J Mol Sci 2024; 25:3758. [PMID: 38612567 PMCID: PMC11011780 DOI: 10.3390/ijms25073758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
Autophagy, a catabolic process orchestrating the degradation of proteins and organelles within lysosomes, is pivotal for maintaining cellular homeostasis. However, its dual role in cancer involves preventing malignant transformation while fostering progression and therapy resistance. Vacuole Membrane Protein 1 (VMP1) is an essential autophagic protein whose expression, per se, triggers autophagy, being present in the whole autophagic flux. In pancreatic cancer, VMP1-whose expression is linked to the Kirsten Rat Sarcoma Virus (KRAS) oncogene-significantly contributes to disease promotion, progression, and chemotherapy resistance. This investigation extends to breast cancer, colon cancer, hepatocellular carcinoma, and more, highlighting VMP1's nuanced nature, contingent on specific tissue contexts. The examination of VMP1's interactions with micro-ribonucleic acids (miRNAs), including miR-21, miR-210, and miR-124, enhances our understanding of its regulatory network in cancer. Additionally, this article discusses VMP1 gene fusions, especially with ribosomal protein S6 kinase B1 (RPS6KB1), shedding light on potential implications for tumor malignancy. By deciphering the molecular mechanisms linking VMP1 to cancer progression, this exploration paves the way for innovative therapeutic strategies to disrupt these pathways and potentially improve treatment outcomes.
Collapse
Affiliation(s)
- Felipe J. Renna
- Instituto de Bioquimica y Medicina Molecular Prof Alberto Boveris (IBIMOL), CONICET, Universidad de Buenos Aires, Buenos Aires C1113AAC, Argentina;
| | - Claudio D. Gonzalez
- Instituto de Investigaciones, IUC, Medicina Traslacional, Hospital Universitario CEMIC, Buenos Aires C1431FWN, Argentina;
| | - Maria I. Vaccaro
- Instituto de Bioquimica y Medicina Molecular Prof Alberto Boveris (IBIMOL), CONICET, Universidad de Buenos Aires, Buenos Aires C1113AAC, Argentina;
- Instituto de Investigaciones, IUC, Medicina Traslacional, Hospital Universitario CEMIC, Buenos Aires C1431FWN, Argentina;
| |
Collapse
|
6
|
Li P, Busam K. Compound clear cell sarcoma with EWSR1::CREM fusion. J Cutan Pathol 2023; 50:1065-1069. [PMID: 36640048 DOI: 10.1111/cup.14390] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 01/01/2023] [Accepted: 01/10/2023] [Indexed: 01/15/2023]
Abstract
Cutaneous clear cell sarcomas may be confused with melanomas as a result of overlapping histopathology and immunohistochemical staining. We report a case of a 41-year-old woman with a purported history of acral melanoma of the great toe. Twenty-one months after excision of the primary tumor, the patient developed a groin mass, diagnosed as metastatic melanoma on excision. Five months later, a biopsy of a lung mass was reported as metastatic melanoma. The patient was referred to our institution for treatment, which prompted molecular testing on the groin metastasis by targeted next-generation sequencing. Molecular testing results revealed TP53 and TERT promoter mutations and the absence of BRAF, KRAS, and KIT mutations; it also revealed an EWSR1::CREM fusion that was confirmed by Archer FusionPlex. The alleged acral melanoma was re-reviewed, showing an invasive amelanotic spindle cell neoplasm in the dermis with neoplastic nests at the dermal-epidermal junction; the tumor cells expressed markers of melanocytic differentiation but were negative for PRAME and BRAF immunohistochemical staining. Molecular testing of the toe and lung metastasis revealed the same EWSR1::CREM fusion. In light of the molecular findings, the diagnosis was revised to a primary acral compound clear cell sarcoma with EWSR1::CREM fusion.
Collapse
Affiliation(s)
- Philippa Li
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Klaus Busam
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
7
|
López-Palacios TP, Andersen JL. Kinase regulation by liquid-liquid phase separation. Trends Cell Biol 2023; 33:649-666. [PMID: 36528418 PMCID: PMC10267292 DOI: 10.1016/j.tcb.2022.11.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 12/23/2022]
Abstract
Liquid-liquid phase separation (LLPS) is emerging as a mechanism of spatiotemporal regulation that could answer long-standing questions about how order is achieved in biochemical signaling. In this review we discuss how LLPS orchestrates kinase signaling, either by creating condensate structures that are sensed by kinases or by direct LLPS of kinases, cofactors, and substrates - thereby acting as a mechanism to compartmentalize kinase-substrate relationships, and in some cases also sequestering the kinase away from inhibitory factors. We also examine the possibility that selective pressure promotes genomic rearrangements that fuse pro-growth kinases to LLPS-prone protein sequences, which in turn drives aberrant kinase activation through LLPS.
Collapse
Affiliation(s)
- Tania P López-Palacios
- Fritz B. Burns Cancer Research Laboratory, Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA
| | - Joshua L Andersen
- Fritz B. Burns Cancer Research Laboratory, Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT, USA.
| |
Collapse
|
8
|
Grenier K, Rivière JB, Bencheikh BOA, Corredor ALG, Shieh BC, Wang H, Fiset PO, Camilleri-Broët S. Routine Clinically Detected Increased ROS1 Transcripts Are Related With ROS1 Expression by Immunohistochemistry and Associated With EGFR Mutations in Lung Adenocarcinoma. JTO Clin Res Rep 2023; 4:100530. [PMID: 37415647 PMCID: PMC10320302 DOI: 10.1016/j.jtocrr.2023.100530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 05/11/2023] [Accepted: 05/16/2023] [Indexed: 07/08/2023] Open
Abstract
Introduction Translocations of the ROS1 gene were found to drive tumorigenesis in 1% to 2% of lung adenocarcinoma. In clinical practice, ROS1 rearrangements are often screened by immunohistochemistry (IHC) before confirmation with either fluorescence in situ hybridization or molecular techniques. This screening test leads to a non-negligible number of cases that have equivocal or positive ROS1 IHC, without ROS1 translocation. Methods In this study, we have analyzed retrospectively 1021 cases of nonsquamous NSCLC having both ROS1 IHC and molecular analysis using next-generation sequencing. Results ROS1 IHC was negative in 938 cases (91.9%), equivocal in 65 cases (6.4%), and positive in 18 cases (1.7%). Among these 83 equivocal or positive cases, only two were ROS1 rearranged, leading to a low predictive positive value of the IHC test (2%). ROS1-positive IHC was correlated with an increased mRNA ROS1 transcripts. Moreover, we have found a mean statistically significant relationship between ROS1 expression and EGFR gene mutations, suggesting a crosstalk mechanism between these oncogenic driver molecules. Conclusion This study demonstrates that ROS1 IHC represents true ROS1 mRNA expression, and raises the question of a potential benefit of combined targeted therapy in EGFR-mutated NSCLC.
Collapse
Affiliation(s)
- Karl Grenier
- Department of Pathology, McGill University Health Center, Montreal, Quebec, Canada
- Department of Laboratory Medicine, Montreal, Quebec, Canada
| | - Jean-Baptiste Rivière
- Department of Laboratory Medicine, Montreal, Quebec, Canada
- Division of Molecular Genetics, McGill University Health Center, Montreal, Quebec, Canada
| | - Bouchra Ouled Amar Bencheikh
- Department of Laboratory Medicine, Montreal, Quebec, Canada
- Division of Molecular Genetics, McGill University Health Center, Montreal, Quebec, Canada
| | - Andrea Liliam Gomez Corredor
- Department of Pathology, McGill University Health Center, Montreal, Quebec, Canada
- Department of Laboratory Medicine, Montreal, Quebec, Canada
- Division of Molecular Genetics, McGill University Health Center, Montreal, Quebec, Canada
| | | | - Hangjun Wang
- Department of Pathology, McGill University Health Center, Montreal, Quebec, Canada
- Department of Laboratory Medicine, Montreal, Quebec, Canada
| | - Pierre Olivier Fiset
- Department of Pathology, McGill University Health Center, Montreal, Quebec, Canada
- Department of Laboratory Medicine, Montreal, Quebec, Canada
| | - Sophie Camilleri-Broët
- Department of Pathology, McGill University Health Center, Montreal, Quebec, Canada
- Department of Laboratory Medicine, Montreal, Quebec, Canada
| |
Collapse
|
9
|
Spencer KR, Portal DE, Aisner J, Stein MN, Malhotra J, Shih W, Chan N, Silk AW, Ganesan S, Goodin S, Gounder M, Lin H, Li J, Cerchio R, Marinaro C, Chen S, Mehnert JM. A phase I trial of riluzole and sorafenib in patients with advanced solid tumors: CTEP #8850. Oncotarget 2023; 14:302-315. [PMID: 37036756 PMCID: PMC10085060 DOI: 10.18632/oncotarget.28403] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 03/21/2023] [Indexed: 04/11/2023] Open
Abstract
BACKGROUND Overexpression of metabotropic glutamate receptor 1 (GRM1) has been implicated in the pathogenesis of multiple cancers. Riluzole, an inhibitor of glutamate release, showed synergistic antitumor activity in combination with the multi-kinase inhibitor sorafenib in preclinical models. This phase I trial identified the toxicity profile, dose-limiting toxicities, maximum tolerated dose (MTD), and pharmacokinetic and pharmacodynamic properties of riluzole combined with sorafenib in patients with advanced cancers. PATIENTS AND METHODS Patients with refractory solid tumors were enrolled utilizing a 3+3 dose-escalation design. Riluzole was given at 100 mg PO BID in combination with sorafenib, beginning at 200 mg PO daily and escalating in 200 mg increments per level in 28-day cycles. Restaging evaluations were performed every 2 cycles. RESULTS 35 patients were enrolled over 4 dose levels. The MTD was declared at dose level 3 (riluzole: 100 mg PO BID; sorafenib: 400 mg AM/200 mg PM). Pharmacokinetic analyses did not reveal definitive evidence of drug-drug interactions. Consistent decreases in phospho-forms of ERK and AKT in tumor tissue analyses with accompanying decrease in GRM1 expression and increase in pro-apoptotic BIM suggest target engagement by the combination. Best responses included a partial response in 1 (2.9%) patient with pancreatic acinar cell carcinoma with a KANK4-RAF1 fusion, and stable disease in 11 (36%) patients. CONCLUSION Combination therapy with riluzole and sorafenib was safe and tolerable in patients with advanced solid tumors. The partial response in a patient with a RAF1 fusion suggests that further exploration in a genomically selected cohort may be warranted.
Collapse
Affiliation(s)
- Kristen R. Spencer
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08903, USA
- Department of Medicine, Division of Medical Oncology, Rutgers Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Daniella E. Portal
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08903, USA
- Department of Medicine, Division of Medical Oncology, Rutgers Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Joseph Aisner
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08903, USA
- Department of Medicine, Division of Medical Oncology, Rutgers Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Mark N. Stein
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08903, USA
- Department of Medicine, Division of Medical Oncology, Rutgers Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Jyoti Malhotra
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08903, USA
- Department of Medicine, Division of Medical Oncology, Rutgers Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Weichung Shih
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08903, USA
- Department of Biostatistics, School of Public Health, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Nancy Chan
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08903, USA
- Department of Medicine, Division of Medical Oncology, Rutgers Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Ann W. Silk
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08903, USA
- Department of Medicine, Division of Medical Oncology, Rutgers Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
- Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Shridar Ganesan
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08903, USA
- Department of Medicine, Division of Medical Oncology, Rutgers Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Susan Goodin
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08903, USA
- Department of Medicine, Division of Medical Oncology, Rutgers Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Murugesan Gounder
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08903, USA
- Department of Pharmacology, Rutgers Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Hongxia Lin
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08903, USA
| | - Jiadong Li
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08903, USA
| | - Robert Cerchio
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Christina Marinaro
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Suzie Chen
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ 08903, USA
- Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Janice M. Mehnert
- Department of Medicine, New York University Grossman School of Medicine, Perlmutter Cancer Center of NYU Langone Health, NY 10016, USA
| |
Collapse
|
10
|
Kaprio H, Siddiqui A, Saustila L, Heuser VD, Gardberg M. The oncogenic properties of the EWSR1::CREM fusion gene are associated with polyamine metabolism. Sci Rep 2023; 13:4884. [PMID: 36966162 PMCID: PMC10039922 DOI: 10.1038/s41598-023-31576-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 03/14/2023] [Indexed: 03/27/2023] Open
Abstract
The EWSR1::CREM fusion gene, caused by a chromosomal translocation t(10;22)(p11;q12), has been discovered in divergent malignancies, ranging from low-grade to highly malignant cancers. The translocation gives rise to a chimeric protein, EWSR1::CREM. The molecular mechanisms behind the oncogenic properties of the EWSR1::CREM protein have not previously been systematically characterized. In this study, we performed transcriptional profiling of the melanoma cell line CHL-1, with depletion of endogenous EWSR1::CREM protein using siRNA mediated knockdown. We found that the expression of 712 genes was altered (Log2 fold-change ≥ 2). We performed pathway analysis to identify EWSR1::CREM mediated pathways and cell studies to examine functional differences brought upon by the knockdown. Altered pathways involved cell cycle and proliferation, this was further validated by the cell studies where cell migration was affected as well. Among the target genes with the greatest downregulation, we discovered ODC1-a well-established oncogenic enzyme that can be pharmacologically inhibited and is essential for polyamine synthesis. We found that the main effects seen upon EWSR1::CREM knockdown can be reproduced by directly silencing ODC1 expression. These findings provide novel insights into pathogenesis of tumors harboring a EWSR1::CREM fusion gene, hopefully facilitating the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Heidi Kaprio
- Department of Pathology, Turku University Hospital, Kiinamyllynkatu 10 D, Turku, Finland.
- Institute of Biomedicine, University of Turku, Turku, Finland.
| | - Arafat Siddiqui
- Department of Obstetrics and Gynecology, Turku University Hospital, Turku, Finland
| | - Lotta Saustila
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Vanina D Heuser
- Department of Pathology, Turku University Hospital, Kiinamyllynkatu 10 D, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Maria Gardberg
- Department of Pathology, Turku University Hospital, Kiinamyllynkatu 10 D, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
| |
Collapse
|
11
|
Javaid S, Patton A, Tinoco G, Oghumu S, Iwenofu OH. Metastatic sporadic paraganglioma with EWSR1::CREM gene fusion: A unique molecular profile that expands the phenotypic diversity of the molecular landscape of the EWSR1::CREM gene fusion positive tumors. Genes Chromosomes Cancer 2023; 62:85-92. [PMID: 36083250 PMCID: PMC10092737 DOI: 10.1002/gcc.23094] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 08/14/2022] [Accepted: 08/29/2022] [Indexed: 12/13/2022] Open
Abstract
Chromosomal translocations with gene fusions are uniquely rare events in paraganglioma, mostly involving UBTF::MAML3 gene fusion. Precedent literature suggests that tumors involving MAML3 gene fusion correlate with poor clinical outcomes. Herein, we report a case of metastatic sporadic paraganglioma harboring EWSR1::CREM gene fusion in a 36-year-old male, that has not been previously described. The patient presented with large paraspinal mass that was resected the same year. Tumor recurred 3-years later and on further work-up, patient was found to have metastases involving both lungs. Histopathologic evaluation of the original primary tumor showed tightly packed irregular nests and cords of cells containing palely eosinophilic cytoplasm. Features considered atypical included: areas of solid growth pattern, coagulative tumor necrosis, focal cellular atypia and angiolymphatic invasion were also identified. By immunohistochemistry, the tumor cells were positive for synaptophysin and chromogranin and negative for keratin. The S100 stain highlights the sustentacular cells and the Ki-67 proliferation index of 15%. The recurrence specimen was similar but showed increased cellularity, atypia, necrosis, and proliferative activity (Ki-67 proliferation index of 35%). CT guided biopsy of the right lung lesion was consistent with metastasis. Next generation sequencing identified EWSR1::CREM fusion. The breakpoints were found in chromosome 22: 29683123 for EWSR1 exon 7 (NM_005243.3) and at chromosome 10:35495823 for CREM exon 6 (NM_001267562.1). Fluorescence in situ hybridization for EWSR1 gene rearrangement was positive. In summary, we report a case of metastatic paraganglioma with EWSR1::CREM gene fusion, not previously described in this entity, and expands on the phenotypic diversity within the genetic landscape of EWSR1::CREM gene fusion positive tumors.
Collapse
Affiliation(s)
- Sehrish Javaid
- Division of Oral and Maxillofacial Pathology, College of Dentistry, The Ohio State University, Columbus, Ohio, USA
| | - Ashley Patton
- Department of Pathology & Laboratory Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Gabriel Tinoco
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Medical Center, Columbus, Ohio, USA.,The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Steve Oghumu
- Division of Oral and Maxillofacial Pathology, College of Dentistry, The Ohio State University, Columbus, Ohio, USA.,The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Obiajulu Hans Iwenofu
- Division of Oral and Maxillofacial Pathology, College of Dentistry, The Ohio State University, Columbus, Ohio, USA.,The James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
12
|
Wang T, Wei L, Lu Q, Shao Y, You S, Yin JC, Wang S, Shao Y, Chen Z, Wang Z. Landscape of potentially targetable receptor tyrosine kinase fusions in diverse cancers by DNA-based profiling. NPJ Precis Oncol 2022; 6:84. [DOI: 10.1038/s41698-022-00325-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 10/19/2022] [Indexed: 11/13/2022] Open
Abstract
AbstractRecurrent fusions of receptor tyrosine kinases (RTKs) are often driving events in tumorigenesis that carry important diagnostic value and are potentially targetable by the increasing number of tyrosine kinase inhibitors (TKIs). Here, we characterized the spectrum of 1324 RTK fusions with intact kinase domains in solid tumors by DNA-based high-throughput sequencing. Overall, the prevalence of RTK fusions were 4.7%, with variable frequencies and diverse genomic structures and fusion partners across cancer types. Cancer types, such as thyroid cancers, urological cancers and neuroendocrine tumors are selective in the RTK fusions they carry, while others exhibit highly complex spectra of fusion events. Notably, most RTKs were promiscuous in terms of the partner genes they recombine with. A large proportion of RTK fusions had one of the breakpoints localized to intergenic regions. Comprehensive genomic profiling revealed differences in co-mutational patterns pre- and post-TKI treatments across various RTK fusions. At baseline, multiple cases were detected with co-occurring RTK fusions or concomitant oncogenic mutations in driver genes, such as KRAS and EGFR. Following TKI resistance, we observed differences in potential on- and off-target resistance mutations among fusion variants. For example, the EML4-ALK v3 variant displayed more complex on-target resistance mechanisms, which might explain the reduced survival outcome compared with the v1 variant. Finally, we identified two lung cancer patients with MET+ and NTRK1+ tumors, respectively, who responded well to crizotinib treatment. Taken together, our findings demonstrate the diagnostic and prognostic values of screening for RTK fusions using DNA-based sequencing in solid tumors.
Collapse
|
13
|
Jain P, Iyer S, Straka J, Surrey LF, Pogoriler J, Han H, Smith T, Busch C, Fox E, Li M, Waanders AJ, Resnick A, Davare MA. Discovery and functional characterization of the oncogenicity and targetability of a novel NOTCH1-ROS1 gene fusion in pediatric angiosarcoma. Cold Spring Harb Mol Case Stud 2022; 8:mcs.a006222. [PMID: 36307212 PMCID: PMC9632357 DOI: 10.1101/mcs.a006222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/09/2022] [Indexed: 01/25/2023] Open
Abstract
Angiosarcomas are rare, malignant soft tissue tumors in children that arise in a wide range of anatomical locations and have limited targeted therapies available. Here, we report a rare case of a pediatric angiosarcoma (pAS) with Li-Fraumeni syndrome (LFS) expressing a novel NOTCH1-ROS1 gene fusion. Although both NOTCH1 and ROS1 are established proto-oncogenes, our study is the first to describe the mechanistic role of NOTCH1-ROS1 fusion arising via intrachromosomal rearrangement. NOTCH1-ROS1 displayed potent neoplastic transformation propensity in vitro, and harbors tumorigenic potential in vivo, where it induced oncogenic activation of the MAPK, PI3K/mTOR, and JAK-STAT signaling pathways in a murine allograft model. We found an unexpected contribution of the NOTCH1 extracellular region in mediating NOTCH1-ROS1 activation and oncogenic function, highlighting the contribution of both NOTCH1 and ROS1 fusion partners in driving tumorigenicity. Interestingly, neither membrane localization nor fusion protein dimerization were found to be essential for NOTCH1-ROS1 fusion oncogenicity. To target NOTCH1-ROS1-driven tumors, we tested both NOTCH1-directed inhibitors and ROS1-targeted tyrosine kinase inhibitors (TKI) in heterologous models (NIH3T3, Ba/F3). Although NOTCH1 inhibitors did not suppress NOTCH1-ROS1-driven oncogenic growth, we found that oral entrectinib treatment effectively suppressed the growth of NOTCH-ROS1-driven tumors. Taken together, we report the first known pAS case with a novel NOTCH1-ROS1 alteration along with a detailed report on the function and therapeutic targeting of NOTCH1-ROS1. Our study highlights the importance of genomic profiling of rare cancers such as pAS to reveal actionable drivers and improve patient outcomes.
Collapse
Affiliation(s)
- Payal Jain
- Center for Data Driven Discovery in Biomedicine, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA;,Division of Neurosurgery, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Sudarshan Iyer
- Department of Pediatrics, Oregon Health and Sciences University, Portland, Oregon 97239, USA
| | - Joshua Straka
- Center for Data Driven Discovery in Biomedicine, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA;,Division of Neurosurgery, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Lea F. Surrey
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA;,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Jennifer Pogoriler
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA;,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Harry Han
- Center for Data Driven Discovery in Biomedicine, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA;,Division of Neurosurgery, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Tiffany Smith
- Center for Data Driven Discovery in Biomedicine, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA;,Division of Neurosurgery, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Christine Busch
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Elizabeth Fox
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Marilyn Li
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA;,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Angela J. Waanders
- Department of Pediatrics, Feinberg School of Medicine Northwestern University, Chicago, Illinois 60611, USA;,Division of Hematology, Oncology, and Stem Cell Transplant, Ann & Robert H Lurie Children's Hospital of Chicago, Chicago, Illinois 60611, USA
| | - Adam Resnick
- Center for Data Driven Discovery in Biomedicine, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA;,Division of Neurosurgery, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Monika A. Davare
- Department of Pediatrics, Oregon Health and Sciences University, Portland, Oregon 97239, USA
| |
Collapse
|
14
|
Kato T, Kusakizako T, Jin C, Zhou X, Ohgaki R, Quan L, Xu M, Okuda S, Kobayashi K, Yamashita K, Nishizawa T, Kanai Y, Nureki O. Structural insights into inhibitory mechanism of human excitatory amino acid transporter EAAT2. Nat Commun 2022; 13:4714. [PMID: 35953475 PMCID: PMC9372063 DOI: 10.1038/s41467-022-32442-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 08/01/2022] [Indexed: 11/18/2022] Open
Abstract
Glutamate is a pivotal excitatory neurotransmitter in mammalian brains, but excessive glutamate causes numerous neural disorders. Almost all extracellular glutamate is retrieved by the glial transporter, Excitatory Amino Acid Transporter 2 (EAAT2), belonging to the SLC1A family. However, in some cancers, EAAT2 expression is enhanced and causes resistance to therapies by metabolic disturbance. Despite its crucial roles, the detailed structural information about EAAT2 has not been available. Here, we report cryo-EM structures of human EAAT2 in substrate-free and selective inhibitor WAY213613-bound states at 3.2 Å and 2.8 Å, respectively. EAAT2 forms a trimer, with each protomer consisting of transport and scaffold domains. Along with a glutamate-binding site, the transport domain possesses a cavity that could be disrupted during the transport cycle. WAY213613 occupies both the glutamate-binding site and cavity of EAAT2 to interfere with its alternating access, where the sensitivity is defined by the inner environment of the cavity. We provide the characterization of the molecular features of EAAT2 and its selective inhibition mechanism that may facilitate structure-based drug design for EAAT2. EAAT2 is an amino acid transporter implicated in glutamate homeostasis in brain and therapy resistance of cancer cells. Here, the authors report cryo-EM structures and reveal inhibitory mechanisms via selective inhibitor WAY213613.
Collapse
Affiliation(s)
- Takafumi Kato
- Department of Biological Science, Graduate School of Science, The University of Tokyo, Tokyo, Japan.,Department of Biochemistry, The University of Oxford, Oxford, UK
| | - Tsukasa Kusakizako
- Department of Biological Science, Graduate School of Science, The University of Tokyo, Tokyo, Japan.
| | - Chunhuan Jin
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Xinyu Zhou
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Ryuichi Ohgaki
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, Osaka, Japan.,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiative (OTRI), Osaka University, Osaka, Japan
| | - LiLi Quan
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Molecular Pharmacology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Minhui Xu
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Suguru Okuda
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Kan Kobayashi
- Department of Biological Science, Graduate School of Science, The University of Tokyo, Tokyo, Japan.,Peptidream Inc, Kawasaki, Japan
| | - Keitaro Yamashita
- Department of Biological Science, Graduate School of Science, The University of Tokyo, Tokyo, Japan.,Structural Studies Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Tomohiro Nishizawa
- Department of Biological Science, Graduate School of Science, The University of Tokyo, Tokyo, Japan.,Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Yoshikatsu Kanai
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, Osaka, Japan. .,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiative (OTRI), Osaka University, Osaka, Japan.
| | - Osamu Nureki
- Department of Biological Science, Graduate School of Science, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
15
|
PPP1R7 Is a Novel Translocation Partner of CBFB via t(2;16)(q37;q22) in Acute Myeloid Leukemia. Genes (Basel) 2022; 13:genes13081367. [PMID: 36011278 PMCID: PMC9407081 DOI: 10.3390/genes13081367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 11/28/2022] Open
Abstract
In a subset of acute myeloid leukemia (AML) cases, the core binding factor beta subunit gene (CBFB) was rearranged via inv(16)(p13.1q22) or t(16;16)(p13.1;q22), in which the smooth muscle myosin heavy chain 11 gene (MYH11) was the partner (CBFB::MYH11). Rare variants of CBFB rearrangement occurring via non-classic chromosomal aberrations have been reported, such as t(1;16), t(2;16), t(3;16), t(5;16), and t(16;19), but the partners of CBFB have not been characterized. We report a case of AML with a complex karyotype, including t(2;16)(q37;q22), in which the protein phosphatase 1 regulatory subunit 7 gene (PPP1R7) at chromosome 2q37 was rearranged with CBFB (CBFB::PPP1R7). This abnormality was inconspicuous by conventional karyotype and interphase fluorescence in situ hybridization (FISH), thus leading to an initial interpretation of inv(16)(p13.1q22); however, metaphase FISH showed that the CBFB rearrangement involved chromosome 2. Using whole genome and Sanger sequencing, the breakpoints were identified as being located in intron 5 of CBFB and intron 7 of PPP1R7. A microhomology of CAG was found in the break and reconnection sites of CBFB and PPP1R7, thus supporting the formation of CBFB::PPP1R7 by microhomology-mediated end joining.
Collapse
|
16
|
Xu ZH, Wang WQ, Liu L, Lou WH. A special subtype: Revealing the potential intervention and great value of KRAS wildtype pancreatic cancer. Biochim Biophys Acta Rev Cancer 2022; 1877:188751. [PMID: 35732240 DOI: 10.1016/j.bbcan.2022.188751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/11/2022] [Accepted: 06/13/2022] [Indexed: 11/22/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the predominant form of pancreatic cancer and has devastating consequences on affected families and society. Its dismal prognosis is attributed to poor specificity of symptoms during early stages. It is widely believed that PDAC patients with the wildtype (WT) KRAS gene benefit more from currently available treatments than those with KRAS mutations. The oncogenic genetic changes alternations generally found in KRAS wildtype PDAC are related to either the KRAS pathway or microsatellite instability/mismatch repair deficiency (MSI/dMMR), which enable the application of tailored treatments based on each patient's genetic characteristics. This review focuses on targeted therapies against alternative tumour mechanisms in KRAS WT PDAC.
Collapse
Affiliation(s)
- Zhi-Hang Xu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wen-Quan Wang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liang Liu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Wen-Hui Lou
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
17
|
Hung MS, Lin YC, Chen FF, Jiang YY, Fang YH, Lu MS, Lin CK, Yang TM, Lung J, Chen CC, Lee KD, Tsai YH. The potential and limitation of targeted chromosomal breakpoint sequencing for the ROS1 fusion gene identification in lung cancer. Am J Cancer Res 2022; 12:2376-2386. [PMID: 35693072 PMCID: PMC9185620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 03/20/2022] [Indexed: 06/15/2023] Open
Abstract
ROS1 fusion genes are rare but important driver genes in lung cancer. Owing to their rarity, many clinicopathological features and treatment responses for each ROS1 fusion variant are still largely unknown and require further investigation. RNA is the preferable template for the ROS1 fusion gene screening, but deterioration of RNA in FFPE often makes the detection challenging. To resolve the difficulty, a targeted chromosomal breakpoint sequencing method was developed for searching the ROS1 fusion gene, and was compared with fluorescence in situ hybridization, immunohistochemistry, RT-qPCR using 260 lung cancer samples of Southern Taiwan. The results showed that ROS1-altered cases were present at low frequencies, did not share distinct clinicopathological features, and often carried other driver mutations. The performance of the targeted sequencing assay was superior to the RT-qPCR in ROS1 fusion gene identification when the cDNAs were from FFPE samples, but long-read DNA sequencing and fresh-frozen samples would be better to revolve all fusion genes. Precise determination of all ROS1 fusion variants and concomitant driver mutations using both genomic DNA and RNA would be required to help improve the treatment of patients with ROS1 alterations.
Collapse
Affiliation(s)
- Ming-Szu Hung
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Chiayi BranchTaiwan
- Department of Medicine, College of Medicine, Chang Gung UniversityTaoyuan, Taiwan
- Department of Respiratory Care, Chang Gung University of Science and Technology, Chiayi CampusChiayi, Taiwan
| | - Yu-Ching Lin
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Chiayi BranchTaiwan
- Department of Medicine, College of Medicine, Chang Gung UniversityTaoyuan, Taiwan
- Department of Respiratory Care, Chang Gung University of Science and Technology, Chiayi CampusChiayi, Taiwan
| | - Fen-Fen Chen
- Department of Pathology, Chang Gung Memorial Hospital, Chiayi BranchTaiwan
| | - Yuan-Yuan Jiang
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Chiayi BranchTaiwan
| | - Yu-Hung Fang
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Chiayi BranchTaiwan
| | - Ming-Shian Lu
- Department of Surgery, Division of Thoracic and Cardiovascular Surgery, Chang Gung Memorial Hospital, Chiayi BranchTaiwan
| | - Chin-Kuo Lin
- Department of Medicine, College of Medicine, Chang Gung UniversityTaoyuan, Taiwan
| | - Tsung-Ming Yang
- Department of Medicine, College of Medicine, Chang Gung UniversityTaoyuan, Taiwan
| | - Jrhau Lung
- Department of Medical Research and Development, Chang Gung Memorial Hospital, Chiayi BranchTaiwan
| | - Chih-Cheng Chen
- Department of Medicine, College of Medicine, Chang Gung UniversityTaoyuan, Taiwan
- Department of Hematology and Oncology, Chang Gung Memorial Hospital, Chiayi BranchTaiwan
| | - Kuan-Der Lee
- Department of Hematology and Oncology, Taipei Medical University HospitalTaipei 110, Taiwan
| | - Ying-Huang Tsai
- Department of Respiratory Care, College of Medicine, Chang Gung UniversityTaoyuan, Taiwan
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Linkou BranchTaiwan
| |
Collapse
|
18
|
Wang N, Li R, Jia H, Xie H, Liu C, Jiang S, Zhang K, Lin P, Yu X. Apaf-1 interacting protein, a new target of microRNA-146a-3p, promotes prostate cancer cell development via the ERK1/2 pathway. Cell Biol Int 2022; 46:1156-1168. [PMID: 35293661 DOI: 10.1002/cbin.11796] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 02/25/2022] [Accepted: 03/13/2022] [Indexed: 11/06/2022]
Abstract
The Apaf-1 interacting protein APIP, a ubiquitously expressed anti-apoptotic molecule, is aberrantly expressed and of great significance in various cancers. However, little is known regarding the potential value and underlying mechanisms of APIP in prostate cancer. Here, we demonstrated that APIP expression is significantly upregulated in prostate cancer cell lines. APIP overexpression promoted tumor cell proliferation and migration and induced ERK1/2 activation. Pharmacological inhibition of ERK1/2 signaling reversed APIP-induced increase in cell proliferation and migration induced by APIP overexpression. Expression of APIP was hampered by miR-146a-3p. A dual luciferase reporter gene assay identified the regulatory relationship between APIP and miR-146a-3p in prostate cancer, suggesting that APIP is a direct target of miR-146a-3p. miR-146a-3p reduced cell proliferation and migration in prostate cancer. Furthermore, miR-146a-3p inhibited ERK1/2 activation. Application of an ERK1/2 inhibitor reversed the increase in cell proliferation and migration induced by miR-146a-3p inhibition. In summary, this study focused on the role of APIP in regulating cell growth and migration, and proposes a theoretical basis for APIP as a promising biomarker in prostate cancer development. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Nan Wang
- Department of Biochemistry & Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, 150086, P.R. China
| | - Rou Li
- Department of Biochemistry & Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, 150086, P.R. China
| | - Huizhen Jia
- Department of Biochemistry & Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, 150086, P.R. China
| | - Hui Xie
- Teaching Experiment Center of Biotechnology, Harbin Medical University, Harbin, Heilongjiang, 150001, P.R. China
| | - Chi Liu
- Department of Biochemistry & Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, 150086, P.R. China
| | - Shan Jiang
- Department of Biochemistry & Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, 150086, P.R. China
| | - Ke Zhang
- Department of Biochemistry & Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, 150086, P.R. China
| | - Ping Lin
- Department of Biochemistry & Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, 150086, P.R. China
| | - Xiaoguang Yu
- Department of Biochemistry & Molecular Biology, Harbin Medical University, Harbin, Heilongjiang, 150086, P.R. China
| |
Collapse
|
19
|
Jarahian M, Marofi F, Maashi MS, Ghaebi M, Khezri A, Berger MR. Re-Expression of Poly/Oligo-Sialylated Adhesion Molecules on the Surface of Tumor Cells Disrupts Their Interaction with Immune-Effector Cells and Contributes to Pathophysiological Immune Escape. Cancers (Basel) 2021; 13:5203. [PMID: 34680351 PMCID: PMC8534074 DOI: 10.3390/cancers13205203] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 12/28/2022] Open
Abstract
Glycans linked to surface proteins are the most complex biological macromolecules that play an active role in various cellular mechanisms. This diversity is the basis of cell-cell interaction and communication, cell growth, cell migration, as well as co-stimulatory or inhibitory signaling. Our review describes the importance of neuraminic acid and its derivatives as recognition elements, which are located at the outermost positions of carbohydrate chains linked to specific glycoproteins or glycolipids. Tumor cells, especially from solid tumors, mask themselves by re-expression of hypersialylated neural cell adhesion molecule (NCAM), neuropilin-2 (NRP-2), or synaptic cell adhesion molecule 1 (SynCAM 1) in order to protect themselves against the cytotoxic attack of the also highly sialylated immune effector cells. More particularly, we focus on α-2,8-linked polysialic acid chains, which characterize carrier glycoproteins such as NCAM, NRP-2, or SynCam-1. This characteristic property correlates with an aggressive clinical phenotype and endows them with multiple roles in biological processes that underlie all steps of cancer progression, including regulation of cell-cell and/or cell-extracellular matrix interactions, as well as increased proliferation, migration, reduced apoptosis rate of tumor cells, angiogenesis, and metastasis. Specifically, re-expression of poly/oligo-sialylated adhesion molecules on the surface of tumor cells disrupts their interaction with immune-effector cells and contributes to pathophysiological immune escape. Further, sialylated glycoproteins induce immunoregulatory cytokines and growth factors through interactions with sialic acid-binding immunoglobulin-like lectins. We describe the processes, which modulate the interaction between sialylated carrier glycoproteins and their ligands, and illustrate that sialic acids could be targets of novel therapeutic strategies for treatment of cancer and immune diseases.
Collapse
Affiliation(s)
- Mostafa Jarahian
- German Cancer Research Center, Toxicology and Chemotherapy Unit Heidelberg, 69120 Heidelberg, Germany;
| | - Faroogh Marofi
- Department of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 5165665931, Iran;
| | - Marwah Suliman Maashi
- Stem Cells and Regenerative Medicine Unit at King Fahad Medical Research Centre, Jeddah 11211, Saudi Arabia;
| | - Mahnaz Ghaebi
- Cancer Gene Therapy Research Center (CGRC), Zanjan University of Medical Sciences, Zanjan 4513956184, Iran;
| | - Abdolrahman Khezri
- Department of Biotechnology, Inland Norway University of Applied Sciences, 2418 Hamar, Norway;
| | - Martin R. Berger
- German Cancer Research Center, Toxicology and Chemotherapy Unit Heidelberg, 69120 Heidelberg, Germany;
| |
Collapse
|
20
|
Afkhami M, Ally F, Pullarkat V, Pillai RK. Genetics and Diagnostic Approach to Lymphoblastic Leukemia/Lymphoma. Cancer Treat Res 2021; 181:17-43. [PMID: 34626353 DOI: 10.1007/978-3-030-78311-2_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Our understanding of the genetics and biology of lymphoblastic leukemia/lymphoma (acute lymphoblastic leukemia, ALL) has advanced rapidly in the past decade with advances in sequencing and other molecular techniques. Besides recurrent chromosomal abnormalities detected by karyotyping or fluorescence in situ hybridization, these leukemias/lymphomas are characterized by a variety of mutations, gene rearrangements as well as copy number alterations. This is particularly true in the case of Philadelphia-like (Ph-like) ALL, a major subset which has the same gene expression signature as Philadelphia chromosome-positive ALL but lacks BCR-ABL1 translocation. Ph-like ALL is associated with a worse prognosis and hence its detection is critical. However, techniques to detect this entity are complex and are not widely available. This chapter discusses various subsets of ALL and describes our approach to the accurate classification and prognostication of these cases.
Collapse
Affiliation(s)
- Michelle Afkhami
- City of Hope Medical Center, 1500 E Duarte Rd., Duarte, CA, 91010, USA.
| | - Feras Ally
- City of Hope Medical Center, 1500 E Duarte Rd., Duarte, CA, 91010, USA
| | - Vinod Pullarkat
- City of Hope Medical Center, 1500 E Duarte Rd., Duarte, CA, 91010, USA
| | - Raju K Pillai
- City of Hope Medical Center, 1500 E Duarte Rd., Duarte, CA, 91010, USA
| |
Collapse
|
21
|
Hyung D, Baek MJ, Lee J, Cho J, Kim HS, Park C, Cho SY. Protein-gene Expression Nexus: Comprehensive characterization of human cancer cell lines with proteogenomic analysis. Comput Struct Biotechnol J 2021; 19:4759-4769. [PMID: 34504668 PMCID: PMC8405889 DOI: 10.1016/j.csbj.2021.08.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 08/13/2021] [Accepted: 08/14/2021] [Indexed: 12/30/2022] Open
Abstract
Researchers have gained new therapeutic insights using multi-omics platform approaches to study DNA, RNA, and proteins of comprehensively characterized human cancer cell lines. To improve our understanding of the molecular features associated with oncogenic modulation in cancer, we proposed a proteogenomic database for human cancer cell lines, called Protein-gene Expression Nexus (PEN). We have expanded the characterization of cancer cell lines to include genetic, mRNA, and protein data of 145 cancer cell lines from various public studies. PEN contains proteomic and phosphoproteomic data on 4,129,728 peptides, 13,862 proteins, 7,138 phosphorylation site-associated genomic variations, 117 studies, and 12 cancer. We analyzed functional characterizations along with the integrated datasets, such as cis/trans association for copy number alteration (CNA), single amino acid variation for coding genes, post-translation modification site variation for Single Amino Acid Variation, and novel peptide expression for noncoding regions and fusion genes. PEN provides a user-friendly interface for searching, browsing, and downloading data and also supports the visualization of genome-wide association between CNA and expression, novel peptide landscape, mRNA-protein abundance, and functional annotation. Together, this dataset and PEN data portal provide a resource to accelerate cancer research using model cancer cell lines. PEN is freely accessible at http://combio.snu.ac.kr/pen.
Collapse
Affiliation(s)
- Daejin Hyung
- National Cancer Center, 323 Ilsan-ro, Goyang-si, Gyeonggi-do 10408, Republic of Korea
| | - Min-Jeong Baek
- National Cancer Center, 323 Ilsan-ro, Goyang-si, Gyeonggi-do 10408, Republic of Korea
| | - Jongkeun Lee
- National Cancer Center, 323 Ilsan-ro, Goyang-si, Gyeonggi-do 10408, Republic of Korea
| | - Juyeon Cho
- National Cancer Center, 323 Ilsan-ro, Goyang-si, Gyeonggi-do 10408, Republic of Korea
| | - Hyoun Sook Kim
- National Cancer Center, 323 Ilsan-ro, Goyang-si, Gyeonggi-do 10408, Republic of Korea
| | - Charny Park
- National Cancer Center, 323 Ilsan-ro, Goyang-si, Gyeonggi-do 10408, Republic of Korea
| | - Soo Young Cho
- National Cancer Center, 323 Ilsan-ro, Goyang-si, Gyeonggi-do 10408, Republic of Korea.,Department of Molecular and Life Science, Hanyang University, Ansan 15588, Republic of Korea
| |
Collapse
|
22
|
Kaprio H, Heuser VD, Orte K, Tukiainen M, Leivo I, Gardberg M. Expression of Transcription Factor CREM in Human Tissues. J Histochem Cytochem 2021; 69:495-509. [PMID: 34261344 PMCID: PMC8329441 DOI: 10.1369/00221554211032008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cyclic AMP element modulator (CREM) is a transcription factor best known for its intricate involvement in spermatogenesis. The CREM gene encodes for multiple protein isoforms, which can enhance or repress transcription of target genes. Recent studies have identified fusion genes, with CREM as a partner gene in many neoplastic diseases. EWSR1-CREM fusion genes have been found in several mesenchymal tumors and in salivary gland carcinoma. These genes encode fusion proteins that include the C-terminal DNA-binding domain of CREM. We used a transcriptomic approach and immunohistochemistry to study the expression of CREM isoforms that include DNA-binding domains across human tissues. We found that CREM protein is widely expressed in almost all normal human tissues. A transcriptomic analysis of normal tissues and cancer showed that transcription of CREM can be altered in tumors, suggesting that also wild-type CREM may be involved in cancer biology. The wide expression of CREM protein in normal human tissues and cancer may limit the utility of immunohistochemistry for identification of tumors with CREM fusions:
Collapse
Affiliation(s)
- Heidi Kaprio
- Department of Pathology, Institute of Biomedicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Vanina D Heuser
- Department of Pathology, Institute of Biomedicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Katri Orte
- Department of Pathology, Institute of Biomedicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Mikko Tukiainen
- Auria Biobank, Turku University Hospital and University of Turku, Turku, Finland
| | - Ilmo Leivo
- Department of Pathology, Institute of Biomedicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Maria Gardberg
- Department of Pathology, Institute of Biomedicine, Turku University Hospital and University of Turku, Turku, Finland
| |
Collapse
|
23
|
Panagopoulos I, Heim S. Interstitial Deletions Generating Fusion Genes. Cancer Genomics Proteomics 2021; 18:167-196. [PMID: 33893073 DOI: 10.21873/cgp.20251] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 12/16/2022] Open
Abstract
A fusion gene is the physical juxtaposition of two different genes resulting in a structure consisting of the head of one gene and the tail of the other. Gene fusion is often a primary neoplasia-inducing event in leukemias, lymphomas, solid malignancies as well as benign tumors. Knowledge about fusion genes is crucial not only for our understanding of tumorigenesis, but also for the diagnosis, prognostication, and treatment of cancer. Balanced chromosomal rearrangements, in particular translocations and inversions, are the most frequent genetic events leading to the generation of fusion genes. In the present review, we summarize the existing knowledge on chromosome deletions as a mechanism for fusion gene formation. Such deletions are mostly submicroscopic and, hence, not detected by cytogenetic analyses but by array comparative genome hybridization (aCGH) and/or high throughput sequencing (HTS). They are found across the genome in a variety of neoplasias. As tumors are increasingly analyzed using aCGH and HTS, it is likely that more interstitial deletions giving rise to fusion genes will be found, significantly impacting our understanding and treatment of cancer.
Collapse
Affiliation(s)
- Ioannis Panagopoulos
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway;
| | - Sverre Heim
- Section for Cancer Cytogenetics, Institute for Cancer Genetics and Informatics, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
24
|
Suehara Y, Kohsaka S, Hayashi T, Akaike K, Kurisaki-Arakawa A, Sato S, Kobayashi E, Mizuno S, Ueno T, Morii T, Okuma T, Kurihara T, Hasegawa N, Sano K, Sasa K, Okubo T, Kim Y, Mano H, Saito T. Identification of a Novel MAN1A1-ROS1 Fusion Gene Through mRNA-based Screening for Tyrosine Kinase Gene Aberrations in a Patient with Leiomyosarcoma. Clin Orthop Relat Res 2021; 479:838-852. [PMID: 33196586 PMCID: PMC8083907 DOI: 10.1097/corr.0000000000001548] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 10/06/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Soft tissue sarcomas are a heterogeneous group of rare malignant tumors. Advanced soft tissue sarcomas have a poor prognosis, and effective systemic therapies have not been established. Tyrosine kinases are increasingly being used as therapeutic targets for a variety of cancers and soft tissue sarcomas. Although complex karyotype sarcomas typically tend to carry more potentially actionable genetic alterations than do translocation-associated sarcomas (fusion gene sarcomas), based on our database review, we found that leiomyosarcoma and malignant peripheral nerve sheath tumors have lower frequencies of potential targets than other nontranslocation soft tissue sarcomas. We theorized that both leiomyosarcoma and malignant peripheral nerve sheath tumors might be included in any unique translocations. Furthermore, if tyrosine kinase imbalances, especially fusion genes, occur in patients with leiomyosarcomas and malignant peripheral nerve sheath tumors, tyrosine kinase inhibitors might be a drug development target for this sarcoma. In this study, we used a tyrosine kinase screening system that could detect an imbalance in mRNA between 5'- and 3'-sides in tyrosine kinase genes to identify potential novel therapeutic tyrosine kinase targets for soft tissue sarcomas. QUESTIONS/PURPOSES (1) Are there novel therapeutic tyrosine kinase targets in tumors from patients with soft tissue sarcomas that are detectable using mRNA screening focusing on imbalance expressions between the 5' and 3' end of the kinase domain? (2) Can potential targets be verified by RNA sequencing and reverse transcription PCR (RT-PCR)? (3) Will potential fusion gene(s) transform cells in in vitro assays? (4) Will tumors in mice that have an identified fusion gene respond to treatment with a therapeutic drug directed at that target? METHODS We used mRNA screening to look for novel tyrosine kinase targets that might be of therapeutic potential. Using functional assays, we verified whether the identified fusion genes would be good therapeutic candidates for soft tissue sarcomas. Additionally, using in vivo assays, we assessed whether suppressing the fusion's kinase activity has therapeutic potential. Study eligibility was based on a patient having high-grade spindle cell and nontranslocation sarcomas, including leiomyosarcoma, malignant peripheral nerve sheath tumor, and high-grade myxofibrosarcoma. Between 2015 and 2019, of the 172 patients with soft tissue sarcomas treated with surgical resection at Juntendo University Hospital, 72 patients had high-grade nontranslocation sarcomas. The analysis was primarily for leiomyosarcoma and malignant peripheral nerve sheath tumors, and there was a limitation of analysis size (reagent limitations) totaling 24 samples at the start of the study. We collected additional samples from a sample bank at the Tokyo Medical and Dental University to increase the number of sarcomas to study. Therefore, in this study, a total of 15 leiomyosarcoma samples, five malignant peripheral nerve sheath tumors samples, and four high-grade myxofibrosarcoma samples were collected to achieve the sample size of 24 patients. To identify tyrosine kinase fusion genes, we designed a NanoString-based assay (NanoString Technologies Inc, Seattle, WA, USA) to query the expression balances regarding transcripts of 90 tyrosine kinases at two points: the 5' end of the kinase domain and within the kinase domain or 3' end of the kinase domain. The tumor's RNA was hybridized to the NanoString probes and analyzed for the expression ratios of outliers from the 3' to 5' end of the kinase domain. Presumed novel fusion events in these positive tumors that were defined by NanoString-based assays were confirmed tyrosine kinase fusion genes by RNA sequencing and confirmatory RT-PCR. Functional analyses consisting of in vitro and in vivo assays were also performed to elucidate whether the identified tyrosine kinase gene fusions were associated with oncogenic abilities and drug responses. RESULTS We identified aberrant expression ratios regarding the 3' to 5' end of the kinase domain ratios in ROS1 transcripts in a leiomyosarcoma in a 90-year-old woman. A novel MAN1A1-ROS1 fusion gene was identified from her thigh tumor through RNA sequencing, which was confirmed with real-time PCR. In functional assays, MAN1A1-ROS1 rearrangement revealed strong transforming potential in 3T3 cells. Moreover, in an in vivo assay, crizotinib, a ROS1 inhibitor, markedly inhibited the growth of MAN1A1-ROS1 rearrangement-induced transformed cells in a dose-dependent manner. CONCLUSION We conducted tyrosine kinase screening to identify new therapeutic targets in soft tissue sarcomas. We found a novel MAN1A1-ROS1 fusion gene that may be a therapeutic target in patients with leiomyosarcoma. This study demonstrates that the mRNA screening system may aid in the development of useful therapeutic options for soft tissue sarcomas. CLINICAL RELEVANCE If novel tyrosine fusions such as MAN1A1-ROS1 fusion can be found in sarcomas from other patients, they could offer avenues for new molecular target therapies for sarcomas that currently do not have effective chemotherapeutic options. Therefore, the establishment of a screening system that includes both genomic and transcript analyses in the clinical setting is needed to verify our discoveries and take the developmental process of treatment to the next step.
Collapse
Affiliation(s)
- Yoshiyuki Suehara
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Shinji Kohsaka
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Takuo Hayashi
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Keisuke Akaike
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Aiko Kurisaki-Arakawa
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Shingo Sato
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Eisuke Kobayashi
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Sho Mizuno
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Toshihide Ueno
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Takeshi Morii
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Tomotake Okuma
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Taisei Kurihara
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Nobuhiko Hasegawa
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Kei Sano
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Keita Sasa
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Taketo Okubo
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Youngji Kim
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Hiroyuki Mano
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Tsuyoshi Saito
- Y. Suehara, K. Akaike, T. Kurihara, N. Hasegawa, K. Sano, K. Sasa, T. Okubo, Y. Kim, Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
- S. Kohsaka, S. Mizuno, T. Ueno, N. Hasegawa, H. Mano, Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- T. Hayashi, A. Kurisaki-Arakawa, T. Saito, Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
- S. Sato, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, Tokyo, Japan
- E. Kobayashi, Division of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
- T. Morii, Department of Orthopedic Surgery, Kyorin University, Faculty of Medicine, Tokyo, Japan
- T. Okuma, Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| |
Collapse
|
25
|
Domingo RA, Vivas-Buitrago T, Jentoft M, Quinones-Hinojosa A. Intracranial Myxoid Mesenchymal Tumor/Myxoid Subtype Angiomatous Fibrous Histiocytoma: Diagnostic and Prognostic Challenges. Neurosurgery 2021; 88:E114-E122. [PMID: 32970137 DOI: 10.1093/neuros/nyaa357] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 06/18/2020] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND IMPORTANCE In the setting of intracranial neoplasms, EWSR1-cAMP Response Element-Binding Protein (CREB) transcription factor family fusions have been described in myxoid mesenchymal tumors, extremely rare entities with a close histopathologic and immunologic resemblance to myxoid subtype angiomatoid fibrous histiocytomas (AFH). Controversy exists on whether these central nervous system lesions are a subtype of myxoid AFH or a completely separate entity, which entitles a distinct clinical behavior and, consequently, a different approach to management. Upon review of the literature, only 14 cases of intracranial tumors harboring an EWSR1-CREB family fusion were identified, with only 3 cases presenting in middle-aged adults, none of which reported an EWSR1-CREM fusion mutation. Significant variability in reported radiographic and histopathological characteristics, as well as in clinical outcomes, was noted. Their similarity with other soft tissue tumors, added to the scarce information on its clinical behavior, represents a great diagnostic and therapeutic challenge to the treating physician. CLINICAL PRESENTATION We present a rare case of EWSR1-CREM mutated intracranial myxoid mesenchymal tumor/myxoid subtype AFH presenting as persistent headaches in a 36-yr-old woman with radiographic evidence of rapid growth and extensive vasogenic edema, for which she underwent surgical resection. CONCLUSION This represents a unique case of EWSR1-CREM mutated intracranial myxoid mesenchymal tumor presenting in adulthood, with evidence of aggressive behavior.
Collapse
Affiliation(s)
- Ricardo A Domingo
- Department of Neurologic Surgery, Mayo Clinic, Jacksonville, Florida
| | | | - Mark Jentoft
- Department of Pathology, Mayo Clinic, Jacksonville, Florida
| | | |
Collapse
|
26
|
Kim JH, Megquier K, Thomas R, Sarver AL, Song JM, Kim YT, Cheng N, Schulte AJ, Linden MA, Murugan P, Oseth L, Forster CL, Elvers I, Swofford R, Turner-Maier J, Karlsson EK, Breen M, Lindblad-Toh K, Modiano JF. Genomically Complex Human Angiosarcoma and Canine Hemangiosarcoma Establish Convergent Angiogenic Transcriptional Programs Driven by Novel Gene Fusions. Mol Cancer Res 2021; 19:847-861. [PMID: 33649193 DOI: 10.1158/1541-7786.mcr-20-0937] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/21/2020] [Accepted: 02/10/2021] [Indexed: 11/16/2022]
Abstract
Sporadic angiosarcomas are aggressive vascular sarcomas whose rarity and genomic complexity present significant obstacles in deciphering the pathogenic significance of individual genetic alterations. Numerous fusion genes have been identified across multiple types of cancers, but their existence and significance remain unclear in sporadic angiosarcomas. In this study, we leveraged RNA-sequencing data from 13 human angiosarcomas and 76 spontaneous canine hemangiosarcomas to identify fusion genes associated with spontaneous vascular malignancies. Ten novel protein-coding fusion genes, including TEX2-PECAM1 and ATP8A2-FLT1, were identified in seven of the 13 human tumors, with two tumors showing mutations of TP53. HRAS and NRAS mutations were found in angiosarcomas without fusions or TP53 mutations. We found 15 novel protein-coding fusion genes including MYO16-PTK2, GABRA3-FLT1, and AKT3-XPNPEP1 in 11 of the 76 canine hemangiosarcomas; these fusion genes were seen exclusively in tumors of the angiogenic molecular subtype that contained recurrent mutations in TP53, PIK3CA, PIK3R1, and NRAS. In particular, fusion genes and mutations of TP53 cooccurred in tumors with higher frequency than expected by random chance, and they enriched gene signatures predicting activation of angiogenic pathways. Comparative transcriptomic analysis of human angiosarcomas and canine hemangiosarcomas identified shared molecular signatures associated with activation of PI3K/AKT/mTOR pathways. Our data suggest that genome instability induced by TP53 mutations might create a predisposition for fusion events that may contribute to tumor progression by promoting selection and/or enhancing fitness through activation of convergent angiogenic pathways in this vascular malignancy. IMPLICATIONS: This study shows that, while drive events of malignant vasoformative tumors of humans and dogs include diverse mutations and stochastic rearrangements that create novel fusion genes, convergent transcriptional programs govern the highly conserved morphologic organization and biological behavior of these tumors in both species.
Collapse
Affiliation(s)
- Jong Hyuk Kim
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota. .,Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.,Institute for Engineering in Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Kate Megquier
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Rachael Thomas
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine & Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
| | - Aaron L Sarver
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.,Institute for Health Informatics, University of Minnesota, Minneapolis, Minnesota
| | - Jung Min Song
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Yoon Tae Kim
- Department of Electrical Engineering and Computer Science, York University, Toronto, Ontario, Canada
| | - Nuojin Cheng
- School of Mathematics, College of Science and Engineering, University of Minnesota, Minneapolis, Minnesota
| | - Ashley J Schulte
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota.,Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Michael A Linden
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.,Department of Laboratory Medicine and Pathology, School of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Paari Murugan
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.,Department of Laboratory Medicine and Pathology, School of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - LeAnn Oseth
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Colleen L Forster
- The University of Minnesota Biological Materials Procurement Network (BioNet), University of Minnesota, Minneapolis, Minnesota
| | - Ingegerd Elvers
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Ross Swofford
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | | | - Elinor K Karlsson
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,University of Massachusetts Medical School, Worcester, Massachusetts
| | - Matthew Breen
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine & Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina.,Cancer Genetics Program, University of North Carolina Lineberger Comprehensive Cancer Center, Raleigh, North Carolina
| | - Kerstin Lindblad-Toh
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Jaime F Modiano
- Animal Cancer Care and Research Program, University of Minnesota, St Paul, Minnesota.,Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.,Institute for Engineering in Medicine, University of Minnesota, Minneapolis, Minnesota.,Department of Laboratory Medicine and Pathology, School of Medicine, University of Minnesota, Minneapolis, Minnesota.,Center for Immunology, University of Minnesota, Minneapolis, Minnesota.,Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
27
|
Jung H, Kim HN, Jang Y, Park CK, Shin SH, Ha SY. Hepatic Angiosarcoma: Clinicopathologic Study With an Investigation of ROS1 Gene Rearrangements. In Vivo 2021; 34:1463-1467. [PMID: 32354947 DOI: 10.21873/invivo.11930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/02/2020] [Accepted: 03/03/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND/AIM Primary hepatic angiosarcoma (PHA) is a rare disease entity with variable morphologic features. Recent findings regarding ROS1 gene rearrangements in PHA may lead to new targeted therapies. PATIENTS AND METHODS Thirteen cases (4 resected specimens and 9 biopsy samples) underwent histologic review and morphologic patterns were classified according to a previous study as 1) sinusoidal, 2) peliotic, 3) vasoformative, and 4) solid (epithelioid/spindled). ROS1 immunohistochemistry and investigation of the presence of a ROS1 fusion gene by reverse transcription-polymerase chain reaction were performed in available cases. RESULTS Eight of 13 cases (62%) showed vasoformative patterns. Three cases (23%) were classified as sinusoidal and two (15%) as solid patterns. Mortality rate was 90% (9/10) except for three patients lost in follow up. Only one patient is still alive and has survived for 8 months with the disease. All cases tested did not have ROS1 expression (0/9) or a ROS1 fusion gene (0/4). CONCLUSION We report 13 cases of PHA with 90% mortality. Vasoformative PHA is the most common histologic type. New findings on ROS1 fusion gene rearrangements could lead to the development of novel targeted therapeutics for PHA patients with dismal prognosis.
Collapse
Affiliation(s)
- Hera Jung
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Han-Na Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yunjeong Jang
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Cheol-Keun Park
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Anatomic Pathology Reference Lab, Seegene Medical Foundation, Seoul, Republic of Korea
| | - So-Hyun Shin
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sang Yun Ha
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
28
|
D’Angelo A, Sobhani N, Chapman R, Bagby S, Bortoletti C, Traversini M, Ferrari K, Voltolini L, Darlow J, Roviello G. Focus on ROS1-Positive Non-Small Cell Lung Cancer (NSCLC): Crizotinib, Resistance Mechanisms and the Newer Generation of Targeted Therapies. Cancers (Basel) 2020; 12:3293. [PMID: 33172113 PMCID: PMC7694780 DOI: 10.3390/cancers12113293] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/02/2020] [Accepted: 11/05/2020] [Indexed: 12/18/2022] Open
Abstract
The treatment of patients affected by non-small cell lung cancer (NSCLC) has been revolutionised by the discovery of druggable mutations. ROS1 (c-ros oncogene) is one gene with druggable mutations in NSCLC. ROS1 is currently targeted by several specific tyrosine kinase inhibitors (TKIs), but only two of these, crizotinib and entrectinib, have received Food and Drug Administration (FDA) approval. Crizotinib is a low molecular weight, orally available TKI that inhibits ROS1, MET and ALK and is considered the gold standard first-line treatment with demonstrated significant activity for lung cancers harbouring ROS1 gene rearrangements. However, crizotinib resistance often occurs, making the treatment of ROS1-positive lung cancers more challenging. A great effort has been undertaken to identify a new generation or ROS1 inhibitors. In this review, we briefly introduce the biology and role of ROS1 in lung cancer and discuss the underlying acquired mechanisms of resistance to crizotinib and the promising new agents able to overcome resistance mechanisms and offer alternative efficient therapies.
Collapse
Affiliation(s)
- Alberto D’Angelo
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, UK; (S.B.); (J.D.)
| | - Navid Sobhani
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Robert Chapman
- University College London Hospitals NHS Foundation Trust, 235 Euston Rd, London NW1 2BU, UK;
| | - Stefan Bagby
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, UK; (S.B.); (J.D.)
| | - Carlotta Bortoletti
- Department of Dermatology, University of Padova, via Vincenzo Gallucci 4, 35121 Padova, Italy;
| | - Mirko Traversini
- Unità Operativa Anatomia Patologica, Ospedale Maggiore Carlo Alberto Pizzardi, AUSL Bologna, Largo Bartolo Nigrisoli 2, 40100 Bologna, Italy;
| | - Katia Ferrari
- Respiratory Medicine, Careggi University Hospital, 50139 Florence, Italy;
| | - Luca Voltolini
- Thoracic Surgery Unit, Careggi University Hospital, Largo Brambilla, 1, 50134 Florence, Italy;
| | - Jacob Darlow
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, UK; (S.B.); (J.D.)
| | - Giandomenico Roviello
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy;
| |
Collapse
|
29
|
Primary Intracranial Mesenchymal Tumor with EWSR1-CREM Gene Fusion: A Case Report and Literature Review. World Neurosurg 2020; 142:318-324. [PMID: 32668333 DOI: 10.1016/j.wneu.2020.07.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/30/2020] [Accepted: 07/02/2020] [Indexed: 01/29/2023]
Abstract
BACKGROUND The prevalence of gene translocation in some mesenchymal tumors can be used as highly specific molecular diagnostic markers in clinic and pathology. EWSR1 is a partner gene in a large, diverse range of mesenchymal tumors. CASE DESCRIPTION This paper describes the case of a 31-year-old man who was diagnosed with a primary intracranial mesenchymal tumor with EWSR1-CREM gene fusion and eventually returned to a normal live with no signs of tumor recurrence or metastasis after treatment, including surgery therapy, radiotherapy, and 6 cycles of vincristine-doxorubicin-cyclophosphamide chemotherapy, even though the classification and grade of the tumor are still controversial. CONCLUSIONS This case is a novel entity of intracranial mesenchymal neoplasm with EWSR1-CREM gene fusion which was confirmed by histopathology, molecular pathology, and next-generation sequencing (NGS). The literature review shows only 5 cases of intracranial tumor harboring EWSR1-CREM gene fusion with similar features. With the further application of molecular pathology and NGS in clinical practice, there will be more intracranial mesenchymal tumor cases with EWSR1-CREM gene fusion found in the future, which may lead to further understanding of the diagnosis and clinical features of this neoplasm.
Collapse
|
30
|
Expanding the Phenotypic Spectrum of Mesenchymal Tumors Harboring the EWSR1-CREM Fusion. Am J Surg Pathol 2020; 43:1622-1630. [PMID: 31305268 DOI: 10.1097/pas.0000000000001331] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
ATF1, CREB1, and CREM constitute the CREB family of transcription factors. The genes encoding these factors are involved in gene fusion events in human tumors. EWSR1-ATF1 and EWSR1-CREB1 are the 2 most characterized fusions, whereas EWSR1-CREM has been less studied. To better understand the phenotypic spectrum of mesenchymal tumors associated with the EWSR1-CREM fusion, we investigated archival cases using fluorescence in situ hybridization and/or RNA sequencing. Among 33 clear cell sarcomas of soft tissue tested, we found 1 specimen, a hand tumor bearing the rearrangements of EWSR1 and CREM, with classic histology and immunophenotype. None of 6 clear cell sarcoma-like tumors of the gastrointestinal tract tested harbored the EWSR1-CREM fusion. Among 11 angiomatoid fibrous histiocytomas, we found that 3 tumors of myxoid variant harbored the rearrangements of EWSR1 and CREM. All 3 tumors occurred in middle-aged men and involved the distal extremities (N=2) and the lung (N=1). Prominent lymphoid cuff, fibrous pseudocapsule, and amianthoid fiber were present in 3, 2, and 2 tumors, respectively, whereas none showed pseudoangiomatoid spaces. All 3 tumors were immunohistochemically positive for epithelial membrane antigen and desmin. These cases suggested a closer relationship between angiomatoid fibrous histiocytoma and a recently proposed novel group of myxoid tumors with CREB family fusions. Our cohort also included 2 unclassifiable sarcomas positive for EWSR1-CREM. One of these was an aggressive pediatric tumor of the abdominal cavity characterized by proliferation of swirling spindle cells immunopositive for cytokeratin and CD34. The other tumor derived from the chest wall of an adult and exhibited a MUC4-positive sclerosing epithelioid fibrosarcoma-like histology. Our study demonstrates that a wider phenotypic spectrum is associated with the EWSR1-CREM fusion than previously reported.
Collapse
|
31
|
Uras IZ, Sexl V, Kollmann K. CDK6 Inhibition: A Novel Approach in AML Management. Int J Mol Sci 2020; 21:ijms21072528. [PMID: 32260549 PMCID: PMC7178035 DOI: 10.3390/ijms21072528] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 03/29/2020] [Accepted: 04/02/2020] [Indexed: 02/01/2023] Open
Abstract
Acute myeloid leukemia (AML) is a complex disease with an aggressive clinical course and high mortality rate. The standard of care for patients has only changed minimally over the past 40 years. However, potentially useful agents have moved from bench to bedside with the potential to revolutionize therapeutic strategies. As such, cell-cycle inhibitors have been discussed as alternative treatment options for AML. In this review, we focus on cyclin-dependent kinase 6 (CDK6) emerging as a key molecule with distinct functions in different subsets of AML. CDK6 exerts its effects in a kinase-dependent and -independent manner which is of clinical significance as current inhibitors only target the enzymatic activity.
Collapse
Affiliation(s)
- Iris Z. Uras
- Department of Pharmacology, Center of Physiology and Pharmacology & Comprehensive Cancer Center (CCC), Medical University of Vienna, 1090 Vienna, Austria;
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria;
| | - Karoline Kollmann
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria;
- Correspondence: ; Tel.: + 43-1-25077-2917
| |
Collapse
|
32
|
The Angiosarcoma Project: enabling genomic and clinical discoveries in a rare cancer through patient-partnered research. Nat Med 2020; 26:181-187. [PMID: 32042194 DOI: 10.1038/s41591-019-0749-z] [Citation(s) in RCA: 183] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 12/23/2019] [Indexed: 11/09/2022]
Abstract
Despite rare cancers accounting for 25% of adult tumors1, they are difficult to study due to the low disease incidence and geographically dispersed patient populations, which has resulted in significant unmet clinical needs for patients with rare cancers. We assessed whether a patient-partnered research approach using online engagement can overcome these challenges, focusing on angiosarcoma, a sarcoma with an annual incidence of 300 cases in the United States. Here we describe the development of the Angiosarcoma Project (ASCproject), an initiative enabling US and Canadian patients to remotely share their clinical information and biospecimens for research. The project generates and publicly releases clinically annotated genomic data on tumor and germline specimens on an ongoing basis. Over 18 months, 338 patients registered for the ASCproject, which comprises a large proportion of all patients with angiosarcoma. Whole-exome sequencing (WES) of 47 tumors revealed recurrently mutated genes that included KDR, TP53, and PIK3CA. PIK3CA-activating mutations were observed predominantly in primary breast angiosarcoma, which suggested a therapeutic rationale. Angiosarcoma of the head, neck, face and scalp (HNFS) was associated with a high tumor mutation burden (TMB) and a dominant ultraviolet damage mutational signature, which suggested that for the subset of patients with angiosarcoma of HNFS, ultraviolet damage may be a causative factor and that immune checkpoint inhibition may be beneficial. Medical record review revealed that two patients with HNFS angiosarcoma had received off-label therapeutic use of antibody to the programmed death-1 protein (anti-PD-1) and had experienced exceptional responses, which highlights immune checkpoint inhibition as a therapeutic avenue for HNFS angiosarcoma. This patient-partnered approach has catalyzed an opportunity to discover the etiology and potential therapies for patients with angiosarcoma. Collectively, this proof-of-concept study demonstrates that empowering patients to directly participate in research can overcome barriers in rare diseases and can enable discoveries.
Collapse
|
33
|
Amino Acid Transporters and Exchangers from the SLC1A Family: Structure, Mechanism and Roles in Physiology and Cancer. Neurochem Res 2020; 45:1268-1286. [DOI: 10.1007/s11064-019-02934-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/10/2019] [Accepted: 12/13/2019] [Indexed: 12/13/2022]
|
34
|
Salvucci M, Zakaria Z, Carberry S, Tivnan A, Seifert V, Kögel D, Murphy BM, Prehn JHM. System-based approaches as prognostic tools for glioblastoma. BMC Cancer 2019; 19:1092. [PMID: 31718568 PMCID: PMC6852738 DOI: 10.1186/s12885-019-6280-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 10/09/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The evasion of apoptosis is a hallmark of cancer. Understanding this process holistically and overcoming apoptosis resistance is a goal of many research teams in order to develop better treatment options for cancer patients. Efforts are also ongoing to personalize the treatment of patients. Strategies to confirm the therapeutic efficacy of current treatments or indeed to identify potential novel additional options would be extremely beneficial to both clinicians and patients. In the past few years, system medicine approaches have been developed that model the biochemical pathways of apoptosis. These systems tools incorporate and analyse the complex biological networks involved. For their successful integration into clinical practice, it is mandatory to integrate systems approaches with routine clinical and histopathological practice to deliver personalized care for patients. RESULTS We review here the development of system medicine approaches that model apoptosis for the treatment of cancer with a specific emphasis on the aggressive brain cancer, glioblastoma. CONCLUSIONS We discuss the current understanding in the field and present new approaches that highlight the potential of system medicine approaches to influence how glioblastoma is diagnosed and treated in the future.
Collapse
Affiliation(s)
- Manuela Salvucci
- Centre for Systems Medicine, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen’s Green, Dublin 2, Ireland
| | - Zaitun Zakaria
- Centre for Systems Medicine, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen’s Green, Dublin 2, Ireland
| | - Steven Carberry
- Centre for Systems Medicine, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen’s Green, Dublin 2, Ireland
| | - Amanda Tivnan
- Centre for Systems Medicine, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen’s Green, Dublin 2, Ireland
| | - Volker Seifert
- Department of Neurosurgery, Frankfurt University Hospital, Frankfurt am Main, Germany
| | - Donat Kögel
- Department of Neurosurgery, Frankfurt University Hospital, Frankfurt am Main, Germany
| | - Brona M. Murphy
- Centre for Systems Medicine, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen’s Green, Dublin 2, Ireland
| | - Jochen H. M. Prehn
- Centre for Systems Medicine, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephen’s Green, Dublin 2, Ireland
| |
Collapse
|
35
|
|
36
|
Hanlon K, Thompson A, Pantano L, Hutchinson JN, Al-Obeidi A, Wang S, Bliss-Moreau M, Helble J, Alexe G, Stegmaier K, Bauer DE, Croker BA. Single-cell cloning of human T-cell lines reveals clonal variation in cell death responses to chemotherapeutics. Cancer Genet 2019; 237:69-77. [PMID: 31447068 DOI: 10.1016/j.cancergen.2019.06.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 04/18/2019] [Accepted: 06/09/2019] [Indexed: 12/12/2022]
Abstract
Genetic modification of human leukemic cell lines using CRISPR-Cas9 has become a staple of gene-function studies. Single-cell cloning of modified cells is frequently used to facilitate studies of gene function. Inherent in this approach is an assumption that the genetic drift, amplified in some cell lines by mutations in DNA replication and repair machinery, as well as non-genetic factors will not introduce significant levels of experimental cellular heterogeneity in clones derived from parental populations. In this study, we characterize the variation in cell death of fifty clonal cell lines generated from human Jurkat and MOLT-4 T-cells edited by CRISPR-Cas9. We demonstrate a wide distribution of sensitivity to chemotherapeutics between non-edited clonal human leukemia T-cell lines, and also following CRISPR-Cas9 editing at the NLRP1 locus, or following transfection with non-targeting sgRNA controls. The cell death sensitivity profile of clonal cell lines was consistent across experiments and failed to revert to the non-clonal parental phenotype. Whole genome sequencing of two clonal cell lines edited by CRISPR-Cas9 revealed unique and shared genetic variants, which had minimal read support in the non-clonal parental population and were not suspected CRISPR-Cas9 off-target effects. These variants included genes related to cell death and drug metabolism. The variation in cell death phenotype of clonal populations of human T-cell lines may be a consequence of T-cell line genetic instability, and to a lesser extent clonal heterogeneity in the parental population or CRISPR-Cas9 off-target effects not predicted by current models. This work highlights the importance of genetic variation between clonal T-cell lines in the design, conduct, and analysis of experiments to investigate gene function after single-cell cloning.
Collapse
Affiliation(s)
- Kathleen Hanlon
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Alex Thompson
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Lorena Pantano
- Department of Biostatistics, Harvard Chan School of Public Health, Boston, MA, United States
| | - John N Hutchinson
- Department of Biostatistics, Harvard Chan School of Public Health, Boston, MA, United States
| | - Arshed Al-Obeidi
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Shu Wang
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Meghan Bliss-Moreau
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Jennifer Helble
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, United States
| | - Gabriela Alexe
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Boston, MA, United States
| | - Kimberly Stegmaier
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Boston, MA, United States
| | - Daniel E Bauer
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States; Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Ben A Croker
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States; Department of Pediatrics, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
37
|
Kim CY, Na K, Park S, Jeong SK, Cho JY, Shin H, Lee MJ, Han G, Paik YK. FusionPro, a Versatile Proteogenomic Tool for Identification of Novel Fusion Transcripts and Their Potential Translation Products in Cancer Cells. Mol Cell Proteomics 2019; 18:1651-1668. [PMID: 31208993 PMCID: PMC6683003 DOI: 10.1074/mcp.ra119.001456] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/23/2019] [Indexed: 01/21/2023] Open
Abstract
Fusion proteoforms are translation products derived from gene fusion. Although very rare, the fusion proteoforms play important roles in biomedical science. For example, fusion proteoforms influence the development of tumors by serving as cancer markers or cell cycle regulators. Although numerous studies have reported bioinformatics tools that can predict fusion transcripts, few proteogenomic tools are available that can predict and identify proteoforms. In this study, we develop a versatile proteogenomic tool "FusionPro," which facilitates the identification of fusion transcripts and their potential translatable peptides. FusionPro provides an independent gene fusion prediction module and can build sequence databases for annotated fusion proteoforms. FusionPro shows greater sensitivity than the available fusion finders when analyzing simulated or real RNA sequencing data sets. We use FusionPro to identify 18 fusion junction peptides and three potential fusion-derived peptides by MS/MS-based analysis of leukemia cell lines (Jurkat and K562) and ovarian cancer tissues from the Clinical Proteomic Tumor Analysis Consortium. Among the identified fusion proteins, we molecularly validate two fusion junction isoforms and a translation product of FAM133B:CDK6. Moreover, sequence analysis suggests that the fusion protein participates in the cell cycle progression. In addition, our prediction results indicate that fusion transcripts often have multiple fusion junctions and that these fusion junctions tend to be distributed in a nonrandom pattern at both the chromosome and gene levels. Thus, FusionPro allows users to detect various types of fusion translation products using a transcriptome-informed approach and to gain a comprehensive understanding of the formation and biological roles of fusion proteoforms.
Collapse
Affiliation(s)
- Chae-Yeon Kim
- ‡Interdisciplinary Program of Integrated OMICS for Biomedical Science, The Graduate School, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea; §Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Keun Na
- §Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Saeram Park
- §Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Seul-Ki Jeong
- §Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Jin-Young Cho
- §Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Heon Shin
- §Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Min Jung Lee
- §Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Gyoonhee Han
- ¶Department of Pharmacy, College of Pharmacy, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Young-Ki Paik
- §Yonsei Proteome Research Center, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea.
| |
Collapse
|
38
|
Khan S, Liu Y, Siddique R, Nabi G, Xue M, Hou H. Impact of chronically alternating light-dark cycles on circadian clock mediated expression of cancer (glioma)-related genes in the brain. Int J Biol Sci 2019; 15:1816-1834. [PMID: 31523185 PMCID: PMC6743288 DOI: 10.7150/ijbs.35520] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 05/15/2019] [Indexed: 12/16/2022] Open
Abstract
Disruption of the circadian rhythm is a risk factor for cancer, while glioma is a leading contributor to mortality worldwide. Substantial efforts are being undertaken to decrypt underlying molecular pathways. Our understanding of the mechanisms through which disrupted circadian rhythm induces glioma development and progression is incomplete. We, therefore, examined changes in the expression of glioma-related genes in the mouse brain after chronic jetlag (CJL) exposure. A total of 22 candidate tumor suppressor (n= 14) and oncogenes (n= 8) were identified and analyzed for their interaction with clock genes. Both the control and CJL groups were investigated for the expression of candidate genes in the nucleus accumbens, hippocampus, prefrontal cortex, hypothalamus, and striatum of wild type, Bmal1-/- and Cry1/2 double knockout male mice. We found significant variations in the expression of candidate tumor suppressor and oncogenes in the brain tissues after CJL treatment in the wild type, Bmal1-/- and Cry1/2 double knockout mice. In response to CJL treatment, some of the genes were regulated in the wild type, Bmal1-/- and Cry1/2 similarly. However, the expression of some of the genes indicated their association with the functional clock. Overall, our result suggests a link between CJL and gliomas risk at least partially dependent on the circadian clock. However, further studies are needed to investigate the molecular mechanism associated with CJL and gliomas.
Collapse
Affiliation(s)
- Suliman Khan
- The Key Laboratory of Aquatic Biodiversity and Conservation of Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Wuhan, Hubei 430074, China
- University of Chinese Academy of Sciences, Beijing, China
- The Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Liu
- The Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, China
| | - Rabeea Siddique
- The Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, China
| | - Ghulam Nabi
- The Key Laboratory of Aquatic Biodiversity and Conservation of Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Mengzhou Xue
- The Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, China
| | - Hongwei Hou
- The Key Laboratory of Aquatic Biodiversity and Conservation of Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
39
|
Targeting ROS1 Rearrangements in Non-small Cell Lung Cancer: Crizotinib and Newer Generation Tyrosine Kinase Inhibitors. Drugs 2019; 79:1277-1286. [DOI: 10.1007/s40265-019-01164-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
40
|
Mini E, Lapucci A, Perrone G, D'Aurizio R, Napoli C, Brugia M, Landini I, Tassi R, Picariello L, Simi L, Mancini I, Messerini L, Magi A, Pinzani P, Mazzei T, Tonelli F, Nobili S. RNA sequencing reveals PNN and KCNQ1OT1 as predictive biomarkers of clinical outcome in stage III colorectal cancer patients treated with adjuvant chemotherapy. Int J Cancer 2019; 145:2580-2593. [PMID: 30973654 DOI: 10.1002/ijc.32326] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 03/13/2019] [Accepted: 04/01/2019] [Indexed: 12/16/2022]
Abstract
Five-year overall survival of stage III colorectal cancer (CRC) patients treated with standard adjuvant chemotherapy (ACHT) is highly variable. Genomic biomarkers and/or transcriptomic profiles identified lack of adequate validation. Aim of our study was to identify and validate molecular biomarkers predictive of ACHT response in stage III CRC patients by a transcriptomic approach. From a series of CRC patients who received ACHT, two stage III extreme cohorts (unfavorable vs. favorable prognosis) were selected. RNA-sequencing was performed from fresh frozen explants. Tumors were characterized for somatic mutations. Validation was performed in stage III CRC patients extracted from two GEO datasets. According to disease-free survival (DFS), 108 differentially expressed genes (104/4 up/downregulated in the unfavorable prognosis group) were identified. Among 104 upregulated genes, 42 belonged to olfactory signaling pathways, 62 were classified as pseudogenes (n = 17), uncharacterized noncoding RNA (n = 10), immune response genes (n = 4), microRNA (n = 1), cancer-related genes (n = 14) and cancer-unrelated genes (n = 16). Three out of four down-regulated genes were cancer-related. Mutational status (i.e., RAS, BRAF, PIK3CA) did not differ among the cohorts. In the validation cohort, multivariate analysis showed high PNN and KCNQ1OT1 expression predictive of shorter DFS in ACHT treated patients (p = 0.018 and p = 0.014, respectively); no difference was observed in untreated patients. This is the first study that identifies by a transcriptomic approach and validates PNN and KCNQ1OT1 as molecular biomarkers predictive of chemotherapy response in stage III CRC patients. After a further validation in an independent cohort, PNN and KCNQ1OT1 evaluation could be proposed to prospectively identify stage III CRC patients benefiting from ACHT.
Collapse
Affiliation(s)
- Enrico Mini
- Department of Health Sciences, University of Florence, Florence, Italy.,DENOTHE Excellence Center, University of Florence, Florence, Italy
| | - Andrea Lapucci
- Department of Health Sciences, University of Florence, Florence, Italy.,DENOTHE Excellence Center, University of Florence, Florence, Italy
| | - Gabriele Perrone
- Department of Health Sciences, University of Florence, Florence, Italy.,DENOTHE Excellence Center, University of Florence, Florence, Italy
| | - Romina D'Aurizio
- Institute of Informatics and Telematics (IIT), National Research Council (CNR), Pisa, Italy
| | - Cristina Napoli
- Department of Health Sciences, University of Florence, Florence, Italy.,DENOTHE Excellence Center, University of Florence, Florence, Italy
| | - Marco Brugia
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Ida Landini
- Department of Health Sciences, University of Florence, Florence, Italy.,DENOTHE Excellence Center, University of Florence, Florence, Italy
| | - Renato Tassi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Lucia Picariello
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Lisa Simi
- Molecular and Clinical Biochemistry Laboratory, Careggi University Hospital, Florence, Italy
| | - Irene Mancini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Luca Messerini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alberto Magi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Pamela Pinzani
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy.,Molecular and Clinical Biochemistry Laboratory, Careggi University Hospital, Florence, Italy
| | - Teresita Mazzei
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Francesco Tonelli
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | - Stefania Nobili
- Department of Health Sciences, University of Florence, Florence, Italy.,DENOTHE Excellence Center, University of Florence, Florence, Italy
| |
Collapse
|
41
|
Weidema M, Versleijen-Jonkers Y, Flucke U, Desar I, van der Graaf W. Targeting angiosarcomas of the soft tissues: A challenging effort in a heterogeneous and rare disease. Crit Rev Oncol Hematol 2019; 138:120-131. [DOI: 10.1016/j.critrevonc.2019.04.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 01/08/2019] [Accepted: 04/09/2019] [Indexed: 02/06/2023] Open
|
42
|
Picco G, Chen ED, Alonso LG, Behan FM, Gonçalves E, Bignell G, Matchan A, Fu B, Banerjee R, Anderson E, Butler A, Benes CH, McDermott U, Dow D, Iorio F, Stronach E, Yang F, Yusa K, Saez-Rodriguez J, Garnett MJ. Functional linkage of gene fusions to cancer cell fitness assessed by pharmacological and CRISPR-Cas9 screening. Nat Commun 2019; 10:2198. [PMID: 31097696 PMCID: PMC6522557 DOI: 10.1038/s41467-019-09940-1] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 04/09/2019] [Indexed: 12/21/2022] Open
Abstract
Many gene fusions are reported in tumours and for most their role remains unknown. As fusions are used for diagnostic and prognostic purposes, and are targets for treatment, it is crucial to assess their function in cancer. To systematically investigate the role of fusions in tumour cell fitness, we utilized RNA-sequencing data from 1011 human cancer cell lines to functionally link 8354 fusion events with genomic data, sensitivity to >350 anti-cancer drugs and CRISPR-Cas9 loss-of-fitness effects. Established clinically-relevant fusions were identified. Overall, detection of functional fusions was rare, including those involving cancer driver genes, suggesting that many fusions are dispensable for tumour fitness. Therapeutically actionable fusions involving RAF1, BRD4 and ROS1 were verified in new histologies. In addition, recurrent YAP1-MAML2 fusions were identified as activators of Hippo-pathway signaling in multiple cancer types. Our approach discriminates functional fusions, identifying new drivers of carcinogenesis and fusions that could have clinical implications. Gene fusions are observed in many cancers but their link to tumour fitness is largely unknown. Here, transcriptomic analysis combined with pharmacological and CRISPR-Cas9 screening of cancer cell lines was used to evaluate the functional linkage between fusions and tumour fitness.
Collapse
Affiliation(s)
- Gabriele Picco
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
| | - Elisabeth D Chen
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
| | - Luz Garcia Alonso
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, CB10 1SD, UK.,Open Targets, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
| | - Fiona M Behan
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK.,Open Targets, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
| | - Emanuel Gonçalves
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
| | - Graham Bignell
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
| | - Angela Matchan
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
| | - Beiyuan Fu
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
| | - Ruby Banerjee
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
| | - Elizabeth Anderson
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
| | - Adam Butler
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
| | - Cyril H Benes
- Massachusetts General Hospital, 55 Fruit Street, Boston, MA, 02114, USA
| | - Ultan McDermott
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK.,AstraZeneca, CRUK Cambridge Institute, Cambridge, CB2 0RE, UK
| | - David Dow
- Open Targets, Wellcome Genome Campus, Cambridge, CB10 1SA, UK.,Research and Development, GlaxoSmithKline, Stevenage, SG1 2NY, UK.,Research and Development, GlaxoSmithKline, Collegeville, PA, 19426-0989, USA
| | - Francesco Iorio
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK.,European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, CB10 1SD, UK.,Open Targets, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
| | - Euan Stronach
- Open Targets, Wellcome Genome Campus, Cambridge, CB10 1SA, UK.,Research and Development, GlaxoSmithKline, Stevenage, SG1 2NY, UK.,Research and Development, GlaxoSmithKline, Collegeville, PA, 19426-0989, USA
| | - Fengtang Yang
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
| | - Kosuke Yusa
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
| | - Julio Saez-Rodriguez
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, CB10 1SD, UK.,Open Targets, Wellcome Genome Campus, Cambridge, CB10 1SA, UK.,Institute for Computational Biomedicine, Faculty of Medicine, Bioquant, Heidelberg University, 69120, Heidelberg, Germany
| | - Mathew J Garnett
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK. .,Open Targets, Wellcome Genome Campus, Cambridge, CB10 1SA, UK.
| |
Collapse
|
43
|
CDK6 coordinates JAK2 V617F mutant MPN via NF-κB and apoptotic networks. Blood 2019; 133:1677-1690. [PMID: 30635286 DOI: 10.1182/blood-2018-08-872648] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 01/07/2019] [Indexed: 01/27/2023] Open
Abstract
Over 80% of patients with myeloproliferative neoplasms (MPNs) harbor the acquired somatic JAK2 V617F mutation. JAK inhibition is not curative and fails to induce a persistent response in most patients, illustrating the need for the development of novel therapeutic approaches. We describe a critical role for CDK6 in MPN evolution. The absence of Cdk6 ameliorates clinical symptoms and prolongs survival. The CDK6 protein interferes with 3 hallmarks of disease: besides regulating malignant stem cell quiescence, it promotes nuclear factor κB (NF-κB) signaling and contributes to cytokine production while inhibiting apoptosis. The effects are not mirrored by palbociclib, showing that the functions of CDK6 in MPN pathogenesis are largely kinase independent. Our findings thus provide a rationale for targeting CDK6 in MPN.
Collapse
|
44
|
Marks EI, Pamarthy S, Dizon D, Birnbaum A, Yakirevich E, Safran H, Carneiro BA. ROS1-GOPC/FIG: a novel gene fusion in hepatic angiosarcoma. Oncotarget 2019; 10:245-251. [PMID: 30719217 PMCID: PMC6349438 DOI: 10.18632/oncotarget.26521] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 12/12/2018] [Indexed: 12/11/2022] Open
Abstract
Hepatic angiosarcoma (HAS) is a rare and highly lethal malignancy with few effective systemic treatments. Relatively little is known about the genetic abnormalities that drive this disease. As a result, there has been minimal progress towards applying targeted therapies to the treatment of HAS. We describe the first reported case of a patient with HAS that harbored a fusion of ROS1 with GOPC/FIG. Similar to other rearrangements involving ROS1, the resulting fusion protein is believed to act as a major driver of carcinogenesis and may be subject to inhibition by drugs that target ROS1 such as crizotinib. We then queried the MSK-IMPACT clinical sequencing cohort and cBioportal datasets, demonstrating the previously unknown prevalence of ROS1-GOPC fusions in soft tissue sarcomas and hepatobiliary cancers. Amplification of these genes was also found to correlate with reduced overall survival. This is followed by a review of the role played by ROS1 rearrangements in cancer, as well as the evidence supporting the use of targeted therapies against the resulting fusion protein. We suggest that testing for ROS1 fusion and, if positive, treatment with a targeted therapy could be considered at the time of diagnosis for patients with angiosarcoma. This report also highlights the need for further investigation into the molecular pathophysiology of this deadly disease.
Collapse
Affiliation(s)
- Eric I. Marks
- Division of Hematology-Oncology, Lifespan Cancer Institute, Warren-Alpert Medical School of Brown University, Providence, RI, USA
| | - Sahithi Pamarthy
- Atrin Pharmaceuticals, Pennsylvania Biotechnology Center, Doylestown, PA, USA
| | - Don Dizon
- Division of Hematology-Oncology, Lifespan Cancer Institute, Warren-Alpert Medical School of Brown University, Providence, RI, USA
| | - Ari Birnbaum
- Division of Hematology-Oncology, Lifespan Cancer Institute, Warren-Alpert Medical School of Brown University, Providence, RI, USA
| | - Evgeny Yakirevich
- Department of Pathology, Lifespan Cancer Institute, Warren-Alpert Medical School of Brown University, Providence, RI, USA
| | - Howard Safran
- Division of Hematology-Oncology, Lifespan Cancer Institute, Warren-Alpert Medical School of Brown University, Providence, RI, USA
| | - Benedito A. Carneiro
- Division of Hematology-Oncology, Lifespan Cancer Institute, Warren-Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
45
|
Ali NM, Niada S, Brini AT, Morris MR, Kurusamy S, Alholle A, Huen D, Antonescu CR, Tirode F, Sumathi V, Latif F. Genomic and transcriptomic characterisation of undifferentiated pleomorphic sarcoma of bone. J Pathol 2018; 247:166-176. [PMID: 30281149 DOI: 10.1002/path.5176] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 08/24/2018] [Accepted: 09/25/2018] [Indexed: 12/13/2022]
Abstract
Undifferentiated pleomorphic sarcoma of bone (UPSb) is a rare primary bone sarcoma that lacks a specific line of differentiation. There is very little information about the genetic alterations leading to tumourigenesis or malignant transformation. Distinguishing between UPSb and other malignant bone sarcomas, including dedifferentiated chondrosarcoma and osteosarcoma, can be challenging due to overlapping features. To explore the genomic and transcriptomic landscape of UPSb tumours, whole-exome sequencing (WES) and RNA sequencing (RNA-Seq) were performed on UPSb tumours. All tumours lacked hotspot mutations in IDH1/2 132 or 172 codons, thereby excluding the diagnosis of dedifferentiated chondrosarcoma. Recurrent somatic mutations in TP53 were identified in four of 14 samples (29%). Moreover, recurrent mutations in histone chromatin remodelling genes, including H3F3A, ATRX and DOT1L, were identified in five of 14 samples (36%), highlighting the potential role of deregulated chromatin remodelling pathways in UPSb tumourigenesis. The majority of recurrent mutations in chromatin remodelling genes identified here are reported in COSMIC, including the H3F3A G34 and K36 hotspot residues. Copy number alteration analysis identified gains and losses in genes that have been previously altered in UPSb or UPS of soft tissue. Eight somatic gene fusions were identified by RNA-Seq, two of which, CLTC-VMP1 and FARP1-STK24, were reported previously in multiple cancers. Five gene fusions were genomically characterised. Hierarchical clustering analysis, using RNA-Seq data, distinctly clustered UPSb tumours from osteosarcoma and other sarcomas, thus molecularly distinguishing UPSb from other sarcomas. RNA-Seq expression profiling analysis and quantitative reverse transcription-polymerase chain reaction showed an elevated expression in FGF23, which can be a potential molecular biomarker for UPSb. To our knowledge, this study represents the first comprehensive WES and RNA-Seq analysis of UPSb tumours revealing novel protein-coding recurrent gene mutations, gene fusions and identifying a potential UPSb molecular biomarker, thereby broadening the understanding of the pathogenic mechanisms and highlighting the possibility of developing novel targeted therapeutics. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Naser M Ali
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Stefania Niada
- Laboratory of Biotechnological Applications, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Anna T Brini
- Laboratory of Biotechnological Applications, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy.,Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, Milan, Italy
| | - Mark R Morris
- Research Institute in Healthcare Science, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, UK
| | - Sathishkumar Kurusamy
- Research Institute in Healthcare Science, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, UK
| | - Abdullah Alholle
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - David Huen
- Research Institute in Healthcare Science, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, UK
| | - Cristina R Antonescu
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Franck Tirode
- Department of Translational Research and Innovation, Centre Léon Bérard, Université Claude Bernard Lyon 1, CNRS 5286, INSERM U1052, Cancer Research Center of Lyon, Lyon, France
| | - Vaiyapuri Sumathi
- Department of Musculoskeletal Pathology, The Royal Orthopaedic Hospital, Robert Aitken Institute of Clinical Research, University of Birmingham, Birmingham, UK
| | - Farida Latif
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
46
|
Abstract
Somatic structural variants undoubtedly play important roles in driving tumourigenesis. This is evident despite the substantial technical challenges that remain in accurately detecting structural variants and their breakpoints in tumours and in spite of our incomplete understanding of the impact of structural variants on cellular function. Developments in these areas of research contribute to the ongoing discovery of structural variation with a clear impact on the evolution of the tumour and on the clinical importance to the patient. Recent large whole genome sequencing studies have reinforced our impression of each tumour as a unique combination of mutations but paradoxically have also discovered similar genome-wide patterns of single-nucleotide and structural variation between tumours. Statistical methods have been developed to deconvolute mutation patterns, or signatures, that recur across samples, providing information about the mutagens and repair processes that may be active in a given tumour. These signatures can guide treatment by, for example, highlighting vulnerabilities in a particular tumour to a particular chemotherapy. Thus, although the complete reconstruction of the full evolutionary trajectory of a tumour genome remains currently out of reach, valuable data are already emerging to improve the treatment of cancer.
Collapse
Affiliation(s)
- Ailith Ewing
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, The University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH42XU, UK
| | - Colin Semple
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, The University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH42XU, UK
| |
Collapse
|
47
|
Detection of specific gene rearrangements by fluorescence in situ hybridization in 16 cases of clear cell sarcoma of soft tissue and 6 cases of clear cell sarcoma-like gastrointestinal tumor. Diagn Pathol 2018; 13:73. [PMID: 30219084 PMCID: PMC6138919 DOI: 10.1186/s13000-018-0752-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 09/03/2018] [Indexed: 12/25/2022] Open
Abstract
Background Clear cell sarcoma of soft tissue (CCSST) and clear cell sarcoma-like gastrointestinal tumor (CCSLGT) are malignant mesenchymal tumors that share some pathological features, but they also have several different characteristics. They are well known to express chimeric fusions of Ewing sarcoma breakpoint region 1 (EWSR1) and cAMP response element-binding protein (CREB) family members; namely, EWSR1-activating transcription factor 1 (ATF1) and EWSR1-CREB1. In addition, recent studies have suggested the presence of other fusions. Methods We used fluorescence in situ hybridization to detect specific rearrangements including EWSR1, ATF1, CREB1, and cAMP response element modulator (CREM) in 16 CCSST and 6 CCSLGT cases. We also used reverse transcription polymerase chain reaction (RT-PCR) to detect specific chimeric fusions of EWSR1-ATF1 and EWSR1-CREB1 using fresh tumor samples in available cases. Results A total of 15 of 16 CCSST cases (93.8%) had EWSR1 rearrangement, of which 11 (68.8%) also had ATF1 rearrangement, suggestive of the presence of EWSR1-ATF1 fusions. One CCSST case (6.3%) was found to have EWSR1 and CREM rearrangements, and 4 of 6 CCSLGT cases (66.7%) had EWSR1 rearrangement, of which 2 (33.3%) showed ATF1 rearrangement and the other 2 cases (33.3%) showed CREB1 rearrangement. These cases most likely had EWSR1-ATF1 and EWSR1-CREB1 fusions, respectively. RT-PCR was performed in 8 available cases, including 6 CCSSTs and 2 CCSLGTs. All CCSSTs showed EWSR1-ATF1 fusions. Among the 2 CCSLGT cases, one had EWSR1-ATF1 fusion and the other had EWSR1-CREB1 fusion. Conclusions Rearrangements of EWSR1 and ATF1 or EWSR1-ATF1 fusion were predominantly found in CCSST, whereas those of EWSR1 and CREB1 or EWSR1-CREB1 tended to be detected in CCSLGT. A novel CREM fusion was also detected in a few cases of CCSST and CCSLGT. The cases in which EWSR1 rearrangement was detected without definitive partner genes should be considered for the presence of CREM rearrangement.
Collapse
|
48
|
Molecular Profiling of Hyalinizing Clear Cell Carcinomas Revealed a Subset of Tumors Harboring a Novel EWSR1-CREM Fusion. Am J Surg Pathol 2018; 42:1182-1189. [DOI: 10.1097/pas.0000000000001114] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
49
|
Gao Q, Liang WW, Foltz SM, Mutharasu G, Jayasinghe RG, Cao S, Liao WW, Reynolds SM, Wyczalkowski MA, Yao L, Yu L, Sun SQ, Chen K, Lazar AJ, Fields RC, Wendl MC, Van Tine BA, Vij R, Chen F, Nykter M, Shmulevich I, Ding L. Driver Fusions and Their Implications in the Development and Treatment of Human Cancers. Cell Rep 2018; 23:227-238.e3. [PMID: 29617662 PMCID: PMC5916809 DOI: 10.1016/j.celrep.2018.03.050] [Citation(s) in RCA: 401] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 02/25/2018] [Accepted: 03/13/2018] [Indexed: 12/11/2022] Open
Abstract
Gene fusions represent an important class of somatic alterations in cancer. We systematically investigated fusions in 9,624 tumors across 33 cancer types using multiple fusion calling tools. We identified a total of 25,664 fusions, with a 63% validation rate. Integration of gene expression, copy number, and fusion annotation data revealed that fusions involving oncogenes tend to exhibit increased expression, whereas fusions involving tumor suppressors have the opposite effect. For fusions involving kinases, we found 1,275 with an intact kinase domain, the proportion of which varied significantly across cancer types. Our study suggests that fusions drive the development of 16.5% of cancer cases and function as the sole driver in more than 1% of them. Finally, we identified druggable fusions involving genes such as TMPRSS2, RET, FGFR3, ALK, and ESR1 in 6.0% of cases, and we predicted immunogenic peptides, suggesting that fusions may provide leads for targeted drug and immune therapy.
Collapse
Affiliation(s)
- Qingsong Gao
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Wen-Wei Liang
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Steven M Foltz
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Gnanavel Mutharasu
- Institute of Signal Processing, Tampere University of Technology, 33101, Tampere, Finland
| | - Reyka G Jayasinghe
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Song Cao
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Wen-Wei Liao
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | | | - Matthew A Wyczalkowski
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Lijun Yao
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Lihua Yu
- H3 Biomedicine, Inc., Cambridge, MA 02139, USA
| | - Sam Q Sun
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Ken Chen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
| | - Alexander J Lazar
- Departments of Pathology, Genomic Medicine, and Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
| | - Ryan C Fields
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA; Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Michael C Wendl
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Mathematics, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Brian A Van Tine
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Ravi Vij
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Feng Chen
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Matti Nykter
- Institute for Biosciences and Medical Technology, University of Tampere, 33520 Tampere, Finland
| | | | - Li Ding
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA; Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63110, USA.
| |
Collapse
|
50
|
Cerrato A, Visconti R, Celetti A. The rationale for druggability of CCDC6-tyrosine kinase fusions in lung cancer. Mol Cancer 2018; 17:46. [PMID: 29455670 PMCID: PMC5817729 DOI: 10.1186/s12943-018-0799-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 02/01/2018] [Indexed: 12/12/2022] Open
Abstract
Gene fusions occur in up to 17% of solid tumours. Oncogenic kinases are often involved in such fusions. In lung cancer, almost 30% of patients carrying an activated oncogene show the fusion of a tyrosine kinase to an heterologous gene. Several genes are partner in the fusion with the three kinases ALK, ROS1 and RET in lung. The impaired function of the partner gene, in combination with the activation of the kinase, may alter the cell signaling and promote the cancer cell addiction to the oncogene. Moreover, the gene that is partner in the fusion to the kinase may affect the response to therapeutics and/or promote resistance in the cancer cells. Few genes are recurrent partners in tyrosine kinase fusions in lung cancer, including CCDC6, a recurrent partner in ROS1 and RET fusions, that can be selected as possible target for new strategies of combined therapy including TKi.
Collapse
Affiliation(s)
- Aniello Cerrato
- Institute for the Experimental Endocrinology and Oncology "Gaetano Salvatore", Italian National Council of Research, Via S. Pansini 5, 80131, Naples, Italy.
| | - Roberta Visconti
- Institute for the Experimental Endocrinology and Oncology "Gaetano Salvatore", Italian National Council of Research, Via S. Pansini 5, 80131, Naples, Italy
| | - Angela Celetti
- Institute for the Experimental Endocrinology and Oncology "Gaetano Salvatore", Italian National Council of Research, Via S. Pansini 5, 80131, Naples, Italy.
| |
Collapse
|