1
|
Giner F, Medina-Ceballos E, López-Reig R, Machado I, López-Guerrero JA, Navarro S, Rubio-Martínez LA, Espino M, Mayordomo-Aranda E, Llombart-Bosch A. The Combined Immunohistochemical Expression of GLI1 and BCOR in Synovial Sarcomas for the Identification of Three Risk Groups and Their Prognostic Outcomes: A Study of 52 Patients. Int J Mol Sci 2024; 25:7615. [PMID: 39062853 PMCID: PMC11276717 DOI: 10.3390/ijms25147615] [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: 06/14/2024] [Revised: 07/05/2024] [Accepted: 07/07/2024] [Indexed: 07/28/2024] Open
Abstract
Synovial sarcoma (SS) is a rare soft-tissue tumor characterized by a monomorphic blue spindle cell histology and variable epithelial differentiation. Morphologically, SSs may be confused with other sarcomas. Systemic treatment is more effective for patients with high-risk SSs, patients with advanced disease, and younger patients. However, further studies are required to find new prognostic biomarkers. Herein, we describe the morphological, molecular, and clinical findings, using a wide immunohistochemical panel, of a series of SS cases. We studied 52 cases confirmed as SSs by morphological diagnosis and/or molecular studies. Clinical data (gender, age, tumor size, tumor location, resection margins, adjuvant treatment, recurrences, metastasis, and survival) were also retrieved for each patient. All the available H&E slides were examined by four pathologists. Three tissue microarrays (TMAs) were constructed for each of the tumors, and a wide immunohistochemical panel was performed. For time-to-event variables, survival analysis was performed using Kaplan-Meier curves and log-rank testing, or Cox regression. Statistical significance was considered at p < 0.05. The mean age of our patients was 40.33, and the median was 40.5 years. We found a predominance of males versus females (1.7:1). The most frequent morphological subtype was monophasic. TRPS1, SS18-SSX, and SSX-C-terminus were positive in 96% of cases. GLI1 expression was strong in six and focal (cytoplasmic) in twenty patients. Moreover, BCOR was expressed in more than half of SSs. Positive expression of both proteins, BCOR and GLI1, was correlated with a worse prognosis. Multivariate analysis was also performed, but only BCOR expression appeared to be significant. The combination of GLI1 and BCOR antibodies can be used to group SSs into three risk groups (low, intermediate, and high risk). We hypothesize that these findings could identify which patients would benefit from receiving adjuvant treatment and which would not. Moreover, these markers could represent therapeutic targets in advanced stages. However, further, larger series of SSs and molecular studies are necessary to corroborate our present findings.
Collapse
Affiliation(s)
- Francisco Giner
- Pathology Department, Hospital Universitari i Politècnic La Fe of Valencia, 46026 Valencia, Spain; (L.A.R.-M.); (E.M.-A.)
- Pathology Department, University of Valencia, 46010 Valencia, Spain; (E.M.-C.); (I.M.); (S.N.); (M.E.); (A.L.-B.)
| | - Emilio Medina-Ceballos
- Pathology Department, University of Valencia, 46010 Valencia, Spain; (E.M.-C.); (I.M.); (S.N.); (M.E.); (A.L.-B.)
| | - Raquel López-Reig
- Molecular Biology Department, Instituto Valenciano de Oncología, 46009 Valencia, Spain; (R.L.-R.); (J.A.L.-G.)
| | - Isidro Machado
- Pathology Department, University of Valencia, 46010 Valencia, Spain; (E.M.-C.); (I.M.); (S.N.); (M.E.); (A.L.-B.)
- Pathology Department, Instituto Valenciano de Oncología and Patologika Laboratory Hospital Quiron Salud, 46010 Valencia, Spain
| | - José Antonio López-Guerrero
- Molecular Biology Department, Instituto Valenciano de Oncología, 46009 Valencia, Spain; (R.L.-R.); (J.A.L.-G.)
- Department of Pathology, Catholic University of Valencia, 46001 Valencia, Spain
- Joint Cancer Research Unit, Centro de Investigación Príncipe Felipe (CIPF), 46012 Valencia, Spain
| | - Samuel Navarro
- Pathology Department, University of Valencia, 46010 Valencia, Spain; (E.M.-C.); (I.M.); (S.N.); (M.E.); (A.L.-B.)
- Pathology Department, Hospital Clínic Universitari, 46010 Valencia, Spain
- Instituto Salud Carlos III (CIBERONC), 28220 Madrid, Spain
| | - Luis Alberto Rubio-Martínez
- Pathology Department, Hospital Universitari i Politècnic La Fe of Valencia, 46026 Valencia, Spain; (L.A.R.-M.); (E.M.-A.)
| | - Mónica Espino
- Pathology Department, University of Valencia, 46010 Valencia, Spain; (E.M.-C.); (I.M.); (S.N.); (M.E.); (A.L.-B.)
| | - Empar Mayordomo-Aranda
- Pathology Department, Hospital Universitari i Politècnic La Fe of Valencia, 46026 Valencia, Spain; (L.A.R.-M.); (E.M.-A.)
| | - Antonio Llombart-Bosch
- Pathology Department, University of Valencia, 46010 Valencia, Spain; (E.M.-C.); (I.M.); (S.N.); (M.E.); (A.L.-B.)
| |
Collapse
|
2
|
Cheng J, Yan Z, Jiang K, Liu C, Xu D, Lyu X, Hu X, Zhang S, Zhou Y, Li J, Zhao Y. Discovery of JN122, a Spiroindoline-Containing Molecule that Inhibits MDM2/p53 Protein-Protein Interaction and Exerts Robust In Vivo Antitumor Efficacy. J Med Chem 2023; 66:16991-17025. [PMID: 38062557 DOI: 10.1021/acs.jmedchem.3c01815] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
MDM2 and MDM4 cooperatively and negatively regulate p53, while this pathway is often hijacked by cancer cells in favor of their survival. Blocking MDM2/p53 interaction with small-molecule inhibitors liberates p53 from MDM2 mediated degradation, which is an attractive strategy for drug discovery. We reported herein structure-based discovery of highly potent spiroindoline-containing MDM2 inhibitor (-)60 (JN122), which also exhibited moderate activities against MDM4/p53 interactions. In a panel of cancer cell lines harboring wild type p53, (-)60 efficiently promoted activation of p53 and its target genes, inhibited cell cycle progression, and induced cell apoptosis. Interestingly, (-)60 also promoted degradation of MDM4. More importantly, (-)60 exhibited good PK properties and exerted robust antitumor efficacies in a systemic mouse xenograft model of MOLM-13. Taken together, our study showcases a class of potent MDM2 inhibitors featuring a novel spiro-indoline scaffold, which is promising for future development targeting cancer cells with wild-type p53.
Collapse
Affiliation(s)
- Jing Cheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, P. R. China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, P. R. China
| | - Ziqin Yan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, P. R. China
| | - Kailong Jiang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, P. R. China
| | - Chen Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, P. R. China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Dehua Xu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, P. R. China
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124000, P. R. China
| | - Xilin Lyu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, P. R. China
| | - Xiaobei Hu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, P. R. China
| | - Shiyan Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, P. R. China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, P. R. China
| | - Yubo Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, P. R. China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, P. R. China
| | - Jia Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, P. R. China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, P. R. China
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124000, P. R. China
| | - Yujun Zhao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, P. R. China
- University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, P. R. China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
- Shandong Provincial Key Laboratory of Biopharmaceuticals, Shandong Academy of Pharmaceutical Sciences, Jinan 250101, P. R. China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, P. R. China
| |
Collapse
|
3
|
Landuzzi L, Manara MC, Pazzaglia L, Lollini PL, Scotlandi K. Innovative Breakthroughs for the Treatment of Advanced and Metastatic Synovial Sarcoma. Cancers (Basel) 2023; 15:3887. [PMID: 37568703 PMCID: PMC10416854 DOI: 10.3390/cancers15153887] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
Synovial sarcoma (SyS) is a rare aggressive soft tissue sarcoma carrying the chromosomal translocation t(X;18), encoding the fusion transcript SS18::SSX. The fusion oncoprotein interacts with both BAF enhancer complexes and polycomb repressor complexes, resulting in genome-wide epigenetic perturbations and a unique altered genetic signature. Over 80% of the patients are initially diagnosed with localized disease and have a 5-year survival rate of 70-80%, but metastatic relapse occurs in 50% of the cases. Advanced, unresectable, or metastatic disease has a 5-year survival rate below 10%, representing a critical issue. This review summarizes the molecular mechanisms behind SyS and illustrates current treatments in front line, second line, and beyond settings. We analyze the use of immune check point inhibitors (ICI) in SyS that do not behave as an ICI-sensitive tumor, claiming the need for predictive genetic signatures and tumor immune microenvironment biomarkers. We highlight the clinical translation of innovative technologies, such as proteolysis targeting chimera (PROTAC) protein degraders or adoptive transfer of engineered immune cells. Adoptive cell transfer of engineered T-cell receptor cells targeting selected cancer/testis antigens has shown promising results against metastatic SyS in early clinical trials and further improvements are awaited from refinements involving immune cell engineering and tumor immune microenvironment enhancement.
Collapse
Affiliation(s)
- Lorena Landuzzi
- Experimental Oncology Laboratory, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (M.C.M.); (L.P.)
| | - Maria Cristina Manara
- Experimental Oncology Laboratory, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (M.C.M.); (L.P.)
| | - Laura Pazzaglia
- Experimental Oncology Laboratory, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (M.C.M.); (L.P.)
| | - Pier-Luigi Lollini
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy;
| | - Katia Scotlandi
- Experimental Oncology Laboratory, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (M.C.M.); (L.P.)
| |
Collapse
|
4
|
Cope BM, Traweek RS, Lazcano R, Keung EZ, Lazar AJ, Roland CL, Nassif EF. Targeting the Molecular and Immunologic Features of Leiomyosarcoma. Cancers (Basel) 2023; 15:2099. [PMID: 37046760 PMCID: PMC10093078 DOI: 10.3390/cancers15072099] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/25/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
Leiomyosarcoma (LMS) is a rare, aggressive mesenchymal tumor with smooth muscle differentiation. LMS is one of the most common histologic subtypes of soft tissue sarcoma; it most frequently occurs in the extremities, retroperitoneum, or uterus. LMS often demonstrates aggressive tumor biology, with a higher risk of developing distant metastatic disease than most sarcoma histologic types. The prognosis is poor, particularly in patients with uterine disease, and there is a need for the development of more effective therapies. Genetically, LMS is karyotypically complex and characterized by a low tumor mutational burden, with frequent alterations in TP53, RB1, PTEN, and DNA damage response pathways that may contribute to resistance against immune-checkpoint blockade monotherapy. The LMS immune microenvironment is highly infiltrated with tumor-associated macrophages and tumor-infiltrating lymphocytes, which may represent promising biomarkers. This review provides an overview of the clinical and pathologic behavior of both soft tissue and uterine LMS and summarizes the genomic and immune characteristics of these tumors and how they may provide opportunities for the development of biomarker-based immune therapies.
Collapse
Affiliation(s)
- Brandon M. Cope
- Department of Surgery, Keesler Medical Center, Biloxi, MS 39534, USA
| | - Raymond S. Traweek
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Rossana Lazcano
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Emily Z. Keung
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Alexander J. Lazar
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- UTHealth Houston Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Christina L. Roland
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Elise F. Nassif
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
5
|
Gambella A, Bertero L, Rondón-Lagos M, Verdun Di Cantogno L, Rangel N, Pitino C, Ricci AA, Mangherini L, Castellano I, Cassoni P. FISH Diagnostic Assessment of MDM2 Amplification in Liposarcoma: Potential Pitfalls and Troubleshooting Recommendations. Int J Mol Sci 2023; 24:ijms24021342. [PMID: 36674856 PMCID: PMC9863600 DOI: 10.3390/ijms24021342] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/04/2023] [Accepted: 01/06/2023] [Indexed: 01/12/2023] Open
Abstract
MDM2 amplification represents the leading oncogenic pathway and diagnostic hallmark of liposarcoma, whose assessment is based on Fluorescence In Situ Hybridization (FISH) analysis. Despite its diagnostic relevance, no univocal interpretation criteria regarding FISH assessments of MDM2 amplification have been established so far, leading to several different approaches and potential diagnostic misinterpretations. This study aims to address the most common issues and proposes troubleshooting guidelines for MDM2 amplification assessments by FISH. We retrospectively retrieved 51 liposarcomas, 25 Lipomas, 5 Spindle Cell Lipoma/Pleomorphic Lipomas, and 2 Atypical Spindle Cell Lipomatous Tumors and the corresponding MDM2 FISH analysis. We observed MDM2 amplification in liposarcomas cases only (43 out of 51 cases) and identified three MDM2-amplified patterns (scattered (50% of cases), clustered (14% of cases), and mixed (36% of cases)) and two nonamplified patterns (low number of signals (82% of cases) and polysomic (18% of cases)). Based on these data and published evidence in the literature, we propose a set of criteria to guide MDM2 amplification analysis in liposarcoma. Kindled by the compelling importance of MDM2 assessments to improve diagnostic and therapeutic liposarcoma management, these suggestions could represent the first step to develop a univocal interpretation model and consensus guidelines.
Collapse
Affiliation(s)
- Alessandro Gambella
- Division of Liver and Transplant Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Medical Sciences, University of Turin, 10124 Turin, Italy
| | - Luca Bertero
- Department of Medical Sciences, University of Turin, 10124 Turin, Italy
| | - Milena Rondón-Lagos
- School of Biological Sciences, Universidad Pedagógica y Tecnológica de Colombia, Tunja 150003, Colombia
| | - Ludovica Verdun Di Cantogno
- Department of Laboratory Medicine, Azienda Ospedaliera Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| | - Nelson Rangel
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
| | - Chiara Pitino
- Department of Medical Sciences, University of Turin, 10124 Turin, Italy
| | | | - Luca Mangherini
- Department of Medical Sciences, University of Turin, 10124 Turin, Italy
| | | | - Paola Cassoni
- Department of Medical Sciences, University of Turin, 10124 Turin, Italy
- Correspondence: ; Tel.: +39-011-633-5588
| |
Collapse
|
6
|
Premalignant Conditions of Bone. JAAOS: GLOBAL RESEARCH AND REVIEWS 2022; 6:01979360-202210000-00004. [PMID: 36227850 PMCID: PMC9575816 DOI: 10.5435/jaaosglobal-d-22-00097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 07/29/2022] [Indexed: 01/10/2023]
Abstract
Development of malignancy is a multifactorial process, and there are multitude of conditions of bone that may predispose patients to malignancy. Etiologies of malignancy include benign osseous conditions, genetic predisposition, and extrinsic conditions. New-onset pain or growth in a previously stable lesion is that should concern for malignant change and should prompt a diagnostic workup for malignancy.
Collapse
|
7
|
Noncanonical roles of p53 in cancer stemness and their implications in sarcomas. Cancer Lett 2022; 525:131-145. [PMID: 34742870 DOI: 10.1016/j.canlet.2021.10.037] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 09/24/2021] [Accepted: 10/25/2021] [Indexed: 12/25/2022]
Abstract
Impairment of the prominent tumor suppressor p53, well known for its canonical role as the "guardian of the genome", is found in almost half of human cancers. More recently, p53 has been suggested to be a crucial regulator of stemness, orchestrating the differentiation of embryonal and adult stem cells, suppressing reprogramming into induced pluripotent stem cells, or inhibiting cancer stemness (i.e., cancer stem cells, CSCs), which underlies the development of therapy-resistant tumors. This review addresses these noncanonical roles of p53 and their implications in sarcoma initiation and progression. Indeed, dysregulation of p53 family proteins is a common event in sarcomas and is associated with poor survival. Additionally, emerging studies have demonstrated that loss of wild-type p53 activity hinders the terminal differentiation of mesenchymal stem cells and leads to the development of aggressive sarcomas. This review summarizes recent findings on the roles of aberrant p53 in sarcoma development and stemness and further describes therapeutic approaches to restore normal p53 activity as a promising anti-CSC strategy to treat refractory sarcomas.
Collapse
|
8
|
Cao P, Yang A, Li P, Xia X, Han Y, Zhou G, Wang R, Yang F, Li Y, Zhang Y, Cui Y, Ji H, Lu L, He F, Zhou G. Genomic gain of RRS1 promotes hepatocellular carcinoma through reducing the RPL11-MDM2-p53 signaling. SCIENCE ADVANCES 2021; 7:7/35/eabf4304. [PMID: 34433556 PMCID: PMC8386927 DOI: 10.1126/sciadv.abf4304] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 07/02/2021] [Indexed: 05/20/2023]
Abstract
Hepatocellular carcinomas (HCCs) are characterized by frequent somatic genomic copy number alterations (CNAs), with most of them biologically unexplored. Here, we performed integrative analyses combining CNAs with the transcriptomic data to reveal the cis- and trans-effects of CNAs in HCC. We identified recurrent genomic gains of chromosome 8q, which exhibit strong trans-effects and are broadly associated with ribosome biogenesis activity. Furthermore, 8q gain-driven overexpression of ribosome biogenesis regulator (RRS1) promotes growth of HCC cells in vitro and in vivo. Mechanistically, RRS1 attenuates ribosomal stress through retaining RPL11 in the nucleolus, which, in turn, potentiates MDM2-mediated ubiquitination and degradation of p53. Clinically, higher RRS1 expression levels predict poor clinical outcomes for patients with HCC, especially in those with intact p53 Our findings established that the chromosome 8q oncogene RRS1 promotes HCC development through attenuating the RPL11-MDM2-p53 pathway and provided new potential targets for treatment of this malignancy.
Collapse
Affiliation(s)
- Pengbo Cao
- State Key Laboratory of Proteomics, National Center for Protein Sciences at Beijing, Beijing Institute of Radiation Medicine, Beijing, China
| | - Aiqing Yang
- State Key Laboratory of Proteomics, National Center for Protein Sciences at Beijing, Beijing Institute of Radiation Medicine, Beijing, China
| | - Peiyao Li
- State Key Laboratory of Proteomics, National Center for Protein Sciences at Beijing, Beijing Institute of Radiation Medicine, Beijing, China
| | - Xia Xia
- State Key Laboratory of Proteomics, National Center for Protein Sciences at Beijing, Beijing Institute of Radiation Medicine, Beijing, China
| | - Yuqing Han
- State Key Laboratory of Proteomics, National Center for Protein Sciences at Beijing, Beijing Institute of Radiation Medicine, Beijing, China
| | - Guangming Zhou
- State Key Laboratory of Proteomics, National Center for Protein Sciences at Beijing, Beijing Institute of Radiation Medicine, Beijing, China
| | - Rui Wang
- State Key Laboratory of Proteomics, National Center for Protein Sciences at Beijing, Beijing Institute of Radiation Medicine, Beijing, China
| | - Fei Yang
- State Key Laboratory of Proteomics, National Center for Protein Sciences at Beijing, Beijing Institute of Radiation Medicine, Beijing, China
| | - Yuanfeng Li
- State Key Laboratory of Proteomics, National Center for Protein Sciences at Beijing, Beijing Institute of Radiation Medicine, Beijing, China
| | - Ying Zhang
- State Key Laboratory of Proteomics, National Center for Protein Sciences at Beijing, Beijing Institute of Radiation Medicine, Beijing, China
| | - Ying Cui
- Affiliated Cancer Hospital of Guangxi Medical University, Nanning City, China
| | - Hongzan Ji
- Department of Gastroenterology and Hepatology, Jinling Hospital, Clinical School of Nanjing University, Nanjing City, China
| | - Lei Lu
- Department of Surgical Oncology, Jingdu Hospital, Nanjing City, China
| | - Fuchu He
- State Key Laboratory of Proteomics, National Center for Protein Sciences at Beijing, Beijing Institute of Lifeomics, Beijing, China.
| | - Gangqiao Zhou
- State Key Laboratory of Proteomics, National Center for Protein Sciences at Beijing, Beijing Institute of Radiation Medicine, Beijing, China.
- Collaborative Innovation Center for Personalized Cancer Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing City, China
- Anhui Medical University, Hefei City, China
- Hebei University, Shijiazhuang City, China
| |
Collapse
|
9
|
Chen L, Li J, Wu X, Zheng Z. Identification of Somatic Genetic Alterations Using Whole-Exome Sequencing of Uterine Leiomyosarcoma Tumors. Front Oncol 2021; 11:687899. [PMID: 34178683 PMCID: PMC8226214 DOI: 10.3389/fonc.2021.687899] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/20/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND The genomic abnormalities associated with uterine leiomyosarcoma (uLMS) have not been fully elucidated to date. OBJECTIVE To understand the pathogenesis of uLMS and to identify driver mutations and potential therapeutic targets in uLMS. METHODS Three matched tumor-constitutional DNA pairs from patients with recurrent uLMS were subjected to whole-exome capture and next-generation sequencing. The role of the selected gene SHARPIN in uLMS was analyzed by the CCK-8 assay and colony formation assay after specific siRNA knockdown. RESULTS We identified four genes with somatic SNVs, namely, SLC39A7, GPR19, ZNF717, and TP53, that could be driver mutations. We observed that 30.7% (4/13) of patients with uLMS had TP53 mutations as analyzed by direct sequencing. Analysis of somatic copy number variants (CNVs) showed regions of chromosomal gain at 1q21-23, 19p13, 17q21, and 17q25, whereas regions of chromosomal loss were observed at 2q35, 2q37, 1p36, 10q26, 6p22, 8q24, 11p15, 11q12, and 9p21. The SHARPIN gene was amplified in two patients and mutated in another (SHARPIN: NM_030974: exon2: c.G264C, p.E88D). Amplification of the SHARPIN gene was associated with shorter PFS and OS in soft tissue sarcoma, as shown by TCGA database analysis. Knockdown of SHARPIN expression was observed to decrease cell growth and colony formation in uterine sarcoma cell lines. CONCLUSIONS Exome sequencing revealed mutational heterogeneity of uLMS. The SHARPIN gene was amplified in uLMS and could be a candidate oncogene.
Collapse
Affiliation(s)
- Lihua Chen
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiajia Li
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaohua Wu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhong Zheng
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
10
|
Chen TWW, Burns J, Jones RL, Huang PH. Optimal Clinical Management and the Molecular Biology of Angiosarcomas. Cancers (Basel) 2020; 12:E3321. [PMID: 33182685 PMCID: PMC7696056 DOI: 10.3390/cancers12113321] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/31/2020] [Accepted: 11/02/2020] [Indexed: 12/12/2022] Open
Abstract
Angiosarcomas comprise less than 3% of all soft tissue sarcomas but have a poor prognosis. Most angiosarcomas occur without obvious risk factors but secondary angiosarcoma could arise after radiotherapy or chronic lymphedema. Surgery remains the standard treatment for localized angiosarcoma but neoadjuvant systemic treatment may improve the curability. For advanced angiosarcoma, anthracyclines and taxanes are the main chemotherapy options. Anti-angiogenic agents have a substantial role but the failure of a randomized phase 3 trial of pazopanib with or without an anti-endoglin antibody brings a challenge to future trials in angiosarcomas. Immune checkpoint inhibitors as single agents or in combination with oncolytic virus may play an important role but the optimal duration remains to be investigated. We also report the current understanding of the molecular pathways involved in angiosarcoma pathogenesis including MYC amplification, activation of angiogenic pathways and different molecular alterations that are associated with angiosarcomas of different aetiology. The success of the patient-partnered Angiosarcoma Project (ASCProject) has provided not only detailed insights into the molecular features of angiosarcomas of different origins but also offers a template for future fruitful collaborations between patients, physicians, and researchers. Lastly, we provide our perspective of future developments in optimizing the clinical management of angiosarcomas.
Collapse
Affiliation(s)
- Tom Wei-Wu Chen
- Department of Oncology, National Taiwan University Hospital and Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Jessica Burns
- Division of Molecular Pathology, The Institute of Cancer Research, London SW3 6JB, UK;
| | - Robin L. Jones
- Sarcoma Unit, The Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London SW3 6JJ, UK;
| | - Paul H. Huang
- Division of Molecular Pathology, The Institute of Cancer Research, London SW3 6JB, UK;
| |
Collapse
|
11
|
Genetic heterogeneity and predictive biomarker for pulmonary sarcomatoid carcinomas. Cancer Genet 2020; 250-251:12-19. [PMID: 33217678 DOI: 10.1016/j.cancergen.2020.11.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 09/02/2020] [Accepted: 11/02/2020] [Indexed: 12/25/2022]
Abstract
PURPOSE The aim of this study is to investigate the genetic heterogeneity (carcinomatous vs. sarcomatous components) and predictive biomarkers in patients with pulmonary sarcomatoid carcinoma (PSC). METHODS Genetic alterations and biomarkers of immunotherapy were performed in a discovery set (n = 6) of PSC. Next-generation sequencing (NGS) on a pan-cancer gene panel was applied to detect the genetic alterations in each component, and the respective mutation profiling and tumor mutation burden (TMB) were compared as well. Immunohistochemistry (IHC) assay with SP263 antibody was used to detect the protein expression of programmed death-ligand 1 (PD-L1) in each component. RESULTS Comparative genetic analysis revealed that the separate carcinomatous and sarcomatous components shared strikingly common mutations. TP53 (4/6, 66.7%) was the most common genetic alteration in 6 PSC patients. MET exon 14 skipping was detected in one case, accounting for 16.7%. An EZR-ROS1 fusion (EZR: intron10-ROS1: intron32) was identified in one case. The TMB of the two components was similar. Nevertheless, significantly higher PD-L1 expression was found in carcinomatous components compared to sarcomatous components. MDM2 amplification was detected in 2/6 (33.3%) of cases and STK11 mutation in 1/6 (16.7%) of cases. CONCLUSIONS PSC containing carcinomatous and sarcomatous components had a mild heterogeneity; the two components may evolve from common ancestral cells. High PD-L1 expression suggests that immunotherapy could be used as a potential therapy for PSC patients, while patients with negative immune-responsive genes need to be screened out. Altogether, these findings further highlight that the detection of genetic alteration and PD-L1 expression plays an important role in treatment of patients with PSC.
Collapse
|
12
|
Han X, Alu A, Xiao Y, Wei Y, Wei X. Hyperprogression: A novel response pattern under immunotherapy. Clin Transl Med 2020; 10:e167. [PMID: 32997401 PMCID: PMC7510779 DOI: 10.1002/ctm2.167] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 02/05/2023] Open
Abstract
Checkpoint blockade therapy has shown significant therapeutic benefits and resulted in durable responses in patients with various tumors. However, accumulating evidence has demonstrated that 4-29% of all patients with cancers with various histologies may suffer from tumor flare following such therapy. This novel tumor response pattern, termed hyperprogression, is a potentially deleterious side effect of checkpoint blockade therapy that accelerates disease progression in a subset of patients. In this review, we describe possible immune checkpoint blockade biomarkers and the epidemiology, different definitions, and predictors of hyperprogression based on the research findings and further present the available evidence supporting pathophysiological hypotheses that might explain hyperprogression during checkpoint blockade therapy. We also compare hyperprogression and pseudoprogression. Finally, we discuss areas requiring further study.
Collapse
Affiliation(s)
- Xue‐jiao Han
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduChina
| | - Aqu Alu
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduChina
| | - Yi‐nan Xiao
- West China School of MedicineWest China HospitalSichuan UniversityChengduChina
| | - Yu‐quan Wei
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduChina
| | - Xia‐wei Wei
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduChina
| |
Collapse
|
13
|
Yu DH, Xu ZY, Mo S, Yuan L, Cheng XD, Qin JJ. Targeting MDMX for Cancer Therapy: Rationale, Strategies, and Challenges. Front Oncol 2020; 10:1389. [PMID: 32850448 PMCID: PMC7419686 DOI: 10.3389/fonc.2020.01389] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 07/01/2020] [Indexed: 12/11/2022] Open
Abstract
The oncogene MDMX, also known as MDM4 is a critical negative regulator of the tumor suppressor p53 and has been implicated in the initiation and progression of human cancers. Increasing evidence indicates that MDMX is often amplified and highly expressed in human cancers, promotes cancer cell growth, and inhibits apoptosis by dampening p53-mediated transcription of its target genes. Inhibiting MDMX-p53 interaction has been found to be effective for restoring the tumor suppressor activity of p53. Therefore, MDMX is becoming one of the most promising molecular targets for developing anticancer therapeutics. In the present review, we mainly focus on the current MDMX-targeting strategies and known MDMX inhibitors, as well as their mechanisms of action and in vitro and in vivo anticancer activities. We also propose other potential targeting strategies for developing more specific and effective MDMX inhibitors for cancer therapy.
Collapse
Affiliation(s)
- De-Hua Yu
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhi-Yuan Xu
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Shaowei Mo
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Yuan
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiang-Dong Cheng
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Jiang-Jiang Qin
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.,Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
14
|
Di Mauro I, Mescam-Mancini L, Chetaille B, Lae M, Pierron G, Dadone-Montaudie B, Bazin A, Bouvier C, Michiels JF, Pedeutour F. MDM2 amplification and fusion gene ss18-ssx in a poorly differentiated synovial sarcoma: A rare but puzzling conjunction. Neoplasia 2020; 22:311-321. [PMID: 32559641 PMCID: PMC7303914 DOI: 10.1016/j.neo.2020.05.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 04/30/2020] [Accepted: 05/17/2020] [Indexed: 01/30/2023]
Abstract
The detection of specific alterations by genetic analyses has been included in the diagnostic criterions of the World Health Organization's classification of soft tissues tumors since 2013. The presence of a SS18 rearrangement is pathognomonic of synovial sarcoma (SS). MDM2 amplification is strongly correlated to well-differentiated or dedifferentiated liposarcoma (DDLPS) in the context of sarcoma. We identified one case of poorly differentiated sarcoma harboring both SS18-SSX2 fusion and MDM2 amplification. The review of the literature showed high discrepancies, concerning the incidence of MDM2 amplification in SS: from 1.4% up to 40%. Our goal was to precisely determine the specific clinico-pathological features of this case and to estimate the frequency and characteristics of the association of SS18-SSX fusion/MDM2 amplification in sarcomas. We performed a retrospective and prospective study in 96 sarcomas, (56 SS and 40 DDLPS), using FISH and/or array-CGH to detect MDM2 amplification and SS18 rearrangement. None of the 96 cases presented both genetic alterations. Among the SS, only the index case (1/57: 1.7 %) presented the double anomaly. We concluded that MDM2 amplification in SS is a very rare event. The final diagnosis of the index case was a SS with SS18-SSX2 and MDM2 amplification as a secondary alteration. If the detection of MDM2 amplification is performed first in a poorly differentiated sarcoma, that may lead to not search other anomalies such as SS18 rearrangement and therefore to an erroneous diagnosis. This observation emphasizes the strong complementarity between histomorphology, immunohistochemistry and molecular studies in sarcoma diagnosis.
Collapse
Affiliation(s)
- Ilaria Di Mauro
- Laboratory of Solid Tumor Genetics, University Hospital of Nice-Côte d'Azur University, Nice, France; Laboratory of Solid Tumor Genetics, Institute for Research on Cancer and Aging of Nice (IRCAN), CNRS UMR 7284/INSERM U1081, Nice, France
| | | | - Bruno Chetaille
- Department of Cancer Biology, Paoli-Calmettes Institute, Marseille, France
| | - Marick Lae
- Department of Pathology, Curie Institute, Paris, France; Department of Pathology, Henri Becquerel Center, INSERM U1245, UniRouen Normandie University, Rouen, France
| | | | - Bérengère Dadone-Montaudie
- Laboratory of Solid Tumor Genetics, University Hospital of Nice-Côte d'Azur University, Nice, France; Laboratory of Solid Tumor Genetics, Institute for Research on Cancer and Aging of Nice (IRCAN), CNRS UMR 7284/INSERM U1081, Nice, France; Central Laboratory of Pathology, University Hospital of Nice-Côte d'Azur University, Nice, France
| | - Audrey Bazin
- Laboratory of Solid Tumor Genetics, University Hospital of Nice-Côte d'Azur University, Nice, France; Laboratory of Solid Tumor Genetics, Institute for Research on Cancer and Aging of Nice (IRCAN), CNRS UMR 7284/INSERM U1081, Nice, France
| | - Corinne Bouvier
- Department of Pathology, Timone Hospital, APHM, Marseille, France
| | - Jean-François Michiels
- Central Laboratory of Pathology, University Hospital of Nice-Côte d'Azur University, Nice, France
| | - Florence Pedeutour
- Laboratory of Solid Tumor Genetics, University Hospital of Nice-Côte d'Azur University, Nice, France; Laboratory of Solid Tumor Genetics, Institute for Research on Cancer and Aging of Nice (IRCAN), CNRS UMR 7284/INSERM U1081, Nice, France.
| |
Collapse
|
15
|
Structure-based designing efficient peptides based on p53 binding site residues to disrupt p53-MDM2/X interaction. Sci Rep 2020; 10:11449. [PMID: 32651397 PMCID: PMC7351717 DOI: 10.1038/s41598-020-67510-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 06/09/2020] [Indexed: 12/23/2022] Open
Abstract
MDM2 and MDMX are known as overexpressed oncoproteins in several wild-type p53 cancer cells. The development of potent and dual antagonist peptides for p53-MDM2/X is a continuous challenge. In this study, we intended to investigate the pivotal structural points respecting the development of potent and dual inhibitors of MDM2/X. Correspondingly, MD simulation was performed on the experimentally confirmed peptides, comprising p53, pDI, pDIQ, PMI, and computationally screened mutant pDI and pDIQ. A follow-up secondary structure analysis showed the last three C-terminal residues provide the helicity reservation of peptides bound to MDM2/X. Furthermore, a delicate residue-residue examination displayed Met 11 and Ser12 in the modified peptides contribute significantly to dual inhibition of MDM2/X. Additionally, the peptides_MDM2/X complexes' ΔGbinding extracted by the umbrella sampling method were in agreement with the pattern of their experimental affinity values. It was concluded the screened pDI mutants were considered as suitable anti-MDM2/X peptides, and the data obtained could be exploited as the theoretical structure-based guide for rational peptide design. Taking account of results, the suitable C-terminal residues of p53-based peptides especially Met11, and Ser12, as well as higher umbrella sampling, generated ΔGbinding to MDM2/X would be considered as the positive structural markers of a promising anti-cancer agent.
Collapse
|
16
|
Hattinger CM, Patrizio MP, Luppi S, Serra M. Pharmacogenomics and Pharmacogenetics in Osteosarcoma: Translational Studies and Clinical Impact. Int J Mol Sci 2020; 21:E4659. [PMID: 32629971 PMCID: PMC7369799 DOI: 10.3390/ijms21134659] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/23/2020] [Accepted: 06/25/2020] [Indexed: 12/14/2022] Open
Abstract
High-grade osteosarcoma (HGOS) is a very aggressive bone tumor which primarily affects adolescents and young adults. Although not advanced as is the case for other cancers, pharmacogenetic and pharmacogenomic studies applied to HGOS have been providing hope for an improved understanding of the biology and the identification of genetic biomarkers, which may impact on clinical care management. Recent developments of pharmacogenetics and pharmacogenomics in HGOS are expected to: i) highlight genetic events that trigger oncogenesis or which may act as drivers of disease; ii) validate research models that best predict clinical behavior; and iii) indicate genetic biomarkers associated with clinical outcome (in terms of treatment response, survival probability and susceptibility to chemotherapy-related toxicities). The generated body of information may be translated to clinical settings, in order to improve both effectiveness and safety of conventional chemotherapy trials as well as to indicate new tailored treatment strategies. Here, we review and summarize the current scientific evidence for each of the aforementioned issues in view of possible clinical applications.
Collapse
Affiliation(s)
| | | | | | - Massimo Serra
- IRCCS Istituto Ortopedico Rizzoli, Laboratory of Experimental Oncology, Pharmacogenomics and Pharmacogenetics Research Unit, 40136 Bologna, Italy; (C.M.H.); (M.P.P.); (S.L.)
| |
Collapse
|
17
|
Smyczyńska U, Strzemecki D, Czarnecka AM, Fendler W, Fiedorowicz M, Wełniak-Kamińska M, Guzowska M, Synoradzki K, Cheda Ł, Rogulski Z, Grieb P. TP53-Deficient Angiosarcoma Expression Profiling in Rat Model. Cancers (Basel) 2020; 12:cancers12061525. [PMID: 32532104 PMCID: PMC7352674 DOI: 10.3390/cancers12061525] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 06/01/2020] [Accepted: 06/03/2020] [Indexed: 12/28/2022] Open
Abstract
Sarcomas are a heterogeneous group of malignant tumors, that develop from mesenchymal cells. Sarcomas are tumors associated with poor prognosis and expected short overall survival. Efforts to improve treatment efficacy and treatment outcomes of advanced and metastatic sarcoma patients have not led to significant improvements in the last decades. In the Tp53C273X/C273X rat model we therefore aimed to characterize specific gene expression pattern of angiosarcomas with a loss of TP53 function. The presence of metabolically active tumors in several locations including the brain, head and neck, extremities and abdomen was confirmed by magnetic resonance imaging (MRI) and positron emission tomography (PET) examinations. Limb angiosarcoma tumors were selected for microarray expression analysis. The most upregulated pathways in angiosarcoma vs all other tissues were related to cell cycle with mitosis and meiosis, chromosome, nucleosome and telomere maintenance as well as DNA replication and recombination. The downregulated genes were responsible for metabolism, including respiratory chain electron transport, tricarboxylic acid (TCA) cycle, fatty acid metabolism and amino-acid catabolism. Our findings demonstrated that the type of developing sarcoma depends on genetic background, underscoring the importance of developing more malignancy susceptibility models in various strains and species to simulate the study of the diverse genetics of human sarcomas.
Collapse
Affiliation(s)
- Urszula Smyczyńska
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, 92-215 Lodz, Poland; (U.S.); (W.F.)
| | - Damian Strzemecki
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland; (D.S.); (M.F.); (M.W.-K.); (M.G.); (K.S.); (P.G.)
| | - Anna M. Czarnecka
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland; (D.S.); (M.F.); (M.W.-K.); (M.G.); (K.S.); (P.G.)
- Department of Soft Tissue, Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
- Correspondence: ; Tel.: +48-22-608-6474
| | - Wojciech Fendler
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, 92-215 Lodz, Poland; (U.S.); (W.F.)
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02284-9168, USA
| | - Michał Fiedorowicz
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland; (D.S.); (M.F.); (M.W.-K.); (M.G.); (K.S.); (P.G.)
- Small Animal Magnetic Resonance Imaging Laboratory, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Marlena Wełniak-Kamińska
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland; (D.S.); (M.F.); (M.W.-K.); (M.G.); (K.S.); (P.G.)
- Small Animal Magnetic Resonance Imaging Laboratory, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Magdalena Guzowska
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland; (D.S.); (M.F.); (M.W.-K.); (M.G.); (K.S.); (P.G.)
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences, 02-776 Warsaw, Poland
| | - Kamil Synoradzki
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland; (D.S.); (M.F.); (M.W.-K.); (M.G.); (K.S.); (P.G.)
| | - Łukasz Cheda
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, 02-093 Warsaw, Poland; (Ł.C.); (Z.R.)
| | - Zbigniew Rogulski
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, 02-093 Warsaw, Poland; (Ł.C.); (Z.R.)
| | - Paweł Grieb
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02-106 Warsaw, Poland; (D.S.); (M.F.); (M.W.-K.); (M.G.); (K.S.); (P.G.)
| |
Collapse
|
18
|
Wu HQ, Baker D, Ovaa H. Small molecules that target the ubiquitin system. Biochem Soc Trans 2020; 48:479-497. [PMID: 32196552 PMCID: PMC7200645 DOI: 10.1042/bst20190535] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 03/02/2020] [Accepted: 03/04/2020] [Indexed: 02/07/2023]
Abstract
Eukaryotic life depends upon the interplay between vast networks of signaling pathways composed of upwards of 109-1010 proteins per cell. The integrity and normal operation of the cell requires that these proteins act in a precise spatial and temporal manner. The ubiquitin system is absolutely central to this process and perturbation of its function contributes directly to the onset and progression of a wide variety of diseases, including cancer, metabolic syndromes, neurodegenerative diseases, autoimmunity, inflammatory disorders, infectious diseases, and muscle dystrophies. Whilst the individual components and the overall architecture of the ubiquitin system have been delineated in some detail, how ubiquitination might be successfully targeted, or harnessed, to develop novel therapeutic approaches to the treatment of disease, currently remains relatively poorly understood. In this review, we will provide an overview of the current status of selected small molecule ubiquitin system inhibitors. We will further discuss the unique challenges of targeting this ubiquitous and highly complex machinery, and explore and highlight potential ways in which these challenges might be met.
Collapse
Affiliation(s)
- Hai Qiu Wu
- Oncode Institute, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
| | - David Baker
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
| | - Huib Ovaa
- Oncode Institute, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
| |
Collapse
|
19
|
Schubert NA, Lowery CD, Bergthold G, Koster J, Eleveld TF, Rodríguez A, Jones DTW, Vassal G, Stancato LF, Pfister SM, Caron HN, Molenaar JJ. Systematic target actionability reviews of preclinical proof-of-concept papers to match targeted drugs to paediatric cancers. Eur J Cancer 2020; 130:168-181. [PMID: 32224415 PMCID: PMC7203547 DOI: 10.1016/j.ejca.2020.01.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/21/2020] [Accepted: 01/27/2020] [Indexed: 01/17/2023]
Abstract
Background Children with cancer are in urgent need of new therapies, as approximately 25% of patients experience a relapse and 20% succumb to their disease. Moreover, the majority of survivors suffer from clinically relevant health problems. Repurposing of targeted agents developed for adult indications could provide novel therapeutic options for paediatric cancer patients. To prioritise targeted drugs for paediatric clinical development, we applied a systematic review methodology to develop a Target Actionability Review (TAR) strategy. These TARs assess the strength and completeness of published preclinical proof-of-concept (PoC) data by structured critical appraisal of and summarising the available scientific literature for a specific target (pathway) and the associated drugs in paediatric tumours. Methods A sensitive literature search in PubMed was performed and relevant papers were identified. For each paper, the individual experimental findings were extracted, marked for paediatric tumour type and categorised into nine separate PoC data modules. Each experimental finding was scored for experimental outcome and quality independently by two reviewers; discrepancies were assessed by a third reviewer and resolved by adjudication. Scores corresponding to one PoC module were merged for each tumour type and visualised in a heat map matrix in the publicly available R2 data portal [r2.amc.nl]. Results and conclusions To test our TAR methodology, we conducted a pilot study on MDM2 and TP53. The heat map generated from analysis of 161 publications provides a rationale to support drug development in specific paediatric solid and brain tumour types. Furthermore, our review highlights tumour types where preclinical data are incomplete or lacking and for which additional preclinical testing is advisable. A new strategy to review literature on targeted compounds in paediatric cancer. Results help to guide and prioritise clinical development of novel targeted agents. Outcomes are visualised in a publicly available, interactive heat map. We applied this unique methodology to MDM2 and TP53 and MDM2 inhibitors.
Collapse
Affiliation(s)
- Nil A Schubert
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | | | | | - Jan Koster
- Department of Oncogenomics, Amsterdam University Medical Centre, Amsterdam, the Netherlands
| | - Thomas F Eleveld
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | | | - David T W Jones
- Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Gilles Vassal
- Department of Clinical Research, Gustave Roussy, Villejuif, France
| | | | - Stefan M Pfister
- Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; Heidelberg University Hospital, Heidelberg, Germany
| | | | - Jan J Molenaar
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.
| |
Collapse
|
20
|
Sinusoidal-type Angiosarcoma of the Liver: Imaging Features and Potential Diagnostic Utility of p53 Immunostaining. Am J Surg Pathol 2019; 43:1728-1731. [DOI: 10.1097/pas.0000000000001349] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
21
|
The PTEN Tumor Suppressor Gene in Soft Tissue Sarcoma. Cancers (Basel) 2019; 11:cancers11081169. [PMID: 31416195 PMCID: PMC6721622 DOI: 10.3390/cancers11081169] [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: 06/15/2019] [Revised: 07/26/2019] [Accepted: 08/08/2019] [Indexed: 02/07/2023] Open
Abstract
Soft tissue sarcoma (STS) is a rare malignancy of mesenchymal origin classified into more than 50 different subtypes with distinct clinical and pathologic features. Despite the poor prognosis in the majority of patients, only modest improvements in treatment strategies have been achieved, largely due to the rarity and heterogeneity of these tumors. Therefore, the discovery of new prognostic and predictive biomarkers, together with new therapeutic targets, is of enormous interest. Phosphatase and tensin homolog (PTEN) is a well-known tumor suppressor that commonly loses its function via mutation, deletion, transcriptional silencing, or protein instability, and is frequently downregulated in distinct sarcoma subtypes. The loss of PTEN function has consequent alterations in important pathways implicated in cell proliferation, survival, migration, and genomic stability. PTEN can also interact with other tumor suppressors and oncogenic signaling pathways that have important implications for the pathogenesis in certain STSs. The aim of the present review is to summarize the biological significance of PTEN in STS and its potential role in the development of new therapeutic strategies.
Collapse
|
22
|
Borbora D, Dutta HK, Devi KR, Mahanta J, Medhi P, Narain K. Long telomeres cooperate with p53, MDM2, and p21 polymorphisms to raise pediatric solid tumor risk. Pediatr Int 2019; 61:759-767. [PMID: 31211452 DOI: 10.1111/ped.13915] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 06/05/2019] [Accepted: 06/14/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND While leukocyte telomere length has been linked with altered risk in adult cancer, limited information is available on its association with risk in pediatric solid tumors. We investigated the association of telomeric alterations with risk of pediatric solid tumors. We also investigated whether altered telomeres cooperated with the TP53 rs1042522, MDM2 rs2279744 and CDKN1A (p21cip1 ) rs1059234 single-nucleotide polymorphisms to modify cancer risk. METHODS A total of 101 tumor patients and 202 controls were recruited for this age- and gender-matched case-control study. Relative telomere length (RTL) was determined in peripheral blood leukocytes using quantitative real-time polymerase chain reaction (PCR), and the polymorphisms were genotyped using PCR-restriction fragment length polymorphism. RESULTS Using median RTL in the healthy controls as a cut-off, children with longer telomeres were at an increased risk of developing a solid tumor (OR, 2.70; P < 0.01). When participants were categorized according to control RTL quartiles, a significant dose-response relationship was observed (χ2 = 10.95; P < 0.001). The risk for tumors increased nearly threefold (P = 0.001) for the triple interaction RTL × TP53 rs1042522 × p21cip1 rs1059234 compared with the maximum effect of any single factor, although the interaction effect was less than additive. The MDM2 rs2279744 GG genotype reduced pediatric solid tumor risk significantly (OR, 0.51). CONCLUSION Combined analysis of telomeres and genetic polymorphisms in the TP53 pathway can provide important clues to understanding pediatric solid tumor etiology.
Collapse
Affiliation(s)
- Debasish Borbora
- Regional Medical Research Centre, NE Region (ICMR), Dibrugarh, Assam, India.,Centre for Biotechnology and Bioinformatics, Dibrugarh University, Dibrugarh, Assam, India.,Department of Biotechnology, Gauhati University, Guwahati, Assam, India
| | - Hemonta K Dutta
- Department of Pediatric surgery, Assam Medical College and Hospital, Dibrugarh, Assam, India
| | - Kangjam Rekha Devi
- Regional Medical Research Centre, NE Region (ICMR), Dibrugarh, Assam, India
| | - Jagadish Mahanta
- Regional Medical Research Centre, NE Region (ICMR), Dibrugarh, Assam, India
| | - Pronita Medhi
- Department of Pathology, Assam Medical College and Hospital, Dibrugarh, Assam, India
| | - Kanwar Narain
- Regional Medical Research Centre, NE Region (ICMR), Dibrugarh, Assam, India
| |
Collapse
|
23
|
Thoenen E, Curl A, Iwakuma T. TP53 in bone and soft tissue sarcomas. Pharmacol Ther 2019; 202:149-164. [PMID: 31276706 DOI: 10.1016/j.pharmthera.2019.06.010] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 06/25/2019] [Indexed: 12/13/2022]
Abstract
Genomic and functional study of existing and emerging sarcoma targets, such as fusion proteins, chromosomal aberrations, reduced tumor suppressor activity, and oncogenic drivers, is broadening our understanding of sarcomagenesis. Among these mechanisms, the tumor suppressor p53 (TP53) plays significant roles in the suppression of bone and soft tissue sarcoma progression. Although mutations in TP53 were thought to be relatively low in sarcomas, modern techniques including whole-genome sequencing have recently illuminated unappreciated alterations in TP53 in osteosarcoma. In addition, oncogenic gain-of-function activities of missense mutant p53 (mutp53) have been reported in sarcomas. Moreover, new targeting strategies for TP53 have been discovered: restoration of wild-type p53 (wtp53) activity through inhibition of TP53 negative regulators, reactivation of the wtp53 activity from mutp53, depletion of mutp53, and targeting of vulnerabilities in cells with TP53 deletions or mutations. These discoveries enable development of novel therapeutic strategies for therapy-resistant sarcomas. We have outlined nine bone and soft tissue sarcomas for which TP53 plays a crucial tumor suppressive role. These include osteosarcoma, Ewing sarcoma, chondrosarcoma, rhabdomyosarcoma (RMS), leiomyosarcoma (LMS), synovial sarcoma, liposarcoma (LPS), angiosarcoma, and undifferentiated pleomorphic sarcoma (UPS).
Collapse
Affiliation(s)
- Elizabeth Thoenen
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66010, USA
| | - Amanda Curl
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66010, USA
| | - Tomoo Iwakuma
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66010, USA; Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66010, USA; Translational Laboratory Oncology Research, Children's Mercy Research Institute, Kansas City, MO 64108, USA.
| |
Collapse
|
24
|
Mirili C, Paydaş S, Guney IB, Ogul A, Gokcay S, Buyuksimsek M, Yetisir AE, Karaalioglu B, Tohumcuoglu M, Seydaoglu G. Assessment of potential predictive value of peripheral blood inflammatory indexes in 26 cases with soft tissue sarcoma treated by pazopanib: a retrospective study. Cancer Manag Res 2019; 11:3445-3453. [PMID: 31114378 PMCID: PMC6485039 DOI: 10.2147/cmar.s191199] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose The aim of this study was to evaluate the prognostic and predictive value of neutrophil-to-lymphocyte ratio (NLR), derived neutrophil-to-lymphocyte ratio (DNLR), lymphocyte-to-monocyte ratio (LMR), and platelet-to-lymphocyte ratio (PLR) in soft tissue sarcoma (STS) cases treated with pazopanib. Materials and methods The study population included 26 STS cases treated with pazopanib for at least 3 months. NLR, DNLR, LMR, and PLR were evaluated at baseline, and at third month of therapy and also compared with response to pazopanib. Median measurements were taken as cutoff for NLR (4.8), DNLR (3.1), LMR (3.6), and PLR (195). The associations between these cutoff values and survival times (progression-free survival [PFS] and overall survival [OS]) were assessed by Kaplan–Meier curves and Cox proportional models. Results Patients with low pretreatment NLR and DNLR had longer OS (P=0.022, P=0.018), but low PLR was found to be associated only with longer OS. Additionally, decrease in NLR and DNLR after 3 months of therapy as compared with pretreatment measurements was found to be associated with an advantage for OS (P=0.021, P=0.010, respectively) and PFS (P=0.005, P=0.001, respectively). Response to pazopanib; changes in NLR, DNLR, LMR, and PLR; and >3 metastatic sites were found to be independent risk factors in univariate analysis, but NLR was the only independent risk factor in multivariate analysis. Conclusion Low pretreatment and decrease in NLR and DNLR values, and regression/stable disease after 3 months of pazopanib are predictive factors for longer OS and PFS.
Collapse
Affiliation(s)
- Cem Mirili
- Department of Medical Oncology, Çukurova University Faculty of Medicine, Adana, Turkey,
| | - Semra Paydaş
- Department of Medical Oncology, Çukurova University Faculty of Medicine, Adana, Turkey,
| | - Isa B Guney
- Department of Nuclear Medicine, Çukurova University Faculty of Medicine, Adana, Turkey
| | - Ali Ogul
- Department of Medical Oncology, Çukurova University Faculty of Medicine, Adana, Turkey,
| | - Serkan Gokcay
- Department of Medical Oncology, Çukurova University Faculty of Medicine, Adana, Turkey,
| | - Mahmut Buyuksimsek
- Department of Medical Oncology, Çukurova University Faculty of Medicine, Adana, Turkey,
| | - Abdullah E Yetisir
- Department of Medical Oncology, Çukurova University Faculty of Medicine, Adana, Turkey,
| | - Bilgin Karaalioglu
- Department of Medical Oncology, Çukurova University Faculty of Medicine, Adana, Turkey,
| | - Mert Tohumcuoglu
- Department of Medical Oncology, Çukurova University Faculty of Medicine, Adana, Turkey,
| | - Gulsah Seydaoglu
- Department of Bioistatistics, Çukurova University Faculty of Medicine, Adana, Turkey
| |
Collapse
|
25
|
Association between TP53 rs1042522 gene polymorphism and the risk of malignant bone tumors: a meta-analysis. Biosci Rep 2019; 39:BSR20181832. [PMID: 30833364 PMCID: PMC6422898 DOI: 10.1042/bsr20181832] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 02/13/2019] [Accepted: 03/01/2019] [Indexed: 12/19/2022] Open
Abstract
TP53 is a tumor suppressor gene which is essential for regulating cell division and preventing tumor formation. Several studies have assessed the associations of TP53 single-nucleotide polymorphisms (SNP) with susceptibility of malignant bone tumors, including osteosarcoma and Ewing sarcoma, but the results are inconsistent. In the present meta-analysis, we aimed to elucidate the associations of TP53 rs1042522 genetic polymorphism with the risk of osteosarcoma or Ewing sarcoma. We systematically searched Medline, PubMed, Web of Science, Embase, and the Cochrane Library databases. Eligible studies assessing the polymorphisms in the TP53 rs1042522 gene and risk of malignant bone tumors were incorporated. The pooled odds ratio (OR) with its 95% confidence intervals (95% CIs) were used to assess these possible associations. Five studies with a total of 567 cases and 935 controls were finally included the meta-analysis. Meta-analysis of TP53 rs1042522 polymorphism was significantly associated with an increased risk of malignant bone tumors (G versus C: OR = 1.27, 95% CI 1.08–1.50, P=0.005; GG versus GC/CC: OR = 1.55, 95% CI 1.21–2.00, P=0.001). Moreover, in a stratified analysis, a statistically significant correlation between this SNP and osteosarcoma risk was also observed. Our results suggest that there are significant associations of TP53 rs1042522 polymorphism with malignant bone tumors risk. More studies based on larger sample sizes and homogeneous samples are warranted to confirm these findings.
Collapse
|
26
|
Pollock RE, Payne JE, Rogers AD, Smith SM, Iwenofu OH, Valerio IL, Zomerlei TA, Howard JH, Dornbos D, Galgano MA, Goulart C, Mendel E, Miller ED, Xu-Welliver M, Martin DD, Haglund KE, Bupathi M, Chen JL, Yeager ND. Multidisciplinary sarcoma care. Curr Probl Surg 2018; 55:517-580. [PMID: 30526918 DOI: 10.1067/j.cpsurg.2018.10.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Raphael E Pollock
- The Ohio State University Comprehensive Cancer Center, Columbus, OH.
| | - Jason E Payne
- The Ohio State University Wexner Medical Center, Columbus, OH
| | - Alan D Rogers
- The Ohio State University Wexner Medical Center, Columbus, OH
| | - Stephen M Smith
- The Ohio State University Wexner Medical Center, Columbus, OH
| | - O Hans Iwenofu
- Department of Pathology & Laboratory Medicine, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Ian L Valerio
- The Ohio State University Wexner Medical Center, Columbus, OH
| | | | | | - David Dornbos
- The Ohio State University Wexner Medical Center, Columbus, OH
| | | | | | - Ehud Mendel
- The Ohio State University Wexner Medical Center, Columbus, OH
| | - Eric D Miller
- The Ohio State University Wexner Medical Center, Columbus, OH
| | | | | | - Karl E Haglund
- The Ohio State University Wexner Medical Center, Columbus, OH
| | | | - James L Chen
- The Ohio State University Wexner Medical Center, Columbus, OH
| | - Nicholas D Yeager
- Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, OH
| |
Collapse
|
27
|
Stolte B, Iniguez AB, Dharia NV, Robichaud AL, Conway AS, Morgan AM, Alexe G, Schauer NJ, Liu X, Bird GH, Tsherniak A, Vazquez F, Buhrlage SJ, Walensky LD, Stegmaier K. Genome-scale CRISPR-Cas9 screen identifies druggable dependencies in TP53 wild-type Ewing sarcoma. J Exp Med 2018; 215:2137-2155. [PMID: 30045945 PMCID: PMC6080915 DOI: 10.1084/jem.20171066] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 03/16/2018] [Accepted: 06/27/2018] [Indexed: 01/06/2023] Open
Abstract
Stolte et al. use genome-scale CRISPR-Cas9 screening technology to identify druggable targets for TP53 wild-type Ewing sarcoma and discover reactivation of p53 through inhibition of MDM2, MDM4, Wip1, or USP7 as therapeutic strategies for the disease. Ewing sarcoma is a pediatric cancer driven by EWS-ETS transcription factor fusion oncoproteins in an otherwise stable genomic background. The majority of tumors express wild-type TP53, and thus, therapies targeting the p53 pathway would benefit most patients. To discover targets specific for TP53 wild-type Ewing sarcoma, we used a genome-scale CRISPR-Cas9 screening approach and identified and validated MDM2, MDM4, USP7, and PPM1D as druggable dependencies. The stapled peptide inhibitor of MDM2 and MDM4, ATSP-7041, showed anti-tumor efficacy in vitro and in multiple mouse models. The USP7 inhibitor, P5091, and the Wip1/PPM1D inhibitor, GSK2830371, decreased the viability of Ewing sarcoma cells. The combination of ATSP-7041 with P5091, GSK2830371, and chemotherapeutic agents showed synergistic action on the p53 pathway. The effects of the inhibitors, including the specific USP7 inhibitor XL-188, were rescued by concurrent TP53 knockout, highlighting the essentiality of intact p53 for the observed cytotoxic activities.
Collapse
Affiliation(s)
- Björn Stolte
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, MA.,Dr. von Hauner Children's Hospital, Department of Pediatrics, University Hospital, LMU Munich, Munich, Germany.,The Broad Institute of MIT and Harvard, Cambridge, MA
| | - Amanda Balboni Iniguez
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, MA.,The Broad Institute of MIT and Harvard, Cambridge, MA
| | - Neekesh V Dharia
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, MA.,The Broad Institute of MIT and Harvard, Cambridge, MA
| | - Amanda L Robichaud
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Amy Saur Conway
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Ann M Morgan
- Department of Pediatric Oncology and the Linde Program in Cancer Chemical Biology, Dana-Farber Cancer Institute, Boston, MA
| | - Gabriela Alexe
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, MA.,The Broad Institute of MIT and Harvard, Cambridge, MA.,Bioinformatics Graduate Program, Boston University, Boston, MA
| | - Nathan J Schauer
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA
| | - Xiaoxi Liu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA
| | - Gregory H Bird
- Department of Pediatric Oncology and the Linde Program in Cancer Chemical Biology, Dana-Farber Cancer Institute, Boston, MA
| | | | | | - Sara J Buhrlage
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA
| | - Loren D Walensky
- Department of Pediatric Oncology and the Linde Program in Cancer Chemical Biology, Dana-Farber Cancer Institute, Boston, MA
| | - Kimberly Stegmaier
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, MA .,The Broad Institute of MIT and Harvard, Cambridge, MA
| |
Collapse
|
28
|
Kato S, Ross JS, Gay L, Dayyani F, Roszik J, Subbiah V, Kurzrock R. Analysis of MDM2 Amplification: Next-Generation Sequencing of Patients With Diverse Malignancies. JCO Precis Oncol 2018; 2018. [PMID: 30148248 PMCID: PMC6106866 DOI: 10.1200/po.17.00235] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Purpose MDM2 amplification can promote tumorigenesis directly or indirectly through p53 inhibition. MDM2 has increasing clinical relevance because inhibitors are under evaluation in clinical trials, and MDM2 amplification is a possible genomic correlate of accelerated progression, known as hyperprogression, after anti–PD-1/PD-L1 immunotherapy. We used next-generation sequencing (NGS) to ascertain MDM2 amplification status across a large number of diverse cancers. Methods We interrogated the molecular profiles of 102,878 patients with diverse malignancies for MDM2 amplification and co-altered genes using clinical-grade NGS (182 to 465 genes). Results MDM2 amplification occurred in 3.5% of patients (3,650 of 102,878). The majority of tumor types had a small subset of patients with MDM2 amplification. Most of these patients (99.0% [3,613/3,650]) had co-alterations that accompanied MDM2 amplification. Various pathways, including those related to tyrosine kinase (37.9% [1,385 of 3,650]), PI3K signaling (25.4% [926 of 3,650]), TP53 (24.9% [910 of 3,650]), and MAPK signaling (23.6% [863 of 3,650]), were involved. Although infrequent, mismatch repair genes and PD-L1 amplification also were co-altered (2.2% [79 of 3,650]). Most patients (97.6% [3,563 of 3,650]) had one or more co-alterations potentially targetable with either a Food and Drug Administration–approved or investigational agent. MDM2 amplifications were less frequently associated with high tumor mutation burden compared with the MDM2 wild-type population (2.9% v 6.5%; P < .001). An illustrative patient who harbored MDM2 amplification and experienced hyperprogression with an immune checkpoint inhibitor is presented. Conclusion MDM2 amplification was found in 3.5% of 102,878 patients, 97.6% of whom harbored genomic co-alterations that were potentially targetable. This study suggests that a small subset of most tumor types have MDM2 amplification as well as pharmacologically tractable co-alterations.
Collapse
Affiliation(s)
- Shumei Kato
- University of California, San Diego, Moores Cancer Center, La Jolla
| | | | | | | | - Jason Roszik
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Vivek Subbiah
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Razelle Kurzrock
- University of California, San Diego, Moores Cancer Center, La Jolla
| |
Collapse
|
29
|
Molecular analyses in the diagnosis and prediction of prognosis in non-GIST soft tissue sarcomas: A systematic review and meta-analysis. Cancer Treat Rev 2018; 66:74-81. [DOI: 10.1016/j.ctrv.2018.04.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 04/10/2018] [Accepted: 04/20/2018] [Indexed: 12/14/2022]
|
30
|
Genetic susceptibility to bone and soft tissue sarcomas: a field synopsis and meta-analysis. Oncotarget 2018; 9:18607-18626. [PMID: 29719630 PMCID: PMC5915097 DOI: 10.18632/oncotarget.24719] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 03/07/2018] [Indexed: 12/18/2022] Open
Abstract
Background The genetic architecture of bone and soft tissue sarcomas susceptibility is yet to be elucidated. We aimed to comprehensively collect and meta-analyze the current knowledge on genetic susceptibility in these rare tumors. Methods We conducted a systematic review and meta-analysis of the evidence on the association between DNA variation and risk of developing sarcomas through searching PubMed, The Cochrane Library, Scopus and Web of Science databases. To evaluate result credibility, summary evidence was graded according to the Venice criteria and false positive report probability (FPRP) was calculated to further validate result noteworthiness. Integrative analysis of genetic and eQTL (expression quantitative trait locus) data was coupled with network and pathway analysis to explore the hypothesis that specific cell functions are involved in sarcoma predisposition. Results We retrieved 90 eligible studies comprising 47,796 subjects (cases: 14,358, 30%) and investigating 1,126 polymorphisms involving 320 distinct genes. Meta-analysis identified 55 single nucleotide polymorphisms (SNPs) significantly associated with disease risk with a high (N=9), moderate (N=38) and low (N=8) level of evidence, findings being classified as noteworthy basically only when the level of evidence was high. The estimated joint population attributable risk for three independent SNPs (rs11599754 of ZNF365/EGR2, rs231775 of CTLA4, and rs454006 of PRKCG) was 37.2%. We also identified 53 SNPs significantly associated with sarcoma risk based on single studies.Pathway analysis enabled us to propose that sarcoma predisposition might be linked especially to germline variation of genes whose products are involved in the function of the DNA repair machinery. Conclusions We built the first knowledgebase on the evidence linking DNA variation to sarcomas susceptibility, which can be used to generate mechanistic hypotheses and inform future studies in this field of oncology.
Collapse
|
31
|
Kitamoto K, Miura Y, Karnan S, Ota A, Konishi H, Hosokawa Y, Sato K. Inhibition of NADPH oxidase 2 induces apoptosis in osteosarcoma: The role of reactive oxygen species in cell proliferation. Oncol Lett 2018; 15:7955-7962. [PMID: 29731909 DOI: 10.3892/ol.2018.8291] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 07/12/2016] [Indexed: 12/31/2022] Open
Abstract
Osteosarcomas (OS) are aggressive tumors that are characterized by dysregulated growth and resistance to apoptosis. Reactive oxygen species (ROS) are thought to be important signal transduction molecules in the regulation of cell growth. ROS-generating nicotinamide adenine dinucleotide phosphate oxidase (NOX) family enzymes have previously been suggested to be involved in neoplastic proliferation. To examine whether NOX-mediated generation of intracellular ROS confers anti-apoptotic activity, and thus a growth advantage, the current study first analyzed the mRNA expression of NOX family members by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) in five human OS cell lines. RT-PCR analysis revealed that NOX2 and NOX4 mRNAs were expressed in all the OS cell lines examined, whereas little or no NOX1 and NOX3 mRNAs were detected. By RT-qPCR, NOX2 mRNA expression levels were demonstrated to be higher than NOX4 mRNA expression levels. The viability of OS cells decreased in a dose-dependent manner with treatment of diphenylene iodonium (DPI), an inhibitor of flavoprotein-dependent oxidase. DPI treatment was observed to reduce intracellular ROS levels by ~50%, and increase the frequency of apoptosis by 30%. Notably, small interfering RNAs (siRNAs) targeting NOX2 significantly suppressed ROS generation; ROS depletion by DPI or NOX2 siRNAs induced apoptosis in OS cells. Together, the results of the present study indicate that NOX2-mediated ROS generation promotes cell survival and ROS depletion leads to apoptosis, thus highlighting the NOX2-ROS signaling pathway as a potential therapeutic target for OS treatment.
Collapse
Affiliation(s)
- Kazumasa Kitamoto
- Department of Orthopedic Surgery, Aichi Medical University School of Medicine, Nagakute, Aichi 480-1195, Japan
| | - Yuji Miura
- Department of Pathophysiology, Aichi Medical University School of Nursing, Nagakute, Aichi 480-1195, Japan
| | - Sivasundaram Karnan
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Aichi 480-1195, Japan
| | - Akinobu Ota
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Aichi 480-1195, Japan
| | - Hiroyuki Konishi
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Aichi 480-1195, Japan
| | - Yoshitaka Hosokawa
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Aichi 480-1195, Japan
| | - Keiji Sato
- Department of Orthopedic Surgery, Aichi Medical University School of Medicine, Nagakute, Aichi 480-1195, Japan
| |
Collapse
|
32
|
Yang P, Chen W, Li X, Eilers G, He Q, Liu L, Wu Y, Wu Y, Yu W, Fletcher JA, Ou WB. Downregulation of cyclin D1 sensitizes cancer cells to MDM2 antagonist Nutlin-3. Oncotarget 2018; 7:32652-63. [PMID: 27129163 PMCID: PMC5078041 DOI: 10.18632/oncotarget.8999] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Accepted: 03/31/2016] [Indexed: 12/14/2022] Open
Abstract
The MDM2-p53 pathway has a prominent oncogenic function in the pathogenesis of various cancers. Nutlin-3, a small-molecule antagonist of MDM2-p53 interaction, inhibits proliferation in cancer cells with wild-type p53. Herein, we evaluate the expression of MDM2, both the full length and a splicing variant MDM2-A, and the sensitivity of Nutlin-3 in different cancer cell lines. Included are seven cell lines with wild-type p53 (four mesothelioma, one breast cancer, one chondrosarcoma, and one leiomyosarcoma), two liposarcoma cell lines harboring MDM2 amplification and wild-type p53, and one mesothelioma cell line harboring a p53 point mutation. Nutlin-3 treatment increased expression of cyclin D1, MDM2, and p53 in cell lines with wild-type p53. Additive effects were observed in cells containing wild-type p53 through coordinated attack on MDM2-p53 binding and cyclin D1 by lentivirual shRNA knockdown or small molecule inhibition, as demonstrated by immunoblots and cell viability analyses. Further results demonstrate that MDM2 binds to cyclin D1, and that an increase in cyclin D1 expression after Nutlin-3 treatment is correlated with expression and ubiquitin E3-ligase activity of MDM2. MDM2 and p53 knockdown experiments demonstrated inhibition of cyclin D1 by MDM2 but not p53. These results indicate that combination inhibition of cyclin D1 and MDM2-p53 binding warrants clinical evaluation as a novel therapeutic strategy in cancer cells harboring wild-type p53.
Collapse
Affiliation(s)
- Peipei Yang
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, China
| | - Weicai Chen
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xuhui Li
- Zhejiang Provincial Key Laboratory of Applied Enzymology, Yangtze Delta Region Institute of Tsinghua University, Jiaxing, Zhejiang, China
| | - Grant Eilers
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Quan He
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, China
| | - Lili Liu
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yeqing Wu
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yuehong Wu
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, China
| | - Wei Yu
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, China
| | - Jonathan A Fletcher
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Wen-Bin Ou
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Applied Enzymology, Yangtze Delta Region Institute of Tsinghua University, Jiaxing, Zhejiang, China.,Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
33
|
Integrative genomic and transcriptomic analysis of leiomyosarcoma. Nat Commun 2018; 9:144. [PMID: 29321523 PMCID: PMC5762758 DOI: 10.1038/s41467-017-02602-0] [Citation(s) in RCA: 213] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 12/13/2017] [Indexed: 02/07/2023] Open
Abstract
Leiomyosarcoma (LMS) is an aggressive mesenchymal malignancy with few therapeutic options. The mechanisms underlying LMS development, including clinically actionable genetic vulnerabilities, are largely unknown. Here we show, using whole-exome and transcriptome sequencing, that LMS tumors are characterized by substantial mutational heterogeneity, near-universal inactivation of TP53 and RB1, widespread DNA copy number alterations including chromothripsis, and frequent whole-genome duplication. Furthermore, we detect alternative telomere lengthening in 78% of cases and identify recurrent alterations in telomere maintenance genes such as ATRX, RBL2, and SP100, providing insight into the genetic basis of this mechanism. Finally, most tumors display hallmarks of "BRCAness", including alterations in homologous recombination DNA repair genes, multiple structural rearrangements, and enrichment of specific mutational signatures, and cultured LMS cells are sensitive towards olaparib and cisplatin. This comprehensive study of LMS genomics has uncovered key biological features that may inform future experimental research and enable the design of novel therapies.
Collapse
|
34
|
Zhang Q, Kanis MJ, Ubago J, Liu D, Scholtens DM, Strohl AE, Lurain JR, Shahabi S, Kong B, Wei JJ. The selected biomarker analysis in 5 types of uterine smooth muscle tumors. Hum Pathol 2017; 76:17-27. [PMID: 29258902 DOI: 10.1016/j.humpath.2017.12.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 11/25/2017] [Accepted: 12/07/2017] [Indexed: 10/18/2022]
Abstract
Uterine smooth muscle tumors (USMTs) consist of a group of histologically heterogeneous and clinically diverse diseases ranging from malignant leiomyosarcoma (LMS) to benign leiomyoma (ULM). The genetic alterations in LMS are complex, with some genetic alterations present in both LMS and other atypical histologic variants of USMT. In this study, we reviewed 119 USMTs with a diagnosis of LMS, smooth muscle tumor of uncertain malignant potential, atypical leiomyomas/leiomyoma with bizarre nuclei, and cellular leiomyoma, as well as 46 ULMs and 60 myometrial controls. We selected 17 biomarkers highly relevant to LMS in 4 tumorigenic pathways including steroid hormone receptors (estrogen receptor [ER] and progesterone receptor [PR]), cell cycle/tumor suppressor genes, AKT pathway markers, and associated oncogenes. ER and PR expression was significantly lower in LMS than smooth muscle tumor of uncertain malignant potential, atypical leiomyomas/leiomyoma with bizarre nuclei, cellular leiomyoma, and ULM (P < .01). Sixty-five percent of LMSs showed complete loss of ER, and 75% of LMSs showed complete loss of PR. All cell cycle genes were differentially expressed in different types of tumor, but significant overlap was noted. More than 75% of LMSs had Ki-67 index greater than 33%, and only 5% in all other types of USMT. Expression of the selected oncogenes varied widely among different types of USMT. PR positivity and p53 had a borderline association with progression-free survival (P = .055 for PR and P = .0847 for p53). Furthermore, high PR expression was significantly associated with a longer overall survival (P = .0163, hazard ratio 0.198). Cell proliferative indices (Ki-67) and sex steroid hormone receptors were the most valuable markers in differentiating LMS from other USMT variants.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Pathology, Northwestern University, Feinberg School of Medicine; Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, PR China
| | - Margaux Jenna Kanis
- Department of Gynecology and Obstetrics, Northwestern University, Feinberg School of Medicine
| | - Julianne Ubago
- Department of Pathology, Northwestern University, Feinberg School of Medicine
| | - Dachao Liu
- Department of Preventive Medicine, Feinberg School of Medicine
| | | | - Anna E Strohl
- Department of Gynecology and Obstetrics, Northwestern University, Feinberg School of Medicine
| | - John R Lurain
- Department of Gynecology and Obstetrics, Northwestern University, Feinberg School of Medicine
| | - Shohreh Shahabi
- Department of Gynecology and Obstetrics, Northwestern University, Feinberg School of Medicine
| | - Beihua Kong
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, Shandong, 250012, PR China
| | - Jian-Jun Wei
- Department of Pathology, Northwestern University, Feinberg School of Medicine; Department of Gynecology and Obstetrics, Northwestern University, Feinberg School of Medicine.
| |
Collapse
|
35
|
Gao P, Seebacher NA, Hornicek F, Guo Z, Duan Z. Advances in sarcoma gene mutations and therapeutic targets. Cancer Treat Rev 2017; 62:98-109. [PMID: 29190505 DOI: 10.1016/j.ctrv.2017.11.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 10/31/2017] [Accepted: 11/01/2017] [Indexed: 12/14/2022]
Abstract
Sarcomas are rare and complex malignancies that have been associated with a poor prognostic outcome. Over the last few decades, traditional treatment with surgery and/or chemotherapy has not significantly improved outcomes for most types of sarcomas. In recent years, there have been significant advances in the understanding of specific gene mutations that are important in driving the pathogenesis and progression of sarcomas. Identification of these new gene mutations, using next-generation sequencing and advanced molecular techniques, has revealed a range of potential therapeutic targets. This, in turn, may lead to the development of novel agents targeted to different sarcoma subtypes. In this review, we highlight the advances made in identifying sarcoma gene mutations, including those of p53, RB, PI3K and IDH genes, as well as novel therapeutic strategies aimed at utilizing these mutant genes. In addition, we discuss a number of preclinical studies and ongoing early clinical trials in sarcoma targeting therapies, as well as gene editing technology, which may provide a better choice for sarcoma patient management.
Collapse
Affiliation(s)
- Peng Gao
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China; UCLA Orthopaedic Surgery, Sarcoma Biology Laboratory, 615 Charles E Young Dr. South, Biomedical Sciences Research Building, Room 410, Los Angeles, CA 90095, USA
| | - Nicole A Seebacher
- UCLA Orthopaedic Surgery, Sarcoma Biology Laboratory, 615 Charles E Young Dr. South, Biomedical Sciences Research Building, Room 410, Los Angeles, CA 90095, USA
| | - Francis Hornicek
- UCLA Orthopaedic Surgery, Sarcoma Biology Laboratory, 615 Charles E Young Dr. South, Biomedical Sciences Research Building, Room 410, Los Angeles, CA 90095, USA
| | - Zheng Guo
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| | - Zhenfeng Duan
- UCLA Orthopaedic Surgery, Sarcoma Biology Laboratory, 615 Charles E Young Dr. South, Biomedical Sciences Research Building, Room 410, Los Angeles, CA 90095, USA.
| |
Collapse
|
36
|
Buchakjian MR, Merritt NM, Moose DL, Dupuy AJ, Tanas MR, Henry MD. A Trp53fl/flPtenfl/fl mouse model of undifferentiated pleomorphic sarcoma mediated by adeno-Cre injection and in vivo bioluminescence imaging. PLoS One 2017; 12:e0183469. [PMID: 28841687 PMCID: PMC5571905 DOI: 10.1371/journal.pone.0183469] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 08/04/2017] [Indexed: 12/24/2022] Open
Abstract
Genetic mouse models of soft tissue sarcoma provide critical insights into disease pathophysiology, which are oftentimes unable to be extracted from human tumor samples or xenograft models. In this study we describe a mouse model of soft tissue sarcoma mediated by adenoviral-Cre recombinase injection into Trp53fl/fl/Ptenfl/fl lox-stop-lox luciferase mice. Injection of adenovirus expressing Cre recombinase, either subcutaneously or intramuscularly in two experimental groups, results in viral infection and gene recombination with 100% penetrance within the first 24 hours following injection. Luciferase expression measured by real-time bioluminescence imaging increases over time, with an initial robust increase following viral injection, followed by a steady rise over the next several weeks as primary tumors develop and grow. Intramuscular injections were more commonly associated with evidence of systemic viral distribution than subcutaneous injections. All mice developed soft tissue sarcomas at the primary injection site, with histological examination identifying 93% of tumors as invasive pleomorphic sarcomas based on microscopic morphology and immunohistochemical expression of sarcoma markers. A lymphocytic infiltrate was present in 64% of the sarcomas in this immunocompetent model and 71% of tumors expressed PD-L1. This is the first report of a viral-Cre mediated Trp53/Pten mouse model of undifferentiated pleomorphic sarcoma. The bioluminescence imaging feature, along with high penetrance of the model and its immunological characteristics, makes it suited for pre-clinical studies of soft tissue sarcoma.
Collapse
Affiliation(s)
- Marisa R. Buchakjian
- Department of Otolaryngology – Head and Neck Surgery, University of Iowa Hospitals & Clinics, Iowa City, Iowa, United States of America
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
- Holden Comprehensive Cancer Center, University of Iowa Hospitals & Clinics, Iowa City, Iowa, United States of America
| | - Nicole M. Merritt
- Department of Pathology, University of Iowa Hospitals & Clinics, Iowa City, Iowa, United States of America
| | - Devon L. Moose
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
| | - Adam J. Dupuy
- Holden Comprehensive Cancer Center, University of Iowa Hospitals & Clinics, Iowa City, Iowa, United States of America
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
| | - Munir R. Tanas
- Holden Comprehensive Cancer Center, University of Iowa Hospitals & Clinics, Iowa City, Iowa, United States of America
- Department of Pathology, University of Iowa Hospitals & Clinics, Iowa City, Iowa, United States of America
- * E-mail:
| | - Michael D. Henry
- Department of Molecular Physiology and Biophysics, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
- Holden Comprehensive Cancer Center, University of Iowa Hospitals & Clinics, Iowa City, Iowa, United States of America
- Department of Pathology, University of Iowa Hospitals & Clinics, Iowa City, Iowa, United States of America
| |
Collapse
|
37
|
Miyazaki M, Aoki M, Oba S, Sakata T, Nakagawa T, Nabeshima K. A rare case of dedifferentiated liposarcoma of the sinonasal cavity: A case report. Mol Clin Oncol 2017; 7:539-542. [PMID: 29046788 PMCID: PMC5639279 DOI: 10.3892/mco.2017.1379] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 05/19/2017] [Indexed: 11/24/2022] Open
Abstract
Sarcoma is an uncommon histopathological presentation of sinonasal tumors, comprising ~15% of all cases; liposarcoma is particularly uncommon. An analysis of the available medical literature revealed no prior reports of dedifferentiated liposarcoma (DDLPS) of the sinonasal cavity. This case report presents a rare case of DDLPS of the sinonasal cavity. A 40-year old six-week pregnant female was admitted with a left nasal obstruction. Endoscopic evaluation of the left nasal cavity revealed a polypoid lesion. A computed tomography scan indicated a mass invading the left nasal cavity, maxillary sinus and anterior ethmoid sinus with focal destruction of the surrounding bone. A biopsy of the tumor was performed and hematoxylin and eosin staining of the tissue sections revealed proliferation of atypical and pleomorphic spindle cells with enlarged or elongated hyperchromatic nuclei and occasional vacuolated cytoplasm arranged in short interlacing fascicles or storiform structures, accompanied by tumor necrosis. These findings were consistent with undifferentiated pleomorphic sarcoma. Immunohistochemically, the tumor cells were positive for cyclin dependent kinase 4, mouse double minute 2 homolog (MDM2) and adipophilin. Fluorescence in situ hybridization (FISH) analysis revealed amplification of the MDM2 gene. Recently, undifferentiated pleomorphic sarcoma without areas of well-differentiated liposarcoma but with MDM2 amplification is regarded as conventional DDLPS. In the present case, the tumor was diagnosed as a DDLPS due to the results of histopathological, immunohistochemical and FISH analysis.
Collapse
Affiliation(s)
- Masaru Miyazaki
- Department of Pathology, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan.,Department of Otorhinolaryngology and Head and Neck Surgery, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan
| | - Mikiko Aoki
- Department of Pathology, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan
| | - Satoru Oba
- Department of Otorhinolaryngology and Head and Neck Surgery, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan
| | - Toshifumi Sakata
- Department of Otorhinolaryngology and Head and Neck Surgery, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan
| | - Takashi Nakagawa
- Department of Otorhinolaryngology and Head and Neck Surgery, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan
| | - Kazuki Nabeshima
- Department of Pathology, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan
| |
Collapse
|
38
|
Abstract
Osteosarcoma (OS) is a malignant bone tumor which is found primarily in adolescents, with the distal femur as the most common location. OS with a jaw primary is present in only about 10% of cases and the risk of recurrence is considered to be decreased in the jaw versus other primary locations. We present a unique case of a patient with localized OS of the jaw with an isolated recurrence in her bone marrow almost 5 years after completion of initial treatment.
Collapse
|
39
|
Peng G, Bojadzieva J, Ballinger ML, Li J, Blackford AL, Mai PL, Savage SA, Thomas DM, Strong LC, Wang W. Estimating TP53 Mutation Carrier Probability in Families with Li-Fraumeni Syndrome Using LFSPRO. Cancer Epidemiol Biomarkers Prev 2017; 26:837-844. [PMID: 28137790 DOI: 10.1158/1055-9965.epi-16-0695] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 01/19/2017] [Accepted: 01/20/2017] [Indexed: 11/16/2022] Open
Abstract
Background: Li-Fraumeni syndrome (LFS) is associated with germline TP53 mutations and a very high lifetime cancer risk. Algorithms that assess a patient's risk of inherited cancer predisposition are often used in clinical counseling. The existing LFS criteria have limitations, suggesting the need for an advanced prediction tool to support clinical decision making for TP53 mutation testing and LFS management.Methods: Based on a Mendelian model, LFSPRO estimates TP53 mutation probability through the Elston-Stewart algorithm and consequently estimates future risk of cancer. With independent datasets of 1,353 tested individuals from 867 families, we evaluated the prediction performance of LFSPRO.Results: LFSPRO accurately predicted TP53 mutation carriers in a pediatric sarcoma cohort from MD Anderson Cancer Center in the United States, the observed to expected ratio (OE) = 1.35 (95% confidence interval, 0.99-1.80); area under the receiver operating characteristic curve (AUC) = 0.85 (0.75-0.93); a population-based sarcoma cohort from the International Sarcoma Kindred Study in Australia, OE = 1.62 (1.03-2.55); AUC = 0.67 (0.54-0.79); and the NCI LFS study cohort, OE = 1.28 (1.17-1.39); AUC = 0.82 (0.78-0.86). LFSPRO also showed higher sensitivity and specificity than the classic LFS and Chompret criteria. LFSPRO is freely available through the R packages LFSPRO and BayesMendel.Conclusions: LFSPRO shows good performance in predicting TP53 mutations in individuals and families in varied situations.Impact: LFSPRO is more broadly applicable than the current clinical criteria and may improve clinical management for individuals and families with LFS. Cancer Epidemiol Biomarkers Prev; 26(6); 837-44. ©2017 AACR.
Collapse
Affiliation(s)
- Gang Peng
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jasmina Bojadzieva
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mandy L Ballinger
- The Kinghorn Cancer Center and Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Jialu Li
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Amanda L Blackford
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Phuong L Mai
- Clinical Genetics Branch, Division of Cancer Genetic and Epidemiology, National Cancer Institute, Bethesda, Maryland
| | - Sharon A Savage
- Clinical Genetics Branch, Division of Cancer Genetic and Epidemiology, National Cancer Institute, Bethesda, Maryland
| | - David M Thomas
- The Kinghorn Cancer Center and Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Louise C Strong
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wenyi Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
40
|
Abstract
Sarcomas are rare malignancies of mesenchymal origin and are broadly divided into soft tissue sarcomas and bone sarcomas. The etiology of these tumors is largely unknown, and most sarcomas are sporadic. A small subset of sarcomas is associated with certain genetic syndromes and environmental factors. Ionizing radiation is the strongest environmental factor linked to sarcoma development.
Collapse
Affiliation(s)
- Jane Y C Hui
- Division of Surgical Oncology, Department of Surgery, University of Minnesota, 420 Delaware Street Southeast, Mayo Mail Code 195, Minneapolis, MN 55455, USA.
| |
Collapse
|
41
|
Gerarduzzi C, de Polo A, Liu XS, El Kharbili M, Little JB, Yuan ZM. Human epidermal growth factor receptor 4 (Her4) Suppresses p53 Protein via Targeting the MDMX-MDM2 Protein Complex: IMPLICATION OF A NOVEL MDMX SER-314 PHOSPHOSITE. J Biol Chem 2016; 291:25937-25949. [PMID: 27777309 DOI: 10.1074/jbc.m116.752303] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 10/03/2016] [Indexed: 12/11/2022] Open
Abstract
Deregulated receptor tyrosine kinase (RTK) signaling is frequently associated with tumorigenesis and therapy resistance, but its underlying mechanisms still need to be elucidated. In this study, we have shown that the RTK human epidermal growth factor receptor 4 (Her4, also known as Erbb4) can inhibit the tumor suppressor p53 by regulating MDMX-mouse double minute 2 homolog (MDM2) complex stability. Upon activation by either overexpression of a constitutively active vector or ligand binding (Neuregulin-1), Her4 was able to stabilize the MDMX-MDM2 complex, resulting in suppression of p53 transcriptional activity, as shown by p53-responsive element-driven luciferase assay and mRNA levels of p53 target genes. Using a phospho-proteomics approach, we functionally identified a novel Her4-induced posttranslational modification on MDMX at Ser-314, a putative phosphorylation site for the CDK4/6 kinase. Remarkably, inhibition of Ser-314 phosphorylation either with Ser-to-Ala substitution or with a specific inhibitor of CDK4/6 kinase blocked Her4-induced stabilization of MDMX-MDM2 and rescued p53 activity. Our study offers insights into the mechanisms of deregulated RTK-induced carcinogenesis and provides the basis for the use of inhibitors targeting RTK-mediated signals for p53 restoration.
Collapse
Affiliation(s)
- Casimiro Gerarduzzi
- From the John B. Little Center for Radiation Sciences, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115
| | - Anna de Polo
- From the John B. Little Center for Radiation Sciences, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115
| | - Xue-Song Liu
- From the John B. Little Center for Radiation Sciences, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115
| | - Manale El Kharbili
- From the John B. Little Center for Radiation Sciences, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115
| | - John B Little
- From the John B. Little Center for Radiation Sciences, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115
| | - Zhi-Min Yuan
- From the John B. Little Center for Radiation Sciences, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115
| |
Collapse
|
42
|
Bilbao-Aldaiturriaga N, Askaiturrieta Z, Granado-Tajada I, Goričar K, Dolžan V, For The Slovenian Osteosarcoma Study Group, Garcia-Miguel P, Garcia de Andoin N, Martin-Guerrero I, Garcia-Orad A. A systematic review and meta-analysis of MDM2 polymorphisms in osteosarcoma susceptibility. Pediatr Res 2016; 80:472-9. [PMID: 27438225 DOI: 10.1038/pr.2016.120] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 04/21/2016] [Indexed: 01/05/2023]
Abstract
Two polymorphisms in the murine double minute 2 (MDM2) gene (rs1690916 and rs2279744) have been associated with the risk of osteosarcoma (OS). When we analyzed these two polymorphisms in two new independents cohorts (Spanish and Slovenian), we found no association. In order to clarify this, we conducted a meta-analysis including six populations, with a total of 246 OS patients and 1,760 controls for rs1690916; and 433 OS patients and 1,959 controls for rs2279744. Pooled odds ratio risks and corresponding 95% CI were estimated to assess the possible associations. Our results showed that these two polymorphisms were not associated with the susceptibility of OS under any genetic model studied. In conclusion, the present meta-analysis indicates that MDM2 rs1690916 and rs2279744 cannot be considered as genetic risk factors for OS susceptibility in the different populations. Therefore, the influence of these two polymorphisms on the risk of OS may be less important than previously suggested. Future studies are needed to confirm these results.
Collapse
Affiliation(s)
- Nerea Bilbao-Aldaiturriaga
- Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Medicine and Odontology, University of the Basque Country, UPV/EHU, Spain
| | - Ziortza Askaiturrieta
- Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Medicine and Odontology, University of the Basque Country, UPV/EHU, Spain
| | - Itsasne Granado-Tajada
- Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Medicine and Odontology, University of the Basque Country, UPV/EHU, Spain
| | - Katja Goričar
- Institute of Biochemistry, Faculty of Medicine, Ljubljana, Slovenia
| | - Vita Dolžan
- Institute of Biochemistry, Faculty of Medicine, Ljubljana, Slovenia
| | | | | | | | - Idoia Martin-Guerrero
- Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Medicine and Odontology, University of the Basque Country, UPV/EHU, Spain
| | - Africa Garcia-Orad
- Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Medicine and Odontology, University of the Basque Country, UPV/EHU, Spain.,BioCruces Health Research Institute, Barakaldo, Spain
| |
Collapse
|
43
|
Serra M, Hattinger CM. The pharmacogenomics of osteosarcoma. THE PHARMACOGENOMICS JOURNAL 2016; 17:11-20. [PMID: 27241064 DOI: 10.1038/tpj.2016.45] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 04/15/2016] [Accepted: 05/04/2016] [Indexed: 12/30/2022]
Abstract
Osteosarcoma (OS), the most common malignant tumor of bone, is presently treated with multidrug neoadjuvant chemotherapy protocols, which allow to cure 60-65% of patients but also induce toxicity events that cannot be predicted or efficiently prevented. The identification and validation of pharmacogenomic biomarkers is, therefore, absolutely warranted to provide the bases for planning personalized treatments with the aim to increase the therapeutic benefits and to avoid or limit unnecessary toxicities. As several targeted therapies against molecular and immunological markers in OS are presently under clinical investigation, it may be speculated that some new agents for innovative treatments may emerge in the next years. However, the real improvement of therapeutic perspectives for OS is strictly connected to the identification of pharmacogenomic biomarkers that may stratify patients in responders or non-responders and identify those individuals with higher susceptibility to treatment-associated toxicity. This review provides an overview of the pharmacogenomic biomarkers identified so far in OS, which appear to be promising candidates for a translation to clinical practice, after further investigation and/or prospective validation.
Collapse
Affiliation(s)
- M Serra
- Laboratory of Experimental Oncology, Orthopaedic Rizzoli Institute, Bologna, Italy
| | - C M Hattinger
- Laboratory of Experimental Oncology, Orthopaedic Rizzoli Institute, Bologna, Italy
| |
Collapse
|
44
|
Hayashi T, Horiuchi A, Sano K, Kanai Y, Yaegashi N, Aburatani H, Konishi I. Biological characterization of soft tissue sarcomas. ANNALS OF TRANSLATIONAL MEDICINE 2016; 3:368. [PMID: 26807423 DOI: 10.3978/j.issn.2305-5839.2015.12.33] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Soft tissue sarcomas are neoplastic malignancies that typically arise in tissues of mesenchymal origin. The identification of novel molecular mechanisms leading to mesenchymal transformation and the establishment of new therapies and diagnostic biomarker has been hampered by several critical factors. First, malignant soft tissue sarcomas are rarely observed in the clinic with fewer than 15,000 newly cases diagnosed each year in the United States. Another complicating factor is that soft tissue sarcomas are extremely heterogeneous as they arise in a multitude of tissues from many different cell lineages. The scarcity of clinical materials coupled with its inherent heterogeneity creates a challenging experimental environment for clinicians and scientists. Faced with these challenges, there has been extremely limited advancement in clinical treatment options available to patients as compared to other malignant tumours. In order to glean insight into the pathobiology of soft tissue sarcomas, scientists are now using mouse models whose genomes have been specifically tailored to carry gene deletions, gene amplifications, and somatic mutations commonly observed in human soft tissue sarcomas. The use of these model organisms has been successful in increasing our knowledge and understanding of how alterations in relevant oncogenic and/or tumour suppressive signal cascades, i.e., interferon-γ (IFN-γ), tumour protein 53 (TP53) and/or retinoblastoma (RB) pathway directly impact sarcomagenesis. It is the goal of many in the physiological community that the use of several mouse models will serve as powerful in vivo tools for further understanding of sarcomagenesis and potentially identify new diagnostic biomarker and therapeutic strategies against human soft tissue sarcomas.
Collapse
Affiliation(s)
- Takuma Hayashi
- 1 Department of Immunology and Infectious Disease, Shinshu University School of Medicine, Nagano, Japan ; 2 Promoting Business using Advanced Technology, Japan Science and Technology Agency (JST), Tokyo, Japan ; 3 Sigma-Aldrich Collaboration Laboratory, Rehovot 76100, Israel ; 4 Horiuchi Ladies Clinic, Nagano, Japan ; 5 Department of Laboratory Medicine, Shinshu University Hospital, Nagano, Japan ; 6 Department of Pathology, Keio University Graduate School of Medicine, Tokyo, Japan ; 7 The International Human Epigenome Consortium (IHEC) and CREST, Japan Science and Technology Agency (JST), Saitama, Japan ; 8 Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Miyagi, Japan ; 9 The Cancer System Laboratory, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan ; 10 Department of Obstetrics and Gynecology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Akiko Horiuchi
- 1 Department of Immunology and Infectious Disease, Shinshu University School of Medicine, Nagano, Japan ; 2 Promoting Business using Advanced Technology, Japan Science and Technology Agency (JST), Tokyo, Japan ; 3 Sigma-Aldrich Collaboration Laboratory, Rehovot 76100, Israel ; 4 Horiuchi Ladies Clinic, Nagano, Japan ; 5 Department of Laboratory Medicine, Shinshu University Hospital, Nagano, Japan ; 6 Department of Pathology, Keio University Graduate School of Medicine, Tokyo, Japan ; 7 The International Human Epigenome Consortium (IHEC) and CREST, Japan Science and Technology Agency (JST), Saitama, Japan ; 8 Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Miyagi, Japan ; 9 The Cancer System Laboratory, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan ; 10 Department of Obstetrics and Gynecology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kenji Sano
- 1 Department of Immunology and Infectious Disease, Shinshu University School of Medicine, Nagano, Japan ; 2 Promoting Business using Advanced Technology, Japan Science and Technology Agency (JST), Tokyo, Japan ; 3 Sigma-Aldrich Collaboration Laboratory, Rehovot 76100, Israel ; 4 Horiuchi Ladies Clinic, Nagano, Japan ; 5 Department of Laboratory Medicine, Shinshu University Hospital, Nagano, Japan ; 6 Department of Pathology, Keio University Graduate School of Medicine, Tokyo, Japan ; 7 The International Human Epigenome Consortium (IHEC) and CREST, Japan Science and Technology Agency (JST), Saitama, Japan ; 8 Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Miyagi, Japan ; 9 The Cancer System Laboratory, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan ; 10 Department of Obstetrics and Gynecology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yae Kanai
- 1 Department of Immunology and Infectious Disease, Shinshu University School of Medicine, Nagano, Japan ; 2 Promoting Business using Advanced Technology, Japan Science and Technology Agency (JST), Tokyo, Japan ; 3 Sigma-Aldrich Collaboration Laboratory, Rehovot 76100, Israel ; 4 Horiuchi Ladies Clinic, Nagano, Japan ; 5 Department of Laboratory Medicine, Shinshu University Hospital, Nagano, Japan ; 6 Department of Pathology, Keio University Graduate School of Medicine, Tokyo, Japan ; 7 The International Human Epigenome Consortium (IHEC) and CREST, Japan Science and Technology Agency (JST), Saitama, Japan ; 8 Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Miyagi, Japan ; 9 The Cancer System Laboratory, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan ; 10 Department of Obstetrics and Gynecology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Nobuo Yaegashi
- 1 Department of Immunology and Infectious Disease, Shinshu University School of Medicine, Nagano, Japan ; 2 Promoting Business using Advanced Technology, Japan Science and Technology Agency (JST), Tokyo, Japan ; 3 Sigma-Aldrich Collaboration Laboratory, Rehovot 76100, Israel ; 4 Horiuchi Ladies Clinic, Nagano, Japan ; 5 Department of Laboratory Medicine, Shinshu University Hospital, Nagano, Japan ; 6 Department of Pathology, Keio University Graduate School of Medicine, Tokyo, Japan ; 7 The International Human Epigenome Consortium (IHEC) and CREST, Japan Science and Technology Agency (JST), Saitama, Japan ; 8 Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Miyagi, Japan ; 9 The Cancer System Laboratory, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan ; 10 Department of Obstetrics and Gynecology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroyuki Aburatani
- 1 Department of Immunology and Infectious Disease, Shinshu University School of Medicine, Nagano, Japan ; 2 Promoting Business using Advanced Technology, Japan Science and Technology Agency (JST), Tokyo, Japan ; 3 Sigma-Aldrich Collaboration Laboratory, Rehovot 76100, Israel ; 4 Horiuchi Ladies Clinic, Nagano, Japan ; 5 Department of Laboratory Medicine, Shinshu University Hospital, Nagano, Japan ; 6 Department of Pathology, Keio University Graduate School of Medicine, Tokyo, Japan ; 7 The International Human Epigenome Consortium (IHEC) and CREST, Japan Science and Technology Agency (JST), Saitama, Japan ; 8 Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Miyagi, Japan ; 9 The Cancer System Laboratory, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan ; 10 Department of Obstetrics and Gynecology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ikuo Konishi
- 1 Department of Immunology and Infectious Disease, Shinshu University School of Medicine, Nagano, Japan ; 2 Promoting Business using Advanced Technology, Japan Science and Technology Agency (JST), Tokyo, Japan ; 3 Sigma-Aldrich Collaboration Laboratory, Rehovot 76100, Israel ; 4 Horiuchi Ladies Clinic, Nagano, Japan ; 5 Department of Laboratory Medicine, Shinshu University Hospital, Nagano, Japan ; 6 Department of Pathology, Keio University Graduate School of Medicine, Tokyo, Japan ; 7 The International Human Epigenome Consortium (IHEC) and CREST, Japan Science and Technology Agency (JST), Saitama, Japan ; 8 Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Miyagi, Japan ; 9 The Cancer System Laboratory, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan ; 10 Department of Obstetrics and Gynecology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
45
|
Chinyengetere F, Sekula DJ, Lu Y, Giustini AJ, Sanglikar A, Kawakami M, Ma T, Burkett SS, Eisenberg BL, Wells WA, Hoopes PJ, Demicco EG, Lazar AJ, Torres KE, Memoli V, Freemantle SJ, Dmitrovsky E. Mice null for the deubiquitinase USP18 spontaneously develop leiomyosarcomas. BMC Cancer 2015; 15:886. [PMID: 26555296 PMCID: PMC4640382 DOI: 10.1186/s12885-015-1883-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 10/30/2015] [Indexed: 11/10/2022] Open
Abstract
Background USP18 (ubiquitin-specific protease 18) removes ubiquitin-like modifier interferon stimulated gene 15 (ISG15) from conjugated proteins. USP18 null mice in a FVB/N background develop tumors as early as 2 months of age. These tumors are leiomyosarcomas and thus represent a new murine model for this disease. Methods Heterozygous USP18 +/− FVB/N mice were bred to generate wild-type, heterozygous and homozygous cohorts. Tumors were characterized immunohistochemically and two cell lines were derived from independent tumors. Cell lines were karyotyped and their responses to restoration of USP18 activity assessed. Drug testing and tumorigenic assays were also performed. USP18 immunohistochemical staining in a large series of human leiomyosacomas was examined. Results USP18 −/− FVB/N mice spontaneously develop tumors predominantly on the back of the neck with most tumors evident between 6–12 months (80 % penetrance). Immunohistochemical characterization of the tumors confirmed they were leiomyosarcomas, which originate from smooth muscle. Restoration of USP18 activity in sarcoma-derived cell lines did not reduce anchorage dependent or independent growth or xenograft tumor formation demonstrating that these cells no longer require USP18 suppression for tumorigenesis. Karyotyping revealed that both tumor-derived cell lines were aneuploid with extra copies of chromosomes 3 and 15. Chromosome 15 contains the Myc locus and MYC is also amplified in human leiomyosarcomas. MYC protein levels were elevated in both murine leiomyosarcoma cell lines. Stabilized P53 protein was detected in a subset of these murine tumors, another feature of human leiomyosarcomas. Immunohistochemical analyses of USP18 in human leiomyosarcomas revealed a range of staining intensities with the highest USP18 expression in normal vascular smooth muscle. USP18 tissue array analysis of primary leiomyosarcomas from 89 patients with a clinical database revealed cases with reduced USP18 levels had a significantly decreased time to metastasis (P = 0.0441). Conclusions USP18 null mice develop leiomyosarcoma recapitulating key features of clinical leiomyosarcomas and patients with reduced-USP18 tumor levels have an unfavorable outcome. USP18 null mice and the derived cell lines represent clinically-relevant models of leiomyosarcoma and can provide insights into both leiomyosarcoma biology and therapy. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1883-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fadzai Chinyengetere
- Department of Pharmacology and Toxicology, Dartmouth, Hanover, NH, USA. .,Geisel School of Medicine, Dartmouth, Hanover, NH, USA. .,Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA.
| | - David J Sekula
- Department of Pharmacology and Toxicology, Dartmouth, Hanover, NH, USA. .,Geisel School of Medicine, Dartmouth, Hanover, NH, USA. .,Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA.
| | - Yun Lu
- Department of Pharmacology and Toxicology, Dartmouth, Hanover, NH, USA. .,Geisel School of Medicine, Dartmouth, Hanover, NH, USA. .,Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA.
| | - Andrew J Giustini
- Geisel School of Medicine, Dartmouth, Hanover, NH, USA. .,Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA. .,Thayer School of Engineering, Dartmouth College, Hanover, NH, USA.
| | | | - Masanori Kawakami
- Department of Pharmacology and Toxicology, Dartmouth, Hanover, NH, USA. .,Geisel School of Medicine, Dartmouth, Hanover, NH, USA. .,Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA.
| | - Tian Ma
- Department of Pharmacology and Toxicology, Dartmouth, Hanover, NH, USA. .,Geisel School of Medicine, Dartmouth, Hanover, NH, USA. .,Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA.
| | - Sandra S Burkett
- Comparative Molecular Cytogenetics Core, Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD, USA.
| | - Burton L Eisenberg
- Department of Surgery, Dartmouth, Hanover, NH, USA. .,Norris Cotton Cancer Center, Lebanon, NH, USA. .,Geisel School of Medicine, Dartmouth, Hanover, NH, USA. .,Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA.
| | - Wendy A Wells
- Department of Pathology, Dartmouth, Hanover, NH, USA. .,Norris Cotton Cancer Center, Lebanon, NH, USA. .,Geisel School of Medicine, Dartmouth, Hanover, NH, USA. .,Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA.
| | - Paul J Hoopes
- Department of Surgery, Dartmouth, Hanover, NH, USA. .,Norris Cotton Cancer Center, Lebanon, NH, USA. .,Geisel School of Medicine, Dartmouth, Hanover, NH, USA. .,Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA. .,Thayer School of Engineering, Dartmouth College, Hanover, NH, USA.
| | | | - Alexander J Lazar
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,Sarcoma Research Center, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Keila E Torres
- Sarcoma Research Center, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Vincent Memoli
- Department of Pathology, Dartmouth, Hanover, NH, USA. .,Norris Cotton Cancer Center, Lebanon, NH, USA. .,Geisel School of Medicine, Dartmouth, Hanover, NH, USA. .,Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA.
| | - Sarah J Freemantle
- Department of Pharmacology and Toxicology, Dartmouth, Hanover, NH, USA. .,Geisel School of Medicine, Dartmouth, Hanover, NH, USA. .,Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA.
| | - Ethan Dmitrovsky
- Department of Pharmacology and Toxicology, Dartmouth, Hanover, NH, USA. .,Department of Medicine, Dartmouth, Hanover, NH, USA. .,Norris Cotton Cancer Center, Lebanon, NH, USA. .,Geisel School of Medicine, Dartmouth, Hanover, NH, USA. .,Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA. .,Present address: MD Anderson Cancer Center, Houston, TX, 77030-4009, USA.
| |
Collapse
|
46
|
Agaram NP, Zhang L, LeLoarer F, Silk T, Sung YS, Scott SN, Kuk D, Qin LX, Berger MF, Antonescu CR, Singer S. Targeted exome sequencing profiles genetic alterations in leiomyosarcoma. Genes Chromosomes Cancer 2015; 55:124-30. [PMID: 26541895 DOI: 10.1002/gcc.22318] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 09/16/2015] [Accepted: 09/16/2015] [Indexed: 12/29/2022] Open
Abstract
Leiomyosarcoma (LMS) belongs to the class of genetically complex sarcomas and shows numerous, often non-recurrent chromosomal imbalances and aberrations. We investigated a group of LMS using NGS platform to identify recurrent genetic abnormalities and possible therapeutic targets. Targeted exome sequencing of 230 cancer-associated genes was performed on 35 primary soft tissue and visceral (extra-uterine) LMS. Sequence data were analyzed to identify single nucleotide variants, small insertions/deletions (indels), and copy number alterations. Key alterations were further investigated using FISH assay. The study group included patients with median age of 64 years and median tumor size of 7 cm. The primary sites included retroperitoneal/intra-abdominal, extremity, truncal, and visceral. Thirty-one tumors were high grade LMS, while four were low grade. Losses of chromosomal regions involving key tumor suppressor genes PTEN (10q), RB1 (13q), CDH1 (16q), and TP53 (17p) were the most frequent genetic events. Gains mainly involved chromosome regions 17p11.2 (MYOCD) and 15q25-26 (IGF1R). The most frequent mutations were identified in the TP53 gene in 13 of 35 (37%) cases. FISH analysis showed amplification of the myocardin (MYOCD) gene in 5 of 25 (20%) cases analyzed. None of the four low grade LMS showed losses or mutations of PTEN or TP53 genes. Genetic complexity is the hallmark of LMS with losses of important tumor suppressor genes being a common feature. MYOCD, a key gene associated with smooth muscle differentiation, is amplified in a subset of both retroperitoneal and extremity LMS. Further studies are necessary to investigate the significance of gains/amplifications in the development of these tumors.
Collapse
Affiliation(s)
- Narasimhan P Agaram
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Lei Zhang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Francois LeLoarer
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Tarik Silk
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Yun-Shao Sung
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sasinya N Scott
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Deborah Kuk
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Li-Xuan Qin
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Michael F Berger
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY.,Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Samuel Singer
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
47
|
Harwood JL, Alexander JH, Mayerson JL, Scharschmidt TJ. Targeted Chemotherapy in Bone and Soft-Tissue Sarcoma. Orthop Clin North Am 2015; 46:587-608. [PMID: 26410647 DOI: 10.1016/j.ocl.2015.06.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Historically surgical intervention has been the mainstay of therapy for bone and soft-tissue sarcomas, augmented with adjuvant radiation for local control. Although cytotoxic chemotherapy revolutionized the treatment of many sarcomas, classic treatment regimens are fraught with side effects while outcomes have plateaued. However, since the approval of imatinib in 2002, research into targeted chemotherapy has increased exponentially. With targeted therapies comes the potential for decreased side effects and more potent, personalized treatment options. This article reviews the evolution of medical knowledge regarding sarcoma, the basic science of sarcomatogenesis, and the major targets and pathways now being studied.
Collapse
Affiliation(s)
- Jared L Harwood
- Department of Orthopaedics, The Ohio State University, 725 Prior Hall, 376 West 10 Avenue, Columbus, OH 43210, USA
| | - John H Alexander
- Department of Orthopaedics, The Ohio State University, 725 Prior Hall, 376 West 10 Avenue, Columbus, OH 43210, USA
| | - Joel L Mayerson
- Department of Orthopaedics, The Ohio State University, 725 Prior Hall, 376 West 10 Avenue, Columbus, OH 43210, USA.
| | - Thomas J Scharschmidt
- Department of Orthopaedics, The Ohio State University, 725 Prior Hall, 376 West 10 Avenue, Columbus, OH 43210, USA
| |
Collapse
|
48
|
Urso L, Calabrese F, Favaretto A, Conte P, Pasello G. Critical review about MDM2 in cancer: Possible role in malignant mesothelioma and implications for treatment. Crit Rev Oncol Hematol 2015; 97:220-30. [PMID: 26358421 DOI: 10.1016/j.critrevonc.2015.08.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 07/02/2015] [Accepted: 08/18/2015] [Indexed: 02/07/2023] Open
Abstract
The tumor suppressor p53 regulates genes involved in DNA repair, metabolism, cell cycle arrest, apoptosis and senescence. p53 is mutated in about 50% of the human cancers, while in tumors with wild-type p53 gene, the protein function may be lost because of overexpression of Murine Double Minute 2 (MDM2). MDM2 targets p53 for ubiquitylation and proteasomal degradation. p53 reactivation through MDM2 inhibitors seems to be a promising strategy to sensitize p53 wild-type cancer cells to apoptosis. Moreover, additional p53-independent molecular functions of MDM2, such as neoangiogenesis promotion, have been suggested. Thus, MDM2 might be a target for anticancer treatment because of its antiapoptotic and proangiogenetic role. Malignant pleural mesothelioma (MPM) is an aggressive asbestos-related tumor where wild-type p53 might be present. The present review gives a complete landscape about the role of MDM2 in cancer pathogenesis, prognosis and treatment, with particular focus on Malignant Pleural Mesothelioma.
Collapse
Affiliation(s)
- Loredana Urso
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Italy
| | - Fiorella Calabrese
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Italy
| | - Adolfo Favaretto
- Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy
| | - PierFranco Conte
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Italy; Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy
| | - Giulia Pasello
- Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy.
| |
Collapse
|
49
|
Lee DH, Forscher C, Di Vizio D, Koeffler HP. Induction of p53-independent apoptosis by ectopic expression of HOXA5 in human liposarcomas. Sci Rep 2015. [PMID: 26219418 PMCID: PMC4518222 DOI: 10.1038/srep12580] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Dedifferentiated liposarcoma (DDLPS) is a highly malignant subtype of human liposarcoma (LPS), whose genomic profile is characterized by chromosomal amplification at 12q13-q22. miR-26a-2 is one of the most frequently amplified genes in the region, and inhibition of its downstream target genes likely contributes to LPS tumorigenesis. Our previous study of LPS predicted homeobox protein A5 (HOXA5) as a target of miR-26a-2, and here we explored further the function of HOXA5, and its relationship with miR-26a-2 in DDLPS cells. Compared to normal human adipocytes, all LPS cell lines showed significant downregulation of HOXA5 (p = 0.046), and inhibition of miR-26a-2 using anti-miR-26a-2 substantially upregulated HOXA5 expression in these LPS cells. Interestingly, overexpression of HOXA5 alone induced very strong apoptotic response of LPS cells. HOXA5-induced apoptosis was p53-independent and caspase-dependent. Surprisingly, overexpression of HOXA5 induced nuclear translocation of RELA (p65), which was not associated with the transcriptional activity of RELA. Rather, nucleolar sequestration of RELA was observed. Overall, our study demonstrated for the first time that the downregulation of HOXA5 in LPS cells, partly by overexpression of miR-26a-2 in DDLPS, confers LPS cells resistance to apoptotic death. Further studies are required to understand the relationship of HOXA5 and the NFκB pathway in LPS cells.
Collapse
Affiliation(s)
- Dhong Hyun Lee
- Division of Hematology and Oncology, Departments of Surgery,Biomedical Sciences and Pathology and Laboratory Medicine, Samuel Oschin Comprehensive Cancer Institute,Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Charles Forscher
- Division of Hematology and Oncology, Departments of Surgery,Biomedical Sciences and Pathology and Laboratory Medicine, Samuel Oschin Comprehensive Cancer Institute,Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Dolores Di Vizio
- 1] Division of Cancer Biology and Therapeutics, Departments of Surgery, Biomedical Sciences and Pathology and Laboratory Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA [2] The Urological Diseases Research Center; Boston Children's Hospital, Boston, MA, Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - H Phillip Koeffler
- 1] Division of Hematology and Oncology, Departments of Surgery,Biomedical Sciences and Pathology and Laboratory Medicine, Samuel Oschin Comprehensive Cancer Institute,Cedars-Sinai Medical Center, Los Angeles, CA, USA [2] National Cancer Institute and Cancer Science Institute, National University of Singapore, Singapore
| |
Collapse
|
50
|
Haupt S, Buckley D, Pang JMB, Panimaya J, Paul PJ, Gamell C, Takano EA, Lee YY, Hiddingh S, Rogers TM, Teunisse AFAS, Herold MJ, Marine JC, Fox SB, Jochemsen A, Haupt Y. Targeting Mdmx to treat breast cancers with wild-type p53. Cell Death Dis 2015; 6:e1821. [PMID: 26181202 PMCID: PMC4650725 DOI: 10.1038/cddis.2015.173] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 05/15/2015] [Accepted: 05/22/2015] [Indexed: 01/07/2023]
Abstract
The function of the tumor suppressor p53 is universally compromised in cancers. It is the most frequently mutated gene in human cancers (reviewed). In cases where p53 is not mutated, alternative regulatory pathways inactivate its tumor suppressive functions. This is primarily achieved through elevation in the expression of the key inhibitors of p53: Mdm2 or Mdmx (also called Mdm4) (reviewed). In breast cancer (BrCa), the frequency of p53 mutations varies markedly between the different subtypes, with basal-like BrCas bearing a high frequency of p53 mutations, whereas luminal BrCas generally express wild-type (wt) p53. Here we show that Mdmx is unexpectedly highly expressed in normal breast epithelial cells and its expression is further elevated in most luminal BrCas, whereas p53 expression is generally low, consistent with wt p53 status. Inducible knockdown (KD) of Mdmx in luminal BrCa MCF-7 cells impedes the growth of these cells in culture, in a p53-dependent manner. Importantly, KD of Mdmx in orthotopic xenograft transplants resulted in growth inhibition associated with prolonged survival, both in a preventative model and also in a treatment model. Growth impediment in response to Mdmx KD was associated with cellular senescence. The growth inhibitory capacity of Mdmx KD was recapitulated in an additional luminal BrCa cell line MPE600, which expresses wt p53. Further, the growth inhibitory capacity of Mdmx KD was also demonstrated in the wt p53 basal-like cell line SKBR7 line. These results identify Mdmx growth dependency in wt p53 expressing BrCas, across a range of subtypes. Based on our findings, we propose that Mdmx targeting is an attractive strategy for treating BrCas harboring wt p53.
Collapse
Affiliation(s)
- S Haupt
- Tumor Suppression Laboratory, Research Division, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - D Buckley
- Tumor Suppression Laboratory, Research Division, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - J-M B Pang
- Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - J Panimaya
- Tumor Suppression Laboratory, Research Division, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - P J Paul
- Tumor Suppression Laboratory, Research Division, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - C Gamell
- Tumor Suppression Laboratory, Research Division, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - E A Takano
- Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - Y Ying Lee
- Tumor Suppression Laboratory, Research Division, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - S Hiddingh
- Tumor Suppression Laboratory, Research Division, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - T-M Rogers
- Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - A F A S Teunisse
- Department of Molecular Cell Biology, University Medical Centre, Leiden, The Netherlands
| | - M J Herold
- 1] Department of Molecular Genetics of Cancer, The Walter and Eliza Hall Institute, Parkville, Victoria, Australia [2] Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - J-C Marine
- Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - S B Fox
- 1] Department of Pathology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia [2] Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - A Jochemsen
- Department of Molecular Cell Biology, University Medical Centre, Leiden, The Netherlands
| | - Y Haupt
- 1] Tumor Suppression Laboratory, Research Division, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia [2] Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia [3] Department of Pathology, University of Melbourne, Parkville, Victoria, Australia [4] Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|