1
|
Lan W, Hong J, Huayun T. Advances in ovarian cancer radiomics: a bibliometric analysis from 2010 to 2024. Front Oncol 2024; 14:1456932. [PMID: 39411123 PMCID: PMC11473287 DOI: 10.3389/fonc.2024.1456932] [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: 06/29/2024] [Accepted: 09/09/2024] [Indexed: 10/19/2024] Open
Abstract
Objective Ovarian cancer, a leading cause of death among gynecological malignancies, often eludes early detection, leading to diagnoses at advanced stages. The objective of this bibliometric analysis is to map the landscape of ovarian cancer radiomics research from 2010 to 2024, emphasizing its growth, global contributions, and the impact of emerging technologies on early diagnosis and treatment strategies. Methods A comprehensive search was conducted using the Web of Science Core Collection (WoSCC), focusing on publications related to radiomics and ovarian cancer within the specified period. Analytical tools such as VOSviewer and CiteSpace were employed to visualize trends, collaborations, and key contributions, while the R programming environment offered further statistical insights. Results From the initial dataset, 149 articles were selected, showing a significant increase in research output, especially in the years 2021-2023. The analysis revealed a dominant contribution from China, with significant inputs from England. Major institutional contributors included the University of Cambridge and GE Healthcare. 'Frontiers in Oncology' emerged as a crucial journal in the field, according to Bradford's Law. Keyword analysis highlighted the focus on advanced imaging techniques and machine learning. Conclusions The steady growth in ovarian cancer radiomics research reflects its critical role in advancing diagnostic and prognostic methodologies, underscoring the potential of radiomics in the shift towards personalized medicine. Despite some methodological challenges, the field's dynamic evolution suggests a promising future for radiomics in enhancing the accuracy of ovarian cancer diagnosis and treatment, contributing to improved patient care and outcomes.
Collapse
Affiliation(s)
| | | | - Tan Huayun
- Department of Obstetrics, Weifang People's Hospital, Shandong Second Medical University, Weifang, China
| |
Collapse
|
2
|
Fujimoto H, Yoshihara M, Rodgers R, Iyoshi S, Mogi K, Miyamoto E, Hayakawa S, Hayashi M, Nomura S, Kitami K, Uno K, Sugiyama M, Koya Y, Yamakita Y, Nawa A, Enomoto A, Ricciardelli C, Kajiyama H. Tumor-associated fibrosis: a unique mechanism promoting ovarian cancer metastasis and peritoneal dissemination. Cancer Metastasis Rev 2024; 43:1037-1053. [PMID: 38546906 PMCID: PMC11300578 DOI: 10.1007/s10555-024-10169-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 01/11/2024] [Indexed: 08/06/2024]
Abstract
Epithelial ovarian cancer (EOC) is often diagnosed in advanced stage with peritoneal dissemination. Recent studies indicate that aberrant accumulation of collagen fibers in tumor stroma has a variety of effects on tumor progression. We refer to remodeled fibrous stroma with altered expression of collagen molecules, increased stiffness, and highly oriented collagen fibers as tumor-associated fibrosis (TAF). TAF contributes to EOC cell invasion and metastasis in the intraperitoneal cavity. However, an understanding of molecular events involved is only just beginning to emerge. Further development in this field will lead to new strategies to treat EOC. In this review, we focus on the recent findings on how the TAF contributes to EOC malignancy. Furthermore, we will review the recent initiatives and future therapeutic strategies for targeting TAF in EOC.
Collapse
Affiliation(s)
- Hiroki Fujimoto
- Department of Obstetrics and Gynaecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide, Australia
| | - Masato Yoshihara
- Department of Obstetrics and Gynaecology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Raymond Rodgers
- School of Biomedicine, Robinson Research Institute, The University of Adelaide, Adelaide, Australia
| | - Shohei Iyoshi
- Department of Obstetrics and Gynaecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Spemann Graduate School of Biology and Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Kazumasa Mogi
- Department of Obstetrics and Gynaecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Emiri Miyamoto
- Department of Obstetrics and Gynaecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Sae Hayakawa
- Department of Obstetrics and Gynaecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Maia Hayashi
- Department of Obstetrics and Gynaecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Satoshi Nomura
- Department of Obstetrics and Gynaecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuhisa Kitami
- Department of Obstetrics and Gynaecology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Kaname Uno
- Department of Obstetrics and Gynaecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University Graduate School of Medicine, Lund, Sweden
| | - Mai Sugiyama
- Bell Research Center-Department of Obstetrics and Gynaecology Collaborative Research, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshihiro Koya
- Bell Research Center-Department of Obstetrics and Gynaecology Collaborative Research, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshihiko Yamakita
- Bell Research Center-Department of Obstetrics and Gynaecology Collaborative Research, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akihiro Nawa
- Bell Research Center-Department of Obstetrics and Gynaecology Collaborative Research, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Atsushi Enomoto
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Carmela Ricciardelli
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide, Australia.
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynaecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
3
|
Ju HY, Youn SY, Kang J, Whang MY, Choi YJ, Han MR. Integrated analysis of spatial transcriptomics and CT phenotypes for unveiling the novel molecular characteristics of recurrent and non-recurrent high-grade serous ovarian cancer. Biomark Res 2024; 12:80. [PMID: 39135097 PMCID: PMC11318304 DOI: 10.1186/s40364-024-00632-7] [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: 06/12/2024] [Accepted: 08/02/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND High-grade serous ovarian cancer (HGSOC), which is known for its heterogeneity, high recurrence rate, and metastasis, is often diagnosed after being dispersed in several sites, with about 80% of patients experiencing recurrence. Despite a better understanding of its metastatic nature, the survival rates of patients with HGSOC remain poor. METHODS Our study utilized spatial transcriptomics (ST) to interpret the tumor microenvironment and computed tomography (CT) to examine spatial characteristics in eight patients with HGSOC divided into recurrent (R) and challenging-to-collect non-recurrent (NR) groups. RESULTS By integrating ST data with public single-cell RNA sequencing data, bulk RNA sequencing data, and CT data, we identified specific cell population enrichments and differentially expressed genes that correlate with CT phenotypes. Importantly, we elucidated that tumor necrosis factor-α signaling via NF-κB, oxidative phosphorylation, G2/M checkpoint, E2F targets, and MYC targets served as an indicator of recurrence (poor prognostic markers), and these pathways were significantly enriched in both the R group and certain CT phenotypes. In addition, we identified numerous prognostic markers indicative of nonrecurrence (good prognostic markers). Downregulated expression of PTGDS was linked to a higher number of seeding sites (≥ 3) in both internal HGSOC samples and public HGSOC TCIA and TCGA samples. Additionally, lower PTGDS expression in the tumor and stromal regions was observed in the R group than in the NR group based on our ST data. Chemotaxis-related markers (CXCL14 and NTN4) and markers associated with immune modulation (DAPL1 and RNASE1) were also found to be good prognostic markers in our ST and radiogenomics analyses. CONCLUSIONS This study demonstrates the potential of radiogenomics, combining CT and ST, for identifying diagnostic and therapeutic targets for HGSOC, marking a step towards personalized medicine.
Collapse
Affiliation(s)
- Hye-Yeon Ju
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, 22012, Korea
| | - Seo Yeon Youn
- Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Jun Kang
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Min Yeop Whang
- Department of Obstetrics and Gynecology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Youn Jin Choi
- Department of Obstetrics and Gynecology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
| | - Mi-Ryung Han
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, 22012, Korea.
- Institute for New Drug Development, College of Life Science and Bioengineering, Incheon National University, Incheon, 22012, Korea.
| |
Collapse
|
4
|
Lu J, Guo Q, Zhang Y, Zhao S, Li R, Fu Y, Feng Z, Wu Y, Li R, Li X, Qiang J, Wu X, Gu Y, Li H. A modified diffusion-weighted magnetic resonance imaging-based model from the radiologist's perspective: improved performance in determining the surgical resectability of advanced high-grade serous ovarian cancer. Am J Obstet Gynecol 2024; 231:117.e1-117.e17. [PMID: 38432417 DOI: 10.1016/j.ajog.2024.02.302] [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: 10/20/2023] [Revised: 02/22/2024] [Accepted: 02/22/2024] [Indexed: 03/05/2024]
Abstract
BACKGROUND Complete resection of all visible lesions during primary debulking surgery is associated with the most favorable prognosis in patients with advanced high-grade serous ovarian cancer. An accurate preoperative assessment of resectability is pivotal for tailored management. OBJECTIVE This study aimed to assess the potential value of a modified model that integrates the original 8 radiologic criteria of the Memorial Sloan Kettering Cancer Center model with imaging features of the subcapsular or diaphragm and mesenteric lesions depicted on diffusion-weighted magnetic resonance imaging and growth patterns of all lesions for predicting the resectability of advanced high-grade serous ovarian cancer. STUDY DESIGN This study included 184 patients with high-grade serous ovarian cancer who underwent preoperative diffusion-weighted magnetic resonance imaging between December 2018 and May 2023 at 2 medical centers. The patient cohort was divided into 3 subsets, namely a study cohort (n=100), an internal validation cohort (n=46), and an external validation cohort (n=38). Preoperative radiologic evaluations were independently conducted by 2 radiologists using both the Memorial Sloan Kettering Cancer Center model and the modified diffusion-weighted magnetic resonance imaging-based model. The morphologic characteristics of the ovarian tumors depicted on magnetic resonance imaging were assessed as either mass-like or infiltrative, and transcriptomic analysis of the primary tumor samples was performed. Univariate and multivariate statistical analyses were performed. RESULTS In the study cohort, both the scores derived using the Memorial Sloan Kettering Cancer Center (intraclass correlation coefficients of 0.980 and 0.959, respectively; both P<.001) and modified diffusion-weighted magnetic resonance imaging-based models (intraclass correlation coefficients of 0.962 and 0.940, respectively; both P<.001) demonstrated excellent intra- and interobserver agreement. The Memorial Sloan Kettering Cancer Center model (odds ratio, 1.825; 95% confidence interval, 1.390-2.395; P<.001) and the modified diffusion-weighted magnetic resonance imaging-based model (odds ratio, 1.776; 95% confidence interval, 1.410-2.238; P<.001) independently predicted surgical resectability. The modified diffusion-weighted magnetic resonance imaging-based model demonstrated improved predictive performance with an area under the curve of 0.867 in the study cohort and 0.806 and 0.913 in the internal and external validation cohorts, respectively. Using the modified diffusion-weighted magnetic resonance imaging-based model, patients with scores of 0 to 2, 3 to 4, 5 to 6, 7 to 10, and ≥11 achieved complete tumor debulking rates of 90.3%, 66.7%, 53.3%, 11.8%, and 0%, respectively. Most patients with incomplete tumor debulking had infiltrative tumors, and both the Memorial Sloan Kettering Cancer Center and the modified diffusion-weighted magnetic resonance imaging-based models yielded higher scores. The molecular differences between the 2 morphologic subtypes were identified. CONCLUSION When compared with the Memorial Sloan Kettering Cancer Center model, the modified diffusion-weighted magnetic resonance imaging-based model demonstrated enhanced accuracy in the preoperative prediction of resectability for advanced high-grade serous ovarian cancer. Patients with scores of 0 to 6 were eligible for primary debulking surgery.
Collapse
Affiliation(s)
- Jing Lu
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qinhao Guo
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ya Zhang
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, Yunnan, China
| | - Shuhui Zhao
- Department of Radiology, Xinhua Hospital affiliated with the Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ruimin Li
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi Fu
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zheng Feng
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yong Wu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Rong Li
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaojie Li
- Department of Radiology, Kunming Second People's Hospital, Kunming, Yunnan, China
| | - Jinwei Qiang
- Department of Radiology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Xiaohua Wu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yajia Gu
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Haiming Li
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
5
|
Lin Z, Ge H, Guo Q, Ren J, Gu W, Lu J, Zhong Y, Qiang J, Gong J, Li H. MRI-based radiomics model to preoperatively predict mesenchymal transition subtype in high-grade serous ovarian cancer. Clin Radiol 2024; 79:e715-e724. [PMID: 38342715 DOI: 10.1016/j.crad.2024.01.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/04/2024] [Accepted: 01/12/2024] [Indexed: 02/13/2024]
Abstract
AIM To develop a magnetic resonance imaging (MRI)-based radiomics model for the preoperative identification of mesenchymal transition (MT) subtype in high-grade serous ovarian cancer (HGSOC). MATERIALS AND METHODS One hundred and eighty-nine patients with histopathologically confirmed HGSOC were enrolled retrospectively. Among the included patients, 55 patients were determined as the MT subtype and the remaining 134 were non-MT subtype. After extracting a total of 204 features from T2-weighted imaging (T2WI) and contrast-enhanced (CE)-T1WI images, the Mann-Whitney U-test, Spearman correlation test, and Boruta algorithm were adopted to select the optimal feature set. Three classifiers, including logistic regression (LR), support vector machine (SVM), and random forest (RF), were trained to develop radiomics models. The performance of established models was evaluated from three aspects: discrimination, calibration, and clinical utility. RESULTS Seven radiomics features relevant to MT subtypes were selected to build the radiomics models. The model based on the RF algorithm showed the best performance in predicting MT subtype, with areas under the curves (AUCs) of 0.866 (95 % confidence interval [CI]: 0.797-0.936) and 0.852 (95 % CI: 0.736-0.967) in the training and testing cohorts, respectively. The calibration curves, supported with Brier scores, indicated very good consistency between observation and prediction. Decision curve analysis (DCA) showed that the RF-based model could provide more net benefit, which suggested favorable utility in clinical application. CONCLUSION The RF-based radiomics model provided accurate identification of MT from the non-MT subtype and may help facilitate personalised management of HGSOC.
Collapse
Affiliation(s)
- Z Lin
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Radiology, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - H Ge
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Q Guo
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - J Ren
- Department of Pharmaceuticals Diagnostics, GE HealthCare, Beijing 100176, China
| | - W Gu
- Department of Pathology, Obstetrics & Gynecology Hospital, Fudan University, Shanghai 200090, China
| | - J Lu
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Y Zhong
- Department of Radiology, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - J Qiang
- Department of Radiology, Jinshan Hospital, Fudan University, Shanghai 201508, China.
| | - J Gong
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - H Li
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
6
|
Hameed M, Yeung J, Boone D, Mallett S, Halligan S. Meta-research: How many diagnostic or prognostic models published in radiological journals are evaluated externally? Eur Radiol 2024; 34:2524-2533. [PMID: 37696974 PMCID: PMC10957714 DOI: 10.1007/s00330-023-10168-3] [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: 12/03/2022] [Revised: 07/03/2023] [Accepted: 07/11/2023] [Indexed: 09/13/2023]
Abstract
OBJECTIVES Prognostic and diagnostic models must work in their intended clinical setting, proven via "external evaluation", preferably by authors uninvolved with model development. By systematic review, we determined the proportion of models published in high-impact radiological journals that are evaluated subsequently. METHODS We hand-searched three radiological journals for multivariable diagnostic/prognostic models 2013-2015 inclusive, developed using regression. We assessed completeness of data presentation to allow subsequent external evaluation. We then searched literature to August 2022 to identify external evaluations of these index models. RESULTS We identified 98 index studies (73 prognostic; 25 diagnostic) describing 145 models. Only 15 (15%) index studies presented an evaluation (two external). No model was updated. Only 20 (20%) studies presented a model equation. Just 7 (15%) studies developing Cox models presented a risk table, and just 4 (9%) presented the baseline hazard. Two (4%) studies developing non-Cox models presented the intercept. Just 20 (20%) articles presented a Kaplan-Meier curve of the final model. The 98 index studies attracted 4224 citations (including 559 self-citations), median 28 per study. We identified just six (6%) subsequent external evaluations of an index model, five of which were external evaluations by researchers uninvolved with model development, and from a different institution. CONCLUSIONS Very few prognostic or diagnostic models published in radiological literature are evaluated externally, suggesting wasted research effort and resources. Authors' published models should present data sufficient to allow external evaluation by others. To achieve clinical utility, researchers should concentrate on model evaluation and updating rather than continual redevelopment. CLINICAL RELEVANCE STATEMENT The large majority of prognostic and diagnostic models published in high-impact radiological journals are never evaluated. It would be more efficient for researchers to evaluate existing models rather than practice continual redevelopment. KEY POINTS • Systematic review of highly cited radiological literature identified few diagnostic or prognostic models that were evaluated subsequently by researchers uninvolved with the original model. • Published radiological models frequently omit important information necessary for others to perform an external evaluation: Only 20% of studies presented a model equation or nomogram. • A large proportion of research citing published models focuses on redevelopment and ignores evaluation and updating, which would be a more efficient use of research resources.
Collapse
Affiliation(s)
- Maira Hameed
- Centre for Medical Imaging, University College London UCL, Charles Bell House, 43-45 Foley Street, London, W1W 7TS, UK
| | - Jason Yeung
- Centre for Medical Imaging, University College London UCL, Charles Bell House, 43-45 Foley Street, London, W1W 7TS, UK
| | - Darren Boone
- Centre for Medical Imaging, University College London UCL, Charles Bell House, 43-45 Foley Street, London, W1W 7TS, UK
| | - Sue Mallett
- Centre for Medical Imaging, University College London UCL, Charles Bell House, 43-45 Foley Street, London, W1W 7TS, UK
| | - Steve Halligan
- Centre for Medical Imaging, University College London UCL, Charles Bell House, 43-45 Foley Street, London, W1W 7TS, UK.
| |
Collapse
|
7
|
Crispin-Ortuzar M, Woitek R, Reinius MAV, Moore E, Beer L, Bura V, Rundo L, McCague C, Ursprung S, Escudero Sanchez L, Martin-Gonzalez P, Mouliere F, Chandrananda D, Morris J, Goranova T, Piskorz AM, Singh N, Sahdev A, Pintican R, Zerunian M, Rosenfeld N, Addley H, Jimenez-Linan M, Markowetz F, Sala E, Brenton JD. Integrated radiogenomics models predict response to neoadjuvant chemotherapy in high grade serous ovarian cancer. Nat Commun 2023; 14:6756. [PMID: 37875466 PMCID: PMC10598212 DOI: 10.1038/s41467-023-41820-7] [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: 05/27/2022] [Accepted: 09/20/2023] [Indexed: 10/26/2023] Open
Abstract
High grade serous ovarian carcinoma (HGSOC) is a highly heterogeneous disease that typically presents at an advanced, metastatic state. The multi-scale complexity of HGSOC is a major obstacle to predicting response to neoadjuvant chemotherapy (NACT) and understanding critical determinants of response. Here we present a framework to predict the response of HGSOC patients to NACT integrating baseline clinical, blood-based, and radiomic biomarkers extracted from all primary and metastatic lesions. We use an ensemble machine learning model trained to predict the change in total disease volume using data obtained at diagnosis (n = 72). The model is validated in an internal hold-out cohort (n = 20) and an independent external patient cohort (n = 42). In the external cohort the integrated radiomics model reduces the prediction error by 8% with respect to the clinical model, achieving an AUC of 0.78 for RECIST 1.1 classification compared to 0.47 for the clinical model. Our results emphasize the value of including radiomics data in integrative models of treatment response and provide methods for developing new biomarker-based clinical trials of NACT in HGSOC.
Collapse
Affiliation(s)
- Mireia Crispin-Ortuzar
- Department of Oncology, University of Cambridge, Cambridge, UK.
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK.
| | - Ramona Woitek
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Department of Radiology, University of Cambridge, Cambridge, UK
- Centre for Medical Image Analysis and AI (MIAAI), Danube Private University, Krems, Austria
| | - Marika A V Reinius
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Elizabeth Moore
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Lucian Beer
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Department of Radiology, University of Cambridge, Cambridge, UK
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Vlad Bura
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Department of Radiology, University of Cambridge, Cambridge, UK
| | - Leonardo Rundo
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Department of Radiology, University of Cambridge, Cambridge, UK
- Department of Information and Electrical Engineering and Applied Mathematics, University of Salerno, Fisciano, SA, Italy
| | - Cathal McCague
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Department of Radiology, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Stephan Ursprung
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Department of Radiology, University of Cambridge, Cambridge, UK
| | - Lorena Escudero Sanchez
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Department of Radiology, University of Cambridge, Cambridge, UK
| | - Paula Martin-Gonzalez
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Florent Mouliere
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- Department of Pathology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | - James Morris
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Teodora Goranova
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Anna M Piskorz
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Naveena Singh
- Department of Cellular Pathology, Barts Health NHS Trust, London, UK
| | - Anju Sahdev
- Department of Radiology, Barts Health NHS Trust, London, UK
| | - Roxana Pintican
- "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Radiology, County Clinical Emergency Hospital, Cluj-Napoca, Romania
| | - Marta Zerunian
- Department of Surgical and Medical Sciences and Translational Medicine, Sapienza University of Rome-Sant'Andrea University Hospital, Rome, Italy
| | - Nitzan Rosenfeld
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Helen Addley
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Department of Radiology, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Mercedes Jimenez-Linan
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Florian Markowetz
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Evis Sala
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Department of Radiology, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Dipartimento di Scienze Radiologiche ed Ematologiche, Universita Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Western Balkans University, Tirana, Albania
| | - James D Brenton
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| |
Collapse
|
8
|
Torkildsen CF, Thomsen LCV, Sande RK, Krakstad C, Stefansson I, Lamark EK, Knappskog S, Bjørge L. Molecular and phenotypic characteristics influencing the degree of cytoreduction in high-grade serous ovarian carcinomas. Cancer Med 2023. [PMID: 37191035 DOI: 10.1002/cam4.6085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 04/23/2023] [Accepted: 05/05/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND High-grade serous ovarian carcinoma (HGSOC) is the deadliest ovarian cancer subtype, and survival relates to initial cytoreductive surgical treatment. The existing tools for surgical outcome prediction remain inadequate for anticipating the outcomes of the complex relationship between tumour biology, clinical phenotypes, co-morbidity and surgical skills. In this genotype-phenotype association study, we combine phenotypic markers with targeted DNA sequencing to discover novel biomarkers to guide the surgical management of primary HGSOC. METHODS Primary tumour tissue samples (n = 97) and matched blood from a phenotypically well-characterised treatment-naïve HGSOC patient cohort were analysed by targeted massive parallel DNA sequencing (next generation sequencing [NGS]) of a panel of 360 cancer-related genes. Association analyses were performed on phenotypic traits related to complete cytoreductive surgery, while logistic regression analysis was applied for the predictive model. RESULTS The positive influence of complete cytoreductive surgery (R0) on overall survival was confirmed (p = 0.003). Before surgery, low volumes of ascitic fluid, lower CA125 levels, higher platelet counts and relatively lower clinical stage at diagnosis were all indicators, alone and combined, for complete cytoreduction (R0). Mutations in either the chromatin remodelling SWI_SNF (p = 0.036) pathway or the histone H3K4 methylation pathway (p = 0.034) correlated with R0. The R0 group also demonstrated higher tumour mutational burden levels (p = 0.028). A predictive model was developed by combining two phenotypes and the mutational status of five genes and one genetic pathway, enabling the prediction of surgical outcomes in 87.6% of the cases in this cohort. CONCLUSION Inclusion of molecular biomarkers adds value to the pre-operative stratification of HGSOC patients. A potential preoperative risk stratification model combining phenotypic traits and single-gene mutational status is suggested, but the set-up needs to be validated in larger cohorts.
Collapse
Affiliation(s)
- Cecilie Fredvik Torkildsen
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Obstetrics and Gynecology, Stavanger University Hospital, Stavanger, Norway
| | - Liv Cecilie Vestrheim Thomsen
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| | - Ragnar Kvie Sande
- Department of Obstetrics and Gynecology, Stavanger University Hospital, Stavanger, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Camilla Krakstad
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| | - Ingunn Stefansson
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Eva Karin Lamark
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| | - Stian Knappskog
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Line Bjørge
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
9
|
Deep learning for the ovarian lesion localization and discrimination between borderline and malignant ovarian tumors based on routine MR imaging. Sci Rep 2023; 13:2770. [PMID: 36797331 PMCID: PMC9935539 DOI: 10.1038/s41598-023-29814-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 02/10/2023] [Indexed: 02/18/2023] Open
Abstract
To establish a deep learning (DL) model in differentiating borderline ovarian tumor (BOT) from epithelial ovarian cancer (EOC) on conventional MR imaging. We retrospectively enrolled 201 patients of 102 pathologically proven BOTs and 99 EOCs at OB/GYN hospital Fudan University, between January 2015 and December 2017. All imaging data were reviewed on picture archiving and communication systems (PACS) server. Both T1-weighted imaging (T1WI) and T2-weighted imaging (T2WI) MR images were used for lesion area determination. We trained a U-net++ model with deep supervision to segment the lesion area on MR images. Then, the segmented regions were fed into a classification model based on DL network to categorize ovarian masses automatically. For ovarian lesion segmentation, the mean dice similarity coefficient (DSC) of the trained U-net++ model in the testing dataset achieved 0.73 [Formula: see text] 0.25, 0.76 [Formula: see text] 0.18, and 0.60 [Formula: see text] 0.24 in the sagittal T2WI, coronal T2WI, and axial T1WI images, respectively. The DL model by combined T2WI computerized network could differentiate BOT from EOC with a significantly higher AUC of 0.87, an accuracy of 83.7%, a sensitivity of 75.0% and a specificity of 87.5%. In comparison, the AUC yielded by radiologist was only 0.75, with an accuracy of 75.5%, a sensitivity of 96.0% and specificity of 54.2% (P < 0.001).The trained DL network model derived from routine MR imaging could help to distinguish BOT from EOC with a high accuracy, which was superior to radiologists' assessment.
Collapse
|
10
|
Lei R, Yu Y, Li Q, Yao Q, Wang J, Gao M, Wu Z, Ren W, Tan Y, Zhang B, Chen L, Lin Z, Yao H. Deep learning magnetic resonance imaging predicts platinum sensitivity in patients with epithelial ovarian cancer. Front Oncol 2022; 12:895177. [PMID: 36505880 PMCID: PMC9727155 DOI: 10.3389/fonc.2022.895177] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 10/18/2022] [Indexed: 11/24/2022] Open
Abstract
Objective The aim of the study is to develop and validate a deep learning model to predict the platinum sensitivity of patients with epithelial ovarian cancer (EOC) based on contrast-enhanced magnetic resonance imaging (MRI). Methods In this retrospective study, 93 patients with EOC who received platinum-based chemotherapy (≥4 cycles) and debulking surgery at the Sun Yat-sen Memorial Hospital from January 2011 to January 2020 were enrolled and randomly assigned to the training and validation cohorts (2:1). Two different models were built based on either the primary tumor or whole volume of the abdomen as the volume of interest (VOI) within the same cohorts, and then a pre-trained convolutional neural network Med3D (Resnet 10 version) was transferred to automatically extract 1,024 features from two MRI sequences (CE-T1WI and T2WI) of each patient to predict platinum sensitivity. The performance of the two models was compared. Results A total of 93 women (mean age, 50.5 years ± 10.5 [standard deviation]) were evaluated (62 in the training cohort and 31 in the validation cohort). The AUCs of the whole abdomen model were 0.97 and 0.98 for the training and validation cohorts, respectively, which was better than the primary tumor model (AUCs of 0.88 and 0.81 in the training and validation cohorts, respectively). In k-fold cross-validation and stratified analysis, the whole abdomen model maintained a stable performance, and the decision function value generated by the model was a prognostic indicator that successfully discriminates high- and low-risk recurrence patients. Conclusion The non-manually segmented whole-abdomen deep learning model based on MRI exhibited satisfactory predictive performance for platinum sensitivity and may assist gynecologists in making optimal treatment decisions.
Collapse
Affiliation(s)
- Ruilin Lei
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China,Department of Gynecological Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China,Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yunfang Yu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China,Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China,Faculty of Medicine, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Qingjian Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China,Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China,Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qinyue Yao
- Cells Vision Medical Technology Inc., Guangzhou, China
| | - Jin Wang
- Cells Vision Medical Technology Inc., Guangzhou, China
| | - Ming Gao
- Department of Radiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhuo Wu
- Department of Radiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wei Ren
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China,Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China,Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yujie Tan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China,Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China,Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Bingzhong Zhang
- Department of Gynecological Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liliang Chen
- Cells Vision Medical Technology Inc., Guangzhou, China
| | - Zhongqiu Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China,Department of Gynecological Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China,*Correspondence: Zhongqiu Lin, ; Herui Yao,
| | - Herui Yao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China,Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China,Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China,Breast Tumor Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China,*Correspondence: Zhongqiu Lin, ; Herui Yao,
| |
Collapse
|
11
|
Li H, Lu J, Deng L, Guo Q, Lin Z, Zhao S, Ge H, Qiang J, Gu Y, Liu Z. Diffusion-Weighted Magnetic Resonance Imaging and Morphological Characteristics Evaluation for Outcome Prediction of Primary Debulking Surgery for Advanced High-Grade Serous Ovarian Carcinoma. J Magn Reson Imaging 2022; 57:1340-1349. [PMID: 36054024 DOI: 10.1002/jmri.28418] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 08/19/2022] [Accepted: 08/19/2022] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Preoperative assessment of whether a successful primary debulking surgery (PDS) can be performed in patients with advanced high-grade serous ovarian carcinoma (HGSOC) remains a challenge. A reliable model to precisely predict resectability is highly demanded. PURPOSE To investigate the value of diffusion-weighted MRI (DW-MRI) combined with morphological characteristics to predict the PDS outcome in advanced HGSOC patients. STUDY TYPE Prospective. SUBJECTS A total of 95 consecutive patients with histopathologically confirmed advanced HGSOC (ranged from 39 to 77 years). FIELDS STRENGTH/SEQUENCE A 3.0 T, readout-segmented echo-planar DWI. ASSESSMENT The MRI morphological characteristics of the primary ovarian tumor, a peritoneal carcinomatosis index (PCI) derived from DWI (DWI-PCI) and histogram analysis of the primary ovarian tumor and the largest peritoneal carcinomatosis were assessed by three radiologists. Three different models were developed to predict the resectability, including a clinicoradiologic model combing MRI morphological characteristic with ascites and CA125 level; DWI-PCI alone; and a fusion model combining the clinical-morphological information and DWI-PCI. STATISTICAL TESTS Multivariate logistic regression analyses, receiver operating characteristic (ROC) curve, net reclassification index (NRI) and integrated discrimination improvement (IDI) were used. A P < 0.05 was considered to be statistically significant. RESULTS Sixty-seven cases appeared as a definite mass, whereas 28 cases as an infiltrative mass. The morphological characteristics and DWI-PCI were independent factors for predicting the resectability, with an AUC of 0.724 and 0.824, respectively. The multivariable predictive model consisted of morphological characteristics, CA-125, and the amount of ascites, with an incremental AUC of 0.818. Combining the application of a clinicoradiologic model and DWI-PCI showed significantly higher AUC of 0.863 than the ones of each of them implemented alone, with a positive NRI and IDI. DATA CONCLUSIONS The combination of two clinical factors, MRI morphological characteristics and DWI-PCI provide a reliable and valuable paradigm for the noninvasive prediction of the outcome of PDS. EVIDENCE LEVEL 2 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Haiming Li
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Guangdong Cardiovascular Institute, Guangzhou, China.,Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jing Lu
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lin Deng
- Department of Radiology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Qinhao Guo
- Department of Radiology, Jinshan Hospital, Fudan University, Shanghai, China.,Department of Gynecological oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zijing Lin
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Radiology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Shuhui Zhao
- Department of Radiology, Xinhua Hospital affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Huijuan Ge
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jinwei Qiang
- Department of Radiology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Yajia Gu
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
12
|
Wang X, Xu C, Grzegorzek M, Sun H. Habitat radiomics analysis of pet/ct imaging in high-grade serous ovarian cancer: Application to Ki-67 status and progression-free survival. Front Physiol 2022; 13:948767. [PMID: 36091379 PMCID: PMC9452776 DOI: 10.3389/fphys.2022.948767] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
Purpose: We aim to develop and validate PET/ CT image-based radiomics to determine the Ki-67 status of high-grade serous ovarian cancer (HGSOC), in which we use the metabolic subregion evolution to improve the prediction ability of the model. At the same time, the stratified effect of the radiomics model on the progression-free survival rate of ovarian cancer patients was illustrated.Materials and methods: We retrospectively reviewed 161 patients with HGSOC from April 2013 to January 2019. 18F-FDG PET/ CT images before treatment, pathological reports, and follow-up data were analyzed. A randomized grouping method was used to divide ovarian cancer patients into a training group and validation group. PET/ CT images were fused to extract radiomics features of the whole tumor region and radiomics features based on the Habitat method. The feature is dimensionality reduced, and meaningful features are screened to form a signature for predicting the Ki-67 status of ovarian cancer. Meanwhile, survival analysis was conducted to explore the hierarchical guidance significance of radiomics in the prognosis of patients with ovarian cancer.Results: Compared with texture features extracted from the whole tumor, the texture features generated by the Habitat method can better predict the Ki-67 state (p < 0.001). Radiomics based on Habitat can predict the Ki-67 expression accurately and has the potential to become a new marker instead of Ki-67. At the same time, the Habitat model can better stratify the prognosis (p < 0.05).Conclusion: We found a noninvasive imaging predictor that could guide the stratification of prognosis in ovarian cancer patients, which is related to the expression of Ki-67 in tumor tissues. This method is of great significance for the diagnosis and treatment of ovarian cancer.
Collapse
Affiliation(s)
- Xinghao Wang
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Chen Xu
- Department of Surgical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Marcin Grzegorzek
- Institute of Medical Informatics, University of Luebeck, Luebeck, Germany
| | - Hongzan Sun
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Hongzan Sun,
| |
Collapse
|
13
|
Kumar A, Wang C, Sheedy SP, McCauley BM, Winham SJ, Ramus SJ, Anglesio MS, Kim B, Torres D, Keeney GL, Cliby WA, Goode EL. Into the future: A pilot study combining imaging with molecular profiling to predict resectability in ovarian cancer. Gynecol Oncol 2022; 166:508-514. [PMID: 35931468 DOI: 10.1016/j.ygyno.2022.07.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 07/17/2022] [Accepted: 07/19/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVE We sought to determine the predictive value of combining tumor molecular subtype and computerized tomography (CT) imaging for surgical outcomes after primary cytoreductive surgery in advanced stage high-grade serous ovarian cancer (HGSOC) patients. METHODS We identified 129 HGSOC patients who underwent pre-operative CT imaging and post-operative tumor mRNA profiling. A continuous CT-score indicative of overall disease burden was defined based on six imaging measurements of anatomic involvement. Molecular subtypes were derived from mRNA profiling of chemo-naïve tumors and classified as mesenchymal (MES) subtype (36%) or non-MES subtype (64%). Fischer exact tests and multivariate logistic regression examined residual disease and surgical complexity. RESULTS Women with higher CT-scores were more likely to have MES subtype tumors (p = 0.014). MES subtypes and a high CT-score were independently predictive of macroscopic disease and high surgical complexity. In multivariate models adjusting for age, stage and American Society of Anesthesiologists (ASA) score, patients with a MES subtype and high CT-score had significantly elevated risk of macroscopic disease (OR = 26.7, 95% CI = [6.42, 187]) and were more likely to undergo high complexity surgery (OR = 9.53, 95% CI = [2.76, 40.6], compared to patients with non-MES tumor and low CT-score. CONCLUSION Preoperative CT imaging combined with tumor molecular subtyping can identify a subset of women unlikely to have resectable disease and likely to require high complexity surgery. Along with other clinical factors, these may refine predictive scores for resection and assist treatment planning. Investigating methods for pre-surgical molecular subtyping is an important next step.
Collapse
Affiliation(s)
- Amanika Kumar
- Department of Obstetrics and Gynecology, Division of Gynecologic Surgery, Mayo Clinic, Rochester, MN, United States.
| | - Chen Wang
- Department of Quantitative Health Sciences, Division of Computational Biology, Mayo Clinic, Rochester, MN, United States
| | - Shannon P Sheedy
- Department of Radiology, Mayo Clinic, Rochester, MN, United States
| | - Bryan M McCauley
- Department of Quantitative Health Sciences, Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN, United States
| | - Stacey J Winham
- Department of Quantitative Health Sciences, Division of Computational Biology, Mayo Clinic, Rochester, MN, United States
| | - Susan J Ramus
- School of Clinical Medicine, Faculty of Medicine, University of NSW Sydney, Sydney, New South Wales, Australia; Adult Cancer Program, Lowy Cancer Research Centre, University of NSW Sydney, Sydney, New South Wales, Australia
| | - Michael S Anglesio
- British Columbia's Ovarian Cancer Research (OVCARE) Program, BC Cancer, Vancouver General Hospital, and University of British Columbia, Vancouver, BC, Canada; Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, BC, Canada
| | - Bohyun Kim
- Department of Radiology, Mayo Clinic, Rochester, MN, United States
| | - Diogo Torres
- Division of Gynecologic Oncology, Ochsner Health Center, New Orleans, LA
| | - Gary L Keeney
- Division of Gynecologic Oncology, Ochsner Health Center, New Orleans, LA
| | - William A Cliby
- Department of Obstetrics and Gynecology, Division of Gynecologic Surgery, Mayo Clinic, Rochester, MN, United States
| | - Ellen L Goode
- Department of Quantitative Health Sciences, Division of Epidemiology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
14
|
Cai SQ, Song ZY, Wu MR, Lu JJ, Sun WW, Wei F, Li HM, Qiang JW, Li YA, Zhu J, Zhou JJ, Zeng MS. Magnetic Resonance Imaging and Diffusion Weighted Imaging-Based Histogram in Predicting Mesenchymal Transition High-Grade Serous Ovarian Cancer. Acad Radiol 2022; 30:1118-1128. [PMID: 35909051 DOI: 10.1016/j.acra.2022.06.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/24/2022] [Accepted: 06/26/2022] [Indexed: 11/01/2022]
Abstract
RATIONALE AND OBJECTIVES To investigate the value of magnetic resonance imaging (MRI) including diffusion-weighted imaging (DWI) findings in predicting mesenchymal transition (MT) high-grade serous ovarian cancer (HGSOC). MATERIALS AND METHODS Patients with HGSOC were enrolled from May 2017 to December 2020, who underwent pelvic MRI including DWI (b = 0,1000 s/mm2) before surgery, and were assigned to the MT HGSOC or non-MT HGSOC group according to histopathology results. Clinical characteristics and MRI features including DWI-based histogram metrics were assessed and compared between the two groups. Univariate and multivariate analyses were performed to identify the significant variables associated with MT HGSOC - these variables were then incorporated into a predictive nomogram, and ROC curve analysis was subsequently carried out to evaluate diagnostic performance. RESULTS A total of 81 consecutive patients were recruited for pelvic MRI before surgery, including 37 (45.7%) MT patients and 44 (54.3%) non-MT patients. At univariate analysis, the features significantly related to MT HGSOC were identified as absence of discrete primary ovarian mass, pouch of Douglas implants, ovarian mass size, tumor volume, mean, SD, median, and 95th percentile apparent diffusion coefficient (ADC) values (all p < 0.05). At multivariate analysis, the absence of discrete primary ovarian mass {odds ratio (OR): 46.477; p = 0.025}, mean ADC value ≤ 1.105 (OR: 1.023; p = 0.009), and median ADC value ≤ 1.038 (OR: 0.982; p = 0.034) were found to be independent risk factors associated with MT HGSOC. The combination of all independent criteria yielded the largest AUC of 0.82 with a sensitivity of 83.87% and specificity of 66.67%, superior to any of the single predictor alone (p ≤ 0.012). The predictive C-index nomogram performance of the combination was 0.82. CONCLUSION The combination of absence of discrete primary ovarian mass, lower mean ADC value, and median ADC value may be helpful for preoperatively predicting MT HGSOC.
Collapse
|
15
|
Radiogenomics: A Valuable Tool for the Clinical Assessment and Research of Ovarian Cancer. J Comput Assist Tomogr 2022; 46:371-378. [DOI: 10.1097/rct.0000000000001279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
16
|
Liu X, Wang T, Zhang G, Hua K, Jiang H, Duan S, Jin J, Zhang H. Two-dimensional and three-dimensional T2 weighted imaging-based radiomic signatures for the preoperative discrimination of ovarian borderline tumors and malignant tumors. J Ovarian Res 2022; 15:22. [PMID: 35115022 PMCID: PMC8815217 DOI: 10.1186/s13048-022-00943-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 12/31/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Ovarian cancer is the most women malignancy in the whole world. It is difficult to differentiate ovarian cancers from ovarian borderline tumors because of some similar imaging findings.Radiomics study may help clinicians to make a proper diagnosis before invasive surgery. PURPOSE To evaluate the ability of T2-weighted imaging (T2WI)-based radiomics to discriminate ovarian borderline tumors (BOTs) from malignancies based on two-dimensional (2D) and three-dimensional (3D) lesion segmentation methods. METHODS A total of 95 patients with pathologically proven ovarian BOTs and 101 patients with malignancies were retrospectively included in this study. We evaluated the diagnostic performance of the signatures derived from T2WI-based radiomics in their ability to differentiate between BOTs and malignancies and compared the performance differences in the 2D and 3D segmentation models. The least absolute shrinkage and selection operator method (Lasso) was used for radiomics feature selection and machine learning processing. RESULTS The radiomics score between BOTs and malignancies in four types of selected T2WI-based radiomics models differed significantly at the statistical level (p < 0.0001). For the classification between BOTs and malignant masses, the 2D and 3D coronal T2WI-based radiomics models yielded accuracy values of 0.79 and 0.83 in the testing group, respectively; the 2D and 3D sagittal fat-suppressed (fs) T2WI-based radiomics models yielded an accuracy of 0.78 and 0.99, respectively. CONCLUSIONS Our results suggest that T2WI-based radiomic features were highly correlated with ovarian tumor subtype classification. 3D-sagittal MRI radiomics features may help clinicians differentiate ovarian BOTs from malignancies with high ACC.
Collapse
Affiliation(s)
- Xuefen Liu
- Department of Radiology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, P.R. China
| | - Tianping Wang
- Department of Radiology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, P.R. China
| | - Guofu Zhang
- Department of Radiology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, P.R. China
| | - Keqin Hua
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, P.R. China
| | - Hua Jiang
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, P.R. China
| | | | - Jun Jin
- Department of Pathology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, P.R. China
| | - He Zhang
- Department of Radiology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, P.R. China.
| |
Collapse
|
17
|
He Y, Liu P, Xie L, Zeng S, Lin H, Zhang B, Liu J. Construction and Verification of a Predictive Model for Risk Factors in Children With Severe Adenoviral Pneumonia. Front Pediatr 2022; 10:874822. [PMID: 35832584 PMCID: PMC9271770 DOI: 10.3389/fped.2022.874822] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 06/08/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE To construct and validate a predictive model for risk factors in children with severe adenoviral pneumonia based on chest low-dose CT imaging and clinical features. METHODS A total of 177 patients with adenoviral pneumonia who underwent low-dose CT examination were collected between January 2019 and August 2019. The assessment criteria for severe pneumonia were divided into mild group (N = 125) and severe group (N = 52). All cases divided into training cohort (N = 125) and validation cohort (N = 52). We constructed a prediction model by drawing a nomogram and verified the predictive efficacy of the model through the ROC curve, calibration curve and decision curve analysis. RESULTS The difference was statistically significant (P < 0.05) between the mild adenovirus pneumonia group and the severe adenovirus pneumonia group in gender, age, weight, body temperature, L/N ratio, LDH, ALT, AST, CK-MB, ADV DNA, bronchial inflation sign, emphysema, ground glass sign, bronchial wall thickening, bronchiectasis, pleural effusion, consolidation score, and lobular inflammation score. Multivariate logistic regression analysis showed that gender, LDH value, emphysema, consolidation score, and lobular inflammation score were severe independent risk factors for adenovirus pneumonia in children. Logistic regression was employed to construct clinical model, imaging semantic feature model, and combined model. The AUC values of the training sets of the three models were 0.85 (0.77-0.94), 0.83 (0.75-0.91), and 0.91 (0.85-0.97). The AUC of the validation set was 0.77 (0.64-0.91), 0.83 (0.71-0.94), and 0.85 (0.73-0.96), respectively. The calibration curve fit good of the three models. The clinical decision curve analysis demonstrates the clinical application value of the nomogram prediction model. CONCLUSION The prediction model based on chest low-dose CT image characteristics and clinical characteristics has relatively clear predictive value in distinguishing mild adenovirus pneumonia from severe adenovirus pneumonia in children and might provide a new method for early clinical prediction of the outcome of adenovirus pneumonia in children.
Collapse
Affiliation(s)
- Yaqiong He
- Department of Radiology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Peng Liu
- Department of Radiology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Leyun Xie
- Department of Pediatrics, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Saizhen Zeng
- Department of Pediatrics, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, China
| | | | - Bing Zhang
- Department of Pediatrics, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Jianbin Liu
- Department of Radiology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, China
| |
Collapse
|
18
|
Development of MRI-Based Radiomics Model to Predict the Risk of Recurrence in Patients With Advanced High-Grade Serous Ovarian Carcinoma. AJR Am J Roentgenol 2021; 217:664-675. [PMID: 34259544 DOI: 10.2214/ajr.20.23195] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE. The purpose of our study was to develop a radiomics model based on preoperative MRI and clinical information for predicting recurrence-free survival (RFS) in patients with advanced high-grade serous ovarian carcinoma (HGSOC). MATERIALS AND METHODS. This retrospective study enrolled 117 patients with HGSOC, including 90 patients with recurrence and 27 without recurrence; 1046 radiomics features were extracted from T2-weighted images and contrast-enhanced T1-weighted images using a manual segmentation method. L1 regularization-based least absolute shrinkage and selection operator (LASSO) regression was performed to select features, and the synthetic minority oversampling technique (SMOTE) was used to balance our dataset. A support vector machine (SVM) classifier was used to build the classification model. To validate the performance of the proposed models, we applied a leave-one-out cross-validation method to train and test the classifier. Cox proportional hazards regression, Harrell concordance index (C-index), and Kaplan-Meier plots analysis were used to evaluate the associations between radiomics signatures and RFS. RESULTS. The fusion radiomics-based model yielded a significantly higher AUC value of 0.85 in evaluating RFS than the model using contrast-enhanced T1-weighted imaging features alone or T2-weighted imaging features alone (AUC = 0.79 and 0.74 and p = .02 and .01, respectively). Kaplan-Meier survival curves showed significant differences between high and low recurrence risk in patients with HGSOC by different models. The fusion model combining radiomics features and clinical information showed higher performance than the clinical model (C-index = 0.62 and 0.60, respectively). CONCLUSION. The proposed MRI-based radiomics signatures may provide a potential way to develop a prediction model and can help identify patients with advanced HGSOC who have a high risk of recurrence.
Collapse
|
19
|
Zhu H, Ai Y, Zhang J, Zhang J, Jin J, Xie C, Su H, Jin X. Preoperative Nomogram for Differentiation of Histological Subtypes in Ovarian Cancer Based on Computer Tomography Radiomics. Front Oncol 2021; 11:642892. [PMID: 33842352 PMCID: PMC8027335 DOI: 10.3389/fonc.2021.642892] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 03/03/2021] [Indexed: 12/27/2022] Open
Abstract
Objectives Non-invasive method to predict the histological subtypes preoperatively is essential for the overall management of ovarian cancer (OC). The feasibility of radiomics in the differentiating of epithelial ovarian cancer (EOC) and non-epithelial ovarian cancer (NEOC) based on computed tomography (CT) images was investigated. Methods Radiomics features were extracted from preoperative CT for 101 patients with pathologically proven OC. Radiomics signature was built using the least absolute shrinkage and selection operator (LASSO) logistic regression. A nomogram was developed with the combination of radiomics features and clinical factors to differentiate EOC and NEOC. Results Eight radiomics features were selected to build a radiomics signature with an area under curve (AUC) of 0.781 (95% confidence interval (CI), 0.666 -0.897) in the discrimination between EOC and NEOC. The AUC of the combined model integrating clinical factors and radiomics features was 0.869 (95% CI, 0.783 -0.955). The nomogram demonstrated that the combined model provides a better net benefit to predict histological subtypes compared with radiomics signature and clinical factors alone when the threshold probability is within a range from 0.43 to 0.97. Conclusions Nomogram developed with CT radiomics signature and clinical factors is feasible to predict the histological subtypes preoperative for patients with OC.
Collapse
Affiliation(s)
- Haiyan Zhu
- Department of Gynecology, Shanghai First Maternal and Infant Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Gynecology, The 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yao Ai
- Department of Radiotherapy Center, The 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jindi Zhang
- Department of Gynecology, The 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ji Zhang
- Department of Radiotherapy Center, The 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Juebin Jin
- Department of Medical Engineering, The 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Congying Xie
- Department of Radiation and Medical Oncology, The 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Radiation and Medical Oncology, The 2nd Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Huafang Su
- Department of Radiation and Medical Oncology, The 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiance Jin
- Department of Gynecology, The 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
20
|
Vieira AC, Antunes N, Damasceno E, Ramalho M, Esteves S, Vaz F, Félix A, Cunha TM. Ovarian carcinoma in patients with BRCA mutation - a correlation between the growing pattern of peritoneal implants evaluated by CT/MRI and the genotype BRCA1 and BRCA2. THE EGYPTIAN JOURNAL OF RADIOLOGY AND NUCLEAR MEDICINE 2020. [DOI: 10.1186/s43055-020-00183-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Ovarian cancer is the leading cause of death from gynecologic cancer. The risk of developing ovarian cancer is significantly increased in patients that carry a genetic mutation of tumor suppressor gene BRCA1 or BRCA2. The majority of BRCA-associated ovarian/fallopian tube cancers are high-grade serous carcinomas (HGSC). The recognition of patterns of disease is crucial to identify distinctive imaging features that could be useful for predicting prognosis and therapeutic response.
Results
An institutional review board-approved retrospective study was performed and included 34patients (23 BRCA-mutated and 11 BRCA wild-type) with HGSC FIGO III/IV who underwent pre-operative or pre-chemotherapy contrast-enhanced CT/MRI of the abdomen and pelvis between January 2003 and December 2017. Three radiologists independently reviewed the imaging studies and looked for qualitative features of the primary tumor and peritoneal metastases (nodular versus infiltrative pattern). Two pathologists also assessed the histopathologic characteristics of the surgical specimens, with emphasis on the growth pattern of metastatic deposits (expansive/nodular and infiltrative) and inflammatory infiltrate (intra- and/or peritumoral).
No significant associations were found between the different groups of patients (BRCA1-mutant HGSC, BRCA2-mutant HGSC. and BRCA wild-type) and CT/MRI features of ovarian tumors, morphology of peritoneal metastasis, and pathologic characteristics.
Conclusion
Identification of specific imaging and pathologic features is important to pursue an optimal personalized cancer treatment strategy and to develop precision medicine in the future.
Collapse
|
21
|
Veeraraghavan H, Vargas HA, Jimenez-Sanchez A, Micco M, Mema E, Lakhman Y, Crispin-Ortuzar M, Huang EP, Levine DA, Grisham RN, Abu-Rustum N, Deasy JO, Snyder A, Miller ML, Brenton JD, Sala E. Integrated Multi-Tumor Radio-Genomic Marker of Outcomes in Patients with High Serous Ovarian Carcinoma. Cancers (Basel) 2020; 12:E3403. [PMID: 33212885 PMCID: PMC7698381 DOI: 10.3390/cancers12113403] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/06/2020] [Accepted: 11/11/2020] [Indexed: 02/06/2023] Open
Abstract
Purpose: Develop an integrated intra-site and inter-site radiomics-clinical-genomic marker of high grade serous ovarian cancer (HGSOC) outcomes and explore the biological basis of radiomics with respect to molecular signaling pathways and the tumor microenvironment (TME). Method: Seventy-five stage III-IV HGSOC patients from internal (N = 40) and external factors via the Cancer Imaging Archive (TCGA) (N = 35) with pre-operative contrast enhanced CT, attempted primary cytoreduction, at least two disease sites, and molecular analysis performed within TCGA were retrospectively analyzed. An intra-site and inter-site radiomics (cluDiss) measure was combined with clinical-genomic variables (iRCG) and compared against conventional (volume and number of sites) and average radiomics (N = 75) for prognosticating progression-free survival (PFS) and platinum resistance. Correlation with molecular signaling and TME derived using a single sample gene set enrichment that was measured. Results: The iRCG model had the best platinum resistance classification accuracy (AUROC of 0.78 [95% CI 0.77 to 0.80]). CluDiss was associated with PFS (HR 1.03 [95% CI: 1.01 to 1.05], p = 0.002), negatively correlated with Wnt signaling, and positively to immune TME. Conclusions: CluDiss and the iRCG prognosticated HGSOC outcomes better than conventional and average radiomic measures and could better stratify patient outcomes if validated on larger multi-center trials.
Collapse
Affiliation(s)
- Harini Veeraraghavan
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Herbert Alberto Vargas
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (H.A.V.); (Y.L.); (E.S.)
| | - Alejandro Jimenez-Sanchez
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Center, Cambridge, Cambridgeshire CB2 0RE, UK; (A.J.-S.); (M.C.-O.); (M.L.M.); (J.D.B.)
| | - Maura Micco
- Radioterapia Oncologica ed Ematologica, Dipartimento Diagnostica per Immagini, Area Diagnostica per Immagini, Radiologica Diagnostica e Interventistica Generale, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy;
| | - Eralda Mema
- Columbia University Medical Center, New York, NY 10032, USA;
| | - Yulia Lakhman
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (H.A.V.); (Y.L.); (E.S.)
| | - Mireia Crispin-Ortuzar
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Center, Cambridge, Cambridgeshire CB2 0RE, UK; (A.J.-S.); (M.C.-O.); (M.L.M.); (J.D.B.)
| | | | - Douglas A. Levine
- Laura and Issac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA;
| | - Rachel N. Grisham
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (R.N.G.); (A.S.)
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Nadeem Abu-Rustum
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Joseph O. Deasy
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Alexandra Snyder
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (R.N.G.); (A.S.)
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Martin L. Miller
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Center, Cambridge, Cambridgeshire CB2 0RE, UK; (A.J.-S.); (M.C.-O.); (M.L.M.); (J.D.B.)
| | - James D. Brenton
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Center, Cambridge, Cambridgeshire CB2 0RE, UK; (A.J.-S.); (M.C.-O.); (M.L.M.); (J.D.B.)
| | - Evis Sala
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (H.A.V.); (Y.L.); (E.S.)
| |
Collapse
|
22
|
Li HM, Tang W, Feng F, Zhao SH, Gu WY, Zhang GF, Qiang JW. Whole solid tumor volume histogram parameters for predicting the recurrence in patients with epithelial ovarian carcinoma: a feasibility study on quantitative DCE-MRI. Acta Radiol 2020; 61:1266-1276. [PMID: 31955611 DOI: 10.1177/0284185119898654] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Preoperative prediction of the recurrence of epithelial ovarian carcinoma (EOC) can guide the clinical treatment and improve the prognosis. However, there are still no reliable predictive biomarkers. PURPOSE To evaluate whether whole solid tumor volume histogram parameters measured from quantitative dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) can predict the recurrence in patients with EOC. MATERIAL AND METHODS We followed up 56 patients with surgical and histopathologically diagnosed EOC who underwent quantitative DCE-MRI scans. The differences of the histogram parameters between patients with and without recurrence were compared. Mann-Whitney U test, Pearson's Chi-squared test, or Fisher's exact test, and receiver operating characteristic (ROC) curves were used for statistical analysis. RESULTS All histogram parameters of Ktrans, kep, and ve were not significantly different between EOC patients with and without recurrence (P>0.05). For 30 patients with high-grade serous ovarian carcinoma (HGSOC), the histogram parameters of Ktrans (mean and 5th, 10th, 25th, 50th, 75th percentiles) and kep (mean and 50th percentile) in 12 patients with recurrence were significantly lower than those in 18 patients without recurrence (all P<0.05). ROC curves showed that the 5th percentile of Ktrans had the largest area under the curve (AUC) of 0.792 for predicting the recurrence in patients with HGSOC. When the threshold value was ≤0.0263/min, the sensitivity, specificity, and accuracy were 100%, 66.7%, and 80%, respectively. CONCLUSION Instead of predicting the recurrence of EOC, whole solid tumor volume quantitative DCE-MRI histogram parameters could predict the recurrence of HGSOC and may be potential biomarkers for the prediction of HGSOC recurrence.
Collapse
Affiliation(s)
- Hai Ming Li
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, PR China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, PR China
- Department of Radiology, Jinshan Hospital, Fudan University, Shanghai, PR China
| | - Wei Tang
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, PR China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, PR China
| | - Feng Feng
- Department of Radiology, Nantong Cancer Hospital, Nantong University, Nantong, Jiangsu, PR China
| | - Shu Hui Zhao
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University, Shanghai, PR China
| | - Wei Yong Gu
- Department of Pathology, Obstetrics & Gynecology Hospital, Fudan University, Shanghai, PR China
| | - Guo Fu Zhang
- Department of Radiology, Obstetrics & Gynecology Hospital, Fudan University, Shanghai, PR China
| | - Jin Wei Qiang
- Department of Radiology, Jinshan Hospital, Fudan University, Shanghai, PR China
| |
Collapse
|
23
|
Martin-Gonzalez P, Crispin-Ortuzar M, Rundo L, Delgado-Ortet M, Reinius M, Beer L, Woitek R, Ursprung S, Addley H, Brenton JD, Markowetz F, Sala E. Integrative radiogenomics for virtual biopsy and treatment monitoring in ovarian cancer. Insights Imaging 2020; 11:94. [PMID: 32804260 PMCID: PMC7431480 DOI: 10.1186/s13244-020-00895-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 07/16/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Ovarian cancer survival rates have not changed in the last 20 years. The majority of cases are High-grade serous ovarian carcinomas (HGSOCs), which are typically diagnosed at an advanced stage with multiple metastatic lesions. Taking biopsies of all sites of disease is infeasible, which challenges the implementation of stratification tools based on molecular profiling. MAIN BODY In this review, we describe how these challenges might be overcome by integrating quantitative features extracted from medical imaging with the analysis of paired genomic profiles, a combined approach called radiogenomics, to generate virtual biopsies. Radiomic studies have been used to model different imaging phenotypes, and some radiomic signatures have been associated with paired molecular profiles to monitor spatiotemporal changes in the heterogeneity of tumours. We describe different strategies to integrate radiogenomic information in a global and local manner, the latter by targeted sampling of tumour habitats, defined as regions with distinct radiomic phenotypes. CONCLUSION Linking radiomics and biological correlates in a targeted manner could potentially improve the clinical management of ovarian cancer. Radiogenomic signatures could be used to monitor tumours during the course of therapy, offering additional information for clinical decision making. In summary, radiogenomics may pave the way to virtual biopsies and treatment monitoring tools for integrative tumour analysis.
Collapse
Affiliation(s)
- Paula Martin-Gonzalez
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, CB2 0RE, UK
| | - Mireia Crispin-Ortuzar
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, CB2 0RE, UK
| | - Leonardo Rundo
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, CB2 0RE, UK
- Department of Radiology, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Maria Delgado-Ortet
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, CB2 0RE, UK
- Department of Radiology, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Marika Reinius
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, CB2 0RE, UK
| | - Lucian Beer
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, CB2 0RE, UK
- Department of Radiology, University of Cambridge, Cambridge, CB2 0QQ, UK
- Department of Biomedical Imaging and Image-guided Therapy, Medical University Vienna, 1090, Vienna, Austria
| | - Ramona Woitek
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, CB2 0RE, UK
- Department of Radiology, University of Cambridge, Cambridge, CB2 0QQ, UK
- Department of Biomedical Imaging and Image-guided Therapy, Medical University Vienna, 1090, Vienna, Austria
| | - Stephan Ursprung
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, CB2 0RE, UK
- Department of Radiology, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Helen Addley
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK
- Department of Radiology, University of Cambridge, Cambridge, CB2 0QQ, UK
- Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - James D Brenton
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, CB2 0RE, UK
| | - Florian Markowetz
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, CB2 0RE, UK
| | - Evis Sala
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, CB2 0RE, UK.
- Department of Radiology, University of Cambridge, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
24
|
Beer L, Sahin H, Bateman NW, Blazic I, Vargas HA, Veeraraghavan H, Kirby J, Fevrier-Sullivan B, Freymann JB, Jaffe CC, Brenton J, Miccó M, Nougaret S, Darcy KM, Maxwell GL, Conrads TP, Huang E, Sala E. Integration of proteomics with CT-based qualitative and radiomic features in high-grade serous ovarian cancer patients: an exploratory analysis. Eur Radiol 2020; 30:4306-4316. [PMID: 32253542 PMCID: PMC7338824 DOI: 10.1007/s00330-020-06755-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 01/21/2020] [Accepted: 02/17/2020] [Indexed: 11/24/2022]
Abstract
OBJECTIVES To investigate the association between CT imaging traits and texture metrics with proteomic data in patients with high-grade serous ovarian cancer (HGSOC). METHODS This retrospective, hypothesis-generating study included 20 patients with HGSOC prior to primary cytoreductive surgery. Two readers independently assessed the contrast-enhanced computed tomography (CT) images and extracted 33 imaging traits, with a third reader adjudicating in the event of a disagreement. In addition, all sites of suspected HGSOC were manually segmented texture features which were computed from each tumor site. Three texture features that represented intra- and inter-site tumor heterogeneity were used for analysis. An integrated analysis of transcriptomic and proteomic data identified proteins with conserved expression between primary tumor sites and metastasis. Correlations between protein abundance and various CT imaging traits and texture features were assessed using the Kendall tau rank correlation coefficient and the Mann-Whitney U test, whereas the area under the receiver operating characteristic curve (AUC) was reported as a metric of the strength and the direction of the association. P values < 0.05 were considered significant. RESULTS Four proteins were associated with CT-based imaging traits, with the strongest correlation observed between the CRIP2 protein and disease in the mesentery (p < 0.001, AUC = 0.05). The abundance of three proteins was associated with texture features that represented intra-and inter-site tumor heterogeneity, with the strongest negative correlation between the CKB protein and cluster dissimilarity (p = 0.047, τ = 0.326). CONCLUSION This study provides the first insights into the potential associations between standard-of-care CT imaging traits and texture measures of intra- and inter-site heterogeneity, and the abundance of several proteins. KEY POINTS • CT-based texture features of intra- and inter-site tumor heterogeneity correlate with the abundance of several proteins in patients with HGSOC. • CT imaging traits correlate with protein abundance in patients with HGSOC.
Collapse
MESH Headings
- Abdominal Cavity/diagnostic imaging
- Adaptor Proteins, Signal Transducing/metabolism
- Aged
- Aged, 80 and over
- Aldehyde Oxidoreductases/metabolism
- Antigens, Neoplasm/metabolism
- Carcinoma, Ovarian Epithelial/diagnostic imaging
- Carcinoma, Ovarian Epithelial/metabolism
- Carcinoma, Ovarian Epithelial/secondary
- Cytokines/metabolism
- Female
- Gene Expression Profiling
- Glucose-6-Phosphate Isomerase/metabolism
- Humans
- LIM Domain Proteins/metabolism
- Mesentery/diagnostic imaging
- Middle Aged
- Neoplasm Grading
- Neoplasm Proteins/metabolism
- Neoplasms, Cystic, Mucinous, and Serous/diagnostic imaging
- Neoplasms, Cystic, Mucinous, and Serous/metabolism
- Neoplasms, Cystic, Mucinous, and Serous/secondary
- Omentum/diagnostic imaging
- Ovarian Neoplasms/diagnostic imaging
- Ovarian Neoplasms/metabolism
- Ovarian Neoplasms/pathology
- Peritoneal Neoplasms/diagnostic imaging
- Peritoneal Neoplasms/metabolism
- Peritoneal Neoplasms/secondary
- Pilot Projects
- Proteomics
- ROC Curve
- Retrospective Studies
- Tomography, X-Ray Computed/methods
Collapse
Affiliation(s)
- Lucian Beer
- Department of Radiology, Cancer Research UK Cambridge Center, Cambridge, CB2 0QQ, UK
| | - Hilal Sahin
- Department of Radiology, Cancer Research UK Cambridge Center, Cambridge, CB2 0QQ, UK
| | - Nicholas W Bateman
- Department of Obstetrics and Gynecology, Gynecologic Cancer Center of Excellence, Walter Reed National Military Medical Center, Uniformed Services University, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
- The John P. Murtha Cancer Center, Walter Reed National Military Medical Center, Uniformed Services University, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
| | - Ivana Blazic
- Department of Radiology, Clinical Hospital Center Zemun, Vukova 9, Belgrade, 11080, Serbia
| | - Hebert Alberto Vargas
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Harini Veeraraghavan
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Justin Kirby
- Cancer Imaging Informatics Lab, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Brenda Fevrier-Sullivan
- Cancer Imaging Informatics Lab, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - John B Freymann
- Cancer Imaging Informatics Lab, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - C Carl Jaffe
- Department of Radiology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - James Brenton
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, Cambridgeshire, UK
- Cancer Research UK Cambridge Centre, Cambridge, Cambridgeshire, UK
| | - Maura Miccó
- Dipartimento Diagnostica per Immagini, Radiologia Diagnostica e Interventistica Generale, Area Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Rome, Italy
| | - Stephanie Nougaret
- Department of Radiology, Montpellier Cancer Institute, INSERM, University of Montpellier, Montpellier, France
| | - Kathleen M Darcy
- Department of Obstetrics and Gynecology, Gynecologic Cancer Center of Excellence, Walter Reed National Military Medical Center, Uniformed Services University, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
- The John P. Murtha Cancer Center, Walter Reed National Military Medical Center, Uniformed Services University, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
| | - G Larry Maxwell
- Department of Obstetrics and Gynecology, Gynecologic Cancer Center of Excellence, Walter Reed National Military Medical Center, Uniformed Services University, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
- The John P. Murtha Cancer Center, Walter Reed National Military Medical Center, Uniformed Services University, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
- Department of Obstetrics and Gynecology, Inova Fairfax Medical Campus, 3300 Gallows Rd., Falls Church, VA, 22042, USA
| | - Thomas P Conrads
- Department of Obstetrics and Gynecology, Gynecologic Cancer Center of Excellence, Walter Reed National Military Medical Center, Uniformed Services University, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
- The John P. Murtha Cancer Center, Walter Reed National Military Medical Center, Uniformed Services University, 8901 Wisconsin Avenue, Bethesda, MD, 20889, USA
- Department of Obstetrics and Gynecology, Inova Fairfax Medical Campus, 3300 Gallows Rd., Falls Church, VA, 22042, USA
- Inova Center for Personalized Health, Inova Schar Cancer Institute, 3300 Gallows Rd., Falls Church, VA, 22042, USA
| | - Erich Huang
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Rockville, MD, 20850, USA
| | - Evis Sala
- Department of Radiology, Cancer Research UK Cambridge Center, Cambridge, CB2 0QQ, UK.
- Department of Radiology, University of Cambridge, Box 218, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
25
|
Hu Y, Taylor-Harding B, Raz Y, Haro M, Recouvreux MS, Taylan E, Lester J, Millstein J, Walts AE, Karlan BY, Orsulic S. Are Epithelial Ovarian Cancers of the Mesenchymal Subtype Actually Intraperitoneal Metastases to the Ovary? Front Cell Dev Biol 2020; 8:647. [PMID: 32766252 PMCID: PMC7380132 DOI: 10.3389/fcell.2020.00647] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 06/29/2020] [Indexed: 12/12/2022] Open
Abstract
Primary ovarian high-grade serous carcinoma (HGSC) has been classified into 4 molecular subtypes: Immunoreactive, Proliferative, Differentiated, and Mesenchymal (Mes), of which the Mes subtype (Mes-HGSC) is associated with the worst clinical outcomes. We propose that Mes-HGSC comprise clusters of cancer and associated stromal cells that detached from tumors in the upper abdomen/omentum and disseminated in the peritoneal cavity, including to the ovary. Using comparative analyses of multiple transcriptomic data sets, we provide the following evidence that the phenotype of Mes-HGSC matches the phenotype of tumors in the upper abdomen/omentum: (1) irrespective of the primary ovarian HGSC molecular subtype, matched upper abdominal/omental metastases were typically of the Mes subtype, (2) the Mes subtype was present at the ovarian site only in patients with concurrent upper abdominal/omental metastases and not in those with HGSC confined to the ovary, and (3) ovarian Mes-HGSC had an expression profile characteristic of stromal cells in the upper abdominal/omental metastases. We suggest that ovarian Mes-HGSC signifies advanced intraperitoneal tumor dissemination to the ovary rather than a subtype of primary ovarian HGSC. This is consistent with the presence of upper abdominal/omental disease, suboptimal debulking, and worst survival previously reported in patients with ovarian Mes-HGSC compared to other molecular subtypes.
Collapse
Affiliation(s)
- Ye Hu
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Barbie Taylor-Harding
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Yael Raz
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Marcela Haro
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Maria Sol Recouvreux
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Enes Taylan
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Jenny Lester
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Joshua Millstein
- Division of Biostatistics, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Ann E Walts
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Beth Y Karlan
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, United States
| | - Sandra Orsulic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
26
|
Pan S, Ding Z, Zhang L, Ruan M, Shan Y, Deng M, Pang P, Shen Q. A Nomogram Combined Radiomic and Semantic Features as Imaging Biomarker for Classification of Ovarian Cystadenomas. Front Oncol 2020; 10:895. [PMID: 32547958 PMCID: PMC7277787 DOI: 10.3389/fonc.2020.00895] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/06/2020] [Indexed: 12/26/2022] Open
Abstract
Objective: To construct and validate a combined Nomogram model based on radiomic and semantic features to preoperatively classify serous and mucinous pathological types in patients with ovarian cystadenoma. Methods: A total of 103 patients with pathology-confirmed ovarian cystadenoma who underwent CT examination were collected from two institutions. All cases divided into training cohort (N = 73) and external validation cohort (N = 30). The CT semantic features were identified by two abdominal radiologists. The preprocessed initial CT images were used for CT radiomic features extraction. The LASSO regression were applied to identify optimal radiomic features and construct the Radscore. A Nomogram model was constructed combining the Radscore and the optimal semantic feature. The model performance was evaluated by ROC analysis, calibration curve and decision curve analysis (DCA). Result: Five optimal features were ultimately selected and contributed to the Radscore construction. Unilocular/multilocular identification was significant difference from semantic features. The Nomogram model showed a better performance in both training cohort (AUC = 0.94, 95%CI 0.86–0.98) and external validation cohort (AUC = 0.92, 95%CI 0.76–0.98). The calibration curve and DCA analysis indicated a better accuracy of the Nomogram model for classification than either Radscore or the loculus alone. Conclusion: The Nomogram model combined radiomic and semantic features could be used as imaging biomarker for classification of serous and mucinous types of ovarian cystadenomas.
Collapse
Affiliation(s)
- Shushu Pan
- Department of Radiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhongxiang Ding
- Department of Radiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lexing Zhang
- Department of Radiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mei Ruan
- Department of Radiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanna Shan
- Department of Radiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Meixiang Deng
- Department of Radiology, Women's Hospital School of Medicine Zhejiang University, Hangzhou, China
| | - Peipei Pang
- Department of Pharmaceuticals Diagnosis, GE Healthcare, Hangzhou, China
| | - Qijun Shen
- Department of Radiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
27
|
Dohan A, Gallix B, Guiu B, Le Malicot K, Reinhold C, Soyer P, Bennouna J, Ghiringhelli F, Barbier E, Boige V, Taieb J, Bouché O, François E, Phelip JM, Borel C, Faroux R, Seitz JF, Jacquot S, Ben Abdelghani M, Khemissa-Akouz F, Genet D, Jouve JL, Rinaldi Y, Desseigne F, Texereau P, Suc E, Lepage C, Aparicio T, Hoeffel C. Early evaluation using a radiomic signature of unresectable hepatic metastases to predict outcome in patients with colorectal cancer treated with FOLFIRI and bevacizumab. Gut 2020; 69:531-539. [PMID: 31101691 DOI: 10.1136/gutjnl-2018-316407] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 04/28/2019] [Accepted: 04/30/2019] [Indexed: 12/12/2022]
Abstract
PURPOSE The objective of this study was to build and validate a radiomic signature to predict early a poor outcome using baseline and 2-month evaluation CT and to compare it to the RECIST1·1 and morphological criteria defined by changes in homogeneity and borders. METHODS This study is an ancillary study from the PRODIGE-9 multicentre prospective study for which 491 patients with metastatic colorectal cancer (mCRC) treated by 5-fluorouracil, leucovorin and irinotecan (FOLFIRI) and bevacizumab had been analysed. In 230 patients, computed texture analysis was performed on the dominant liver lesion (DLL) at baseline and 2 months after chemotherapy. RECIST1·1 evaluation was performed at 6 months. A radiomic signature (Survival PrEdiction in patients treated by FOLFIRI and bevacizumab for mCRC using contrast-enhanced CT TextuRe Analysis (SPECTRA) Score) combining the significant predictive features was built using multivariable Cox analysis in 120 patients, then locked, and validated in 110 patients. Overall survival (OS) was estimated with the Kaplan-Meier method and compared between groups with the logrank test. An external validation was performed in another cohort of 40 patients from the PRODIGE 20 Trial. RESULTS In the training cohort, the significant predictive features for OS were: decrease in sum of the target liver lesions (STL), (adjusted hasard-ratio(aHR)=13·7, p=1·93×10-7), decrease in kurtosis (ssf=4) (aHR=1·08, p=0·001) and high baseline density of DLL, (aHR=0·98, p<0·001). Patients with a SPECTRA Score >0·02 had a lower OS in the training cohort (p<0·0001), in the validation cohort (p<0·0008) and in the external validation cohort (p=0·0027). SPECTRA Score at 2 months had the same prognostic value as RECIST at 6 months, while non-response according to RECIST1·1 at 2 months was not associated with a lower OS in the validation cohort (p=0·238). Morphological response was not associated with OS (p=0·41). CONCLUSION A radiomic signature (combining decrease in STL, density and computed texture analysis of the DLL) at baseline and 2-month CT was able to predict OS, and identify good responders better than RECIST1.1 criteria in patients with mCRC treated by FOLFIRI and bevacizumab as a first-line treatment. This tool should now be validated by further prospective studies. TRIAL REGISTRATION Clinicaltrial.gov identifier of the PRODIGE 9 study: NCT00952029.Clinicaltrial.gov identifier of the PRODIGE 20 study: NCT01900717.
Collapse
Affiliation(s)
- Anthony Dohan
- Radiologie A, Assistance Publique - Hôpitaux de Paris, Cochin Hospital, Paris, France.,Medical School, Université de Paris, Paris, France.,Radiology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Benoit Gallix
- Radiology, McGill University Health Centre, Montreal, Quebec, Canada.,IRCAD, Institut Hospitalo-Universitaire, Strasbourg, France.,Medical School, Université de Strasbourg, Strasbourg, France
| | - Boris Guiu
- Radiology, Hopital Saint-Eloi, Montpellier, Languedoc-Roussillon, France.,Medical School, Université de Montpellier, Montpellier, France
| | - Karine Le Malicot
- Biostatistics, FFCD, Dijon, France.,EPICAD, INSERM LNC-UMR 1231, University of Burgundy and Franche-Comté, Dijon, France
| | - Caroline Reinhold
- Radiology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Philippe Soyer
- Radiologie A, Assistance Publique - Hôpitaux de Paris, Cochin Hospital, Paris, France.,Medical School, Université de Paris, Paris, France
| | - Jaafar Bennouna
- Gastroenterology and Digestive Oncology, Centre Hospitalier Universitaire de Nantes, Nantes, Pays de la Loire, France
| | | | - Emilie Barbier
- Biostatistics, FFCD, Dijon, France.,EPICAD, INSERM LNC-UMR 1231, University of Burgundy and Franche-Comté, Dijon, France
| | - Valérie Boige
- Oncologic Medicine, Institut Gustave Roussy, Villejuif, France
| | - Julien Taieb
- Medical School, Université de Paris, Paris, France.,Hepatogastroenterology and GI Oncology, Assistance Publique - Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France
| | - Olivier Bouché
- Gastrointestinal Oncology Unit, CHU Reims, Reims, France
| | - Eric François
- Pôle de Médecine, Centre Antoine-Lacassagne, Nice, France
| | - Jean-Marc Phelip
- Hepatogastroenterology, Saint Etienne University Hospital, Hôpital Nord, Saint Priest en Jarez, France
| | | | - Roger Faroux
- Gastroenterology, Hospital of La Roche sur Yon, La Roche sur Yon, France
| | - Jean-Francois Seitz
- Hepatogastroenterology and Oncology, Hopital de la Timone, Marseille, Provence-Alpes-Côte d'Azu, France
| | - Stéphane Jacquot
- Oncology, Centre de Cancérologie du Grand Montpellier, Montpellier, France
| | | | | | - Dominique Genet
- Medical Oncology, Clinique Francois Chenieux, Limoges, France
| | - Jean Louis Jouve
- Hepatogastroenterology, University Hospital Le Bocage, Dijon, France
| | - Yves Rinaldi
- Digestive Oncology, Hopital Européen, Marseilles, France
| | | | - Patrick Texereau
- Gastroenterology, Centre Hospitalier de Mont-de-Marsan, Mont-de-Marsan, Aquitaine, France
| | - Etienne Suc
- Medical oncology, Clinique Saint Jean de Languedoc, Toulouse, Midi-Pyrénées, France
| | - Come Lepage
- EPICAD, INSERM LNC-UMR 1231, University of Burgundy and Franche-Comté, Dijon, France.,Hepatogastroenterology, University Hospital Le Bocage, Dijon, France
| | - Thomas Aparicio
- Medical School, Université de Paris, Paris, France.,Gastroenterology and Digestive Oncology Department, Assistance Publique - Hôpitaux de Paris, Saint-Louis Hospital, Paris, France
| | - Christine Hoeffel
- Radiology, Hopital Maison Blanche, Reims, Champagne-Ardenne, France.,CRESTIC, Université de Reims, Reims, URCA, France
| | | |
Collapse
|
28
|
Lo Gullo R, Daimiel I, Morris EA, Pinker K. Combining molecular and imaging metrics in cancer: radiogenomics. Insights Imaging 2020; 11:1. [PMID: 31901171 PMCID: PMC6942081 DOI: 10.1186/s13244-019-0795-6] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 09/25/2019] [Indexed: 02/07/2023] Open
Abstract
Background Radiogenomics is the extension of radiomics through the combination of genetic and radiomic data. Because genetic testing remains expensive, invasive, and time-consuming, and thus unavailable for all patients, radiogenomics may play an important role in providing accurate imaging surrogates which are correlated with genetic expression, thereby serving as a substitute for genetic testing. Main body In this article, we define the meaning of radiogenomics and the difference between radiomics and radiogenomics. We provide an up-to-date review of the radiomics and radiogenomics literature in oncology, focusing on breast, brain, gynecological, liver, kidney, prostate and lung malignancies. We also discuss the current challenges to radiogenomics analysis. Conclusion Radiomics and radiogenomics are promising to increase precision in diagnosis, assessment of prognosis, and prediction of treatment response, providing valuable information for patient care throughout the course of the disease, given that this information is easily obtainable with imaging. Larger prospective studies and standardization will be needed to define relevant imaging biomarkers before they can be implemented into the clinical workflow.
Collapse
Affiliation(s)
- Roberto Lo Gullo
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, 300 E 66th St, New York, NY, 10065, USA.
| | - Isaac Daimiel
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, 300 E 66th St, New York, NY, 10065, USA
| | - Elizabeth A Morris
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, 300 E 66th St, New York, NY, 10065, USA
| | - Katja Pinker
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, 300 E 66th St, New York, NY, 10065, USA.,Department of Biomedical Imaging and Image-guided Therapy, Molecular and Gender Imaging Service, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Wien, Austria
| |
Collapse
|
29
|
Himoto Y, Cybulska P, Shitano F, Sala E, Zheng J, Capanu M, Nougaret S, Nikolovski I, Vargas HA, Wang W, Mueller JJ, Chi DS, Lakhman Y. Does the method of primary treatment affect the pattern of first recurrence in high-grade serous ovarian cancer? Gynecol Oncol 2019; 155:192-200. [PMID: 31521322 PMCID: PMC6837278 DOI: 10.1016/j.ygyno.2019.08.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 08/10/2019] [Accepted: 08/11/2019] [Indexed: 01/26/2023]
Abstract
PURPOSE To determine if the primary treatment approach (primary debulking surgery (PDS) versus neoadjuvant chemotherapy and interval debulking surgery (NACT-IDS)) influences the pattern of first recurrence in patients with completely cytoreduced advanced high-grade serous ovarian carcinoma (HGSOC). MATERIALS AND METHODS This retrospective study included 178 patients with newly diagnosed stage IIIC-IV HGSOC, complete gross resection during PDS (n = 124) or IDS (n = 54) from January 2008-March 2013, and baseline and first recurrence contrast-enhanced computed tomography scans. Clinical characteristics and number of disease sites at baseline were analyzed for associations with time to recurrence. In 135 patients who experienced recurrence, the overlap in disease locations between baseline and recurrence and the number of new disease locations at recurrence were analyzed according to the primary treatment approach. RESULTS At univariate and multivariate analyses, NACT-IDS was associated with more overlapping locations between baseline and first recurrence (p ≤ 0.003) and fewer recurrences in new anatomic locations (p ≤ 0.043) compared with PDS. The same results were found in a subgroup that received intra-peritoneal adjuvant chemotherapy after either treatment approach. At univariate analysis, patient age, primary treatment approach, adjuvant chemotherapy route, and number of disease locations at baseline were associated with time to recurrence (p ≤ 0.009). At multivariate analysis, older patient age, NACT-IDS, and greater disease locations at baseline remained significant (p ≤ 0.018). CONCLUSION The distribution of disease at the time of first recurrence varied with the choice of primary treatment. Compared to patients treated with PDS, patients who underwent NACT-IDS experienced recurrence more often in the same locations as the original disease.
Collapse
Affiliation(s)
- Yuki Himoto
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Paulina Cybulska
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Fuki Shitano
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Evis Sala
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Junting Zheng
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Marinela Capanu
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Stephanie Nougaret
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ines Nikolovski
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Hebert A Vargas
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Wei Wang
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jennifer J Mueller
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Dennis S Chi
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yulia Lakhman
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
30
|
Lu H, Arshad M, Thornton A, Avesani G, Cunnea P, Curry E, Kanavati F, Liang J, Nixon K, Williams ST, Hassan MA, Bowtell DDL, Gabra H, Fotopoulou C, Rockall A, Aboagye EO. A mathematical-descriptor of tumor-mesoscopic-structure from computed-tomography images annotates prognostic- and molecular-phenotypes of epithelial ovarian cancer. Nat Commun 2019; 10:764. [PMID: 30770825 PMCID: PMC6377605 DOI: 10.1038/s41467-019-08718-9] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 01/24/2019] [Indexed: 12/11/2022] Open
Abstract
The five-year survival rate of epithelial ovarian cancer (EOC) is approximately 35-40% despite maximal treatment efforts, highlighting a need for stratification biomarkers for personalized treatment. Here we extract 657 quantitative mathematical descriptors from the preoperative CT images of 364 EOC patients at their initial presentation. Using machine learning, we derive a non-invasive summary-statistic of the primary ovarian tumor based on 4 descriptors, which we name "Radiomic Prognostic Vector" (RPV). RPV reliably identifies the 5% of patients with median overall survival less than 2 years, significantly improves established prognostic methods, and is validated in two independent, multi-center cohorts. Furthermore, genetic, transcriptomic and proteomic analysis from two independent datasets elucidate that stromal phenotype and DNA damage response pathways are activated in RPV-stratified tumors. RPV and its associated analysis platform could be exploited to guide personalized therapy of EOC and is potentially transferrable to other cancer types.
Collapse
Affiliation(s)
- Haonan Lu
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
- Cancer Imaging Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Mubarik Arshad
- Cancer Imaging Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Andrew Thornton
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Giacomo Avesani
- Cancer Imaging Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Paula Cunnea
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Ed Curry
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Fahdi Kanavati
- Cancer Imaging Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Jack Liang
- Cancer Imaging Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Katherine Nixon
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Sophie T Williams
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Mona Ali Hassan
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - David D L Bowtell
- Peter MacCallum Cancer Centre, Melbourne, 3010, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, 3010, VIC, Australia
| | - Hani Gabra
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
- Early Clinical Development, iMED Biotech Unit, AstraZeneca, Cambridge, SG8 6HB, UK
| | - Christina Fotopoulou
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Andrea Rockall
- Cancer Imaging Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
- Department of Radiology, Imperial College Healthcare NHS Trust, London, W12 0HS, UK
- Department of Radiology, The Royal Marsden NHS Foundation Trust, London, SW3 6JJ, UK
| | - Eric O Aboagye
- Cancer Imaging Centre, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, W12 0HS, UK.
| |
Collapse
|
31
|
Elsherif SB, Faria SC, Lall C, Iyer R, Bhosale PR. Ovarian Cancer Genetics and Implications for Imaging and Therapy. J Comput Assist Tomogr 2019; 43:835-845. [DOI: 10.1097/rct.0000000000000932] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
32
|
Rastogi A, Maheshwari S, Shinagare AB, Baheti AD. Computed Tomography Advances in Oncoimaging. Semin Roentgenol 2018; 53:147-156. [PMID: 29861006 DOI: 10.1053/j.ro.2018.02.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Ashita Rastogi
- Department of Radiodiagnosis, Tata Memorial Centre, Mumbai, India
| | - Sharad Maheshwari
- Department of Radiology, Kokilaben Dhirubhai Ambani Hospital, Mumbai, India
| | - Atul B Shinagare
- Department of Radiology, Harvard Medical School, Dana-Farber Cancer Institute, Boston, MA
| | - Akshay D Baheti
- Department of Radiodiagnosis, Tata Memorial Centre, Mumbai, India.
| |
Collapse
|
33
|
Jansen RW, van Amstel P, Martens RM, Kooi IE, Wesseling P, de Langen AJ, Menke-Van der Houven van Oordt CW, Jansen BHE, Moll AC, Dorsman JC, Castelijns JA, de Graaf P, de Jong MC. Non-invasive tumor genotyping using radiogenomic biomarkers, a systematic review and oncology-wide pathway analysis. Oncotarget 2018; 9:20134-20155. [PMID: 29732009 PMCID: PMC5929452 DOI: 10.18632/oncotarget.24893] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 02/26/2018] [Indexed: 12/12/2022] Open
Abstract
With targeted treatments playing an increasing role in oncology, the need arises for fast non-invasive genotyping in clinical practice. Radiogenomics is a rapidly evolving field of research aimed at identifying imaging biomarkers useful for non-invasive genotyping. Radiogenomic genotyping has the advantage that it can capture tumor heterogeneity, can be performed repeatedly for treatment monitoring, and can be performed in malignancies for which biopsy is not available. In this systematic review of 187 included articles, we compiled a database of radiogenomic associations and unraveled networks of imaging groups and gene pathways oncology-wide. Results indicated that ill-defined tumor margins and tumor heterogeneity can potentially be used as imaging biomarkers for 1p/19q codeletion in glioma, relevant for prognosis and disease profiling. In non-small cell lung cancer, FDG-PET uptake and CT-ground-glass-opacity features were associated with treatment-informing traits including EGFR-mutations and ALK-rearrangements. Oncology-wide gene pathway analysis revealed an association between contrast enhancement (imaging) and the targetable VEGF-signalling pathway. Although the need of independent validation remains a concern, radiogenomic biomarkers showed potential for prognosis prediction and targeted treatment selection. Quantitative imaging enhanced the potential of multiparametric radiogenomic models. A wealth of data has been compiled for guiding future research towards robust non-invasive genomic profiling.
Collapse
Affiliation(s)
- Robin W Jansen
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Paul van Amstel
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Roland M Martens
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Irsan E Kooi
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands
| | - Pieter Wesseling
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands.,Department of Pathology, Princess Máxima Center for Pediatric Oncology and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Adrianus J de Langen
- Department of Respiratory Diseases, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Bernard H E Jansen
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Annette C Moll
- Department of Ophthalmology, VU University Medical Center, Amsterdam, The Netherlands
| | - Josephine C Dorsman
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands
| | - Jonas A Castelijns
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Pim de Graaf
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Marcus C de Jong
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
34
|
Pinker K, Shitano F, Sala E, Do RK, Young RJ, Wibmer AG, Hricak H, Sutton EJ, Morris EA. Background, current role, and potential applications of radiogenomics. J Magn Reson Imaging 2017; 47:604-620. [PMID: 29095543 DOI: 10.1002/jmri.25870] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Revised: 09/17/2017] [Accepted: 09/19/2017] [Indexed: 12/17/2022] Open
Abstract
With the genomic revolution in the early 1990s, medical research has been driven to study the basis of human disease on a genomic level and to devise precise cancer therapies tailored to the specific genetic makeup of a tumor. To match novel therapeutic concepts conceived in the era of precision medicine, diagnostic tests must be equally sufficient, multilayered, and complex to identify the relevant genetic alterations that render cancers susceptible to treatment. With significant advances in training and medical imaging techniques, image analysis and the development of high-throughput methods to extract and correlate multiple imaging parameters with genomic data, a new direction in medical research has emerged. This novel approach has been termed radiogenomics. Radiogenomics aims to correlate imaging characteristics (ie, the imaging phenotype) with gene expression patterns, gene mutations, and other genome-related characteristics and is designed to facilitate a deeper understanding of tumor biology and capture the intrinsic tumor heterogeneity. Ultimately, the goal of radiogenomics is to develop imaging biomarkers for outcome that incorporate both phenotypic and genotypic metrics. Due to the noninvasive nature of medical imaging and its ubiquitous use in clinical practice, the field of radiogenomics is rapidly evolving and initial results are encouraging. In this article, we briefly discuss the background and then summarize the current role and the potential of radiogenomics in brain, liver, prostate, gynecological, and breast tumors. LEVEL OF EVIDENCE 5 Technical Efficacy: Stage 2 J. Magn. Reson. Imaging 2017;47:604-620.
Collapse
Affiliation(s)
- Katja Pinker
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Department of Biomedical Imaging and Image-guided Therapy, Division of Molecular and Gender Imaging, Medical University of Vienna, Austria
| | - Fuki Shitano
- Department of Radiology, Body Imaging Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Evis Sala
- Department of Radiology, Body Imaging Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Richard K Do
- Department of Radiology, Body Imaging Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Robert J Young
- Department of Radiology, Neuroradiology Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Andreas G Wibmer
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Hedvig Hricak
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Elizabeth J Sutton
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Elizabeth A Morris
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
35
|
Vargas HA, Veeraraghavan H, Micco M, Nougaret S, Lakhman Y, Meier AA, Sosa R, Soslow RA, Levine DA, Weigelt B, Aghajanian C, Hricak H, Deasy J, Snyder A, Sala E. A novel representation of inter-site tumour heterogeneity from pre-treatment computed tomography textures classifies ovarian cancers by clinical outcome. Eur Radiol 2017; 27:3991-4001. [PMID: 28289945 PMCID: PMC5545058 DOI: 10.1007/s00330-017-4779-y] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 01/17/2017] [Accepted: 02/14/2017] [Indexed: 02/06/2023]
Abstract
PURPOSE To evaluate the associations between clinical outcomes and radiomics-derived inter-site spatial heterogeneity metrics across multiple metastatic lesions on CT in patients with high-grade serous ovarian cancer (HGSOC). METHODS IRB-approved retrospective study of 38 HGSOC patients. All sites of suspected HGSOC involvement on preoperative CT were manually segmented. Gray-level correlation matrix-based textures were computed from each tumour site, and grouped into five clusters using a Gaussian Mixture Model. Pairwise inter-site similarities were computed, generating an inter-site similarity matrix (ISM). Inter-site texture heterogeneity metrics were computed from the ISM and compared to clinical outcomes. RESULTS Of the 12 inter-site texture heterogeneity metrics evaluated, those capturing the differences in texture similarities across sites were associated with shorter overall survival (inter-site similarity entropy, similarity level cluster shade, and inter-site similarity level cluster prominence; p ≤ 0.05) and incomplete surgical resection (similarity level cluster shade, inter-site similarity level cluster prominence and inter-site cluster variance; p ≤ 0.05). Neither the total number of disease sites per patient nor the overall tumour volume per patient was associated with overall survival. Amplification of 19q12 involving cyclin E1 gene (CCNE1) predominantly occurred in patients with more heterogeneous inter-site textures. CONCLUSION Quantitative metrics non-invasively capturing spatial inter-site heterogeneity may predict outcomes in patients with HGSOC. KEY POINTS • Calculating inter-site texture-based heterogeneity metrics was feasible • Metrics capturing texture similarities across HGSOC sites were associated with overall survival • Heterogeneity metrics were also associated with incomplete surgical resection of HGSOC.
Collapse
Affiliation(s)
- Hebert Alberto Vargas
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
| | - Harini Veeraraghavan
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Maura Micco
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Stephanie Nougaret
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
- Service de Radiologie, Institut Régional du Cancer de Montpellier, Montpellier, France
- INSERM, U1194, Institut de Recherche en Cancérologie de Montpellier (IRCM), Montpellier, France
| | - Yulia Lakhman
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Andreas A Meier
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Ramon Sosa
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Robert A Soslow
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Douglas A Levine
- Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Carol Aghajanian
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Hedvig Hricak
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Joseph Deasy
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Alexandra Snyder
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Evis Sala
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| |
Collapse
|
36
|
Ohsuga T, Yamaguchi K, Kido A, Murakami R, Abiko K, Hamanishi J, Kondoh E, Baba T, Konishi I, Matsumura N. Distinct preoperative clinical features predict four histopathological subtypes of high-grade serous carcinoma of the ovary, fallopian tube, and peritoneum. BMC Cancer 2017; 17:580. [PMID: 28851311 PMCID: PMC5576247 DOI: 10.1186/s12885-017-3573-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 08/21/2017] [Indexed: 11/10/2022] Open
Abstract
Background The Cancer Genome Atlas Research Network reported that high-grade serous carcinoma (HGSC) can be classified based on gene expression profiles into four subtypes, termed “immunoreactive,” “differentiated,” “proliferative,” and “mesenchymal.” We previously established a novel histopathological classification of HGSC, corresponding to the gene expression subtypes: immune reactive (IR), papillo-glandular (PG), solid and proliferative (SP), and mesenchymal transition (MT). The purpose of this study is to identify distinct clinical findings among the four pathological subtypes of HGSC, as well as to predict pathological subtype based on preoperative images. Methods We retrospectively assessed 65 HGSC cases (IR: 17, PG: 7, SP: 14, MT: 27) and analyzed preoperative images. Results All IR cases originated from either the ovary or fallopian tube (P = 0.0269). Significantly more IR cases were diagnosed at earlier stages (P = 0.0013), and IR cases displayed lower levels of ascites (P = 0.0014), fewer peritoneal lesions (P = 0.0080), a sporadic pattern of peritoneal lesions (P = 0.0016), a lower incidence of omental cake (P = 0.0416), and fewer distant metastases (P = 0.0146) compared with the other subtypes. MT cases were more likely to be of peritoneal origin (P = 0.0202), presented at advanced stages with higher levels of ascites (P = 0.0008, 0.0052, respectively), and more frequently had a diffuse pattern of peritoneal lesions (P = 0.0059), omental cake (P = 0.0179), and distant metastasis (P = 0.0053). A decision tree analysis estimated the histopathological subtypes based on preoperative images, with a sensitivity of 67.3%. Conclusions Pathological subtypes of HGSC have distinct clinical behaviors, and preoperative images enable better prediction of pathological subtype. These findings may lead to individualized treatment plans if the effect of treatment based on the HGSC subtype is elucidated. Electronic supplementary material The online version of this article (10.1186/s12885-017-3573-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Takuma Ohsuga
- Department of Gynecology and Obstetrics, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Ken Yamaguchi
- Department of Gynecology and Obstetrics, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Aki Kido
- Department of Diagnostic Imaging and Nuclear Medicine, Kyoto University, Kyoto, Japan
| | - Ryusuke Murakami
- Department of Gynecology and Obstetrics, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kaoru Abiko
- Department of Gynecology and Obstetrics, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Junzo Hamanishi
- Department of Gynecology and Obstetrics, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Eiji Kondoh
- Department of Gynecology and Obstetrics, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Tsukasa Baba
- Department of Gynecology and Obstetrics, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Ikuo Konishi
- Department of Gynecology and Obstetrics, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.,National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Noriomi Matsumura
- Department of Gynecology and Obstetrics, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| |
Collapse
|
37
|
Vargas HA, Huang EP, Lakhman Y, Ippolito JE, Bhosale P, Mellnick V, Shinagare AB, Anello M, Kirby J, Fevrier-Sullivan B, Freymann J, Jaffe CC, Sala E. Radiogenomics of High-Grade Serous Ovarian Cancer: Multireader Multi-Institutional Study from the Cancer Genome Atlas Ovarian Cancer Imaging Research Group. Radiology 2017. [PMID: 28641043 DOI: 10.1148/radiol.2017161870] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Purpose To evaluate interradiologist agreement on assessments of computed tomography (CT) imaging features of high-grade serous ovarian cancer (HGSOC), to assess their associations with time-to-disease progression (TTP) and HGSOC transcriptomic profiles (Classification of Ovarian Cancer [CLOVAR]), and to develop an imaging-based risk score system to predict TTP and CLOVAR profiles. Materials and Methods This study was a multireader, multi-institutional, institutional review board-approved, HIPAA-compliant retrospective analysis of 92 patients with HGSOC (median age, 61 years) with abdominopelvic CT before primary cytoreductive surgery available through the Cancer Imaging Archive. Eight radiologists from the Cancer Genome Atlas Ovarian Cancer Imaging Research Group developed and independently recorded the following CT features: characteristics of primary ovarian mass(es), presence of definable mesenteric implants and infiltration, presence of other implants, presence and distribution of peritoneal spread, presence and size of pleural effusions and ascites, lymphadenopathy, and distant metastases. Interobserver agreement for CT features was assessed, as were univariate and multivariate associations with TTP and CLOVAR mesenchymal profile (worst prognosis). Results Interobserver agreement for some features was strong (eg, α = .78 for pleural effusion and ascites) but was lower for others (eg, α = .08 for intraparenchymal splenic metastases). Presence of peritoneal disease in the right upper quadrant (P = .0003), supradiaphragmatic lymphadenopathy (P = .0004), more peritoneal disease sites (P = .0006), and nonvisualization of a discrete ovarian mass (P = .0037) were associated with shorter TTP. More peritoneal disease sites (P = .0025) and presence of pouch of Douglas implants (P = .0045) were associated with CLOVAR mesenchymal profile. Combinations of imaging features contained predictive signal for TTP (concordance index = 0.658; P = .0006) and CLOVAR profile (mean squared deviation = 1.776; P = .0043). Conclusion These results provide some evidence of the clinical and biologic validity of these image features. Interobserver agreement is strong for some features, but could be improved for others. © RSNA, 2017 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Hebert Alberto Vargas
- From the Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, Room C278, New York, NY 10065 (H.A.V., Y.L., E.S.); Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, Md (E.P.H.); Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Mo (J.E.I., V.M.); Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center, Houston, Tex (P.B.); Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, Boston, Mass (A.S.); Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.S.); Department of Radiology, Magee-Womens Hospital of the University of Pittsburgh Medical Center, Pittsburgh, Pa (M.A.); Leidos Biomedical Research, National Cancer Institute, National Institutes of Health, Frederick, Md (J.K., B.F.S., J.F.); and Department of Radiology, Boston University School of Medicine, Boston, Mass (C.C.J.)
| | - Erich P Huang
- From the Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, Room C278, New York, NY 10065 (H.A.V., Y.L., E.S.); Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, Md (E.P.H.); Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Mo (J.E.I., V.M.); Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center, Houston, Tex (P.B.); Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, Boston, Mass (A.S.); Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.S.); Department of Radiology, Magee-Womens Hospital of the University of Pittsburgh Medical Center, Pittsburgh, Pa (M.A.); Leidos Biomedical Research, National Cancer Institute, National Institutes of Health, Frederick, Md (J.K., B.F.S., J.F.); and Department of Radiology, Boston University School of Medicine, Boston, Mass (C.C.J.)
| | - Yulia Lakhman
- From the Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, Room C278, New York, NY 10065 (H.A.V., Y.L., E.S.); Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, Md (E.P.H.); Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Mo (J.E.I., V.M.); Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center, Houston, Tex (P.B.); Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, Boston, Mass (A.S.); Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.S.); Department of Radiology, Magee-Womens Hospital of the University of Pittsburgh Medical Center, Pittsburgh, Pa (M.A.); Leidos Biomedical Research, National Cancer Institute, National Institutes of Health, Frederick, Md (J.K., B.F.S., J.F.); and Department of Radiology, Boston University School of Medicine, Boston, Mass (C.C.J.)
| | - Joseph E Ippolito
- From the Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, Room C278, New York, NY 10065 (H.A.V., Y.L., E.S.); Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, Md (E.P.H.); Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Mo (J.E.I., V.M.); Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center, Houston, Tex (P.B.); Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, Boston, Mass (A.S.); Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.S.); Department of Radiology, Magee-Womens Hospital of the University of Pittsburgh Medical Center, Pittsburgh, Pa (M.A.); Leidos Biomedical Research, National Cancer Institute, National Institutes of Health, Frederick, Md (J.K., B.F.S., J.F.); and Department of Radiology, Boston University School of Medicine, Boston, Mass (C.C.J.)
| | - Priya Bhosale
- From the Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, Room C278, New York, NY 10065 (H.A.V., Y.L., E.S.); Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, Md (E.P.H.); Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Mo (J.E.I., V.M.); Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center, Houston, Tex (P.B.); Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, Boston, Mass (A.S.); Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.S.); Department of Radiology, Magee-Womens Hospital of the University of Pittsburgh Medical Center, Pittsburgh, Pa (M.A.); Leidos Biomedical Research, National Cancer Institute, National Institutes of Health, Frederick, Md (J.K., B.F.S., J.F.); and Department of Radiology, Boston University School of Medicine, Boston, Mass (C.C.J.)
| | - Vincent Mellnick
- From the Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, Room C278, New York, NY 10065 (H.A.V., Y.L., E.S.); Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, Md (E.P.H.); Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Mo (J.E.I., V.M.); Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center, Houston, Tex (P.B.); Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, Boston, Mass (A.S.); Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.S.); Department of Radiology, Magee-Womens Hospital of the University of Pittsburgh Medical Center, Pittsburgh, Pa (M.A.); Leidos Biomedical Research, National Cancer Institute, National Institutes of Health, Frederick, Md (J.K., B.F.S., J.F.); and Department of Radiology, Boston University School of Medicine, Boston, Mass (C.C.J.)
| | - Atul B Shinagare
- From the Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, Room C278, New York, NY 10065 (H.A.V., Y.L., E.S.); Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, Md (E.P.H.); Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Mo (J.E.I., V.M.); Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center, Houston, Tex (P.B.); Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, Boston, Mass (A.S.); Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.S.); Department of Radiology, Magee-Womens Hospital of the University of Pittsburgh Medical Center, Pittsburgh, Pa (M.A.); Leidos Biomedical Research, National Cancer Institute, National Institutes of Health, Frederick, Md (J.K., B.F.S., J.F.); and Department of Radiology, Boston University School of Medicine, Boston, Mass (C.C.J.)
| | - Maria Anello
- From the Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, Room C278, New York, NY 10065 (H.A.V., Y.L., E.S.); Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, Md (E.P.H.); Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Mo (J.E.I., V.M.); Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center, Houston, Tex (P.B.); Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, Boston, Mass (A.S.); Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.S.); Department of Radiology, Magee-Womens Hospital of the University of Pittsburgh Medical Center, Pittsburgh, Pa (M.A.); Leidos Biomedical Research, National Cancer Institute, National Institutes of Health, Frederick, Md (J.K., B.F.S., J.F.); and Department of Radiology, Boston University School of Medicine, Boston, Mass (C.C.J.)
| | - Justin Kirby
- From the Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, Room C278, New York, NY 10065 (H.A.V., Y.L., E.S.); Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, Md (E.P.H.); Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Mo (J.E.I., V.M.); Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center, Houston, Tex (P.B.); Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, Boston, Mass (A.S.); Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.S.); Department of Radiology, Magee-Womens Hospital of the University of Pittsburgh Medical Center, Pittsburgh, Pa (M.A.); Leidos Biomedical Research, National Cancer Institute, National Institutes of Health, Frederick, Md (J.K., B.F.S., J.F.); and Department of Radiology, Boston University School of Medicine, Boston, Mass (C.C.J.)
| | - Brenda Fevrier-Sullivan
- From the Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, Room C278, New York, NY 10065 (H.A.V., Y.L., E.S.); Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, Md (E.P.H.); Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Mo (J.E.I., V.M.); Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center, Houston, Tex (P.B.); Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, Boston, Mass (A.S.); Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.S.); Department of Radiology, Magee-Womens Hospital of the University of Pittsburgh Medical Center, Pittsburgh, Pa (M.A.); Leidos Biomedical Research, National Cancer Institute, National Institutes of Health, Frederick, Md (J.K., B.F.S., J.F.); and Department of Radiology, Boston University School of Medicine, Boston, Mass (C.C.J.)
| | - John Freymann
- From the Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, Room C278, New York, NY 10065 (H.A.V., Y.L., E.S.); Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, Md (E.P.H.); Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Mo (J.E.I., V.M.); Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center, Houston, Tex (P.B.); Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, Boston, Mass (A.S.); Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.S.); Department of Radiology, Magee-Womens Hospital of the University of Pittsburgh Medical Center, Pittsburgh, Pa (M.A.); Leidos Biomedical Research, National Cancer Institute, National Institutes of Health, Frederick, Md (J.K., B.F.S., J.F.); and Department of Radiology, Boston University School of Medicine, Boston, Mass (C.C.J.)
| | - C Carl Jaffe
- From the Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, Room C278, New York, NY 10065 (H.A.V., Y.L., E.S.); Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, Md (E.P.H.); Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Mo (J.E.I., V.M.); Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center, Houston, Tex (P.B.); Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, Boston, Mass (A.S.); Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.S.); Department of Radiology, Magee-Womens Hospital of the University of Pittsburgh Medical Center, Pittsburgh, Pa (M.A.); Leidos Biomedical Research, National Cancer Institute, National Institutes of Health, Frederick, Md (J.K., B.F.S., J.F.); and Department of Radiology, Boston University School of Medicine, Boston, Mass (C.C.J.)
| | - Evis Sala
- From the Department of Radiology, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, Room C278, New York, NY 10065 (H.A.V., Y.L., E.S.); Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, Md (E.P.H.); Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Mo (J.E.I., V.M.); Department of Diagnostic Radiology, University of Texas MD Anderson Cancer Center, Houston, Tex (P.B.); Department of Imaging, Dana Farber Cancer Institute, Harvard Medical School, Boston, Mass (A.S.); Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.S.); Department of Radiology, Magee-Womens Hospital of the University of Pittsburgh Medical Center, Pittsburgh, Pa (M.A.); Leidos Biomedical Research, National Cancer Institute, National Institutes of Health, Frederick, Md (J.K., B.F.S., J.F.); and Department of Radiology, Boston University School of Medicine, Boston, Mass (C.C.J.)
| |
Collapse
|
38
|
Nougaret S, Lakhman Y, Gönen M, Goldman DA, Miccò M, D'Anastasi M, Johnson SA, Juluru K, Arnold AG, Sosa RE, Soslow RA, Vargas HA, Hricak H, Kauff ND, Sala E. High-Grade Serous Ovarian Cancer: Associations between BRCA Mutation Status, CT Imaging Phenotypes, and Clinical Outcomes. Radiology 2017. [PMID: 28628421 DOI: 10.1148/radiol.2017161697] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Purpose To investigate the associations between BRCA mutation status and computed tomography (CT) phenotypes of high-grade serous ovarian cancer (HGSOC) and to evaluate CT indicators of cytoreductive outcome and survival in patients with BRCA-mutant HGSOC and those with BRCA wild-type HGSOC. Materials and Methods This HIPAA-compliant, institutional review board-approved retrospective study included 108 patients (33 with BRCA mutant and 75 with BRCA wild-type HGSOC) who underwent CT before primary debulking. Two radiologists independently reviewed the CT findings for various qualitative CT features. Associations between CT features, BRCA mutation status, cytoreductive outcome, and progression-free survival (PFS) were evaluated by using logistic regression and Cox proportional hazards regression, respectively. Results Peritoneal disease (PD) pattern, presence of PD in gastrohepatic ligament, mesenteric involvement, and supradiaphragmatic lymphadenopathy at CT were associated with BRCA mutation status (multiple regression: P < .001 for each CT feature). While clinical and CT features were not associated with cytoreductive outcome for patients with BRCA-mutant HGSOC, presence of PD in lesser sac (odds ratio [OR] = 2.40) and left upper quadrant (OR = 1.19), mesenteric involvement (OR = 7.10), and lymphadenopathy in supradiaphragmatic (OR = 2.83) and suprarenal para-aortic (OR = 4.79) regions were associated with higher odds of incomplete cytoreduction in BRCA wild-type HGSOC (multiple regression: P < .001 each CT feature). Mesenteric involvement at CT was associated with significantly shorter PFS for both patients with BRCA-mutant HGSOC (multiple regression: hazard ratio [HR] = 26.7 P < .001) and those with BRCA wild-type HGSOC (univariate analysis: reader 1, HR = 2.42, P < .001; reader 2, HR = 2.61; P < .001). Conclusion Qualitative CT features differed between patients with BRCA-mutant HGSOC and patients with BRCA wild-type HGSOC. CT indicators of cytoreductive outcome varied according to BRCA mutation status. Mesenteric involvement at CT was an indicator of significantly shorter PFS for both patients with BRCA-mutant HGSOC and those with BRCA wild-type HGSOC. © RSNA, 2017 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Stephanie Nougaret
- From the Department of Radiology (S.N., Y.L., M.M., M.D., S.A.J., K.J., R.E.S., H.A.V., H.H., E.S.), Department of Epidemiology and Biostatistics (M.G., D.A.G.), Clinical Genetics Service, Department of Medicine (A.G.A., N.D.K.), and Department of Pathology (R.A.S.), Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Yulia Lakhman
- From the Department of Radiology (S.N., Y.L., M.M., M.D., S.A.J., K.J., R.E.S., H.A.V., H.H., E.S.), Department of Epidemiology and Biostatistics (M.G., D.A.G.), Clinical Genetics Service, Department of Medicine (A.G.A., N.D.K.), and Department of Pathology (R.A.S.), Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Mithat Gönen
- From the Department of Radiology (S.N., Y.L., M.M., M.D., S.A.J., K.J., R.E.S., H.A.V., H.H., E.S.), Department of Epidemiology and Biostatistics (M.G., D.A.G.), Clinical Genetics Service, Department of Medicine (A.G.A., N.D.K.), and Department of Pathology (R.A.S.), Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Debra A Goldman
- From the Department of Radiology (S.N., Y.L., M.M., M.D., S.A.J., K.J., R.E.S., H.A.V., H.H., E.S.), Department of Epidemiology and Biostatistics (M.G., D.A.G.), Clinical Genetics Service, Department of Medicine (A.G.A., N.D.K.), and Department of Pathology (R.A.S.), Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Maura Miccò
- From the Department of Radiology (S.N., Y.L., M.M., M.D., S.A.J., K.J., R.E.S., H.A.V., H.H., E.S.), Department of Epidemiology and Biostatistics (M.G., D.A.G.), Clinical Genetics Service, Department of Medicine (A.G.A., N.D.K.), and Department of Pathology (R.A.S.), Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Melvin D'Anastasi
- From the Department of Radiology (S.N., Y.L., M.M., M.D., S.A.J., K.J., R.E.S., H.A.V., H.H., E.S.), Department of Epidemiology and Biostatistics (M.G., D.A.G.), Clinical Genetics Service, Department of Medicine (A.G.A., N.D.K.), and Department of Pathology (R.A.S.), Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Sarah A Johnson
- From the Department of Radiology (S.N., Y.L., M.M., M.D., S.A.J., K.J., R.E.S., H.A.V., H.H., E.S.), Department of Epidemiology and Biostatistics (M.G., D.A.G.), Clinical Genetics Service, Department of Medicine (A.G.A., N.D.K.), and Department of Pathology (R.A.S.), Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Krishna Juluru
- From the Department of Radiology (S.N., Y.L., M.M., M.D., S.A.J., K.J., R.E.S., H.A.V., H.H., E.S.), Department of Epidemiology and Biostatistics (M.G., D.A.G.), Clinical Genetics Service, Department of Medicine (A.G.A., N.D.K.), and Department of Pathology (R.A.S.), Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Angela G Arnold
- From the Department of Radiology (S.N., Y.L., M.M., M.D., S.A.J., K.J., R.E.S., H.A.V., H.H., E.S.), Department of Epidemiology and Biostatistics (M.G., D.A.G.), Clinical Genetics Service, Department of Medicine (A.G.A., N.D.K.), and Department of Pathology (R.A.S.), Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Ramon E Sosa
- From the Department of Radiology (S.N., Y.L., M.M., M.D., S.A.J., K.J., R.E.S., H.A.V., H.H., E.S.), Department of Epidemiology and Biostatistics (M.G., D.A.G.), Clinical Genetics Service, Department of Medicine (A.G.A., N.D.K.), and Department of Pathology (R.A.S.), Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Robert A Soslow
- From the Department of Radiology (S.N., Y.L., M.M., M.D., S.A.J., K.J., R.E.S., H.A.V., H.H., E.S.), Department of Epidemiology and Biostatistics (M.G., D.A.G.), Clinical Genetics Service, Department of Medicine (A.G.A., N.D.K.), and Department of Pathology (R.A.S.), Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Hebert Alberto Vargas
- From the Department of Radiology (S.N., Y.L., M.M., M.D., S.A.J., K.J., R.E.S., H.A.V., H.H., E.S.), Department of Epidemiology and Biostatistics (M.G., D.A.G.), Clinical Genetics Service, Department of Medicine (A.G.A., N.D.K.), and Department of Pathology (R.A.S.), Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Hedvig Hricak
- From the Department of Radiology (S.N., Y.L., M.M., M.D., S.A.J., K.J., R.E.S., H.A.V., H.H., E.S.), Department of Epidemiology and Biostatistics (M.G., D.A.G.), Clinical Genetics Service, Department of Medicine (A.G.A., N.D.K.), and Department of Pathology (R.A.S.), Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Noah D Kauff
- From the Department of Radiology (S.N., Y.L., M.M., M.D., S.A.J., K.J., R.E.S., H.A.V., H.H., E.S.), Department of Epidemiology and Biostatistics (M.G., D.A.G.), Clinical Genetics Service, Department of Medicine (A.G.A., N.D.K.), and Department of Pathology (R.A.S.), Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Evis Sala
- From the Department of Radiology (S.N., Y.L., M.M., M.D., S.A.J., K.J., R.E.S., H.A.V., H.H., E.S.), Department of Epidemiology and Biostatistics (M.G., D.A.G.), Clinical Genetics Service, Department of Medicine (A.G.A., N.D.K.), and Department of Pathology (R.A.S.), Memorial Sloan-Kettering Cancer Center, New York, NY
| |
Collapse
|
39
|
Giardino A, Gupta S, Olson E, Sepulveda K, Lenchik L, Ivanidze J, Rakow-Penner R, Patel MJ, Subramaniam RM, Ganeshan D. Role of Imaging in the Era of Precision Medicine. Acad Radiol 2017; 24:639-649. [PMID: 28131497 DOI: 10.1016/j.acra.2016.11.021] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 11/07/2016] [Accepted: 11/29/2016] [Indexed: 12/17/2022]
Abstract
Precision medicine is an emerging approach for treating medical disorders, which takes into account individual variability in genetic and environmental factors. Preventive or therapeutic interventions can then be directed to those who will benefit most from targeted interventions, thereby maximizing benefits and minimizing costs and complications. Precision medicine is gaining increasing recognition by clinicians, healthcare systems, pharmaceutical companies, patients, and the government. Imaging plays a critical role in precision medicine including screening, early diagnosis, guiding treatment, evaluating response to therapy, and assessing likelihood of disease recurrence. The Association of University Radiologists Radiology Research Alliance Precision Imaging Task Force convened to explore the current and future role of imaging in the era of precision medicine and summarized its finding in this article. We review the increasingly important role of imaging in various oncological and non-oncological disorders. We also highlight the challenges for radiology in the era of precision medicine.
Collapse
Affiliation(s)
- Angela Giardino
- Department of Imaging, Dana-Farber Cancer Institute, Boston, Massachusetts; Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Supriya Gupta
- Department of Radiology and Imaging, Medical College of Georgia, 1120 15th St, Augusta, GA 30912.
| | - Emmi Olson
- Radiology Resident, University of California San Diego, San Diego, California
| | | | - Leon Lenchik
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Jana Ivanidze
- Department of Diagnostic Radiology, Weill Cornell Medicine, New York, New York
| | - Rebecca Rakow-Penner
- Department of Radiology, University of California San Diego, San Diego, California
| | - Midhir J Patel
- Department of Radiology, University of South Florida, Tampa, Florida
| | - Rathan M Subramaniam
- Cyclotron and Molecular Imaging Program, Department of Radiology, UT Southwestern Medical Center, Dallas, Texas
| | | |
Collapse
|
40
|
Wang R, Luo Y, Yang S, Lin J, Gao D, Zhao Y, Liu J, Shi X, Wang X. Hyaluronic acid-modified manganese-chelated dendrimer-entrapped gold nanoparticles for the targeted CT/MR dual-mode imaging of hepatocellular carcinoma. Sci Rep 2016; 6:33844. [PMID: 27653258 PMCID: PMC5032118 DOI: 10.1038/srep33844] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 09/02/2016] [Indexed: 12/18/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common malignant tumor of the liver. The early and effective diagnosis has always been desired. Herein, we present the preparation and characterization of hyaluronic acid (HA)-modified, multifunctional nanoparticles (NPs) targeting CD44 receptor-expressing cancer cells for computed tomography (CT)/magnetic resonance (MR) dual-mode imaging. We first modified amine-terminated generation 5 poly(amidoamine) dendrimers (G5.NH2) with an Mn chelator, 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA), fluorescein isothiocyanate (FI), and HA. Then, gold nanoparticles (AuNPs) were entrapped within the above raw product, denoted as G5.NH2-FI-DOTA-HA. The designed multifunctional NPs were formed after further Mn chelation and purification and were denoted as {(Au0)100G5.NH2-FI-DOTA(Mn)-HA}. These NPs were characterized via several different techniques. We found that the {(Au0)100G5.NH2-FI-DOTA(Mn)-HA} NPs exhibited good water dispersibility, stability under different conditions, and cytocompatibility within a given concentration range. Because both AuNPs and Mn were present in the product, {(Au0)100G5.NH2-FI-DOTA(Mn)-HA} displayed a high X-ray attenuation intensity and favorable r1 relaxivity, which are advantageous properties for targeted CT/MR dual-mode imaging. This approach was used to image HCC cells in vitro and orthotopically transplanted HCC tumors in a unique in vivo model through the CD44 receptor-mediated endocytosis pathway. This work introduces a novel strategy for preparing multifunctional NPs via dendrimer nanotechnology.
Collapse
Affiliation(s)
- Ruizhi Wang
- Shanghai Institute of Medical Imaging, Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Yu Luo
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, P. R. China
| | - Shuohui Yang
- Shanghai Institute of Medical Imaging, Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Jiang Lin
- Shanghai Institute of Medical Imaging, Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Dongmei Gao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Yan Zhao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Jinguo Liu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Xiangyang Shi
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, P. R. China
| | - Xiaolin Wang
- Shanghai Institute of Medical Imaging, Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai 200032, P. R. China
| |
Collapse
|
41
|
Invasion Patterns of Metastatic Extrauterine High-grade Serous Carcinoma With BRCA Germline Mutation and Correlation With Clinical Outcomes. Am J Surg Pathol 2016; 40:404-9. [PMID: 26574845 DOI: 10.1097/pas.0000000000000556] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Characteristic histopathologic features have been described in high-grade serous carcinoma associated with BRCA abnormalities (HGSC-BRCA), which are known to have relatively favorable clinical outcomes. The aim of this study was to evaluate the clinical significance of invasion patterns in metastatic HGSC-BRCA cases. Of the 37 cases of advanced-stage HGSC with known BRCA1 or BRCA2 germline mutation retrieved from our institutional files, 23 patients had a germline mutation of BRCA1 and 14 had a BRCA2 mutation. The pattern of invasion at metastatic sites was recorded and classified as a pushing pattern (either predominantly or exclusively), an exclusively micropapillary infiltrative pattern, or an infiltrative pattern composed of papillae, micropapillae, glands, and nests (mixed infiltrative pattern). Histologic evaluation of metastases was performed without knowledge of genotype or clinical outcome. Clinical data were abstracted from medical records. Median age was 56 years (range, 31 to 73 y). All patients presented at stage IIIC or IV and underwent complete surgical staging followed by chemotherapy. All 37 HGSC-BRCA cases showed either pushing pattern metastases (30; 81%) or infiltrative micropapillary metastases (7; 19%). No HGSC-BRCA case exhibited metastases composed solely of mixed infiltrative patterns. Among the 7 infiltrative micropapillary cases, 6 had a BRCA1 germline mutation versus 1 with a BRCA2 mutation. The median time of follow-up was 26 months (range, 13 to 49 mo). All 7 patients with infiltrative micropapillary metastases either experienced recurrence or died of disease (5 recurrences and 2 deaths), which was significantly worse than what was seen in patients with predominantly pushing pattern metastases, of whom 16 of 30 (53%) experienced recurrence (n=14) or died of disease (n=2) (P=0.03). In conclusion, the recognition of different invasion patterns of metastatic extrauterine HGSC-BRCA has prognostic implications. The infiltrative micropapillary pattern is associated with poor outcomes and is more frequently seen in BRCA1-associated HGSC than in BRCA2 cases.
Collapse
|
42
|
Abstract
This review will make familiar with new concepts in ovarian cancer and their impact on radiological practice. Disseminated peritoneal spread and ascites are typical of the most common (70-80 %) cancer type, high-grade serous ovarian cancer. Other cancer subtypes differ in origin, precursors, and imaging features. Expert sonography allows excellent risk assessment in adnexal masses. Owing to its high specificity, complementary MRI improves characterization of indeterminate lesions. Major changes in the new FIGO staging classification include fusion of fallopian tube and primary ovarian cancer and the subcategory stage IIIA1 for retroperitoneal lymph node metastases only. Inguinal lymph nodes, cardiophrenic lymph nodes, and umbilical metastases are classified as distant metastases (stage IVB). In multidisciplinary conferences (MDC), CT has been used to predict the success of cytoreductive surgery. Resectability criteria have to be specified and agreed on in MDC. Limitations in detection of metastases may be overcome using advanced MRI techniques.
Collapse
Affiliation(s)
- Rosemarie Forstner
- />Department of Radiology, Landeskliniken Salzburg, Paracelsus Medical University, Müllner Hauptstr. 48, 5020 Salzburg, Austria
| | - Matthias Meissnitzer
- />Department of Radiology, Landeskliniken Salzburg, Paracelsus Medical University, Müllner Hauptstr. 48, 5020 Salzburg, Austria
| | - Teresa Margarida Cunha
- />Serviço de Radiologia, Instituto Português de Oncologia de Lisboa Francisco Gentil, Rua Prof. Lima Basto, 1099-023 Lisbon, Portugal
| |
Collapse
|
43
|
Yamamoto M, Tsujikawa T, Fujita Y, Chino Y, Kurokawa T, Kiyono Y, Okazawa H, Yoshida Y. Metabolic tumor burden predicts prognosis of ovarian cancer patients who receive platinum-based adjuvant chemotherapy. Cancer Sci 2016; 107:478-85. [PMID: 26789906 PMCID: PMC4832857 DOI: 10.1111/cas.12890] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 01/07/2016] [Accepted: 01/12/2016] [Indexed: 01/21/2023] Open
Abstract
Volumetric parameters of positron emission tomography–computed tomography using 18F‐fludeoxyglucose (18F‐FDG PET/CT) that comprehensively reflect both metabolic activity and tumor burden are capable of predicting survival in several cancers. The aim of this study was to investigate the predictive performance of metabolic tumor burden measured by 18F‐FDG PET/CT in ovarian cancer patients who received platinum‐based adjuvant chemotherapy after cytoreductive surgery. Included in this study were 37 epithelial ovarian cancer patients. Metabolic tumor burden in terms of metabolic tumor volume (MTV) and total lesion glycolysis (TLG), clinical stage, histological type, residual tumor after primary cytoreductive surgery, baseline serum carbohydrate antigen 125 (CA125) level, and the maximum standardized uptake value (SUVmax) were determined, and compared for their performance in predicting progression‐free survival (PFS). Metabolic tumor volume correlated with CA125 (r = 0.547, P < 0.001), and TLG correlated with SUVmax and CA125 (SUVmax, r = 0.437, P = 0.007; CA125, r = 0.593, P < 0.001). Kaplan–Meier analysis showed a significant difference in PFS between the groups categorized by TLG (P = 0.043; log–rank test). Univariate analysis indicated that TLG was a statistically significant risk factor for poor PFS. Multivariate analysis adjusted according to the clinicopathological features was carried out for MTV, TLG, SUVmax, tumor size, and CA125. Only TLG showed a significant difference (P = 0.038), and a 3.915‐fold increase in the hazard ratio of PFS. Both MTV and TLG (especially TLG) could serve as potential surrogate biomarkers for recurrence in patients who undergo primary cytoreductive surgery followed by platinum‐based chemotherapy, and could identify patients at high risk of recurrence who need more aggressive treatment.
Collapse
Affiliation(s)
- Makoto Yamamoto
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Tetsuya Tsujikawa
- Biomedical Imaging Research Center, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Yuko Fujita
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Yoko Chino
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Tetsuji Kurokawa
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Yasushi Kiyono
- Biomedical Imaging Research Center, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Hidehiko Okazawa
- Biomedical Imaging Research Center, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Yoshio Yoshida
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| |
Collapse
|
44
|
Au KK, Josahkian JA, Francis JA, Squire JA, Koti M. Current state of biomarkers in ovarian cancer prognosis. Future Oncol 2015; 11:3187-95. [DOI: 10.2217/fon.15.251] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
High-grade serous ovarian cancer remains one of the most lethal malignancies in women. Despite recent advances in surgical and pharmaceutical therapies, survival rates remain poor. A major impediment in management of this disease, that continues to contribute to poor overall survival rates, is resistance to standard carboplatin-paclitaxel combination chemotherapies. In addition to tumor cell intrinsic mechanisms leading to drug resistance, there is increasing awareness of the crucial role of the tumor microenvironment in mediating natural immune defense mechanisms and selective pressures that appear to facilitate chemotherapy sensitivity. We provide an overview of some of the promising new genetic and immunological biomarkers in ovarian cancer and discuss their biology and their likely clinical utility in future ovarian cancer management.
Collapse
Affiliation(s)
- Katrina K Au
- Department of Biomedical & Molecular Sciences, Queen's University, 99 University Ave., Kingston, ON, K7L 3N6, Canada
| | - Juliana A Josahkian
- Departments of Genetics & Pathology, Faculdade de Medicina de Ribeirão Preto, São Paulo, Brazil
| | - Julie-Ann Francis
- Department of Obstetrics & Gynecology, Kingston General Hospital, 76 Stuart St, Kingston, ON, K7L 2V7, Canada
| | - Jeremy A Squire
- Departments of Genetics & Pathology, Faculdade de Medicina de Ribeirão Preto, São Paulo, Brazil
| | - Madhuri Koti
- Department of Biomedical & Molecular Sciences, Queen's University, 99 University Ave., Kingston, ON, K7L 3N6, Canada
- Department of Obstetrics & Gynecology, Kingston General Hospital, 76 Stuart St, Kingston, ON, K7L 2V7, Canada
| |
Collapse
|
45
|
|