1
|
Hsieh FS, Nguyen DPM, Heltberg MS, Wu CC, Lee YC, Jensen MH, Chen SH. Plausible, robust biological oscillations through allelic buffering. Cell Syst 2024; 15:1018-1032.e12. [PMID: 39504970 DOI: 10.1016/j.cels.2024.10.002] [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: 12/30/2023] [Revised: 08/12/2024] [Accepted: 10/10/2024] [Indexed: 11/08/2024]
Abstract
Biological oscillators can specify time- and dose-dependent functions via dedicated control of their oscillatory dynamics. However, how biological oscillators, which recurrently activate noisy biochemical processes, achieve robust oscillations remains unclear. Here, we characterize the long-term oscillations of p53 and its negative feedback regulator Mdm2 in single cells after DNA damage. Whereas p53 oscillates regularly, Mdm2 from a single MDM2 allele exhibits random unresponsiveness to ∼9% of p53 pulses. Using allelic-specific imaging of MDM2 activity, we show that MDM2 alleles buffer each other to maintain p53 pulse amplitude. Removal of MDM2 allelic buffering cripples the robustness of p53 amplitude, thereby elevating p21 levels and cell-cycle arrest. In silico simulations support that allelic buffering enhances the robustness of biological oscillators and broadens their plausible biochemical space. Our findings show how allelic buffering ensures robust p53 oscillations, highlighting the potential importance of allelic buffering for the emergence of robust biological oscillators during evolution. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Feng-Shu Hsieh
- Lab for Cell Dynamics, Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan
| | - Duy P M Nguyen
- Lab for Cell Dynamics, Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan
| | - Mathias S Heltberg
- Niels Bohr Institute, University of Copenhagen, Copenhagen 2100, Denmark
| | - Chia-Chou Wu
- Lab for Cell Dynamics, Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan; National Center for Theoretical Sciences, Physics Division, Complex Systems, Taipei 10617, Taiwan
| | - Yi-Chen Lee
- Lab for Cell Dynamics, Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan
| | - Mogens H Jensen
- Niels Bohr Institute, University of Copenhagen, Copenhagen 2100, Denmark
| | - Sheng-Hong Chen
- Lab for Cell Dynamics, Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan; National Center for Theoretical Sciences, Physics Division, Complex Systems, Taipei 10617, Taiwan.
| |
Collapse
|
2
|
Abdullazade S, Behrens HM, Krüger S, Haag J, Röcken C. MDM2 amplification is rare in gastric cancer. Virchows Arch 2023; 483:795-807. [PMID: 37821635 DOI: 10.1007/s00428-023-03674-8] [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: 07/19/2023] [Revised: 09/21/2023] [Accepted: 10/02/2023] [Indexed: 10/13/2023]
Abstract
The MDM2 proto-oncogene (MDM2) is a primary negative regulator of p53. The latter is frequently mutated in gastric cancer (GC). In the present study, we aimed to validate gene amplification, protein expression, and the putative tumor biological function of MDM2 in a well-characterized Western GC cohort. MDM2 amplification and protein expression were studied in a cohort of 327 GCs by fluorescence in situ hybridization (FISH) and immunohistochemistry. Gene amplification and protein expression were correlated with diverse clinicopathological patient characteristics including patient outcome. Immunohistochemically, 97 GCs (29.7%) were categorized as MDM2 positive and 230 GCs (70.3%) as negative. An amplification of MDM2 was found in 11 (3.4%) cases without evidence of intratumoral heterogeneity. Nine of these eleven (81.8%) cases showed MDM2 protein expression. MDM2 amplification correlated significantly with MDM2 protein expression (p < 0.001). On a case-by-case analysis, MDM2-amplified cases showed varied histological phenotypes and were most commonly microsatellite stable; EBV, HER2, and MET negative; and FGFR2 positive. A single case harbored both, MDM2 amplification and TP53 mutation. MDM2 amplification and MDM2 expression, respectively, did not correlate with overall or tumor-specific survival. Our targeted analysis of MDM2 in a well-characterized cohort of GC patients showed that MDM2 amplification is rare, of no specific histological phenotype, and may not be always mutually exclusive with TP53 mutations. Given the low number of cases, currently, no diagnostic or therapeutic recommendation related to MDM2 amplification can be given for GC of Western origin.
Collapse
Affiliation(s)
- Samir Abdullazade
- Dept. of Pathology, Christian-Albrechts-University, University Hospital Schleswig-Holstein, Arnold-Heller-Str. 3, Haus U33, D-24105, Kiel, Germany
| | - Hans-Michael Behrens
- Dept. of Pathology, Christian-Albrechts-University, University Hospital Schleswig-Holstein, Arnold-Heller-Str. 3, Haus U33, D-24105, Kiel, Germany
| | - Sandra Krüger
- Dept. of Pathology, Christian-Albrechts-University, University Hospital Schleswig-Holstein, Arnold-Heller-Str. 3, Haus U33, D-24105, Kiel, Germany
| | - Jochen Haag
- Dept. of Pathology, Christian-Albrechts-University, University Hospital Schleswig-Holstein, Arnold-Heller-Str. 3, Haus U33, D-24105, Kiel, Germany
| | - Christoph Röcken
- Dept. of Pathology, Christian-Albrechts-University, University Hospital Schleswig-Holstein, Arnold-Heller-Str. 3, Haus U33, D-24105, Kiel, Germany.
| |
Collapse
|
3
|
Raters VM, Gebauer F, Löser H, Schröder W, Schlösser HA, Fuchs H, Bruns C, Quaas A, Zander T. Combined score of Y chromosome loss and T-cell infiltration improves UICC based stratification of esophageal adenocarcinoma. Front Oncol 2023; 13:1249172. [PMID: 38045001 PMCID: PMC10693404 DOI: 10.3389/fonc.2023.1249172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 11/02/2023] [Indexed: 12/05/2023] Open
Abstract
Background Staging, especially clinical lymph node staging in esophageal adenocarcinoma has only moderate sensitivity and specificity. Therefore, we evaluated combined molecular markers to predict prognosis. Patients and methods 890 tumor tissue samples were obtained from patients who underwent surgery for esophageal adenocarcinoma with curative intent. These were stained by tissue micro array for 48 markers which are associated with tumorigenesis and correlated with clinical data (TNM-staging, overall survival) by multivariate Cox regression. Results Two markers (preserved Y chromosome and high grade of (CD3+) T-cell infiltration) were found to be significantly and independently associated with better overall survival. We formed a score (called CY score) from the two markers. The more markers are positive and thus the higher the score (ranging from 0 to 2), the better the overall survival, independently of UICC. Moreover, we developed a combination score of the UICC and CY score based on cluster analysis. Patients with a UICC stage of III with the presence of both traits (CY=2) can be assigned to a better prognosis group (group II), whereas patients with a UICC stage of I without both traits (CY=0) must be assigned to a worse prognosis group (group II). Therefore, patients in stage I with adverse molecular signature might benefit of multimodal therapy. Conclusion In summary, the CY score adds prognostic information to the UICC stage based on tumor biology in esophageal adenocarcinoma and warrants further evaluations in independent clinical cohorts.
Collapse
Affiliation(s)
- Verena Maria Raters
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Gastrointestinal Cancer Group Cologne GCGC, University of Cologne, Cologne, Germany
| | - Florian Gebauer
- Department of General, Visceral and Cancer Surgery, Gastrointestinal Cancer Group Cologne GCGC, University of Cologne, Cologne, Germany
| | - Heike Löser
- Institute of Pathology, Gastrointestinal Cancer Group Cologne GCGC, University of Cologne, Cologne, Germany
| | - Wolfgang Schröder
- Department of General, Visceral and Cancer Surgery, Gastrointestinal Cancer Group Cologne GCGC, University of Cologne, Cologne, Germany
| | - Hans Anton Schlösser
- Department of General, Visceral and Cancer Surgery, Gastrointestinal Cancer Group Cologne GCGC, University of Cologne, Cologne, Germany
| | - Hans Fuchs
- Department of General, Visceral and Cancer Surgery, Gastrointestinal Cancer Group Cologne GCGC, University of Cologne, Cologne, Germany
| | - Christiane Bruns
- Department of General, Visceral and Cancer Surgery, Gastrointestinal Cancer Group Cologne GCGC, University of Cologne, Cologne, Germany
| | - Alexander Quaas
- Institute of Pathology, Gastrointestinal Cancer Group Cologne GCGC, University of Cologne, Cologne, Germany
| | - Thomas Zander
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Gastrointestinal Cancer Group Cologne GCGC, University of Cologne, Cologne, Germany
| |
Collapse
|
4
|
Menon AA, Deshpande V, Suster D. MDM2 for the practicing pathologist: a primer. J Clin Pathol 2023; 76:285-290. [PMID: 36898827 DOI: 10.1136/jcp-2022-208687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2023] [Indexed: 03/12/2023]
Abstract
The mouse double minute 2 (MDM2) gene is located on the long arm of chromosome 12 and is the primary negative regulator of p53. The MDM2 gene encodes an E3 ubiquitin-protein ligase that mediates the ubiquitination of p53, leading to its degradation. MDM2 enhances tumour formation by inactivating the p53 tumour suppressor protein. The MDM2 gene also has many p53-independent functions. Alterations of MDM2 may occur through various mechanisms and contribute to the pathogenesis of many human tumours and some non-neoplastic diseases. Detection of MDM2 amplification is used in the clinical practice setting to help diagnose multiple tumour types, including lipomatous neoplasms, low-grade osteosarcomas and intimal sarcoma, among others. It is generally a marker of adverse prognosis, and MDM2-targeted therapies are currently in clinical trials. This article provides a concise overview of the MDM2 gene and discusses practical diagnostic applications pertaining to human tumour biology.
Collapse
Affiliation(s)
- Aswathy Ashok Menon
- Department of Pathology, Neuberg Anand Reference Laboratory, Bengaluru, Karnataka, India
| | - Vikram Deshpande
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - David Suster
- Department of Pathology, Rutgers University New Jersey Medical School, Newark, New Jersey, USA
| |
Collapse
|
5
|
Sinha A, Zou Y, Patel AS, Yoo S, Jiang F, Sato T, Kong R, Watanabe H, Zhu J, Massion PP, Borczuk AC, Powell CA. Early-Stage Lung Adenocarcinoma MDM2 Genomic Amplification Predicts Clinical Outcome and Response to Targeted Therapy. Cancers (Basel) 2022; 14:cancers14030708. [PMID: 35158979 PMCID: PMC8833784 DOI: 10.3390/cancers14030708] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/18/2022] [Accepted: 01/28/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Invasive subtypes of lung adenocarcinoma (LUAD) show MDM2 amplification that is associated with poor survival. Mouse double minute 2 (MDM2) is frequently amplified in lung adenocarcinoma (LUAD) and is a negative regulator of p53, which binds to p53 and regulates its activity and stability. Genomic amplification and overexpression of MDM2, together with genetic alterations in p53, leads to genomic and genetic heterogeneity in LUAD that represents a therapeutic target. In vitro assays in a panel of LUAD cell lines showed that tumor cell response to MDM2-targeted therapy is associated with MDM2 amplification. Abstract Lung cancer is the most common cause of cancer-related deaths in both men and women, accounting for one-quarter of total cancer-related mortality globally. Lung adenocarcinoma is the major subtype of non-small cell lung cancer (NSCLC) and accounts for around 40% of lung cancer cases. Lung adenocarcinoma is a highly heterogeneous disease and patients often display variable histopathological morphology, genetic alterations, and genomic aberrations. Recent advances in transcriptomic and genetic profiling of lung adenocarcinoma by investigators, including our group, has provided better stratification of this heterogeneous disease, which can facilitate devising better treatment strategies suitable for targeted patient cohorts. In a recent study we have shown gene expression profiling identified novel clustering of early stage LUAD patients and correlated with tumor invasiveness and patient survival. In this study, we focused on copy number alterations in LUAD patients. SNP array data identified amplification at chromosome 12q15 on MDM2 locus and protein overexpression in a subclass of LUAD patients with an invasive subtype of the disease. High copy number amplification and protein expression in this subclass correlated with poor overall survival. We hypothesized that MDM2 copy number and overexpression predict response to MDM2-targeted therapy. In vitro functional data on a panel of LUAD cells showed that MDM2-targeted therapy effectively suppresses cell proliferation, migration, and invasion in cells with MDM2 amplification/overexpression but not in cells without MDM2 amplification, independent of p53 status. To determine the key signaling mechanisms, we used RNA sequencing (RNA seq) to examine the response to therapy in MDM2-amplified/overexpressing p53 mutant and wild-type LUAD cells. RNA seq data shows that in MDM2-amplified/overexpression with p53 wild-type condition, the E2F → PEG10 → MMPs pathway is operative, while in p53 mutant genetic background, MDM2-targeted therapy abrogates tumor progression in LUAD cells by suppressing epithelial to mesenchymal transition (EMT) signaling. Our study provides a potentially clinically relevant strategy of selecting LUAD patients for MDM2-targeted therapy that may provide for increased response rates and, thus, better survival.
Collapse
Affiliation(s)
- Abhilasha Sinha
- Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (A.S.); (A.S.P.); (F.J.); (T.S.); (R.K.); (H.W.)
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Yong Zou
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (Y.Z.); (P.P.M.)
| | - Ayushi S. Patel
- Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (A.S.); (A.S.P.); (F.J.); (T.S.); (R.K.); (H.W.)
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
| | | | - Feng Jiang
- Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (A.S.); (A.S.P.); (F.J.); (T.S.); (R.K.); (H.W.)
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Takashi Sato
- Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (A.S.); (A.S.P.); (F.J.); (T.S.); (R.K.); (H.W.)
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
- Department of Respiratory Medicine, Kitasato University School of Medicine, Sagamihara 252-0374, Japan
| | - Ranran Kong
- Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (A.S.); (A.S.P.); (F.J.); (T.S.); (R.K.); (H.W.)
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Department of Thoracic Surgery, The Second Affiliated Hospital of Medical School, Xi’an Jiaotong University, Xi’an 710004, China
| | - Hideo Watanabe
- Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (A.S.); (A.S.P.); (F.J.); (T.S.); (R.K.); (H.W.)
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jun Zhu
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Sema4, 333 Ludlow St., Stamford, CT 06902, USA;
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Icahn Institute for Data Science and Genomic Technology, New York, NY 10029, USA
| | - Pierre P. Massion
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (Y.Z.); (P.P.M.)
| | - Alain C. Borczuk
- Department of Pathology, Weill Cornell Medicine, New York, NY 10021, USA;
| | - Charles A. Powell
- Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (A.S.); (A.S.P.); (F.J.); (T.S.); (R.K.); (H.W.)
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Correspondence: ; Tel.: +1-212-241-5656
| |
Collapse
|
6
|
Qin J, Ng CS, He P, Lin X, Lin X, Hou P. Pulmonary artery intimal sarcoma - A primeval or rediscovered tumor? A report of 14 new cases with literature review. Pathol Res Pract 2021; 224:153548. [PMID: 34280751 DOI: 10.1016/j.prp.2021.153548] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/06/2021] [Accepted: 07/08/2021] [Indexed: 11/28/2022]
Abstract
Pulmonary artery sarcomas (PAS) are rare with many being undifferentiated pleomorphic or spindle cell (UPSC) sarcomas with variable atypia. The term pulmonary artery intimal sarcoma (PAIS) was rarely coined in the early literature and sometimes used for luminal sarcomas. With the advent of immunohistochemistry and molecular genetics, many of these UPSC sarcomas were found to frequently overpress MDM2 and/or CDK4 and PDGFRA with genetic alterations in 12q12-15 and 4q12, where the MDM2, CDK4 and PDGFRA genes are located. These recent developments enabled refinement in diagnosis of PAIS. We diagnosed 14 cases of PAIS (6 males and 8 females, mean age 44 years) in 2015 - 2020 in our institution. Six were initially misdiagnosed as thromboembolism and the remaining pulmonary artery tumors. The tumors were pulmonary artery intraluminal polypoid masses with histology of spindle cell sarcomas exhibiting immunohistochemical positivity for MDM2 (100%) and CDK4 (79%) with MDM2 gene amplification (100%). Ten surgically treated patients fared better than four other biopsy only and not surgically treated patients, who died of disease within 5-11 months. PAIS needs to be differentiated from other spindle cell tumors and those exhibiting MDM2 gene amplification, especially dedifferentiated liposarcoma. The use of biopsy to provide diagnostic material poses a sampling error problem and correlation with clinical, radiologic, histologic, immunophenotypic and genotypic features are essential for accurate diagnosis and early surgical intervention of PAIS.
Collapse
Affiliation(s)
- Jilong Qin
- Department of Pathology, First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang West Road, Guangzhou 510120, China
| | - Chi Sing Ng
- Department of Pathology, St. Teresa's Hospital, 327 Prince Edward Road, Kowloon, Hong Kong, China.
| | - Ping He
- Department of Pathology, First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang West Road, Guangzhou 510120, China
| | - Xiaodong Lin
- Department of Pathology, First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang West Road, Guangzhou 510120, China
| | - Xina Lin
- Department of Pathology, First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang West Road, Guangzhou 510120, China
| | - Peng Hou
- PET-CT Center, First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang West Road, Guangzhou 510120, China
| |
Collapse
|
7
|
Ye J, Zhang L, Li Z, Lin R, Song Y, Ni H, Gou X, Xie R. High Expression of MDM2 and the p53 Protein is Predictive Biomarkers for Poor Prognosis of Oesophageal Squamous Cell Carcinoma. Cancer Manag Res 2021; 13:2733-2744. [PMID: 33790647 PMCID: PMC8005260 DOI: 10.2147/cmar.s280326] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/20/2021] [Indexed: 12/24/2022] Open
Abstract
Background and Objective In the present study, we detected the expression of MDM2 and p53 in oesophageal squamous cell carcinoma (OSCC) specimens, studied their relationship with the survival of OSCC patients, and explored the potential of MDM2 and p53 to serve as predictive OSCC tumour markers. Patients and Methods Through immunohistochemistry and fluorescence in situ hybridization (FISH), we detected the expression of MDM2 and the p53 protein in 157 OSCC specimens that met the inclusion and exclusion criteria. After scoring the results, Pearson’s chi-square test and Cox regression were used for analysis. Results The results showed that the rates of high MDM2 and p53 expression in OSCC tissues were 60.5% and 51.0%, respectively. The expression levels of MDM2 and p53 in OSCC were significantly positively correlated (p<0.001, r=0.414). In addition, the pathological metastasis (M) status and MDM2 protein expression in OSCC were significantly correlated (p=0.027), and high expression of the p53 protein was positively correlated with OSCC transfer (p=0.005), pathological node status (p=0.008), and clinical stage (p=0.003). Kaplan-Meier survival analysis showed that the high expression of MDM2 and p53 was significantly related to the poor prognosis of OSCC. Moreover, subgroup analysis of the TNM staging of OSCC patients showed that the high expression of MDM2 and p53 was significantly correlated with poor OS and DFS of OSCC patients in either stage I–II or III–IV patients. Both univariate and Cox multivariate analyses showed that p53 and MDM2 can be used as independent factors for the prognosis of OSCC patients. Finally, our FISH detection results for MDM2 showed that the high expression of MDM2 was significantly correlated with the amplification of MDM2 (p=0.015). Conclusion This study shows that MDM2 and p53 can be used as independent predictors of the prognosis of patients with oesophageal squamous cell carcinoma.
Collapse
Affiliation(s)
- Juan Ye
- Department of Head and Neck Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People's Republic of China.,Department of Experimental Research, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, People's Republic of China
| | - Lin Zhang
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, People's Republic of China.,Department of Clinical Laboratory, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, People's Republic of China.,Department of Clinical Laboratory, Yunfu People's Hospital, Yunfu, Guangdong, People's Republic of China
| | - Zhongwen Li
- Department of Head and Neck Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
| | - Runduan Lin
- Department of Clinical Laboratory, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, People's Republic of China
| | - Yiling Song
- Department of Clinical Laboratory, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, People's Republic of China
| | - Huanhe Ni
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, People's Republic of China
| | - Xiaoxia Gou
- Department of Head and Neck Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
| | - Rongzhang Xie
- Department of Clinical Laboratory, Yunfu People's Hospital, Yunfu, Guangdong, People's Republic of China
| |
Collapse
|
8
|
Jiang D, Chen L, Huang J, Wang H, Song Q, Shi P, Wang H, Hou Y. Mouse double minute 2 amplification in oesophageal squamous cell carcinoma is associated with better outcome. Histopathology 2020; 77:963-973. [PMID: 32652667 DOI: 10.1111/his.14208] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 07/07/2020] [Indexed: 12/29/2022]
Abstract
AIMS This study aimed to determine the frequency of mouse double minute 2 (MDM2) amplification in oesophageal squamous cell carcinomas (ESCC) and to clarify its prognostic significance. METHODS AND RESULTS We investigated MDM2 amplification on tissue microarrays using fluorescence in-situ hybridisation and analysed its correlations with clinicopathological features and outcomes in 515 Chinese ESCC patients. MDM2 amplifications were found in 37 of 515 ESCC patients (7.2%). They were significantly negatively correlated with tumour size (P = 0.045), disease progression (P = 0.002) and death (P = 0.003). Univariate analysis showed that the following clinicopathological factors were associated with disease-free survival (DFS) and overall survival (OS): differentiation (P = 0.025 for DFS and P = 0.061 for OS), vessel invasion (P = 0.001 and P = 0.002), nerve invasion (P = 0.009 and P = 0.001), clinical stage (P < 0.001 and P < 0.001) and MDM2 amplification (P = 0.012 and P = 0.014). Multivariate Cox regression analysis showed that MDM2 amplification was an independent prognostic factor for improved outcomes (P = 0.023 for DFS, P = 0.027 for OS) and the clinical stage was an independent prognostic factor for poor outcomes (P < 0.001). When survival analyses were conducted at different clinical stages, MDM2 amplification was associated with longer DFS and OS in stages I-II ESCC (P = 0.003 for DFS and P = 0.003 for OS), but there was no significant survival difference in stages III-IVa ESCC. CONCLUSIONS MDM2 amplification was significantly correlated with an improved patient outcome, especially in stage I and II disease, and was verified as an independent prognostic factor in our patients. Therefore, MDM2 amplification may be a potential biomarker for risk stratification of the lower stages of ESCC.
Collapse
Affiliation(s)
- Dongxian Jiang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lingli Chen
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jie Huang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hao Wang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qi Song
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Peng Shi
- Center for Evidence-based Medicine, Fudan University, Shanghai, China.,Center for Data Management and Statistics, Children's Hospital of Fudan University, Shanghai, China
| | - Haixing Wang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Pathology, School of Basic Medical Sciences and Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
9
|
Du G, Zhou J, Cheng L, Ma X, Gui Y, Tan B. High Expression of miR-206 Predicts Adverse Outcomes: A Potential Therapeutic Target for Esophageal Cancer. Comb Chem High Throughput Screen 2020; 22:599-611. [PMID: 31648633 DOI: 10.2174/1386207322666191018145825] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/11/2019] [Accepted: 09/23/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND MicroRNA-206 (miR-206) inhibits cell proliferation, invasion and migration in a variety of tumors, but the prognostic value of its Esophageal Cancer (EC) remains unclear. OBJECTIVE To study the role of miR-206 in EC. METHODS The datasets of RNA-Seq, miRNA-Seq, methylation, copy number variation (CNV), and clinical follow-up information were download from The Cancer Genome Atlas (TCGA). After integration and standardization, the prognostic value and potential function of miR-206 were analyzed. The important roles of miR-206 expression in EC genetic and epigenetic mechanisms were analyzed by RNA-Seq, miRNA-Seq, and methylation data. The potential mechanism of CNV in different miR-206 expression groups was analyzed using GISTIC. RESULTS High expression of miR-206 was associated with poor outcome of EC (OS: p=0.005, AUC=0.69, N=178). Transforming growth factor β (TGF-β) signaling pathway, Wnt signaling pathway, mitogen-activated protein kinases (MAPK) signaling pathway, mammalian target of rapamycin (mTOR) signaling pathway were inhibited in high expression group. the aberrant methylation sites in the high and low expression groups were mainly distributed in the promoter region containing CpG islands, and there were different copy number patterns in the H and L samples, and the genes in the differential copy number were mainly enriched in cancer-related pathways, such as thyroid cancer, central carbon metabolism. CONCLUSION This study explored the unique genomic and epigenetic landscape associated with the expression of miR-206, provided evidence of mir-206 as a prognostic biomarker or a potential therapeutic target for EC patients.
Collapse
Affiliation(s)
- Guobo Du
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong City, Sichuan 637000, China
| | - Jing Zhou
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Nanchong City, Sichuan 637000, China
| | - Long Cheng
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong City, Sichuan 637000, China
| | - Xiaojie Ma
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong City, Sichuan 637000, China
| | - Yan Gui
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong City, Sichuan 637000, China
| | - Bangxian Tan
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong City, Sichuan 637000, China
| |
Collapse
|
10
|
Roy S, Laroche-Clary A, Verbeke S, Derieppe MA, Italiano A. MDM2 Antagonists Induce a Paradoxical Activation of Erk1/2 through a P53-Dependent Mechanism in Dedifferentiated Liposarcomas: Implications for Combinatorial Strategies. Cancers (Basel) 2020; 12:cancers12082253. [PMID: 32806555 PMCID: PMC7465494 DOI: 10.3390/cancers12082253] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/03/2020] [Accepted: 08/09/2020] [Indexed: 12/13/2022] Open
Abstract
The MDM2 gene is amplified in dedifferentiated liposarcoma (DDLPS). Treatment with MDM2 antagonists is a promising strategy to treat DDLPS; however, drug resistance is a major limitation when these drugs are used as a single agent. This study examined the impact of MDM2 antagonists on the mitogen-activated protein kinase (MAPK) pathway in DDLPS and investigated the potential synergistic activity of a MAPK kinase (MEK) inhibitor in combination with MDM2 antagonists. We identified a synergistic effect and identified the mechanism behind it. Combination effects of MDM2 antagonists and a MEK inhibitor were analyzed in a patient-derived xenograft mouse model and in DDLPS and leiomyosarcoma cell lines using different cell proliferation assays and immunoblot analysis. MDM2 antagonist (RG7388)-resistant IB115 [P4] cells and p53-silenced DDLPS cells were also established to understand the importance of functional p53. We found that MDM2 antagonists induced an upregulation of phosphorylated extracellular signal-regulated kinase (p-ERK) in DDLPS cells. The upregulation of p-ERK occurred due to mitochondrial translocation of p53, which resulted in increased production of reactive oxygen species, causing the activation of receptor tyrosine kinases (RTKs). Activated RTKs led to the activation of the downstream MEK/ERK signaling pathway. Treatment with a MEK inhibitor resulted in decreased expression of p-ERK, causing significant anti-tumor synergy when combined with MDM2 antagonists. Our results provide a framework for designing clinical studies of combination therapies in DDLPS patients.
Collapse
Affiliation(s)
- Shomereeta Roy
- Sarcoma Unit, Institut Bergonié, 33000 Bordeaux, France; (S.R.); (A.L.-C.); (S.V.)
- University of Bordeaux, 33400 Talence, France
| | - Audrey Laroche-Clary
- Sarcoma Unit, Institut Bergonié, 33000 Bordeaux, France; (S.R.); (A.L.-C.); (S.V.)
- Sarcoma Unit, INSERM U1218, Institut Bergonié, 33000 Bordeaux, France
| | - Stephanie Verbeke
- Sarcoma Unit, Institut Bergonié, 33000 Bordeaux, France; (S.R.); (A.L.-C.); (S.V.)
- Sarcoma Unit, INSERM U1218, Institut Bergonié, 33000 Bordeaux, France
| | | | - Antoine Italiano
- Sarcoma Unit, Institut Bergonié, 33000 Bordeaux, France; (S.R.); (A.L.-C.); (S.V.)
- University of Bordeaux, 33400 Talence, France
- Sarcoma Unit, INSERM U1218, Institut Bergonié, 33000 Bordeaux, France
- Department of Medical Oncology, Institut Bergonié, 33000 Bordeaux, France
- Correspondence:
| |
Collapse
|
11
|
Comiskey DF, Montes M, Khurshid S, Singh RK, Chandler DS. SRSF2 Regulation of MDM2 Reveals Splicing as a Therapeutic Vulnerability of the p53 Pathway. Mol Cancer Res 2019; 18:194-203. [PMID: 31662450 DOI: 10.1158/1541-7786.mcr-19-0541] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 09/06/2019] [Accepted: 10/25/2019] [Indexed: 11/16/2022]
Abstract
MDM2 is an oncogene and critical negative regulator of tumor suppressor p53. Genotoxic stress causes alternative splicing of MDM2 transcripts, which leads to alterations in p53 activity and contributes to tumorigenesis. MDM2-ALT1 is one of the alternatively spliced transcripts predominantly produced in response to genotoxic stress, and is comprised of terminal coding exons 3 and 12. Previously, we found that SRSF1 induces MDM2-ALT1 by promoting MDM2 exon 11 skipping. Here we report that splicing regulator SRSF2 antagonizes the regulation of SRSF1 by facilitating the inclusion of exon 11 through binding at two conserved exonic splicing enhancers. Overexpression of SRSF2 reduced the generation of MDM2-ALT1 under genotoxic stress, whereas SRSF2 knockdown induced the expression of MDM2-ALT1 in the absence of genotoxic stress. Blocking the exon 11 SRSF2-binding sites using oligonucleotides promoted MDM2-ALT1 splicing and induced p53 protein expression, and apoptosis in p53 wild-type cells. The regulation of MDM2 splicing by SRSF2 is also conserved in mice, as mutation of one SRSF2-binding site in Mdm2 exon 11, using CRISPR-Cas9, increased the expression of the MDM2-ALT1 homolog Mdm2-MS2. IMPLICATIONS: Taken together, the data indicate that modulating MDM2 splicing may be a useful tool for fine-tuning p53 activity in response to genotoxic stress.
Collapse
Affiliation(s)
- Daniel F Comiskey
- Department of Pediatrics, The Ohio State University, Columbus, Ohio.,Center for Childhood Cancer, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio.,Center for RNA Biology, Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - Matías Montes
- Department of Pediatrics, The Ohio State University, Columbus, Ohio.,Center for Childhood Cancer, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio.,Center for RNA Biology, Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - Safiya Khurshid
- Department of Pediatrics, The Ohio State University, Columbus, Ohio.,Center for Childhood Cancer, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio.,Center for RNA Biology, Wexner Medical Center, The Ohio State University, Columbus, Ohio
| | - Ravi K Singh
- Department of Pediatrics, The Ohio State University, Columbus, Ohio.,Center for Childhood Cancer, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio.,Center for RNA Biology, Wexner Medical Center, The Ohio State University, Columbus, Ohio.,Department of Pathology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Dawn S Chandler
- Department of Pediatrics, The Ohio State University, Columbus, Ohio. .,Center for Childhood Cancer, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio.,Center for RNA Biology, Wexner Medical Center, The Ohio State University, Columbus, Ohio
| |
Collapse
|
12
|
Sawada R, Maehara R, Oshikiri T, Nakamura T, Itoh T, Kodama Y, Kakeji Y, Zen Y. MDM2 copy number increase: a poor prognostic, molecular event in esophageal squamous cell carcinoma. Hum Pathol 2019; 89:1-9. [PMID: 31004651 DOI: 10.1016/j.humpath.2019.04.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/07/2019] [Accepted: 04/08/2019] [Indexed: 12/09/2022]
Abstract
The present study aimed to elucidate the clinicopathological significance of molecular alterations in MDM2 in esophageal squamous cell carcinoma (ESCC). A total of 399 resected cases of ESCC were examined by dual-color in situ hybridization for MDM2 and immunohistochemistry for p53 using tissue microarrays. Clinicopathological features were correlated with the MDM2 status. Among 362 cases with a successful dual-color in situ hybridization analysis, 19 (5%) and 13 (4%) had MDM2 amplification and chromosome 12 polysomy, respectively, and these were examined as an MDM2-positive group. A comparison between amplified and polysomic cases revealed that the latter were more strongly associated with preoperative chemotherapy than the former. Sixteen (50%) of 32 MDM2-positive cases had positive results in all tissue cores examined, indicating diffuse MDM2 alterations. Cases with the diffuse alteration of MDM2 were characterized by an advanced pT stage and extensive vascular infiltration. The relationship between MDM2 copy number increases and p53 mutations was weak, with the overexpression of p53 being similarly detected in MDM2-positive and MDM2-negative cases (59% versus 49%; P = .267). Overall survival was shorter in patients with MDM2-positive ESCC than in those without MDM2 alterations (P = .033). The poor prognostic value of MDM2 alterations became more obvious when only diffusely altered cases were counted (P = .005). In conclusion, the present study revealed that MDM2 copy number increases occurred in 9% of ESCC cases, and MDM2 alterations, particularly diffuse abnormalities, were associated with a poor prognosis. MDM2-altered ESCC may achieve beneficial effects from MDM2-targeted therapy.
Collapse
Affiliation(s)
- Ryuichiro Sawada
- Department of Diagnostic Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; Department of Gastrointestinal Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
| | - Ritsuko Maehara
- Department of Gastrointestinal Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
| | - Taro Oshikiri
- Department of Gastrointestinal Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
| | - Tetsu Nakamura
- Department of Gastrointestinal Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
| | - Tomoo Itoh
- Department of Diagnostic Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
| | - Yuzo Kodama
- Department of Gastroenterology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
| | - Yoshihiro Kakeji
- Department of Gastrointestinal Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan.
| | - Yoh Zen
- Department of Diagnostic Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; Institute of Liver Studies, King's College Hospital & King's College London, London SE5 9RS, United Kingdom.
| |
Collapse
|
13
|
Dembla V, Somaiah N, Barata P, Hess K, Fu S, Janku F, Karp DD, Naing A, Piha-Paul SA, Subbiah V, Tsimberidou AM, Shaw K, Meric-Bernstam F, Hong DS. Prevalence of MDM2 amplification and coalterations in 523 advanced cancer patients in the MD Anderson phase 1 clinic. Oncotarget 2018; 9:33232-33243. [PMID: 30237864 PMCID: PMC6145698 DOI: 10.18632/oncotarget.26075] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 08/20/2018] [Indexed: 01/03/2023] Open
Abstract
Background TP53 is the most commonly mutated gene in cancer and codes for the best studied tumor suppressor, p53. MDM2 is involved in the negative regulation of p53 and itself serves as an oncogene, reported to be overexpressed in several cancer tumor types. In this retrospective study, we assessed the occurrence of MDM2 amplification among patients with various types of cancers and its association with clinical factors, other genetic aberrations, and response to targeted therapy in a phase I clinical trial setting. Methods Samples from patients with advanced solid tumors who had been referred to the MD Anderson phase I clinical trials program between January 2011 and January 2016 were collected and analyzed for MDM2 amplification using FoundationOne's genomic profiling assay. Patients whose tumors expressed MDM2 amplification were compared to those with tumors of the same histologic types without MDM2 amplification. Results We tested tumors from 523 patients, of which 23 (4.4%) had MDM2 amplification. The highest prevalence of MDM2 amplification was in sarcoma (57%), breast cancer (13%) and bladder cancer (9%). Six patients with liposarcoma were treated on phase I protocol with an MDM2 inhibitor. The most common molecular aberrations co-occurring with MDM2 amplification was CDK4 amplification (70%). TP53 mutation was also detected in 7 patients (30%). Conclusion MDM2 amplification was most commonly associated with liposarcoma. Concomitant alterations in additional genes such as CDK4 amplification and TP53 mutations, along with variable responses to targeted therapies including MDM2 inhibitors, suggest that further combinational studies are needed to target this population.
Collapse
Affiliation(s)
- Vikas Dembla
- Department of Investigational Cancer Therapeutics (Phase 1 Program), The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Neeta Somaiah
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Pedro Barata
- Department of Solid Tumors, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Kenneth Hess
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Siqing Fu
- Department of Investigational Cancer Therapeutics (Phase 1 Program), The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Filip Janku
- Department of Investigational Cancer Therapeutics (Phase 1 Program), The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Daniel D Karp
- Department of Investigational Cancer Therapeutics (Phase 1 Program), The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Aung Naing
- Department of Investigational Cancer Therapeutics (Phase 1 Program), The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sarina Anne Piha-Paul
- Department of Investigational Cancer Therapeutics (Phase 1 Program), The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Vivek Subbiah
- Department of Investigational Cancer Therapeutics (Phase 1 Program), The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Apostolia M Tsimberidou
- Department of Investigational Cancer Therapeutics (Phase 1 Program), The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kenna Shaw
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics (Phase 1 Program), The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - David S Hong
- Department of Investigational Cancer Therapeutics (Phase 1 Program), The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
14
|
Robinson L, Ilyas M, Ragunath K, Card T, Abradu-Berchie B, Kaye P. MDM2 expression in the progression of Barrett's oesophagus. Histopathology 2018; 72:1230-1233. [PMID: 29427358 DOI: 10.1111/his.13480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Leora Robinson
- Nottingham University Hospitals and University of Nottingham, Nottingham Digestive Diseases Biomedical Research Centre, Nottingham, UK
| | - Mohammad Ilyas
- Nottingham University Hospitals and University of Nottingham, Nottingham Digestive Diseases Biomedical Research Centre, Nottingham, UK
| | - Krish Ragunath
- Nottingham University Hospitals and University of Nottingham, Nottingham Digestive Diseases Biomedical Research Centre, Nottingham, UK
| | - Tim Card
- Nottingham University Hospitals and University of Nottingham, Nottingham Digestive Diseases Biomedical Research Centre, Nottingham, UK
| | - Berice Abradu-Berchie
- Nottingham University Hospitals and University of Nottingham, Nottingham Digestive Diseases Biomedical Research Centre, Nottingham, UK
| | - Philip Kaye
- Nottingham University Hospitals and University of Nottingham, Nottingham Digestive Diseases Biomedical Research Centre, Nottingham, UK
| |
Collapse
|
15
|
Lee KTW, Gopalan V, Lam AK. Somatic DNA Copy-Number Alterations Detection for Esophageal Adenocarcinoma Using Digital Polymerase Chain Reaction. Methods Mol Biol 2018; 1756:195-212. [PMID: 29600372 DOI: 10.1007/978-1-4939-7734-5_18] [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] [Indexed: 08/30/2023]
Abstract
Somatic copy-number alterations are commonly found in cancer and play key roles in activating oncogenes and deactivating tumor suppressor genes. Digital polymerase chain reaction is an effective way to detect the changes in copy number. In esophageal adenocarcinoma, detection of somatic copy-number alterations could predict the prognosis of patients as well as the response to therapy. This chapter will review the methods involved in digital polymerase chain reaction for the research or potential clinical applications in esophageal adenocarcinoma.
Collapse
Affiliation(s)
- Katherine T W Lee
- Cancer Molecular Pathology of School of Medicine, Griffith University, Gold Coast, Australia
| | - Vinod Gopalan
- Cancer Molecular Pathology of School of Medicine, Griffith University, Gold Coast, Australia
| | - Alfred K Lam
- Cancer Molecular Pathology of School of Medicine, Griffith University, Gold Coast, Australia.
| |
Collapse
|
16
|
Tisato V, Voltan R, Gonelli A, Secchiero P, Zauli G. MDM2/X inhibitors under clinical evaluation: perspectives for the management of hematological malignancies and pediatric cancer. J Hematol Oncol 2017; 10:133. [PMID: 28673313 PMCID: PMC5496368 DOI: 10.1186/s13045-017-0500-5] [Citation(s) in RCA: 199] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 06/20/2017] [Indexed: 02/07/2023] Open
Abstract
The two murine double minute (MDM) family members MDM2 and MDMX are at the center of an intense clinical assessment as molecular target for the management of cancer. Indeed, the two proteins act as regulators of P53, a well-known key controller of the cell cycle regulation and cell proliferation that, when altered, plays a direct role on cancer development and progression. Several evidence demonstrated that functional aberrations of P53 in tumors are in most cases the consequence of alterations on the MDM2 and MDMX regulatory proteins, in particular in patients with hematological malignancies where TP53 shows a relatively low frequency of mutation while MDM2 and MDMX are frequently found amplified/overexpressed. The pharmacological targeting of these two P53-regulators in order to restore or increase P53 expression and activity represents therefore a strategy for cancer therapy. From the discovery of the Nutlins in 2004, several compounds have been developed and reported with the ability of targeting the P53-MDM2/X axis by inhibiting MDM2 and/or MDMX. From natural compounds up to small molecules and stapled peptides, these MDM2/X pharmacological inhibitors have been extensively studied, revealing different biological features and different rate of efficacy when tested in in vitro and in vivo experimental tumor models. The data/evidence coming from the preclinical experimentation have allowed the identification of the most promising molecules and the setting of clinical studies for their evaluation as monotherapy or in therapeutic combination with conventional chemotherapy or with innovative therapeutic protocols in different tumor settings. Preliminary results have been recently published reporting data about safety, tolerability, potential side effects, and efficacy of such therapeutic approaches. In this light, the aim of this review is to give an updated overview about the state of the art of the clinical evaluation of MDM2/X inhibitor compounds with a special attention to hematological malignancies and to the potential for the management of pediatric cancers.
Collapse
Affiliation(s)
- Veronica Tisato
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Via Fossato di Mortara 66, 44121, Ferrara, Italy.
| | - Rebecca Voltan
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Via Fossato di Mortara 66, 44121, Ferrara, Italy
| | - Arianna Gonelli
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Via Fossato di Mortara 66, 44121, Ferrara, Italy
| | - Paola Secchiero
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Via Fossato di Mortara 66, 44121, Ferrara, Italy
| | - Giorgio Zauli
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Via Fossato di Mortara 66, 44121, Ferrara, Italy
| |
Collapse
|
17
|
Jia Y, Chen L, Jia Q, Dou X, Xu N, Liao DJ. The well-accepted notion that gene amplification contributes to increased expression still remains, after all these years, a reasonable but unproven assumption. J Carcinog 2016; 15:3. [PMID: 27298590 PMCID: PMC4895059 DOI: 10.4103/1477-3163.182809] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 04/25/2016] [Indexed: 02/06/2023] Open
Abstract
“Gene amplification causes overexpression” is a longstanding and well-accepted concept in cancer genetics. However, raking the whole literature, we find only statistical analyses showing a positive correlation between gene copy number and expression level, but do not find convincing experimental corroboration for this notion, for most of the amplified oncogenes in cancers. Since an association does not need to be an actual causal relation, in our opinion, this widespread notion still remains a reasonable but unproven assumption awaiting experimental verification.
Collapse
Affiliation(s)
- Yuping Jia
- Animal Facilities, Shandong Academy of Pharmaceutical Sciences, Ji'nan, Shandong 250101, USA
| | - Lichan Chen
- Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Qingwen Jia
- Animal Facilities, Shandong Academy of Pharmaceutical Sciences, Ji'nan, Shandong 250101, USA
| | - Xixi Dou
- Animal Facilities, Shandong Academy of Pharmaceutical Sciences, Ji'nan, Shandong 250101, USA
| | - Ningzhi Xu
- Laboratory of Cell and Molecular Biology, Cancer Institute, Chinese Academy of Medical Science, Beijing 100021, China
| | - Dezhong Joshua Liao
- Department of Pathology, Guizhou Medical University Hospital, Guizhou, Guiyang 550004, P.R. China
| |
Collapse
|