1
|
Wang J, Walker RL, Hornicek FJ, Shi H, Duan Z. Inhibition of discoidin domain receptor 1 as a new therapeutic strategy for osteosarcoma. FASEB J 2024; 38:e70239. [PMID: 39641588 DOI: 10.1096/fj.202401508rr] [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: 07/09/2024] [Revised: 11/18/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
Osteosarcoma is the most common type of bone cancer. Some patients eventually develop recurrent or metastatic diseases and treatment options are extremely limited. Discoidin domain receptor 1 (DDR1) is a unique collagen-activated tyrosine kinase that participates in various human diseases, including cancer. DDR1 promotes adhesion, proliferation, differentiation, migration, and metastasis of cancer cells. The purpose of this study is to assess the expression, clinical prognostic relationship and functional roles of DDR1 in osteosarcoma. The correlation between DDR1 expression in tumor tissues and clinicopathological features, and prognosis was assessed via immunohistochemical staining of a unique tissue microarray (TMA) constructed from osteosarcoma specimens. DDR1-specific siRNA and a highly selective DDR1 inhibitor, 7rh, were applied to determine the impact of DDR1 expression on osteosarcoma cell growth and proliferation. Furthermore, the effect of DDR1 inhibition on clonogenicity was evaluated using a clonogenic assay, and a 3D cell culture model was used to mimic DDR1 effects in an in vivo environment. The results demonstrate that higher DDR1 expression significantly correlates with recurrence, metastasis, and shorter overall survival in osteosarcoma patients. The expression of DDR1 is also inversely correlated to the response to neoadjuvant chemotherapy. Therapeutically, DDR1 knockdown with siRNA or selective inhibition with 7rh decreases the proliferation and growth of osteosarcoma cells. In conclusion, our study supports DDR1 expression as an independent predictor of poor prognosis and a promising therapeutic target for osteosarcoma.
Collapse
Affiliation(s)
- Jinglu Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Robert L Walker
- Sarcoma Biology Laboratory, Department of Orthopedic Surgery, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Francis J Hornicek
- Sarcoma Biology Laboratory, Department of Orthopedic Surgery, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Huirong Shi
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhenfeng Duan
- Sarcoma Biology Laboratory, Department of Orthopedic Surgery, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
2
|
Li Z, Li X, Seebacher NA, Liu X, Wu W, Yu S, Hornicek FJ, Huang C, Duan Z. CDK12 is a promising therapeutic target for the transcription cycle and DNA damage response in metastatic osteosarcoma. Carcinogenesis 2024; 45:786-798. [PMID: 39082894 DOI: 10.1093/carcin/bgae051] [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: 02/12/2024] [Revised: 07/19/2024] [Accepted: 07/30/2024] [Indexed: 10/11/2024] Open
Abstract
Osteosarcoma (OS) is a bone malignant tumor affecting children, adolescents, and young adults. Currently, osteosarcoma is treated with chemotherapy regimens established over 40 years ago. The investigation of novel therapeutic strategies for the treatment of osteosarcoma remains an important clinical need. Cyclin-dependent kinases (CDKs) have been considered promising molecular targets in cancer therapy. Among these, CDK12 has been shown to play a crucial role in the pathogenesis of malignancies, but its clinical significance and biological mechanisms in osteosarcoma remain unclear. In the present study, we aim to determine the expression and function of CDK12 and evaluate its prognostic and therapeutic value in metastatic osteosarcoma. We found that overexpression of CDK12 was associated with high tumor grade, tumor progression and reduced patient survival. The underlying mechanism revealed that knockdown of CDK12 expression with small interfering RNA or functional inhibition with the CDK12-targeting agent THZ531 effectively exhibited time- and dose-dependent cytotoxicity. Downregulation of CDK12 paused transcription by reducing RNAP II phosphorylation, interfered with DNA damage repair with increased γH2AX, and decreased cell proliferation through the PI3K-AKT pathway. This was accompanied by the promotion of apoptosis, as evidenced by enhanced Bax expression and reduced Bcl-xL expression. Furthermore, the CDK12 selective inhibitor THZ531 also hindered ex vivo 3D spheroid formation, growth of in vitro 2D cell colony, and prevented cell mobility. Our findings highlight the clinical importance of CDK12 as a potentially valuable prognostic biomarker and therapeutic target in metastatic osteosarcoma.
Collapse
Affiliation(s)
- Zihao Li
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 10021China
| | - Xiaoyang Li
- Department of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 10021China
| | - Nicole A Seebacher
- Department of Oncology, University of Oxford OX3 9DU, Oxford, UK
- St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Xu Liu
- Department of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 10021China
| | - Wence Wu
- Department of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 10021China
| | - Shengji Yu
- Department of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 10021China
| | - Francis J Hornicek
- Sarcoma Biology Laboratory, Department of Orthopaedics, Sylvester Comprehensive Cancer Center, and the University of Miami Miller School of Medicine, Miami, FL, 33136USA
| | - Changzhi Huang
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 10021China
| | - Zhenfeng Duan
- Sarcoma Biology Laboratory, Department of Orthopaedics, Sylvester Comprehensive Cancer Center, and the University of Miami Miller School of Medicine, Miami, FL, 33136USA
| |
Collapse
|
3
|
Yan P, Wang J, Yue B, Wang X. Unraveling molecular aberrations and pioneering therapeutic strategies in osteosarcoma. Biochim Biophys Acta Rev Cancer 2024; 1879:189171. [PMID: 39127243 DOI: 10.1016/j.bbcan.2024.189171] [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: 02/04/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
Osteosarcoma, a rare primary bone cancer, presents diverse molecular aberrations that underscore its complexity. Despite the persistent endeavors by researchers, the limited amelioration in the five-year survival rate indicates that current therapeutic strategies prove inadequate in addressing the clinical necessities. Advancements in molecular profiling have facilitated an enhanced comprehension of the biology of osteosarcoma, offering a promising outlook for treatment. There is an urgent need to develop innovative approaches to address the complex challenges of osteosarcoma, ultimately contributing to enhanced patient outcomes. This review explores the nexus between osteosarcoma and cancer predisposition syndromes, intricacies in its somatic genome, and clinically actionable alterations. This review covers treatment strategies, including surgery, chemotherapy, immune checkpoint inhibitors (ICIs), and tyrosine kinase inhibitors (TKIs). Innovative treatment modalities targeting diverse pathways, including multi-target tyrosine kinases, cell cycle, PI3K/mTOR pathway, and DNA damage repair (DDR), offer promising interventions. This review also covers promising avenues, including antibody-drug conjugates (ADCs) and immunotherapy strategies, such as cytokines, adoptive cellular therapy (ACT), ICIs, and cancer vaccines. This comprehensive exploration contributes to a holistic understanding, offering guidance for clinical applications to advance the management of osteosarcoma.
Collapse
Affiliation(s)
- Peng Yan
- Department of Orthopedic Oncology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, China
| | - Jie Wang
- Department of Orthopedic Oncology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, China
| | - Bin Yue
- Department of Orthopedic Oncology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, China.
| | - Xinyi Wang
- Department of Pharmacy, the Affiliated Hospital of Qingdao University, Qingdao, Shandong 266100, China.
| |
Collapse
|
4
|
Zhao SJ, Prior D, Heske CM, Vasquez JC. Therapeutic Targeting of DNA Repair Pathways in Pediatric Extracranial Solid Tumors: Current State and Implications for Immunotherapy. Cancers (Basel) 2024; 16:1648. [PMID: 38730598 PMCID: PMC11083679 DOI: 10.3390/cancers16091648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 04/21/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
DNA damage is fundamental to tumorigenesis, and the inability to repair DNA damage is a hallmark of many human cancers. DNA is repaired via the DNA damage repair (DDR) apparatus, which includes five major pathways. DDR deficiencies in cancers give rise to potential therapeutic targets, as cancers harboring DDR deficiencies become increasingly dependent on alternative DDR pathways for survival. In this review, we summarize the DDR apparatus, and examine the current state of research efforts focused on identifying vulnerabilities in DDR pathways that can be therapeutically exploited in pediatric extracranial solid tumors. We assess the potential for synergistic combinations of different DDR inhibitors as well as combinations of DDR inhibitors with chemotherapy. Lastly, we discuss the immunomodulatory implications of targeting DDR pathways and the potential for using DDR inhibitors to enhance tumor immunogenicity, with the goal of improving the response to immune checkpoint blockade in pediatric solid tumors. We review the ongoing and future research into DDR in pediatric tumors and the subsequent pediatric clinical trials that will be critical to further elucidate the efficacy of the approaches targeting DDR.
Collapse
Affiliation(s)
- Sophia J. Zhao
- Department of Pediatric Hematology/Oncology, Yale University School of Medicine, New Haven, CT 06510, USA; (S.J.Z.); (D.P.)
| | - Daniel Prior
- Department of Pediatric Hematology/Oncology, Yale University School of Medicine, New Haven, CT 06510, USA; (S.J.Z.); (D.P.)
| | - Christine M. Heske
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Juan C. Vasquez
- Department of Pediatric Hematology/Oncology, Yale University School of Medicine, New Haven, CT 06510, USA; (S.J.Z.); (D.P.)
| |
Collapse
|
5
|
Pusch FF, Dorado García H, Xu R, Gürgen D, Bei Y, Brückner L, Röefzaad C, von Stebut J, Bardinet V, Chamorro Gonzalez R, Eggert A, Schulte JH, Hundsdörfer P, Seifert G, Haase K, Schäfer BW, Wachtel M, Kühl AA, Ortiz MV, Wengner AM, Scheer M, Henssen AG. Elimusertib has Antitumor Activity in Preclinical Patient-Derived Pediatric Solid Tumor Models. Mol Cancer Ther 2024; 23:507-519. [PMID: 38159110 PMCID: PMC10985474 DOI: 10.1158/1535-7163.mct-23-0094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 09/12/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024]
Abstract
The small-molecule inhibitor of ataxia telangiectasia and Rad3-related protein (ATR), elimusertib, is currently being tested clinically in various cancer entities in adults and children. Its preclinical antitumor activity in pediatric malignancies, however, is largely unknown. We here assessed the preclinical activity of elimusertib in 38 cell lines and 32 patient-derived xenograft (PDX) models derived from common pediatric solid tumor entities. Detailed in vitro and in vivo molecular characterization of the treated models enabled the evaluation of response biomarkers. Pronounced objective response rates were observed for elimusertib monotherapy in PDX, when treated with a regimen currently used in clinical trials. Strikingly, elimusertib showed stronger antitumor effects than some standard-of-care chemotherapies, particularly in alveolar rhabdomysarcoma PDX. Thus, elimusertib has strong preclinical antitumor activity in pediatric solid tumor models, which may translate to clinically meaningful responses in patients.
Collapse
Affiliation(s)
- Fabian F. Pusch
- Experimental and Clinical Research Center (ECRC) of the MDC and Charité Berlin, Berlin, Germany
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Heathcliff Dorado García
- Experimental and Clinical Research Center (ECRC) of the MDC and Charité Berlin, Berlin, Germany
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Robin Xu
- Experimental and Clinical Research Center (ECRC) of the MDC and Charité Berlin, Berlin, Germany
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Dennis Gürgen
- Experimental Pharmacology and Oncology (EPO), Berlin, Germany
| | - Yi Bei
- Experimental and Clinical Research Center (ECRC) of the MDC and Charité Berlin, Berlin, Germany
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Lotte Brückner
- Experimental and Clinical Research Center (ECRC) of the MDC and Charité Berlin, Berlin, Germany
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Max-Delbrück-Centrum für Molekulare Medizin (BIMSB/BIH), Berlin, Germany
| | - Claudia Röefzaad
- Experimental and Clinical Research Center (ECRC) of the MDC and Charité Berlin, Berlin, Germany
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jennifer von Stebut
- Experimental and Clinical Research Center (ECRC) of the MDC and Charité Berlin, Berlin, Germany
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Victor Bardinet
- Experimental and Clinical Research Center (ECRC) of the MDC and Charité Berlin, Berlin, Germany
| | - Rocío Chamorro Gonzalez
- Experimental and Clinical Research Center (ECRC) of the MDC and Charité Berlin, Berlin, Germany
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Angelika Eggert
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Johannes H. Schulte
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Pediatric Oncology and Hematology, University Hospital Tübingen, Tübingen, Germany
| | - Patrick Hundsdörfer
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Helios Klinikum Berlin-Buch, Berlin, Germany
| | - Georg Seifert
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Kerstin Haase
- Experimental and Clinical Research Center (ECRC) of the MDC and Charité Berlin, Berlin, Germany
| | | | | | - Anja A. Kühl
- iPATH.Berlin—Core Unit Immunopathology for Experimental Models, Charité Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Michael V. Ortiz
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York City, New York
| | | | - Monika Scheer
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Anton G. Henssen
- Experimental and Clinical Research Center (ECRC) of the MDC and Charité Berlin, Berlin, Germany
- Department of Pediatric Oncology and Hematology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Max-Delbrück-Centrum für Molekulare Medizin (BIMSB/BIH), Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
- German Cancer Consortium (DKTK), partner site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
6
|
Tang S, Roberts RD, Cheng L, Li L. Osteosarcoma Multi-Omics Landscape and Subtypes. Cancers (Basel) 2023; 15:4970. [PMID: 37894336 PMCID: PMC10605601 DOI: 10.3390/cancers15204970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/12/2023] [Accepted: 09/17/2023] [Indexed: 10/29/2023] Open
Abstract
Osteosarcoma (OS) is the most common primary bone malignancy that exhibits remarkable histologic diversity and genetic heterogeneity. The complex nature of osteosarcoma has confounded precise molecular categorization, prognosis, and prediction for this disease. In this study, we performed a comprehensive multiplatform analysis on 86 osteosarcoma tumors, including somatic copy-number alteration, gene expression and methylation, and identified three molecularly distinct and clinically relevant subtypes of osteosarcoma. The subgrouping criteria was validated on another cohort of osteosarcoma tumors. Previously unappreciated osteosarcoma-type-specific changes in specific genes' copy number, expression and methylation were revealed based on the subgrouping. The subgrouping and novel gene signatures provide insights into refining osteosarcoma therapy and relationships to other types of cancer.
Collapse
Affiliation(s)
- Shan Tang
- College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA;
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA;
| | - Ryan D. Roberts
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA;
| | - Lijun Cheng
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA;
| | - Lang Li
- College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA;
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA;
| |
Collapse
|
7
|
Marinoff AE, Spurr LF, Fong C, Li YY, Forrest SJ, Ward A, Doan D, Corson L, Mauguen A, Pinto N, Maese L, Colace S, Macy ME, Kim A, Sabnis AJ, Applebaum MA, Laetsch TW, Glade-Bender J, Weiser DA, Anderson M, Crompton BD, Meyers P, Zehir A, MacConaill L, Lindeman N, Nowak JA, Ladanyi M, Church AJ, Cherniack AD, Shukla N, Janeway KA. Clinical Targeted Next-Generation Panel Sequencing Reveals MYC Amplification Is a Poor Prognostic Factor in Osteosarcoma. JCO Precis Oncol 2023; 7:e2200334. [PMID: 36996377 PMCID: PMC10531050 DOI: 10.1200/po.22.00334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/11/2022] [Accepted: 01/09/2023] [Indexed: 04/01/2023] Open
Abstract
PURPOSE Osteosarcoma risk stratification, on the basis of the presence of metastatic disease at diagnosis and histologic response to chemotherapy, has remained unchanged for four decades, does not include genomic features, and has not facilitated treatment advances. We report on the genomic features of advanced osteosarcoma and provide evidence that genomic alterations can be used for risk stratification. MATERIALS AND METHODS In a primary analytic patient cohort, 113 tumor and 69 normal samples from 92 patients with high-grade osteosarcoma were sequenced with OncoPanel, a targeted next-generation sequencing assay. In this primary cohort, we assessed the genomic landscape of advanced disease and evaluated the correlation between recurrent genomic events and outcome. We assessed whether prognostic associations identified in the primary cohort were maintained in a validation cohort of 86 patients with localized osteosarcoma tested with MSK-IMPACT. RESULTS In the primary cohort, 3-year overall survival (OS) was 65%. Metastatic disease, present in 33% of patients at diagnosis, was associated with poor OS (P = .04). The most frequently altered genes in the primary cohort were TP53, RB1, MYC, CCNE1, CCND3, CDKN2A/B, and ATRX. Mutational signature 3 was present in 28% of samples. MYC amplification was associated with a worse 3-year OS in both the primary cohort (P = .015) and the validation cohort (P = .012). CONCLUSION The most frequently occurring genomic events in advanced osteosarcoma were similar to those described in prior reports. MYC amplification, detected with clinical targeted next-generation sequencing panel tests, is associated with poorer outcomes in two independent cohorts.
Collapse
Affiliation(s)
- Amanda E. Marinoff
- Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA
- Harvard Medical School, Boston, MA
- Pediatric Hematology/Oncology, UCSF Benioff Children's Hospital, San Francisco, CA
| | - Liam F. Spurr
- Broad Institute of Harvard and MIT, Boston, MA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Christina Fong
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Yvonne Y. Li
- Harvard Medical School, Boston, MA
- Broad Institute of Harvard and MIT, Boston, MA
| | - Suzanne J. Forrest
- Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA
- Harvard Medical School, Boston, MA
| | - Abigail Ward
- Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA
| | - Duong Doan
- Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA
| | - Laura Corson
- Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA
| | - Audrey Mauguen
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Navin Pinto
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, University of Washington, Seattle, WA
| | - Luke Maese
- University of Utah, Huntsman Cancer Institute, and Primary Children's Hospital, Salt Lake City, UT
| | - Susan Colace
- Pediatric Hematology/Oncology/Blood and Marrow Transplant, Nationwide Children's Hospital, Columbus, OH
| | - Margaret E. Macy
- Department of Pediatric Hematology/Oncology, University of Colorado and The Center for Cancer and Blood Disorders, Colorado Children's Hospital, Denver, CO
| | - AeRang Kim
- Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC
| | - Amit J. Sabnis
- Pediatric Hematology/Oncology, UCSF Benioff Children's Hospital, San Francisco, CA
| | | | - Theodore W. Laetsch
- Division of Oncology, Department of Pediatrics, Children’s Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA
| | - Julia Glade-Bender
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Daniel A. Weiser
- Department of Pediatric Hematology/Oncology, Children's Hospital at Montefiore, New York, NY
| | - Megan Anderson
- Harvard Medical School, Boston, MA
- Department of Orthopedic Surgery, Boston Children's Hospital, Boston, MA
| | - Brian D. Crompton
- Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA
- Harvard Medical School, Boston, MA
| | - Paul Meyers
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ahmet Zehir
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Laura MacConaill
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Neal Lindeman
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Jonathan A. Nowak
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Marc Ladanyi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alanna J. Church
- Harvard Medical School, Boston, MA
- Department of Pathology, Boston Children's Hospital, Boston, MA
| | - Andrew D. Cherniack
- Harvard Medical School, Boston, MA
- Broad Institute of Harvard and MIT, Boston, MA
| | - Neerav Shukla
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Katherine A. Janeway
- Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA
- Harvard Medical School, Boston, MA
| |
Collapse
|
8
|
Chen W, Lin Y, Huang J, Yan Z, Cao H. A novel risk score model based on glycolysis-related genes and a prognostic model for predicting overall survival of osteosarcoma patients. J Orthop Res 2022; 40:2372-2381. [PMID: 34997639 DOI: 10.1002/jor.25259] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/15/2021] [Accepted: 01/05/2022] [Indexed: 02/04/2023]
Abstract
This study aims to construct a novel risk score model based on glycolysis-related genes in osteosarcoma and to build and validate a prognostic model for predicting overall survival of patients with osteosarcoma. The transcriptome data and corresponding clinical data of patients with osteosarcoma were obtained from The Cancer Genome Atlas (TCGA) as the training set, and from Gene Expression Omnibus (GEO) database as the validation set. Univariate Cox regression analysis was used to screen the prognostic glycolysis-related genes. The risk coefficient of each glycolysis-related gene was calculated using LASSO regression analysis. Using the median risk score as the cut-off point, patients were divided into high-risk and low-risk groups. Kaplan-Meier survival analysis was used to determine whether there was a significant difference in the overall survival between the two groups. The nomogram was constructed according to the results of multivariate Cox regression. The C-index was calculated, the calibration chart, clinical decision curve and receiver operating characteristic curve were drawn to evaluate the predictive performance of the nomogram. We performed Gene Ontology and Kyoto encyclopedia of genes and genomics enrichment analysis to explore the potential mechanism of prognostic-related glycolysis genes in osteosarcoma. A total of 88 and 53 cases were obtained from the TCGA and GEO database, respectively. A total of 10 key glycolytic genes related to prognosis were screened out. The Kaplan-Meier survival curve revealed that the overall survival of the high-risk group was significantly shorter than that of the low-risk group. The C indices of the training set and the verification set were 0.882 and 0.828, respectively. Our findings will provide further understanding of clinical prognostic outcomes of osteosarcoma patients.
Collapse
Affiliation(s)
- Wenhao Chen
- Department of Pediatric Surgery, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China.,Department of Pediatric Orthopedics, Fujian Provincial Children's Hospital, Fuzhou, Fujian, China
| | - Yuxiang Lin
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Jianping Huang
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Zhiyu Yan
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Hua Cao
- Department of Cardiac Surgery, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China.,Department of Cardiac Surgery, Fujian Provincial Children's Hospital, Fuzhou, Fujian, China
| |
Collapse
|
9
|
Taiana E, Bandini C, Favasuli VK, Ronchetti D, Silvestris I, Puccio N, Todoerti K, Erratico S, Giannandrea D, Bolli N, Amodio N, Ciarrocchi A, Chiaramonte R, Torrente Y, Piva R, Neri A. Activation of long non-coding RNA NEAT1 leads to survival advantage of multiple myeloma cells by supporting a positive regulatory loop with DNA repair proteins. Haematologica 2022; 108:219-233. [PMID: 36073514 PMCID: PMC9827177 DOI: 10.3324/haematol.2022.281167] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Indexed: 02/05/2023] Open
Abstract
Long non-coding RNA NEAT1 is the core structural component of the nuclear paraspeckle (PS) organelles and it has been found to be deregulated in multiple myeloma (MM) patients. Experimental evidence indicated that NEAT1 silencing negatively impacts proliferation and viability of MM cells, both in vitro and in vivo, suggesting a role in DNA damage repair (DDR). In order to elucidate the biological and molecular relevance of NEAT1 upregulation in MM disease we exploited the CRISPR/Cas9 synergistic activation mediator genome editing system to engineer the AMO-1 MM cell line and generate two clones that para-physiologically transactivate NEAT1 at different levels. NEAT1 overexpression is associated with oncogenic and prosurvival advantages in MM cells exposed to nutrient starvation or a hypoxic microenvironment, which are stressful conditions often associated with more aggressive disease phases. Furthermore, we highlighted the NEAT1 involvement in virtually all DDR processes through, at least, two different mechanisms. On one side NEAT1 positively regulates the posttranslational stabilization of essential PS proteins, which are involved in almost all DDR systems, thus increasing their availability within cells. On the other hand, NEAT1 plays a crucial role as a major regulator of a molecular axis that includes ATM and the catalytic subunit of DNA-PK kinase proteins, and their direct targets pRPA32 and pCHK2. Overall, we provided novel important insightsthe role of NEAT1 in supporting MM cells adaptation to stressful conditions by improving the maintenance of DNA integrity. Taken together, our results suggest that NEAT1, and probably PS organelles, could represent a potential therapeutic target for MM treatment.
Collapse
Affiliation(s)
- Elisa Taiana
- Hematology, Fondazione Cà Granda IRCCS Policlinico, Milan,Department of Oncology and Hemato-oncology, University of Milan, Milan,E. Taiana
| | - Cecilia Bandini
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin,Città Della Salute e della Scienza Hospital, Turin
| | - Vanessa Katia Favasuli
- Hematology, Fondazione Cà Granda IRCCS Policlinico, Milan,Department of Oncology and Hemato-oncology, University of Milan, Milan
| | - Domenica Ronchetti
- Hematology, Fondazione Cà Granda IRCCS Policlinico, Milan,Department of Oncology and Hemato-oncology, University of Milan, Milan
| | - Ilaria Silvestris
- Hematology, Fondazione Cà Granda IRCCS Policlinico, Milan,Department of Oncology and Hemato-oncology, University of Milan, Milan
| | - Noemi Puccio
- Hematology, Fondazione Cà Granda IRCCS Policlinico, Milan,Department of Oncology and Hemato-oncology, University of Milan, Milan
| | - Katia Todoerti
- Hematology, Fondazione Cà Granda IRCCS Policlinico, Milan
| | - Silvia Erratico
- Novystem Spa, Milan,Stem Cell Laboratory, Department of Pathophysiology and Transplantation, University of Milan, Centro Dino Ferrari, Unit of Neurology, Fondazione Cà Granda IRCCS Policlinico, Milan
| | | | - Niccolò Bolli
- Hematology, Fondazione Cà Granda IRCCS Policlinico, Milan,Department of Oncology and Hemato-oncology, University of Milan, Milan
| | - Nicola Amodio
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro
| | - Alessia Ciarrocchi
- Laboratory of Translational Research, Azienda Unità Sanitaria Locale-IRCCS Reggio Emilia, Reggio Emilia, Italy
| | | | - Yvan Torrente
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, University of Milan, Centro Dino Ferrari, Unit of Neurology, Fondazione Cà Granda IRCCS Policlinico, Milan
| | - Roberto Piva
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin,Città Della Salute e della Scienza Hospital, Turin
| | - Antonino Neri
- Hematology, Fondazione Cà Granda IRCCS Policlinico, Milan,Department of Oncology and Hemato-oncology, University of Milan, Milan,°Current address: Scientific Directorate, Azienda USL-IRCCS Reggio Emilia, Italy
| |
Collapse
|
10
|
Alemi F, Malakoti F, Vaghari-Tabari M, Soleimanpour J, Shabestani N, Sadigh AR, Khelghati N, Asemi Z, Ahmadi Y, Yousefi B. DNA damage response signaling pathways as important targets for combination therapy and chemotherapy sensitization in osteosarcoma. J Cell Physiol 2022; 237:2374-2386. [PMID: 35383920 DOI: 10.1002/jcp.30721] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/13/2022] [Accepted: 02/25/2022] [Indexed: 11/08/2022]
Abstract
Osteosarcoma (OS) is the most common bone malignancy that occurs most often in young adults, and adolescents with a survival rate of 20% in its advanced stages. Nowadays, increasing the effectiveness of common treatments used in OS has become one of the main problems for clinicians due to cancer cells becoming resistant to chemotherapy. One of the most important mechanisms of resistance to chemotherapy is through increasing the ability of DNA repair because most chemotherapy drugs damage the DNA of cancer cells. DNA damage response (DDR) is a signal transduction pathway involved in preserving the genome stability upon exposure to endogenous and exogenous DNA-damaging factors such as chemotherapy agents. There is evidence that the suppression of DDR may reduce chemoresistance and increase the effectiveness of chemotherapy in OS. In this review, we aim to summarize these studies to better understand the role of DDR in OS chemoresistance in pursuit of overcoming the obstacles to the success of chemotherapy.
Collapse
Affiliation(s)
- Forough Alemi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Faezeh Malakoti
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mostafa Vaghari-Tabari
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jafar Soleimanpour
- Department of Orthopedics Surgery, Shohada Teaching Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Shabestani
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aydin R Sadigh
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nafiseh Khelghati
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Yasin Ahmadi
- Department of Medical Laboratory Sciences, Faculty of Science, Komar University of Science and Technology, Soleimania, Kurdistan Region, Iraq
| | - Bahman Yousefi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
11
|
Identification of LTF as a Prognostic Biomarker for Osteosarcoma. JOURNAL OF ONCOLOGY 2022; 2022:4656661. [PMID: 35096061 PMCID: PMC8799371 DOI: 10.1155/2022/4656661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/17/2021] [Accepted: 12/23/2021] [Indexed: 11/17/2022]
Abstract
Osteosarcoma remains a major health problem in teenagers. However, its pathogenesis mechanism remains not fully elucidated. This study aims to identify the prognostic biomarkers for osteosarcoma. In this study, we selected genes with a median absolute deviation (MAD) value of the top 5000 in the GSE32981 dataset for subsequent analysis. Weighted correlation network analysis (WGCNA) was used to construct a coexpression network. WGCNA showed that the tan module and midnight blue module were highly correlated with origin and metastases of osteosarcoma, respectively. Enrichment analysis was conducted using genes in the tan module and midnight blue module. A gene coexpression network was constructed by calculating the Spearman correlation coefficients. Four key genes (LTF, C10orf107, HIST1H2AK, and NEXN) were identified to be correlated with the prognosis of osteosarcoma patients. LTF has the highest AUC value, and its effect on osteosarcoma cells was then evaluated. The effect of LTF overexpression on proliferation, migration, and invasion of MG63 and 143B cells was detected by the CCK-8 assay, transwell cell migration assay, and transwell invasion assay, respectively. The overexpression of LTF promoted the proliferation, migration, and invasion of MG63 and 143B cells. In conclusion, LTF may serve as a prognostic biomarker for osteosarcoma.
Collapse
|
12
|
Abstract
Improving the survival of patients with osteosarcoma has long proved challenging, although the treatment of this disease is on the precipice of advancement. The increasing feasibility of molecular profiling together with the creation of both robust model systems and large, well-annotated tissue banks has led to an increased understanding of osteosarcoma biology. The historical invariability of survival outcomes and the limited number of agents known to be active in the treatment of this disease facilitate clinical trials designed to identify efficacious novel therapies using small cohorts of patients. In addition, trial designs will increasingly consider the genetic background of the tumour through biomarker-based patient selection, thereby enriching for clinical activity. Indeed, osteosarcoma cells are known to express a number of surface proteins that might be of therapeutic relevance, including B7-H3, GD2 and HER2, which can be targeted using antibody-drug conjugates and/or adoptive cell therapies. In addition, immune-checkpoint inhibition might augment the latter approach by helping to overcome the immunosuppressive tumour microenvironment. In this Review, we provide a brief overview of current osteosarcoma therapy before focusing on the biological insights from the molecular profiling and preclinical modelling studies that have opened new therapeutic opportunities in this disease.
Collapse
Affiliation(s)
- Jonathan Gill
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Richard Gorlick
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
13
|
Drug Resistance in Osteosarcoma: Emerging Biomarkers, Therapeutic Targets and Treatment Strategies. Cancers (Basel) 2021; 13:cancers13122878. [PMID: 34207685 PMCID: PMC8228414 DOI: 10.3390/cancers13122878] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 06/05/2021] [Indexed: 01/02/2023] Open
Abstract
Simple Summary Despite the adoption of aggressive, multimodal treatment schedules, the cure rate of high-grade osteosarcoma (HGOS) has not significantly improved in the last 30 years. The most relevant problem preventing improvement in HGOS prognosis is drug resistance. Therefore, validated novel biomarkers that help to identify those patients who could benefit from innovative treatment options and the development of drugs enabling personalized therapeutic protocols are necessary. The aim of this review was to give an overview on the most relevant emerging drug resistance-related biomarkers, therapeutic targets and new agents or novel candidate treatment strategies, which have been highlighted and suggested for HGOS to improve the success rate of clinical trials. Abstract High-grade osteosarcoma (HGOS), the most common primary malignant tumor of bone, is a highly aggressive neoplasm with a cure rate of approximately 40–50% in unselected patient populations. The major clinical problems opposing the cure of HGOS are the presence of inherent or acquired drug resistance and the development of metastasis. Since the drugs used in first-line chemotherapy protocols for HGOS and clinical outcome have not significantly evolved in the past three decades, there is an urgent need for new therapeutic biomarkers and targeted treatment strategies, which may increase the currently available spectrum of cure modalities. Unresponsive or chemoresistant (refractory) HGOS patients usually encounter a dismal prognosis, mostly because therapeutic options and drugs effective for rescue treatments are scarce. Tailored treatments for different subgroups of HGOS patients stratified according to drug resistance-related biomarkers thus appear as an option that may improve this situation. This review explores drug resistance-related biomarkers, therapeutic targets and new candidate treatment strategies, which have emerged in HGOS. In addition to consolidated biomarkers, specific attention has been paid to the role of non-coding RNAs, tumor-derived extracellular vesicles, and cancer stem cells as contributors to drug resistance in HGOS, in order to highlight new candidate markers and therapeutic targets. The possible use of new non-conventional drugs to overcome the main mechanisms of drug resistance in HGOS are finally discussed.
Collapse
|
14
|
Sadoughi F, Maleki Dana P, Asemi Z, Yousefi B. DNA damage response and repair in osteosarcoma: Defects, regulation and therapeutic implications. DNA Repair (Amst) 2021; 102:103105. [PMID: 33836418 DOI: 10.1016/j.dnarep.2021.103105] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 03/20/2021] [Indexed: 01/03/2023]
Abstract
Osteosarcoma (OS) is the most common primary bone malignancy in children and adolescents which has the survival rate of 20% in its advanced stages. Osteosarcomas are mostly resistance to our common treatments. DNA damage response (DDR) is a specialized multistep process containing abundant proteins which are necessary for the survival of any cell and organism. DDR machinery detects a diversity of DNA lesions and inhibits the cell cycle progression if these lesions are not repairable. DDR is involved in aging, age-related diseases, and cancer. In recent years, DDR inhibitors have gained the attention of researches due to their potentials in offering novel therapeutic targets and improving the response of many cancers to either chemo- or radio-therapy. In this regard, we tried to gather a great body of evidence about the role of DDR ingredients in osteosarcoma's initiation/progression, prognosis, and treatment.
Collapse
Affiliation(s)
- Fatemeh Sadoughi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| | - Parisa Maleki Dana
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
15
|
Bowler E, Skwarska A, Wilson JD, Ramachandran S, Bolland H, Easton A, Ostheimer C, Hwang MS, Leszczynska KB, Conway SJ, Hammond EM. Pharmacological Inhibition of ATR Can Block Autophagy through an ATR-Independent Mechanism. iScience 2020; 23:101668. [PMID: 33134898 PMCID: PMC7588853 DOI: 10.1016/j.isci.2020.101668] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 09/25/2020] [Accepted: 10/07/2020] [Indexed: 12/26/2022] Open
Abstract
Inhibition of the ATR kinase has emerged as a therapeutically attractive means to target cancer since the development of potent inhibitors, which are now in clinical testing. We investigated a potential link between ATR inhibition and the autophagy process in esophageal cancer cells using four ATR inhibitors including two in clinical testing. The response to pharmacological ATR inhibitors was compared with genetic systems to investigate the ATR dependence of the effects observed. The ATR inhibitor, VX-970, was found to lead to an accumulation of p62 and LC3-II indicative of a blocked autophagy. This increase in p62 occurred post-transcriptionally and in all the cell lines tested. However, our data indicate that the accumulation of p62 occurred in an ATR-independent manner and was instead an off-target response to the ATR inhibitor. This study has important implications for the clinical response to pharmacological ATR inhibition, which in some cases includes the blockage of autophagy.
Collapse
Affiliation(s)
- Elizabeth Bowler
- Oxford Institute for Radiation Oncology, Department of Oncology, The University of Oxford, Oxford OX3 7DQ, UK
| | - Anna Skwarska
- Oxford Institute for Radiation Oncology, Department of Oncology, The University of Oxford, Oxford OX3 7DQ, UK
| | - Joseph D. Wilson
- Oxford Institute for Radiation Oncology, Department of Oncology, The University of Oxford, Oxford OX3 7DQ, UK
| | - Shaliny Ramachandran
- Oxford Institute for Radiation Oncology, Department of Oncology, The University of Oxford, Oxford OX3 7DQ, UK
| | - Hannah Bolland
- Oxford Institute for Radiation Oncology, Department of Oncology, The University of Oxford, Oxford OX3 7DQ, UK
| | - Alistair Easton
- Translational Histopathology Lab, Oxford Cancer Centre, Oxford OX3 7DQ, UK
| | - Christian Ostheimer
- Oxford Institute for Radiation Oncology, Department of Oncology, The University of Oxford, Oxford OX3 7DQ, UK
| | - Ming-Shih Hwang
- Oxford Institute for Radiation Oncology, Department of Oncology, The University of Oxford, Oxford OX3 7DQ, UK
| | - Katarzyna B. Leszczynska
- Oxford Institute for Radiation Oncology, Department of Oncology, The University of Oxford, Oxford OX3 7DQ, UK
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Stuart J. Conway
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, UK
| | - Ester M. Hammond
- Oxford Institute for Radiation Oncology, Department of Oncology, The University of Oxford, Oxford OX3 7DQ, UK
| |
Collapse
|