1
|
Wei L, Li Y, Chen J, Wang Y, Wu J, Yang H, Zhang Y. Alternative splicing in ovarian cancer. Cell Commun Signal 2024; 22:507. [PMID: 39425166 PMCID: PMC11488268 DOI: 10.1186/s12964-024-01880-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 10/06/2024] [Indexed: 10/21/2024] Open
Abstract
Ovarian cancer is the second leading cause of gynecologic cancer death worldwide, with only 20% of cases detected early due to its elusive nature, limiting successful treatment. Most deaths occur from the disease progressing to advanced stages. Despite advances in chemo- and immunotherapy, the 5-year survival remains below 50% due to high recurrence and chemoresistance. Therefore, leveraging new research perspectives to understand molecular signatures and identify novel therapeutic targets is crucial for improving the clinical outcomes of ovarian cancer. Alternative splicing, a fundamental mechanism of post-transcriptional gene regulation, significantly contributes to heightened genomic complexity and protein diversity. Increased awareness has emerged about the multifaceted roles of alternative splicing in ovarian cancer, including cell proliferation, metastasis, apoptosis, immune evasion, and chemoresistance. We begin with an overview of altered splicing machinery, highlighting increased expression of spliceosome components and associated splicing factors like BUD31, SF3B4, and CTNNBL1, and their relationships to ovarian cancer. Next, we summarize the impact of specific variants of CD44, ECM1, and KAI1 on tumorigenesis and drug resistance through diverse mechanisms. Recent genomic and bioinformatics advances have enhanced our understanding. By incorporating data from The Cancer Genome Atlas RNA-seq, along with clinical information, a series of prognostic models have been developed, which provided deeper insights into how the splicing influences prognosis, overall survival, the immune microenvironment, and drug sensitivity and resistance in ovarian cancer patients. Notably, novel splicing events, such as PIGV|1299|AP and FLT3LG|50,941|AP, have been identified in multiple prognostic models and are associated with poorer and improved prognosis, respectively. These novel splicing variants warrant further functional characterization to unlock the underlying molecular mechanisms. Additionally, experimental evidence has underscored the potential therapeutic utility of targeting alternative splicing events, exemplified by the observation that knockdown of splicing factor BUD31 or antisense oligonucleotide-induced BCL2L12 exon skipping promotes apoptosis of ovarian cancer cells. In clinical settings, bevacizumab, a humanized monoclonal antibody that specifically targets the VEGF-A isoform, has demonstrated beneficial effects in the treatment of patients with advanced epithelial ovarian cancer. In conclusion, this review constitutes the first comprehensive and detailed exposition of the intricate interplay between alternative splicing and ovarian cancer, underscoring the significance of alternative splicing events as pivotal determinants in cancer biology and as promising avenues for future diagnostic and therapeutic intervention.
Collapse
Affiliation(s)
- Liwei Wei
- Medical School, Faculty of Medicine, Tianjin University, Tianjin, 300072, China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310030, China
| | - Yisheng Li
- College of Pharmacy, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310030, China
| | - Jiawang Chen
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang, 325101, China
| | - Yuanmei Wang
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310030, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jianmin Wu
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310030, China
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Huanming Yang
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310030, China.
| | - Yi Zhang
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310030, China.
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| |
Collapse
|
2
|
Ni M, Zhou J, Gong W, Jiang R, Li X, Dai W, Yin Z, Chen Z, Zheng Z, Zhu J. Proteomic analysis reveals CAAP1 negatively correlates with platinum resistance in ovarian cancer. J Proteomics 2023; 277:104864. [PMID: 36870674 DOI: 10.1016/j.jprot.2023.104864] [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: 01/09/2023] [Revised: 02/21/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023]
Abstract
The present study sought to investigate the correlation between CAAP1 and platinum resistance in ovarian cancer and to preliminarily explore the potential biological function of CAAP1. Proteomic analysis was used to analyze differentially expressed proteins in platinum-sensitive and -resistant tissue samples of ovarian cancer. The Kaplan-Meier plotter was used for prognostic analysis. Immunohistochemistry assay and chi-square test were employed to explore the relationship between CAAP1 and platinum resistance in tissue samples. Lentivirus transfection, immunoprecipitation-mass spectrometry, and bioinformatics analysis were used to determine the potential biological function of CAAP1. Based on results, the expression level of CAAP1 was significantly higher in platinum-sensitive tissues compared to that in resistant tissues. Chi-square test demonstrated that there is a negative correlation between high expression of CAAP1 and platinum resistance. Overexpression of CAAP1 increased cis‑platinum sensitivity of the A2780/DDP cell line likely via the mRNA splicing pathway by interacting with the splicing factor AKAP17A. In summary, there is a negative correlation between high expression of CAAP1 and platinum resistance. CAAP1 might be a potential biomarker for platinum resistance in ovarian cancer. SIGNIFICANCE: Platinum resistance is a key factor affecting the survival of ovarian cancer patients. Understanding the mechanisms of platinum resistance is highly important for ovarian cancer management. Here, we performed the DIA- and DDA-based proteomics to analyze differentially expressed proteins in tissue and cell samples of ovarian cancer. We found that the protein identified as CAAP1, which was first reported to be involved in the regulation of apoptosis, may be negatively correlates with platinum resistance in ovarian cancer. In addition, we also found that CAAP1 enhanced the sensitivity of platinum-resistant cells to cis‑platinum via the mRNA splicing pathway by interacting with the splicing factor AKAP17A. Our data would be useful to reveal novel molecular mechanisms of platinum resistance in ovarian cancer.
Collapse
Affiliation(s)
- Maowei Ni
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Jie Zhou
- Center for Medicinal Resources Research, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Wangang Gong
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Ruibin Jiang
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Xia Li
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Wumin Dai
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Zhuomin Yin
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Zhongbo Chen
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Zhiguo Zheng
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| | - Jianqing Zhu
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| |
Collapse
|
3
|
Hirano H, Abe Y, Nojima Y, Aoki M, Shoji H, Isoyama J, Honda K, Boku N, Mizuguchi K, Tomonaga T, Adachi J. Temporal dynamics from phosphoproteomics using endoscopic biopsy specimens provides new therapeutic targets in stage IV gastric cancer. Sci Rep 2022; 12:4419. [PMID: 35338158 PMCID: PMC8956597 DOI: 10.1038/s41598-022-08430-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 03/08/2022] [Indexed: 11/09/2022] Open
Abstract
Phosphoproteomic analysis expands our understanding of cancer biology. However, the feasibility of phosphoproteomic analysis using endoscopically collected tumor samples, especially with regards to dynamic changes upon drug treatment, remains unknown in stage IV gastric cancer. Here, we conducted a phosphoproteomic analysis using paired endoscopic biopsy specimens of pre- and post-treatment tumors (Ts) and non-tumor adjacent tissues (NATs) obtained from 4 HER2-positive gastric cancer patients who received trastuzumab-based treatment and from pre-treatment Ts and NATs of 4 HER2-negative gastric cancer patients. Our analysis identified 14,622 class 1 phosphosites with 12,749 quantified phosphosites and revealed molecular changes by HER2 positivity and treatment. An inhibitory signature of the ErbB signaling was observed in the post-treatment HER2-positive T group compared with the pre-treatment HER2-positive T group. Phosphoproteomic profiles obtained by a case-by-case review using paired pre- and post-treatment HER2-positive T could be utilized to discover predictive or resistant biomarkers. Furthermore, these data nominated therapeutic kinase targets which were exclusively activated in the patient unresponded to the treatment. The present study suggests that a phosphoproteomic analysis of endoscopic biopsy specimens provides information on dynamic molecular changes which can individually characterize biologic features upon drug treatment and identify therapeutic targets in stage IV gastric cancer.
Collapse
Affiliation(s)
- Hidekazu Hirano
- Laboratory of Proteome Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, 567-0085, Japan.,Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, 567-0085, Japan.,Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, 104-0045, Japan.,Department of Medicine, Keio University Graduate School of Medicine, Tokyo, 160-8582, Japan
| | - Yuichi Abe
- Laboratory of Proteome Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, 567-0085, Japan.,Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, 567-0085, Japan.,Division of Molecular Diagnostics, Aichi Cancer Center Research Institute, Nagoya, 464-8681, Japan
| | - Yosui Nojima
- Laboratory of Bioinformatics, Artificial Intelligence Center for Health and Biomedical Research (ArCHER), National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, 567-0085, Japan.,Center for Mathematical Modeling and Data Science, Osaka University, Osaka, 560-8531, Japan
| | - Masahiko Aoki
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, 104-0045, Japan.,Kyoto Innovation Center for Next Generation Clinical Trials and iPS Cell Therapy (Ki-CONNECT), Kyoto University Hospital, Kyoto, 606-8507, Japan
| | - Hirokazu Shoji
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Junko Isoyama
- Laboratory of Proteome Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, 567-0085, Japan.,Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, 567-0085, Japan
| | - Kazufumi Honda
- Department of Biomarkers for Early Detection of Cancer, National Cancer Center Research Institute, Tokyo, 104-0045, Japan.,Department of Bioregulation, Nippon Medical School, Bunkyo-ku, Tokyo, 113-8602, Japan
| | - Narikazu Boku
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, 104-0045, Japan.,Department of Medical Oncology and General Medicine, IMSUT Hospital, Institute of Medical Science, University of Tokyo, Tokyo, 108-8639, Japan
| | - Kenji Mizuguchi
- Laboratory of Bioinformatics, Artificial Intelligence Center for Health and Biomedical Research (ArCHER), National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, 567-0085, Japan.,Institute for Protein Research, Osaka University, Osaka, 565-0871, Japan
| | - Takeshi Tomonaga
- Laboratory of Proteome Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, 567-0085, Japan. .,Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, 567-0085, Japan.
| | - Jun Adachi
- Laboratory of Proteome Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, 567-0085, Japan. .,Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, 567-0085, Japan. .,Laboratory of Clinical and Analytical Chemistry, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, 567-0085, Japan.
| |
Collapse
|
4
|
Yao S, Yuan C, Shi Y, Qi Y, Sridha R, Dai M, Cai H. Alternative Splicing: A New Therapeutic Target for Ovarian Cancer. Technol Cancer Res Treat 2022; 21:15330338211067911. [PMID: 35343831 PMCID: PMC8966091 DOI: 10.1177/15330338211067911] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background: Increasing evidences have shown that abnormal alternative splicing (AS) events are closely related to the prognosis of various tumors. However, the role of AS in ovarian cancer (OV) is poorly understood. This study aims to explore the correlation between AS and the prognosis of OV and establish a prognostic model for OV. Methods: We downloaded the RNA-seq data of OV from The Cancer Genome Atlas databases and assessed cancer-specific AS through the SpliceSeq software. Then systemically investigated the overall survival (OS)-related AS and splicing factors (SFs) by bioinformatics analysis. The nomogram was established based on the clinical information, and the clinical practicability of the nomogram was verified through the calibration curve. Finally, a splicing correlation network was constructed to reveal the relationship between OS-related AS and SFs. Results: A total of 48,049 AS events were detected from 10,582 genes, of which 1523 were significantly associated with OS. The area under the curve of the final prediction model was 0.785, 0.681, and 0.781 in 1, 3, and 5 years, respectively. Moreover, the nomogram showed high calibration and discrimination in OV patients. Spearman correlation analysis was used to determine 8 SFs significantly related to survival, including major facilitator superfamily domain containing 11, synaptotagmin binding cytoplasmic RNA interacting protein, DEAH-box helicase 35, CWC15, integrator complex subunit 1, LUC7 like 2, cell cycle and apoptosis regulator 1, and heterogeneous nuclear ribonucleoprotein A2/B1. Conclusion: This study provides a prognostic model related to AS in OV, and constructs an AS-clinicopathological nomogram, which provides the possibility to predict the long-term prognosis of OV patients. We have explored the wealth of RNA splicing networks and regulation patterns related to the prognosis of OV, which provides a large number of biomarkers and potential targets for the treatment of OV. Put forward the potential possibility of interfering with the AS of OV in the comprehensive treatment of OV.
Collapse
Affiliation(s)
- Shijie Yao
- 89674Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| | - Cheng Yuan
- 89674Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| | - Yuying Shi
- 89674Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| | - Yuwen Qi
- 89674Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| | - Radhakrishnan Sridha
- Cancer Science Institute of Singapore, 37580National University of Singapore, Singapore, Singapore
| | - Mengyuan Dai
- 89674Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| | - Hongbing Cai
- 89674Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.,Hubei Cancer Clinical Study Center, Wuhan, China
| |
Collapse
|
5
|
Sabnis RW. Novel PRMT5 Inhibitors for Treating Cancer. ACS Med Chem Lett 2021; 12:1537-1538. [PMID: 34676035 DOI: 10.1021/acsmedchemlett.1c00512] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Indexed: 12/19/2022] Open
Affiliation(s)
- Ram W. Sabnis
- Smith, Gambrell & Russell LLP, 1230 Peachtree Street NE, Suite 3100, Atlanta, Georgia 30309, United States
| |
Collapse
|
6
|
Califano D, Gallo D, Rampioni Vinciguerra GL, De Cecio R, Arenare L, Signoriello S, Russo D, Ferrandina G, Citron F, Losito NS, Gargiulo P, Simeon V, Scambia G, Cecere SC, Montella M, Colombo N, Tognon G, Bignotti E, Zannoni GF, Canzonieri V, Ciucci A, Spina A, Scognamiglio G, Del Sesto M, Schettino C, Piccirillo MC, Perrone F, Chiodini P, Pignata S, Baldassarre G. Evaluation of Angiogenesis-Related Genes as Prognostic Biomarkers of Bevacizumab Treated Ovarian Cancer Patients: Results from the Phase IV MITO16A/ManGO OV-2 Translational Study. Cancers (Basel) 2021; 13:cancers13205152. [PMID: 34680301 PMCID: PMC8533892 DOI: 10.3390/cancers13205152] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 01/07/2023] Open
Abstract
Simple Summary The possibility to identify, with appropriate biomarkers, patients that might mostly benefit from any given treatment is the basis of personalized oncology. Cancer biomarkers should be properly identified and validated on a large number of patients possibly enrolled in dedicated clinical trials. Here, we report the first molecular results of the MITO16A-ManGo-OV2 phase IV trial that was specifically designed to identify prognostic biomarkers of survival in epithelial ovarian cancer patients treated in first line with carboplatin-paclitaxel plus Bevacizumab (NCT01706120), a treatment for which validated predictive or prognostic biomarkers are still lacking. With this work we propose not only novel possible biomarkers for Bevacizumab-treated patients but also a way through which they can be properly collected, analyzed and statistically evaluated in the frame of large multicenter clinical trials. Abstract Background. Epithelial ovarian cancer (EOC) is a rare, highly lethal disease. In a subset of high grade EOC patients, maintenance therapy with the antiangiogenic drug Bevacizumab (BEV) is a valuable option. To date, no validated predictive or prognostic biomarkers exist for selecting EOC patients that might benefit from BEV treatment. Methods. Immunohistochemistry and RT-qPCR evaluated the expression of seven angiogenesis-related proteins and of a twelve microRNAs angio-signature in EOC patients, treated in first line with chemotherapy plus BEV (MITO16A/ManGO OV-2 phase IV trial). Centralized statistical analyses assessed the associations between each biomarker, clinical prognostic factors and survival outcomes. Results. High miR-484 expression was associated with longer progression-free and overall survival. Notably, the combined expression of miR-484 and its target VEGFB identified a subset of patients that might mostly benefit from BEV treatment. No other significant correlations were found between the other analyzed biomarkers and patients’ survival. The application of a shrinkage procedure to adjust for over-fitting hazard ratio estimates reduced the association significance. Conclusions. The analysis of angiogenesis related biomarkers in EOC patients homogenously treated with BEV in first line provides novel insight in their prognostic value and suggests that some of them might merit to be tested as predictive markers of drug activity in dedicated randomized trials.
Collapse
Affiliation(s)
- Daniela Califano
- Microenvironment Molecular Targets Unit, Istituto Nazionale Tumori IRCCS, Fondazione G. Pascale, 80131 Napoli, Italy; (D.C.); (D.R.); (A.S.)
| | - Daniela Gallo
- Department of Woman and Child Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (D.G.); (G.F.); (G.S.); (G.F.Z.); (A.C.)
| | - Gian Luca Rampioni Vinciguerra
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, 33081 Aviano, Italy; (G.L.R.V.); (F.C.)
| | - Rossella De Cecio
- Pathology Unit, Istituto Nazionale Tumori IRCCS, Fondazione G. Pascale, 80131 Napoli, Italy; (R.D.C.); (N.S.L.); (G.S.); (M.D.S.)
| | - Laura Arenare
- Clinical Trials Unit, Istituto Nazionale Tumori IRCCS, Fondazione G. Pascale, 80131 Napoli, Italy; (L.A.); (P.G.); (C.S.); (M.C.P.); (F.P.)
| | - Simona Signoriello
- Department of Mental Health and Public Medicine, Section of Statistics, Università degli Studi della Campania Luigi Vanvitelli, 80138 Napoli, Italy; (S.S.); (V.S.); (P.C.)
| | - Daniela Russo
- Microenvironment Molecular Targets Unit, Istituto Nazionale Tumori IRCCS, Fondazione G. Pascale, 80131 Napoli, Italy; (D.C.); (D.R.); (A.S.)
| | - Gabriella Ferrandina
- Department of Woman and Child Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (D.G.); (G.F.); (G.S.); (G.F.Z.); (A.C.)
- Istituto di Ginecologia e Ostetricia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Francesca Citron
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, 33081 Aviano, Italy; (G.L.R.V.); (F.C.)
| | - Nunzia Simona Losito
- Pathology Unit, Istituto Nazionale Tumori IRCCS, Fondazione G. Pascale, 80131 Napoli, Italy; (R.D.C.); (N.S.L.); (G.S.); (M.D.S.)
| | - Piera Gargiulo
- Clinical Trials Unit, Istituto Nazionale Tumori IRCCS, Fondazione G. Pascale, 80131 Napoli, Italy; (L.A.); (P.G.); (C.S.); (M.C.P.); (F.P.)
| | - Vittorio Simeon
- Department of Mental Health and Public Medicine, Section of Statistics, Università degli Studi della Campania Luigi Vanvitelli, 80138 Napoli, Italy; (S.S.); (V.S.); (P.C.)
| | - Giovanni Scambia
- Department of Woman and Child Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (D.G.); (G.F.); (G.S.); (G.F.Z.); (A.C.)
- Istituto di Ginecologia e Ostetricia, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Sabrina Chiara Cecere
- Urogynaecological Medical Oncology, Istituto Nazionale Tumori IRCCS, Fondazione G. Pascale, 80131 Napoli, Italy; (S.C.C.); (S.P.)
| | - Marco Montella
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, Università degli Studi della Campania Luigi Vanvitelli, 80138 Naples, Italy;
| | - Nicoletta Colombo
- Gynecologic Cancer Program, Università degli Studi di Milano, 20126 Bicocca, Italy;
| | - Germana Tognon
- Division of Obstetrics and Gynecology, ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (G.T.); (E.B.)
| | - Eliana Bignotti
- Division of Obstetrics and Gynecology, ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (G.T.); (E.B.)
| | - Gian Franco Zannoni
- Department of Woman and Child Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (D.G.); (G.F.); (G.S.); (G.F.Z.); (A.C.)
| | - Vincenzo Canzonieri
- Pathology Unit, IRCCS CRO Aviano, National Cancer Institute, 33081 Aviano, Italy;
| | - Alessandra Ciucci
- Department of Woman and Child Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; (D.G.); (G.F.); (G.S.); (G.F.Z.); (A.C.)
| | - Anna Spina
- Microenvironment Molecular Targets Unit, Istituto Nazionale Tumori IRCCS, Fondazione G. Pascale, 80131 Napoli, Italy; (D.C.); (D.R.); (A.S.)
| | - Giosuè Scognamiglio
- Pathology Unit, Istituto Nazionale Tumori IRCCS, Fondazione G. Pascale, 80131 Napoli, Italy; (R.D.C.); (N.S.L.); (G.S.); (M.D.S.)
| | - Michele Del Sesto
- Pathology Unit, Istituto Nazionale Tumori IRCCS, Fondazione G. Pascale, 80131 Napoli, Italy; (R.D.C.); (N.S.L.); (G.S.); (M.D.S.)
| | - Clorinda Schettino
- Clinical Trials Unit, Istituto Nazionale Tumori IRCCS, Fondazione G. Pascale, 80131 Napoli, Italy; (L.A.); (P.G.); (C.S.); (M.C.P.); (F.P.)
| | - Maria Carmela Piccirillo
- Clinical Trials Unit, Istituto Nazionale Tumori IRCCS, Fondazione G. Pascale, 80131 Napoli, Italy; (L.A.); (P.G.); (C.S.); (M.C.P.); (F.P.)
| | - Francesco Perrone
- Clinical Trials Unit, Istituto Nazionale Tumori IRCCS, Fondazione G. Pascale, 80131 Napoli, Italy; (L.A.); (P.G.); (C.S.); (M.C.P.); (F.P.)
| | - Paolo Chiodini
- Department of Mental Health and Public Medicine, Section of Statistics, Università degli Studi della Campania Luigi Vanvitelli, 80138 Napoli, Italy; (S.S.); (V.S.); (P.C.)
| | - Sandro Pignata
- Urogynaecological Medical Oncology, Istituto Nazionale Tumori IRCCS, Fondazione G. Pascale, 80131 Napoli, Italy; (S.C.C.); (S.P.)
| | - Gustavo Baldassarre
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, National Cancer Institute, 33081 Aviano, Italy; (G.L.R.V.); (F.C.)
- Correspondence: ; Tel.: +39-0434-659-759
| |
Collapse
|