1
|
Oldan JD, Schroeder JA, Hoffman-Censits J, Rathmell WK, Milowsky MI, Solnes LB, Nimmagadda S, Gorin MA, Khandani AH, Rowe SP. PET/Computed Tomography Transformation of Oncology: Kidney and Urinary Tract Cancers. PET Clin 2024; 19:197-206. [PMID: 38199916 DOI: 10.1016/j.cpet.2023.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
Renal cell carcinoma (RCC) and urothelial carcinoma (UC) are two of the most common genitourinary malignancies. 2-deoxy-2-[18F]fluoro-d-glucose (18F-FDG) can play an important role in the evaluation of patients with RCC and UC. In addition to the clinical utility of 18F-FDG PET to evaluate for metastatic RCC or UC, the shift in molecular imaging to focus on specific ligand-receptor interactions should provide novel diagnostic and therapeutic opportunities in genitourinary malignancies. In combination with the rise of artificial intelligence, our ability to derive imaging biomarkers that are associated with treatment selection, response assessment, and overall patient prognostication will only improve.
Collapse
Affiliation(s)
- Jorge D Oldan
- Molecular Imaging and Therapeutics, Department of Radiology, University of North Carolina, Chapel Hill, NC, USA
| | - Jennifer A Schroeder
- Molecular Imaging and Therapeutics, Department of Radiology, University of North Carolina, Chapel Hill, NC, USA
| | - Jean Hoffman-Censits
- Department of Medical Oncology and Urology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - W Kimryn Rathmell
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Matthew I Milowsky
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Lilja B Solnes
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sridhar Nimmagadda
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael A Gorin
- Milton and Carroll Petrie Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amir H Khandani
- Molecular Imaging and Therapeutics, Department of Radiology, University of North Carolina, Chapel Hill, NC, USA
| | - Steven P Rowe
- Molecular Imaging and Therapeutics, Department of Radiology, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
2
|
Urso L, Bauckneht M, Albano D, Chondrogiannis S, Grassetto G, Lanfranchi F, Dondi F, Fornarini G, Lazzeri M, Evangelista L. The evolution of PET imaging in renal, bladder, upper urinary tract urothelial, testicular and penile carcinoma - Today's impact, tomorrow's potential. Expert Rev Med Devices 2024; 21:55-72. [PMID: 38072680 DOI: 10.1080/17434440.2023.2293919] [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: 08/26/2023] [Accepted: 12/07/2023] [Indexed: 02/05/2024]
Abstract
INTRODUCTION The advancement of hybrid PET/CT or PET/MRI imaging for non-prostate genitourinary cancers has not experienced the rapid progress of prostate cancer. Nevertheless, these neoplasms are aggressive and reliable imaging plays a pivotal role in enhancing patients' quality of life and prognosis. AREAS COVERED the main evidence regarding [18F]FDG and non-[18F]FDG PET/CT or PET/MRI in non-prostate uro-oncological malignancies are summarized and discussed. Moreover, potential future directions concerning PET imaging in these neoplasms are debated, with the aim to stimulate future research projects covering these fields. EXPERT OPINION In Renal Cell Carcinoma (RCC), [18F]FDG PET/CT demonstrates varying efficacy in staging, restaging, and prognostic stratification, but PSMA PET/CT is emerging as a potential game-changer, particularly in advanced, high-grade aggressive clear cell RCC. [18F]FDG PET/CT may see an increased use in N and M-staging of bladder cancer, as well as for detecting recurrence and response to neoadjuvant chemotherapy. Preliminary data regarding [68Ga]-FAPI also looks promising in this context. [18F]FDG PET/MRI could be useful for the T-staging of bladder cancer, while upper tract urothelial carcinoma still lacks of molecular imaging literature reports. In testicular and penile cancer [18F]FDG PET/CT has demonstrated its usefulness in several clinical settings, although experiences with non-[18F]FDG radiotracers are lacking.
Collapse
Affiliation(s)
- Luca Urso
- Department of Nuclear Medicine - PET/CT Center, S. Maria Della Misericordia Hospital, Rovigo, Italy
| | - Matteo Bauckneht
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico, San Martino, Genova, Italy
- Department of Health Sciences (DISSAL), University of Genoa, Genova, Italy
| | - Domenico Albano
- Nuclear Medicine Department, ASST Spedali Civili of Brescia, University of Brescia, Brescia, Italy
| | - Sotirios Chondrogiannis
- Department of Nuclear Medicine - PET/CT Center, S. Maria Della Misericordia Hospital, Rovigo, Italy
| | - Gaia Grassetto
- Department of Nuclear Medicine - PET/CT Center, S. Maria Della Misericordia Hospital, Rovigo, Italy
| | - Francesco Lanfranchi
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico, San Martino, Genova, Italy
- Department of Health Sciences (DISSAL), University of Genoa, Genova, Italy
| | - Francesco Dondi
- Nuclear Medicine Department, ASST Spedali Civili of Brescia, University of Brescia, Brescia, Italy
| | - Giuseppe Fornarini
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico, San Martino, Genova, Italy
| | - Massimo Lazzeri
- Department of Urology, IRCCS Humanitas Clinical and Research Hospital, Rozzano, Italy
| | - Laura Evangelista
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Nuclear Medicine Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| |
Collapse
|
3
|
Feuerecker B, Heimer MM, Geyer T, Fabritius MP, Gu S, Schachtner B, Beyer L, Ricke J, Gatidis S, Ingrisch M, Cyran CC. Artificial Intelligence in Oncological Hybrid Imaging. Nuklearmedizin 2023; 62:296-305. [PMID: 37802057 DOI: 10.1055/a-2157-6810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2023]
Abstract
BACKGROUND Artificial intelligence (AI) applications have become increasingly relevant across a broad spectrum of settings in medical imaging. Due to the large amount of imaging data that is generated in oncological hybrid imaging, AI applications are desirable for lesion detection and characterization in primary staging, therapy monitoring, and recurrence detection. Given the rapid developments in machine learning (ML) and deep learning (DL) methods, the role of AI will have significant impact on the imaging workflow and will eventually improve clinical decision making and outcomes. METHODS AND RESULTS The first part of this narrative review discusses current research with an introduction to artificial intelligence in oncological hybrid imaging and key concepts in data science. The second part reviews relevant examples with a focus on applications in oncology as well as discussion of challenges and current limitations. CONCLUSION AI applications have the potential to leverage the diagnostic data stream with high efficiency and depth to facilitate automated lesion detection, characterization, and therapy monitoring to ultimately improve quality and efficiency throughout the medical imaging workflow. The goal is to generate reproducible, structured, quantitative diagnostic data for evidence-based therapy guidance in oncology. However, significant challenges remain regarding application development, benchmarking, and clinical implementation. KEY POINTS · Hybrid imaging generates a large amount of multimodality medical imaging data with high complexity and depth.. · Advanced tools are required to enable fast and cost-efficient processing along the whole radiology value chain.. · AI applications promise to facilitate the assessment of oncological disease in hybrid imaging with high quality and efficiency for lesion detection, characterization, and response assessment. The goal is to generate reproducible, structured, quantitative diagnostic data for evidence-based oncological therapy guidance.. · Selected applications in three oncological entities (lung, prostate, and neuroendocrine tumors) demonstrate how AI algorithms may impact imaging-based tasks in hybrid imaging and potentially guide clinical decision making..
Collapse
Affiliation(s)
- Benedikt Feuerecker
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
- German Cancer Research Center (DKFZ), Partner site Munich, DKTK German Cancer Consortium, Munich, Germany
| | - Maurice M Heimer
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Thomas Geyer
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | | | - Sijing Gu
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | | | - Leonie Beyer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Jens Ricke
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Sergios Gatidis
- Department of Radiology, University Hospital Tübingen, Tübingen, Germany
- MPI, Max Planck Institute for Intelligent Systems, Tübingen, Germany
| | - Michael Ingrisch
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Clemens C Cyran
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
4
|
Wang Z, Zhang X, Wang X, Li J, Zhang Y, Zhang T, Xu S, Jiao W, Niu H. Deep learning techniques for imaging diagnosis of renal cell carcinoma: current and emerging trends. Front Oncol 2023; 13:1152622. [PMID: 37727213 PMCID: PMC10505614 DOI: 10.3389/fonc.2023.1152622] [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: 01/28/2023] [Accepted: 08/11/2023] [Indexed: 09/21/2023] Open
Abstract
This study summarizes the latest achievements, challenges, and future research directions in deep learning technologies for the diagnosis of renal cell carcinoma (RCC). This is the first review of deep learning in RCC applications. This review aims to show that deep learning technologies hold great promise in the field of RCC diagnosis, and we look forward to more research results to meet us for the mutual benefit of renal cell carcinoma patients. Medical imaging plays an important role in the early detection of renal cell carcinoma (RCC), as well as in the monitoring and evaluation of RCC during treatment. The most commonly used technologies such as contrast enhanced computed tomography (CECT), ultrasound and magnetic resonance imaging (MRI) are now digitalized, allowing deep learning to be applied to them. Deep learning is one of the fastest growing fields in the direction of medical imaging, with rapidly emerging applications that have changed the traditional medical treatment paradigm. With the help of deep learning-based medical imaging tools, clinicians can diagnose and evaluate renal tumors more accurately and quickly. This paper describes the application of deep learning-based imaging techniques in RCC assessment and provides a comprehensive review.
Collapse
Affiliation(s)
- Zijie Wang
- Department of Vascular Intervention, ShengLi Oilfield Center Hospital, Dongying, China
| | - Xiaofei Zhang
- Department of Education and Training, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xinning Wang
- Department of Urology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jianfei Li
- Extenics Specialized Committee, Chinese Association of Artificial Intelligence (ESCCAAI), Beijing, China
| | - Yuhao Zhang
- Department of Urology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Tianwei Zhang
- Department of Urology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shang Xu
- Department of Urology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wei Jiao
- Department of Urology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Haitao Niu
- Department of Urology, Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
5
|
Dai J, Wang H, Xu Y, Chen X, Tian R. Clinical application of AI-based PET images in oncological patients. Semin Cancer Biol 2023; 91:124-142. [PMID: 36906112 DOI: 10.1016/j.semcancer.2023.03.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 02/28/2023] [Accepted: 03/07/2023] [Indexed: 03/11/2023]
Abstract
Based on the advantages of revealing the functional status and molecular expression of tumor cells, positron emission tomography (PET) imaging has been performed in numerous types of malignant diseases for diagnosis and monitoring. However, insufficient image quality, the lack of a convincing evaluation tool and intra- and interobserver variation in human work are well-known limitations of nuclear medicine imaging and restrict its clinical application. Artificial intelligence (AI) has gained increasing interest in the field of medical imaging due to its powerful information collection and interpretation ability. The combination of AI and PET imaging potentially provides great assistance to physicians managing patients. Radiomics, an important branch of AI applied in medical imaging, can extract hundreds of abstract mathematical features of images for further analysis. In this review, an overview of the applications of AI in PET imaging is provided, focusing on image enhancement, tumor detection, response and prognosis prediction and correlation analyses with pathology or specific gene mutations in several types of tumors. Our aim is to describe recent clinical applications of AI-based PET imaging in malignant diseases and to focus on the description of possible future developments.
Collapse
Affiliation(s)
- Jiaona Dai
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hui Wang
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuchao Xu
- School of Nuclear Science and Technology, University of South China, Hengyang City 421001, China
| | - Xiyang Chen
- Division of Vascular Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Rong Tian
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
6
|
Feuerecker B, Heimer MM, Geyer T, Fabritius MP, Gu S, Schachtner B, Beyer L, Ricke J, Gatidis S, Ingrisch M, Cyran CC. Artificial Intelligence in Oncological Hybrid Imaging. ROFO-FORTSCHR RONTG 2023; 195:105-114. [PMID: 36170852 DOI: 10.1055/a-1909-7013] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Artificial intelligence (AI) applications have become increasingly relevant across a broad spectrum of settings in medical imaging. Due to the large amount of imaging data that is generated in oncological hybrid imaging, AI applications are desirable for lesion detection and characterization in primary staging, therapy monitoring, and recurrence detection. Given the rapid developments in machine learning (ML) and deep learning (DL) methods, the role of AI will have significant impact on the imaging workflow and will eventually improve clinical decision making and outcomes. METHODS AND RESULTS The first part of this narrative review discusses current research with an introduction to artificial intelligence in oncological hybrid imaging and key concepts in data science. The second part reviews relevant examples with a focus on applications in oncology as well as discussion of challenges and current limitations. CONCLUSION AI applications have the potential to leverage the diagnostic data stream with high efficiency and depth to facilitate automated lesion detection, characterization, and therapy monitoring to ultimately improve quality and efficiency throughout the medical imaging workflow. The goal is to generate reproducible, structured, quantitative diagnostic data for evidence-based therapy guidance in oncology. However, significant challenges remain regarding application development, benchmarking, and clinical implementation. KEY POINTS · Hybrid imaging generates a large amount of multimodality medical imaging data with high complexity and depth.. · Advanced tools are required to enable fast and cost-efficient processing along the whole radiology value chain.. · AI applications promise to facilitate the assessment of oncological disease in hybrid imaging with high quality and efficiency for lesion detection, characterization, and response assessment. The goal is to generate reproducible, structured, quantitative diagnostic data for evidence-based oncological therapy guidance.. · Selected applications in three oncological entities (lung, prostate, and neuroendocrine tumors) demonstrate how AI algorithms may impact imaging-based tasks in hybrid imaging and potentially guide clinical decision making.. CITATION FORMAT · Feuerecker B, Heimer M, Geyer T et al. Artificial Intelligence in Oncological Hybrid Imaging. Fortschr Röntgenstr 2023; 195: 105 - 114.
Collapse
Affiliation(s)
- Benedikt Feuerecker
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany.,German Cancer Research Center (DKFZ), Partner site Munich, DKTK German Cancer Consortium, Munich, Germany
| | - Maurice M Heimer
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Thomas Geyer
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | | | - Sijing Gu
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | | | - Leonie Beyer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Jens Ricke
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Sergios Gatidis
- Department of Radiology, University Hospital Tübingen, Tübingen, Germany.,MPI, Max Planck Institute for Intelligent Systems, Tübingen, Germany
| | - Michael Ingrisch
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Clemens C Cyran
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
7
|
Yin Q, Xu H, Zhong Y, Ni J, Hu S. Diagnostic performance of MRI, SPECT, and PET in detecting renal cell carcinoma: a systematic review and meta-analysis. BMC Cancer 2022; 22:163. [PMID: 35148700 PMCID: PMC8840296 DOI: 10.1186/s12885-022-09239-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 01/24/2022] [Indexed: 11/30/2022] Open
Abstract
Background Renal cell carcinoma (RCC) is one of the most common malignancies worldwide. Noninvasive imaging techniques, such as magnetic resonance imaging (MRI), single photon emission computed tomography (SPECT), and positron emission tomography (PET), have been involved in increasing evolution to detect RCC. This meta-analysis aims to compare to compare the performance of MRI, SPECT, and PET in the detection of RCC in humans, and to provide evidence for decision-making in terms of further research and clinical settings. Methods Electronic databases including PubMed, Web of Science, Embase, and Cochrane Library were systemically searched. The keywords such as “magnetic resonance imaging”, “MRI”, “single-photon emission computed tomography”, “SPECT”, “positron emission tomography”, “PET”, “renal cell carcinoma” were used for the search. Studies concerning MRI, SPECT, and PET for the detection of RCC were included. Pooled sensitivity, specificity, and the area under the summary receiver operating characteristic (SROC) curve (AUC), etc. were calculated. Results A total of 44 articles were finally detected for inclusion in this study. The pooled sensitivities of MRI, 18F-FDG PET and 18F-FDG PET/CT were 0.80, 0.83, and 0.89, respectively. Their respective overall specificities were 0.90, 0.86, and 0.88. The pooled sensitivity and specificity of MRI studies at 1.5 T were 0.86 and 0.94, respectively. With respect to prospective PET studies, the pooled sensitivity, specificity and AUC were 0.90, 0.93 and 0.97, respectively. In the detection of primary RCC, PET studies manifested a pooled sensitivity, specificity, and AUC of 0.77, 0.80, and 0.84, respectively. The pooled sensitivity, specificity, and AUC of PET/CT studies in detecting primary RCC were 0.80, 0.85, and 0.89. Conclusion Our study manifests that MRI and PET/CT present better diagnostic value for the detection of RCC in comparison with PET. MRI is superior in the diagnosis of primary RCC.
Collapse
Affiliation(s)
- Qihua Yin
- Department of Radiology, Wuxi No. 2 People's Hospital Affiliated to Nanjing Medical University, Address: No. 68, Zhongshan Rd., Wuxi, 214002, Jiangsu Province, China
| | - Huiting Xu
- Department of Radiology, Wuxi No. 2 People's Hospital Affiliated to Nanjing Medical University, Address: No. 68, Zhongshan Rd., Wuxi, 214002, Jiangsu Province, China
| | - Yanqi Zhong
- Department of Radiology, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Road, Wuxi, 214122, China
| | - Jianming Ni
- Department of Radiology, Wuxi No. 2 People's Hospital Affiliated to Nanjing Medical University, Address: No. 68, Zhongshan Rd., Wuxi, 214002, Jiangsu Province, China. .,Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China.
| | - Shudong Hu
- Department of Radiology, Affiliated Hospital of Jiangnan University, No. 1000, Hefeng Road, Wuxi, 214122, China.
| |
Collapse
|
8
|
de la Pinta C, Castillo ME, Collado M, Galindo-Pumariño C, Peña C. Radiogenomics: Hunting Down Liver Metastasis in Colorectal Cancer Patients. Cancers (Basel) 2021; 13:5547. [PMID: 34771709 PMCID: PMC8582778 DOI: 10.3390/cancers13215547] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 02/07/2023] Open
Abstract
Radiomics is a developing new discipline that analyzes conventional medical images to extract quantifiable data that can be mined for new biomarkers that show the biology of pathological processes at microscopic levels. These data can be converted into image-based signatures to improve diagnostic, prognostic and predictive accuracy in cancer patients. The combination of radiomics and molecular data, called radiogenomics, has clear implications for cancer patients' management. Though some studies have focused on radiogenomics signatures in hepatocellular carcinoma patients, only a few have examined colorectal cancer metastatic lesions in the liver. Moreover, the need to differentiate between liver lesions is fundamental for accurate diagnosis and treatment. In this review, we summarize the knowledge gained from radiomics and radiogenomics studies in hepatic metastatic colorectal cancer patients and their use in early diagnosis, response assessment and treatment decisions. We also investigate their value as possible prognostic biomarkers. In addition, the great potential of image mining to provide a comprehensive view of liver niche formation is examined thoroughly. Finally, new challenges and current limitations for the early detection of the liver premetastatic niche, based on radiomics and radiogenomics, are also discussed.
Collapse
Affiliation(s)
- Carolina de la Pinta
- Radiation Oncology Department, Ramón y Cajal University Hospital, IRYCIS, Alcalá University, 28034 Madrid, Spain
| | - María E. Castillo
- Medical Oncology Department, Ramón y Cajal University Hospital, IRYCIS, Alcalá University, 28034 Madrid, Spain; (M.E.C.); (M.C.); (C.G.-P.)
- Centro de Investigación Biomédica en Red de Cancer (CIBERONC), 28029 Madrid, Spain
| | - Manuel Collado
- Medical Oncology Department, Ramón y Cajal University Hospital, IRYCIS, Alcalá University, 28034 Madrid, Spain; (M.E.C.); (M.C.); (C.G.-P.)
| | - Cristina Galindo-Pumariño
- Medical Oncology Department, Ramón y Cajal University Hospital, IRYCIS, Alcalá University, 28034 Madrid, Spain; (M.E.C.); (M.C.); (C.G.-P.)
- Centro de Investigación Biomédica en Red de Cancer (CIBERONC), 28029 Madrid, Spain
| | - Cristina Peña
- Medical Oncology Department, Ramón y Cajal University Hospital, IRYCIS, Alcalá University, 28034 Madrid, Spain; (M.E.C.); (M.C.); (C.G.-P.)
- Centro de Investigación Biomédica en Red de Cancer (CIBERONC), 28029 Madrid, Spain
| |
Collapse
|
9
|
Ursprung S, Priest AN, Zaccagna F, Qian W, Machin A, Stewart GD, Warren AY, Eisen T, Welsh SJ, Gallagher FA, Barrett T. Multiparametric MRI for assessment of early response to neoadjuvant sunitinib in renal cell carcinoma. PLoS One 2021; 16:e0258988. [PMID: 34699525 PMCID: PMC8547646 DOI: 10.1371/journal.pone.0258988] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 10/08/2021] [Indexed: 11/29/2022] Open
Abstract
PURPOSE To detect early response to sunitinib treatment in metastatic clear cell renal cancer (mRCC) using multiparametric MRI. METHOD Participants with mRCC undergoing pre-surgical sunitinib therapy in the prospective NeoSun clinical trial (EudraCtNo: 2005-004502-82) were imaged before starting treatment, and after 12 days of sunitinib therapy using morphological MRI sequences, advanced diffusion-weighted imaging, measurements of R2* (related to hypoxia) and dynamic contrast-enhanced imaging. Following nephrectomy, participants continued treatment and were followed-up with contrast-enhanced CT. Changes in imaging parameters before and after sunitinib were assessed with the non-parametric Wilcoxon signed-rank test and the log-rank test was used to assess effects on survival. RESULTS 12 participants fulfilled the inclusion criteria. After 12 days, the solid and necrotic tumor volumes decreased by 28% and 17%, respectively (p = 0.04). However, tumor-volume reduction did not correlate with progression-free or overall survival (PFS/OS). Sunitinib therapy resulted in a reduction in median solid tumor diffusivity D from 1298x10-6 to 1200x10-6mm2/s (p = 0.03); a larger decrease was associated with a better RECIST response (p = 0.02) and longer PFS (p = 0.03) on the log-rank test. An increase in R2* from 19 to 28s-1 (p = 0.001) was observed, paralleled by a decrease in Ktrans from 0.415 to 0.305min-1 (p = 0.01) and a decrease in perfusion fraction from 0.34 to 0.19 (p<0.001). CONCLUSIONS Physiological imaging confirmed efficacy of the anti-angiogenic agent 12 days after initiating therapy and demonstrated response to treatment. The change in diffusivity shortly after starting pre-surgical sunitinib correlated to PFS in mRCC undergoing nephrectomy, however, no parameter predicted OS. TRIAL REGISTRATION EudraCtNo: 2005-004502-82.
Collapse
Affiliation(s)
- Stephan Ursprung
- University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
| | - Andrew N. Priest
- University of Cambridge, Cambridge, United Kingdom
- Addenbrooke’s Hospital, Cambridge University Hospital NHS Foundation Trust, Cambridge, United Kingdom
| | | | - Wendi Qian
- University of Cambridge, Cambridge, United Kingdom
- Cambridge Cancer Trial Centre, Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Andrea Machin
- University of Cambridge, Cambridge, United Kingdom
- Cambridge Cancer Trial Centre, Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Grant D. Stewart
- University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
- Addenbrooke’s Hospital, Cambridge University Hospital NHS Foundation Trust, Cambridge, United Kingdom
| | - Anne Y. Warren
- University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
- Addenbrooke’s Hospital, Cambridge University Hospital NHS Foundation Trust, Cambridge, United Kingdom
| | - Timothy Eisen
- University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
- Addenbrooke’s Hospital, Cambridge University Hospital NHS Foundation Trust, Cambridge, United Kingdom
| | - Sarah J. Welsh
- University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
- Addenbrooke’s Hospital, Cambridge University Hospital NHS Foundation Trust, Cambridge, United Kingdom
| | - Ferdia A. Gallagher
- University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
| | - Tristan Barrett
- University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
- Addenbrooke’s Hospital, Cambridge University Hospital NHS Foundation Trust, Cambridge, United Kingdom
| |
Collapse
|
10
|
CT-based peritumoral radiomics signatures for malignancy grading of clear cell renal cell carcinoma. Abdom Radiol (NY) 2021; 46:2690-2698. [PMID: 33427908 DOI: 10.1007/s00261-020-02890-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/26/2020] [Accepted: 11/28/2020] [Indexed: 10/22/2022]
Abstract
OBJECTIVE To evaluate the efficiency of CT-based peritumoral radiomics signatures of clear cell renal cell carcinoma (ccRCC) for malignancy grading in preoperative prediction. MATERIALS AND METHODS 203 patients with pathologically confirmed as ccRCC were retrospectively enrolled in this study. All patients were categorized into training set (n = 122) and validation set (n = 81). For each patient, two types of volumes of interest (VOI) were masked on CT images. One type of VOIs was defined as the tumor mass volume (TMV), which was masked by radiologists delineating the outline of all contiguous slices of the entire tumor, while the other type defined as the peritumoral tumor volume (PTV), which was automatically created by an image morphological method. 1760 radiomics features were calculated from each VOI, and then the discriminative radiomics features were selected by Pearson correlation analysis for reproducibility and redundancy. These selected features were investigated their validity for building radiomics signatures by mRMR feature ranking method. Finally, the top ranked features, which were used as radiomics signatures, were input into a classifier for malignancy grading. The prediction performance was evaluated by receiver operating characteristic (ROC) curve in an independent validation cohort. RESULTS The radiomics signatures of PTV showed a better performance on malignancy grade prediction of ccRCC with AUC of 0.807 (95% CI 0.800-0.834) in train data and 0.848 (95% CI 0.760-0.936) in validation data, while the radiomics signatures of TMV with AUC of 0.773 (95% CI 0.744-0.802) in train data and 0.810 (95% CI 0.706-0.914) in validation data. CONCLUSION The CT-based peritumoral radiomics signature is a potential way to be used as a noninvasive tool to preoperatively predict the malignancy grades of ccRCC.
Collapse
|
11
|
Ganeshan B, Miles K, Afaq A, Punwani S, Rodriguez M, Wan S, Walls D, Hoy L, Khan S, Endozo R, Shortman R, Hoath J, Bhargava A, Hanson M, Francis D, Arulampalam T, Dindyal S, Chen SH, Ng T, Groves A. Texture Analysis of Fractional Water Content Images Acquired during PET/MRI: Initial Evidence for an Association with Total Lesion Glycolysis, Survival and Gene Mutation Profile in Primary Colorectal Cancer. Cancers (Basel) 2021; 13:2715. [PMID: 34072712 PMCID: PMC8199380 DOI: 10.3390/cancers13112715] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/19/2021] [Accepted: 05/21/2021] [Indexed: 01/07/2023] Open
Abstract
To assess the capability of fractional water content (FWC) texture analysis (TA) to generate biologically relevant information from routine PET/MRI acquisitions for colorectal cancer (CRC) patients. Thirty consecutive primary CRC patients (mean age 63.9, range 42-83 years) prospectively underwent FDG-PET/MRI. FWC tumor parametric images generated from Dixon MR sequences underwent TA using commercially available research software (TexRAD). Data analysis comprised (1) identification of functional imaging correlates for texture features (TF) with low inter-observer variability (intraclass correlation coefficient: ICC > 0.75), (2) evaluation of prognostic performance for FWC-TF, and (3) correlation of prognostic imaging signatures with gene mutation (GM) profile. Of 32 FWC-TF with ICC > 0.75, 18 correlated with total lesion glycolysis (TLG, highest: rs = -0.547, p = 0.002). Using optimized cut-off values, five MR FWC-TF identified a good prognostic group with zero mortality (lowest: p = 0.017). For the most statistically significant prognostic marker, favorable prognosis was significantly associated with a higher number of GM per patient (medians: 7 vs. 1.5, p = 0.009). FWC-TA derived from routine PET/MRI Dixon acquisitions shows good inter-operator agreement, generates biological relevant information related to TLG, GM count, and provides prognostic information that can unlock new clinical applications for CRC patients.
Collapse
Affiliation(s)
- Balaji Ganeshan
- Research Department of Imaging, Division of Medicine, University College London (UCL), London WC1E 6BT, UK; (K.M.); (S.P.); (D.W.); (L.H.); (J.H.); (S.-H.C.); (A.G.)
| | - Kenneth Miles
- Research Department of Imaging, Division of Medicine, University College London (UCL), London WC1E 6BT, UK; (K.M.); (S.P.); (D.W.); (L.H.); (J.H.); (S.-H.C.); (A.G.)
| | - Asim Afaq
- Imaging Division, Surgery and Cancer Board, University College London Hospitals (UCLH) NHS Foundation Trust, University College Hospital (UCH), London NW1 2BU, UK; (A.A.); (M.R.); (S.W.); (S.K.); (R.E.); (R.S.); (S.D.)
- Department of Radiology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Shonit Punwani
- Research Department of Imaging, Division of Medicine, University College London (UCL), London WC1E 6BT, UK; (K.M.); (S.P.); (D.W.); (L.H.); (J.H.); (S.-H.C.); (A.G.)
| | - Manuel Rodriguez
- Imaging Division, Surgery and Cancer Board, University College London Hospitals (UCLH) NHS Foundation Trust, University College Hospital (UCH), London NW1 2BU, UK; (A.A.); (M.R.); (S.W.); (S.K.); (R.E.); (R.S.); (S.D.)
| | - Simon Wan
- Imaging Division, Surgery and Cancer Board, University College London Hospitals (UCLH) NHS Foundation Trust, University College Hospital (UCH), London NW1 2BU, UK; (A.A.); (M.R.); (S.W.); (S.K.); (R.E.); (R.S.); (S.D.)
| | - Darren Walls
- Research Department of Imaging, Division of Medicine, University College London (UCL), London WC1E 6BT, UK; (K.M.); (S.P.); (D.W.); (L.H.); (J.H.); (S.-H.C.); (A.G.)
| | - Luke Hoy
- Research Department of Imaging, Division of Medicine, University College London (UCL), London WC1E 6BT, UK; (K.M.); (S.P.); (D.W.); (L.H.); (J.H.); (S.-H.C.); (A.G.)
| | - Saif Khan
- Imaging Division, Surgery and Cancer Board, University College London Hospitals (UCLH) NHS Foundation Trust, University College Hospital (UCH), London NW1 2BU, UK; (A.A.); (M.R.); (S.W.); (S.K.); (R.E.); (R.S.); (S.D.)
| | - Raymond Endozo
- Imaging Division, Surgery and Cancer Board, University College London Hospitals (UCLH) NHS Foundation Trust, University College Hospital (UCH), London NW1 2BU, UK; (A.A.); (M.R.); (S.W.); (S.K.); (R.E.); (R.S.); (S.D.)
| | - Robert Shortman
- Imaging Division, Surgery and Cancer Board, University College London Hospitals (UCLH) NHS Foundation Trust, University College Hospital (UCH), London NW1 2BU, UK; (A.A.); (M.R.); (S.W.); (S.K.); (R.E.); (R.S.); (S.D.)
| | - John Hoath
- Research Department of Imaging, Division of Medicine, University College London (UCL), London WC1E 6BT, UK; (K.M.); (S.P.); (D.W.); (L.H.); (J.H.); (S.-H.C.); (A.G.)
| | - Aman Bhargava
- Institute of Health Barts and London Medical School, Queen Mary University of London (QMUL), London E1 2AD, UK;
| | - Matthew Hanson
- Division of Cancer and Clinical Support, Barking, Havering and Redbridge University Hospitals NHS Trust, Queens and King George Hospitals, Essex IG3 8YB, UK;
| | - Daren Francis
- Department of Colorectal Surgery, Royal Free London NHS Foundation Trust, Barnet and Chase Farm Hospitals, London NW3 2QG, UK;
| | - Tan Arulampalam
- Department of Surgery, East Suffolk and North Essex NHS Foundation Trust, Colchester General Hospital, Colchester CO4 5JL, UK;
| | - Sanjay Dindyal
- Imaging Division, Surgery and Cancer Board, University College London Hospitals (UCLH) NHS Foundation Trust, University College Hospital (UCH), London NW1 2BU, UK; (A.A.); (M.R.); (S.W.); (S.K.); (R.E.); (R.S.); (S.D.)
| | - Shih-Hsin Chen
- Research Department of Imaging, Division of Medicine, University College London (UCL), London WC1E 6BT, UK; (K.M.); (S.P.); (D.W.); (L.H.); (J.H.); (S.-H.C.); (A.G.)
- Department of Nuclear Medicine, Keelung Chang Gung Memorial Hospital, Keelung 204, Taiwan
| | - Tony Ng
- School of Cancer & Pharmaceutical Sciences, King’s College London (KCL), London WC2R 2LS, UK;
| | - Ashley Groves
- Research Department of Imaging, Division of Medicine, University College London (UCL), London WC1E 6BT, UK; (K.M.); (S.P.); (D.W.); (L.H.); (J.H.); (S.-H.C.); (A.G.)
| |
Collapse
|
12
|
Brodie A, Dai N, Teoh JYC, Decaestecker K, Dasgupta P, Vasdev N. Artificial intelligence in urological oncology: An update and future applications. Urol Oncol 2021; 39:379-399. [PMID: 34024704 DOI: 10.1016/j.urolonc.2021.03.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 12/20/2020] [Accepted: 03/21/2021] [Indexed: 01/16/2023]
Abstract
There continues to be rapid developments and research in the field of Artificial Intelligence (AI) in Urological Oncology worldwide. In this review we discuss the basics of AI, application of AI per tumour group (Renal, Prostate and Bladder Cancer) and application of AI in Robotic Urological Surgery. We also discuss future applications of AI being developed with the benefits to patients with Urological Oncology.
Collapse
Affiliation(s)
- Andrew Brodie
- Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Nick Dai
- Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Jeremy Yuen-Chun Teoh
- S.H. Ho Urology Centre, Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China
| | | | - Prokar Dasgupta
- Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Nikhil Vasdev
- Hertfordshire and Bedfordshire Urological Cancer Centre, Department of Urology, Lister Hospital, Stevenage, United Kingdom; School of Medicine and Life Sciences, University of Hertfordshire, Hatfield, United Kingdom.
| |
Collapse
|
13
|
Hah YS, Koo KC. Immunology and Immunotherapeutic Approaches for Advanced Renal Cell Carcinoma: A Comprehensive Review. Int J Mol Sci 2021; 22:ijms22094452. [PMID: 33923219 PMCID: PMC8123195 DOI: 10.3390/ijms22094452] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 12/25/2022] Open
Abstract
Renal cell carcinoma (RCC) is a malignant tumor associated with various tumor microenvironments (TMEs). The immune system is activated by the development of cancer and drives T cell anti-tumor response. CD8 T cells are known to improve clinical outcomes and sensitivity to immunotherapy, and play a crucial role against tumors. In contrast, tumor-associated macrophages (TAMs) suppress immunity against malignancy and lead to tumor progression. TAMs are promoted from damaged TMEs and mount proinflammatory responses to pathogens. Initial immunotherapy consists of interferon-α and interleukin-2. However, response to such therapy is unclear in most patients, and it is associated with high levels of toxicity. Immune checkpoint inhibitors (ICIs), which up-regulate immune responses by blocking the programed cell death protein 1 (PD-1) receptor, the ligand of PD-1, or cytotoxic T-lymphocyte-associated protein 4 T cells, have led to a new era of immunotherapy. Furthermore, combination strategies with ICIs have proven effective through several randomized controlled trials. We expect the next generation of immunotherapy to lead to better outcomes based on ongoing trials and inspire new therapeutic strategies.
Collapse
Affiliation(s)
- Yoon-Soo Hah
- Department of Urology, Catholic University of Daegu School of Medicine, Daegu 42472, Korea;
| | - Kyo-Chul Koo
- Department of Urology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06229, Korea
- Correspondence: ; Tel.: +82-2-2019-3470
| |
Collapse
|
14
|
Xiao J, Rahbar H, Hippe DS, Rendi MH, Parker EU, Shekar N, Hirano M, Cheung KJ, Partridge SC. Dynamic contrast-enhanced breast MRI features correlate with invasive breast cancer angiogenesis. NPJ Breast Cancer 2021; 7:42. [PMID: 33863924 PMCID: PMC8052427 DOI: 10.1038/s41523-021-00247-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 03/15/2021] [Indexed: 02/02/2023] Open
Abstract
Angiogenesis is a critical component of breast cancer development, and identification of imaging-based angiogenesis assays has prognostic and treatment implications. We evaluated the association of semi-quantitative kinetic and radiomic breast cancer features on dynamic contrast-enhanced (DCE)-MRI with microvessel density (MVD), a marker for angiogenesis. Invasive breast cancer kinetic features (initial peak percent enhancement [PE], signal enhancement ratio [SER], functional tumor volume [FTV], and washout fraction [WF]), radiomics features (108 total features reflecting tumor morphology, signal intensity, and texture), and MVD (by histologic CD31 immunostaining) were measured in 27 patients (1/2016-7/2017). Lesions with high MVD levels demonstrated higher peak SER than lesions with low MVD (mean: 1.94 vs. 1.61, area under the receiver operating characteristic curve [AUC] = 0.79, p = 0.009) and higher WF (mean: 50.6% vs. 22.5%, AUC = 0.87, p = 0.001). Several radiomics texture features were also promising for predicting increased MVD (maximum AUC = 0.84, p = 0.002). Our study suggests DCE-MRI can non-invasively assess breast cancer angiogenesis, which could stratify biology and optimize treatments.
Collapse
Affiliation(s)
- Jennifer Xiao
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Habib Rahbar
- Department of Radiology, University of Washington, Seattle, WA, USA
- Breast Imaging, Seattle Cancer Care Alliance, Seattle, WA, USA
| | - Daniel S Hippe
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Mara H Rendi
- Department of Pathology, University of Washington, Seattle, WA, USA
| | | | - Neal Shekar
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Michael Hirano
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Kevin J Cheung
- Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA, USA
- Translational Research Program, Public Health Sciences and Human Biology Divisions, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Savannah C Partridge
- Department of Radiology, University of Washington, Seattle, WA, USA.
- Breast Imaging, Seattle Cancer Care Alliance, Seattle, WA, USA.
| |
Collapse
|
15
|
Barua S, Elhalawani H, Volpe S, Al Feghali KA, Yang P, Ng SP, Elgohari B, Granberry RC, Mackin DS, Gunn GB, Hutcheson KA, Chambers MS, Court LE, Mohamed ASR, Fuller CD, Lai SY, Rao A. Computed Tomography Radiomics Kinetics as Early Imaging Correlates of Osteoradionecrosis in Oropharyngeal Cancer Patients. Front Artif Intell 2021; 4:618469. [PMID: 33898983 PMCID: PMC8063205 DOI: 10.3389/frai.2021.618469] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 03/04/2021] [Indexed: 01/08/2023] Open
Abstract
Osteoradionecrosis (ORN) is a major side-effect of radiation therapy in oropharyngeal cancer (OPC) patients. In this study, we demonstrate that early prediction of ORN is possible by analyzing the temporal evolution of mandibular subvolumes receiving radiation. For our analysis, we use computed tomography (CT) scans from 21 OPC patients treated with Intensity Modulated Radiation Therapy (IMRT) with subsequent radiographically-proven ≥ grade II ORN, at three different time points: pre-IMRT, 2-months, and 6-months post-IMRT. For each patient, radiomic features were extracted from a mandibular subvolume that developed ORN and a control subvolume that received the same dose but did not develop ORN. We used a Multivariate Functional Principal Component Analysis (MFPCA) approach to characterize the temporal trajectories of these features. The proposed MFPCA model performs the best at classifying ORN vs. Control subvolumes with an area under curve (AUC) = 0.74 [95% confidence interval (C.I.): 0.61–0.90], significantly outperforming existing approaches such as a pre-IMRT features model or a delta model based on changes at intermediate time points, i.e., at 2- and 6-month follow-up. This suggests that temporal trajectories of radiomics features derived from sequential pre- and post-RT CT scans can provide markers that are correlates of RT-induced mandibular injury, and consequently aid in earlier management of ORN.
Collapse
Affiliation(s)
- Souptik Barua
- Department of Electrical and Computer Engineering, Rice University, Houston, TX, United States.,Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, United States
| | - Hesham Elhalawani
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Stefania Volpe
- Department of Radiation Oncology, European Institute of Oncology IRCSS, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Karine A Al Feghali
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Pei Yang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sweet Ping Ng
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Baher Elgohari
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Robin C Granberry
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Dennis S Mackin
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - G Brandon Gunn
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Katherine A Hutcheson
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mark S Chambers
- Department of Oncologic Dentistry and Prosthodontics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Laurence E Court
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Abdallah S R Mohamed
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Clifton D Fuller
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Stephen Y Lai
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Arvind Rao
- Department of Electrical and Computer Engineering, Rice University, Houston, TX, United States.,Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, United States.,Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
16
|
Tomaszewski MR, Gillies RJ. The Biological Meaning of Radiomic Features. Radiology 2021; 298:505-516. [PMID: 33399513 PMCID: PMC7924519 DOI: 10.1148/radiol.2021202553] [Citation(s) in RCA: 329] [Impact Index Per Article: 82.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/30/2020] [Accepted: 08/17/2020] [Indexed: 02/06/2023]
Abstract
An earlier incorrect version appeared online. This article was corrected on February 10, 2021.
Collapse
Affiliation(s)
- Michal R. Tomaszewski
- From the Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr, Tampa, FL 33612
| | - Robert J. Gillies
- From the Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr, Tampa, FL 33612
| |
Collapse
|
17
|
Tegel BR, Huber S, Savic LJ, Lin M, Gebauer B, Pollak J, Chapiro J. Quantification of contrast-uptake as imaging biomarker for disease progression of renal cell carcinoma after tumor ablation. Acta Radiol 2020; 61:1708-1716. [PMID: 32216452 DOI: 10.1177/0284185120909964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The prognosis of patients with renal cell carcinoma (RCC) depends greatly on the presence of extra-renal metastases. PURPOSE To investigate the value of total tumor volume (TTV) and enhancing tumor volume (ETV) as three-dimensional (3D) quantitative imaging biomarkers for disease aggressiveness in patients with RCC. MATERIAL AND METHODS Retrospective, HIPAA-compliant, IRB-approved study including 37 patients with RCC treated with image-guided thermal ablation during 2007-2015. TNM stage, RENAL Nephrometry Score, largest tumor diameter, TTV, and ETV were assessed on cross-sectional imaging at baseline and correlated with outcome measurements. The primary outcome was time-to-occurrence of extra-renal metastases and the secondary outcome was progression-free survival (PFS). Correlation was assessed using a Cox regression model and differences in outcomes were shown by Kaplan-Meier plots with significance and odds ratios (OR) calculated by Log-rank test/generalized Wilcoxon and continuity-corrected Woolf logit method. RESULTS Patients with a TTV or ETV > 5 cm3 were more likely to develop distant metastases compared to patients with TTV (OR 6.69, 95% confidence interval [CI] 0.33-134.4, P=0.022) or ETV (OR 8.48, 95% CI 0.42-170.1, P=0.016) < 5 cm3. Additionally, PFS was significantly worse in patients with larger ETV (P = 0.039; median PFS 51.87 months vs. 69.97 months). In contrast, stratification by median value of the established, caliper-based measurements showed no significant correlation with outcome parameters. CONCLUSION ETV, as surrogate of lesion vascularity, is a sensitive imaging biomarker for occurrence of extra-renal metastatic disease and PFS in patients with RCC.
Collapse
Affiliation(s)
- Bruno R Tegel
- Yale School of Medicine, Department of Radiology and Biomedical Imaging, New Haven, CT, USA
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität Berlin and Berlin Institute of Health, Institute of Radiology, Berlin, Germany
| | - Steffen Huber
- Yale School of Medicine, Department of Radiology and Biomedical Imaging, New Haven, CT, USA
| | - Lynn J Savic
- Yale School of Medicine, Department of Radiology and Biomedical Imaging, New Haven, CT, USA
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität Berlin and Berlin Institute of Health, Institute of Radiology, Berlin, Germany
| | - MingDe Lin
- U/S Imaging and Interventions, Philips Research North America, Cambridge, MA, USA
| | - Bernhard Gebauer
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt Universität Berlin and Berlin Institute of Health, Institute of Radiology, Berlin, Germany
| | - Jeffrey Pollak
- Yale School of Medicine, Department of Radiology and Biomedical Imaging, New Haven, CT, USA
| | - Julius Chapiro
- Yale School of Medicine, Department of Radiology and Biomedical Imaging, New Haven, CT, USA
| |
Collapse
|
18
|
Association of tumor grade, enhancement on multiphasic CT and microvessel density in patients with clear cell renal cell carcinoma. Abdom Radiol (NY) 2020; 45:3184-3192. [PMID: 31650375 DOI: 10.1007/s00261-019-02271-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE Clear cell renal cell carcinoma (ccRCC) comprises nearly 90% of all diagnosed RCC subtypes and has the worst prognosis and highest metastatic potential. The strongest prognostic factors for patients with ccRCC include histological subtype and Fuhrman grade, which are incorporated into prognostic models. Since ccRCC is a highly vascularized tumor, there may be differences in enhancement patterns on multidetector CT (MDCT) due to the hemodynamics and microvessel density (MVD) of the lesions. This may provide a noninvasive method to characterize incidentally detected low- and high-grade ccRCCs on MDCT. The purpose of our study was to determine the correlation between MDCT enhancement parameters, ccRCC MVD, and Fuhrman grade to determine its utility and value in assessing tumor vascularity and grade in vivo. METHODS In this retrospective, HIPAA-compliant, institutional review board-approved study with waiver of informed consent, 127 consecutive patients with 89 low-grade (LG), and 43 high-grade (HG) ccRCCs underwent preoperative four-phase MDCT. A 3D volume of interest (VOI) was obtained for every tumor and absolute enhancement and the wash-in/wash-out of enhancement for each phase was assessed. Immunohistochemistry on resected specimens was used to quantify MVD. Linear regression and Pearson correlation were used to investigate the strength of the association between 3D VOI enhancement and MVD. Stepwise logistic regression analysis determined independent predictors of HG ccRCC. Cut-off values and odds Ratio (OR) with 95% CIs were reported. The clinical, radiomic, and pathologic features with the highest performance in the stepwise logistic regression analysis were evaluated using receiver operator characteristics (ROC) and area under the curve (AUC). RESULTS Absolute enhancement in the nephrographic phase < 52.1 Hounsfield Units (HU) (HR 0.979, 95% CI 0.964-0.994, p value = 0.006), lesion size > 4.3 cm (HR 1.450, 95% CI 1.211-1.738, p value < 0.001), and an intratumoral MVD < 15% (HR 0.932, 95% CI 0.867-1.002, p value = 0.058) were independent predictors of HG ccRCC with an AUC of 0.818 (95% CI 0.725-0.911). HG ccRCCs had a significant association between 3D VOI enhancement and MVD in each post-contrast phase (r2 = 0.238 to 0.455, p < 0.05). CONCLUSIONS Absolute enhancement of the entire lesion obtained from a 3D VOI in the nephrographic phase on preoperative MDCT can provide quantitative data that are a significant, independent predictor of a high-grade clear cell RCC and can be used to assess tumor vascularity and grade in vivo.
Collapse
|
19
|
Enhanced Computed Tomography-Based Radiomics Signature Combined With Clinical Features in Evaluating Nuclear Grading of Renal Clear Cell Carcinoma. J Comput Assist Tomogr 2020; 44:730-736. [PMID: 32558771 DOI: 10.1097/rct.0000000000001041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
OBJECTIVE The aim of the study was to explore the value of enhanced computed tomography (CT)-based radiomics signature combined with clinical features in evaluating nuclear grading of clear cell renal cell carcinoma (ccRCC). METHODS One hundred one patients with ccRCC were classified into low- and high-grade group, and the data were divided into training set and verification set. Radiomics signatures were constructed in the training set in enhanced 3 stages and the combination of them. The predictive nomogram was constructed. The classification efficiency and the clinical practicability of the integrated radiomics model were evaluated. RESULTS The classification efficiency of enhanced 3-stage integrated histology model was higher than that of each single-phase model. The predictive nomogram incorporated the best radiomics signature, and the independent clinical risk factors showed good performance. A decision curve analysis curve shows that the net benefit of the combined model. CONCLUSIONS It is feasible to evaluate the nuclear grading of ccRCC based on enhanced CT radiomics signature combined with clinical features.
Collapse
|
20
|
Han Y, Chai F, Wei J, Yue Y, Cheng J, Gu D, Zhang Y, Tong T, Sheng W, Hong N, Ye Y, Wang Y, Tian J. Identification of Predominant Histopathological Growth Patterns of Colorectal Liver Metastasis by Multi-Habitat and Multi-Sequence Based Radiomics Analysis. Front Oncol 2020; 10:1363. [PMID: 32923388 PMCID: PMC7456817 DOI: 10.3389/fonc.2020.01363] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 06/29/2020] [Indexed: 12/21/2022] Open
Abstract
Purpose: Developing an MRI-based radiomics model to effectively and accurately predict the predominant histopathologic growth patterns (HGPs) of colorectal liver metastases (CRLMs). Materials and Methods: In this study, 182 resected and histopathological proven CRLMs of chemotherapy-naive patients from two institutions, including 123 replacement CRLMs and 59 desmoplastic CRLMs, were retrospectively analyzed. Radiomics analysis was performed on two regions of interest (ROI), the tumor zone and the tumor-liver interface (TLI) zone. Decision tree (DT) algorithm was used for radiomics modeling on each MR sequence, and fused radiomics model was constructed by combining the radiomics signature of each sequence. The clinical and combination models were developed through multivariate logistic regression method. The performance of the developed models was assessed by receiver operating characteristic (ROC) curves with indicators of area under curve (AUC), accuracy, sensitivity, and specificity. A nomogram was constructed to evaluate the discrimination, calibration, and usefulness. Results: The fused radiomicstumor and radiomicsTLI models showed better performance than any single sequence and clinical model. In addition, the radiomicsTLI model exhibited better performance than radiomicstumor model (AUC of 0.912 vs. 0.879) in internal validation cohort. The combination model showed good discrimination, and the AUC of nomogram was 0.971, 0.909, and 0.905 in the training, internal validation, and external validation cohorts, respectively. Conclusion: MRI-based radiomics method has high potential in predicting the predominant HGPs of CRLM. Preoperative non-invasive identification of predominant HGPs could further explore the ability of HGPs as a potential biomarker for clinical treatment strategy, reflecting different biological pathways.
Collapse
Affiliation(s)
- Yuqi Han
- School of Life Science and Technology, Xidian University, Xi'an, China
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- Beijing Key Laboratory of Molecular Imaging, Beijing, China
| | - Fan Chai
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Jingwei Wei
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- Beijing Key Laboratory of Molecular Imaging, Beijing, China
| | - Yali Yue
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jin Cheng
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Dongsheng Gu
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- Beijing Key Laboratory of Molecular Imaging, Beijing, China
| | - Yinli Zhang
- Department of Pathology, Peking University People's Hospital, Beijing, China
| | - Tong Tong
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Weiqi Sheng
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Nan Hong
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Yingjiang Ye
- Department of Gastrointestinal Surgery, Peking University People' Hospital, Beijing, China
| | - Yi Wang
- Department of Radiology, Peking University People's Hospital, Beijing, China
| | - Jie Tian
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- Beijing Key Laboratory of Molecular Imaging, Beijing, China
- Beijing Advanced Innovation Centre for Big Data-Based Precision Medicine, School of Medicine, Beihang University, Beijing, China
- Engineering Research Centre of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China
| |
Collapse
|
21
|
Radiomics Applications in Renal Tumor Assessment: A Comprehensive Review of the Literature. Cancers (Basel) 2020; 12:cancers12061387. [PMID: 32481542 PMCID: PMC7352711 DOI: 10.3390/cancers12061387] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/22/2020] [Accepted: 05/26/2020] [Indexed: 12/21/2022] Open
Abstract
Radiomics texture analysis offers objective image information that could otherwise not be obtained by radiologists′ subjective radiological interpretation. We investigated radiomics applications in renal tumor assessment and provide a comprehensive review. A detailed search of original articles was performed using the PubMed-MEDLINE database until 20 March 2020 to identify English literature relevant to radiomics applications in renal tumor assessment. In total, 42 articles were included in the analysis and divided into four main categories: renal mass differentiation, nuclear grade prediction, gene expression-based molecular signatures, and patient outcome prediction. The main area of research involves accurately differentiating benign and malignant renal masses, specifically between renal cell carcinoma (RCC) subtypes and from angiomyolipoma without visible fat and oncocytoma. Nuclear grade prediction may enhance proper patient selection for risk-stratified treatment. Radiomics-predicted gene mutations may serve as surrogate biomarkers for high-risk disease, while predicting patients’ responses to targeted therapies and their outcomes will help develop personalized treatment algorithms. Studies generally reported the superiority of radiomics over expert radiological interpretation. Radiomics provides an alternative to subjective image interpretation for improving renal tumor diagnostic accuracy. Further incorporation of clinical and imaging data into radiomics algorithms will augment tumor prediction accuracy and enhance individualized medicine.
Collapse
|
22
|
Shen L, Yin Q. Data maximum dispersion classifier in projection space for high-dimension low-sample-size problems. Knowl Based Syst 2020. [DOI: 10.1016/j.knosys.2019.105420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
23
|
Vaugier L, Ferrer L, Mengue L, Jouglar E. Radiomics for radiation oncologists: are we ready to go? BJR Open 2020; 2:20190046. [PMID: 33178967 PMCID: PMC7594896 DOI: 10.1259/bjro.20190046] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 03/06/2020] [Accepted: 03/09/2020] [Indexed: 12/19/2022] Open
Abstract
Radiomics have emerged as an exciting field of research over the past few years, with very wide potential applications in personalised and precision medicine of the future. Radiomics-based approaches are still however limited in daily clinical practice in oncology. This review focus on how radiomics could be incorporated into the radiation therapy pipeline, and globally help the radiation oncologist, from the tumour diagnosis to follow-up after treatment. Radiomics could impact on all steps of the treatment pipeline, once the limitations in terms of robustness and reproducibility are overcome. Major ongoing efforts should be made to collect and share data in the most standardised manner possible.
Collapse
Affiliation(s)
- Loïg Vaugier
- Department of Radiation Oncology, Institut de Cancérologie de l'Ouest, Nantes - Saint Herblain, France
| | - Ludovic Ferrer
- Department of Medical Physics, Institut de Cancérologie de l'Ouest, Nantes - Saint Herblain, France
| | - Laurence Mengue
- Department of Radiation Oncology, Institut de Cancérologie de l'Ouest, Nantes - Saint Herblain, France
| | - Emmanuel Jouglar
- Department of Radiation Oncology, Institut de Cancérologie de l'Ouest, Nantes - Saint Herblain, France
| |
Collapse
|
24
|
Yuan Q, Zhang H, Deng T, Tang S, Yuan X, Tang W, Xie Y, Ge H, Wang X, Zhou Q, Xiao X. Role of Artificial Intelligence in Kidney Disease. Int J Med Sci 2020; 17:970-984. [PMID: 32308551 PMCID: PMC7163364 DOI: 10.7150/ijms.42078] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 03/17/2020] [Indexed: 12/17/2022] Open
Abstract
Artificial intelligence (AI), as an advanced science technology, has been widely used in medical fields to promote medical development, mainly applied to early detections, disease diagnoses, and management. Owing to the huge number of patients, kidney disease remains a global health problem. Challenges remain in its diagnosis and treatment. AI could take individual conditions into account, produce suitable decisions and promise to make great strides in kidney disease management. Here, we review the current studies of AI applications in kidney disease in alerting systems, diagnostic assistance, guiding treatment and evaluating prognosis. Although the number of studies related to AI applications in kidney disease is small, the potential of AI in the management of kidney disease is well recognized by clinicians; AI will greatly enhance clinicians' capacity in their clinical practice in the future.
Collapse
Affiliation(s)
- Qiongjing Yuan
- Department of Nephrology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China
| | - Haixia Zhang
- Department of Nephrology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China.,Department of Nephrology, Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, Suzhou, Jiangsu 215000, China
| | - Tianci Deng
- Department of Nephrology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China
| | - Shumei Tang
- Department of Nephrology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China
| | - Xiangning Yuan
- Department of Nephrology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China
| | - Wenbin Tang
- Department of Nephrology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China
| | - Yanyun Xie
- Department of Nephrology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China
| | - Huipeng Ge
- Department of Nephrology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China
| | - Xiufen Wang
- Department of Nephrology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China
| | - Qiaoling Zhou
- Department of Nephrology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China
| | - Xiangcheng Xiao
- Department of Nephrology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China
| |
Collapse
|
25
|
Cook GJR, Goh V. What can artificial intelligence teach us about the molecular mechanisms underlying disease? Eur J Nucl Med Mol Imaging 2019; 46:2715-2721. [PMID: 31190176 PMCID: PMC6879441 DOI: 10.1007/s00259-019-04370-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 05/23/2019] [Indexed: 12/24/2022]
Abstract
While molecular imaging with positron emission tomography or single-photon emission computed tomography already reports on tumour molecular mechanisms on a macroscopic scale, there is increasing evidence that there are multiple additional features within medical images that can further improve tumour characterization, treatment prediction and prognostication. Early reports have already revealed the power of radiomics to personalize and improve patient management and outcomes. What remains unclear is how these additional metrics relate to underlying molecular mechanisms of disease. Furthermore, the ability to deal with increasingly large amounts of data from medical images and beyond in a rapid, reproducible and transparent manner is essential for future clinical practice. Here, artificial intelligence (AI) may have an impact. AI encompasses a broad range of 'intelligent' functions performed by computers, including language processing, knowledge representation, problem solving and planning. While rule-based algorithms, e.g. computer-aided diagnosis, have been in use for medical imaging since the 1990s, the resurgent interest in AI is related to improvements in computing power and advances in machine learning (ML). In this review we consider why molecular and cellular processes are of interest and which processes have already been exposed to AI and ML methods as reported in the literature. Non-small-cell lung cancer is used as an exemplar and the focus of this review as the most common tumour type in which AI and ML approaches have been tested and to illustrate some of the concepts.
Collapse
Affiliation(s)
- Gary J R Cook
- Cancer Imaging Department, School of Biomedical Engineering and Imaging Sciences, King's College London, London, SE1 7EH, UK.
- King's College London & Guy's and St Thomas' PET Centre, St Thomas' Hospital, London, UK.
| | - Vicky Goh
- Cancer Imaging Department, School of Biomedical Engineering and Imaging Sciences, King's College London, London, SE1 7EH, UK
- Radiology Department, Guy's and St Thomas' Hospitals NHS Trust, London, UK
| |
Collapse
|
26
|
Sun Z, Li Y, Wang Y, Fan X, Xu K, Wang K, Li S, Zhang Z, Jiang T, Liu X. Radiogenomic analysis of vascular endothelial growth factor in patients with diffuse gliomas. Cancer Imaging 2019; 19:68. [PMID: 31639060 PMCID: PMC6805458 DOI: 10.1186/s40644-019-0256-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 09/25/2019] [Indexed: 01/02/2023] Open
Abstract
OBJECTIVE To predict vascular endothelial growth factor (VEGF) expression in patients with diffuse gliomas using radiomic analysis. MATERIALS AND METHODS Preoperative magnetic resonance images were retrospectively obtained from 239 patients with diffuse gliomas (World Health Organization grades II-IV). The patients were randomly assigned to a training group (n = 160) or a validation group (n = 79) at a 2:1 ratio. For each patient, a total of 431 radiomic features were extracted. The minimum redundancy maximum relevance (mRMR) algorithm was used for feature selection. A machine-learning model for predicting VEGF status was then developed using the selected features and a support vector machine classifier. The predictive performance of the model was evaluated in both groups using receiver operating characteristic curve analysis, and correlations between selected features were assessed. RESULTS Nine radiomic features were selected to generate a VEGF-associated radiomic signature of diffuse gliomas based on the mRMR algorithm. This radiomic signature consisted of two first-order statistics or related wavelet features (Entropy and Minimum) and seven textural features or related wavelet features (including Cluster Tendency and Long Run Low Gray Level Emphasis). The predictive efficiencies measured by the area under the curve were 74.1% in the training group and 70.2% in the validation group. The overall correlations between the 9 radiomic features were low in both groups. CONCLUSIONS Radiomic analysis facilitated efficient prediction of VEGF status in diffuse gliomas, suggesting that using tumor-derived radiomic features for predicting genomic information is feasible.
Collapse
Affiliation(s)
- Zhiyan Sun
- Beijing Neurosurgical Institute, Capital Medical University, 6 Tiantanxili, Beijing, 100050, China
| | - Yiming Li
- Beijing Neurosurgical Institute, Capital Medical University, 6 Tiantanxili, Beijing, 100050, China
| | - Yinyan Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xing Fan
- Beijing Neurosurgical Institute, Capital Medical University, 6 Tiantanxili, Beijing, 100050, China
| | - Kaibin Xu
- Chinese Academy of Sciences, Institute of Automation, Beijing, China
| | - Kai Wang
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shaowu Li
- Beijing Neurosurgical Institute, Capital Medical University, 6 Tiantanxili, Beijing, 100050, China
| | - Zhong Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Tao Jiang
- Beijing Neurosurgical Institute, Capital Medical University, 6 Tiantanxili, Beijing, 100050, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, China
| | - Xing Liu
- Beijing Neurosurgical Institute, Capital Medical University, 6 Tiantanxili, Beijing, 100050, China.
| |
Collapse
|
27
|
Lu Y, Peng W, Song J, Chen T, Wang X, Hou Z, Yan Z, Koh TS. On the potential use of dynamic contrast-enhanced (DCE) MRI parameters as radiomic features of cervical cancer. Med Phys 2019; 46:5098-5109. [PMID: 31523829 DOI: 10.1002/mp.13821] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 07/30/2019] [Accepted: 09/05/2019] [Indexed: 12/26/2022] Open
Abstract
PURPOSE To evaluate whether the analysis of high-temporal resolution DCE-MRI by various tracer kinetic models could yield useful radiomic features in discriminating cervix carcinoma and normal cervix tissue. METHODS Forty-three patients (median age 51 yr; range 26-78 yr) diagnosed with cervical cancer based on postoperative pathology were enrolled in this study with informed consent. DCE-MRI data with temporal resolution of 2 s were acquired and analyzed using the Tofts (TOFTS), extended Tofts (EXTOFTS), conventional two-compartment (CC), adiabatic tissue homogeneity (ATH), and distributed parameter (DP) models. Ability of all kinetic parameters in distinguishing tumor from normal tissue was assessed using Mann-Whitney U test and receiver operating characteristic (ROC) curves. Repeatability of parameter estimates due to sampling of arterial input functions (AIFs) was also studied using intraclass correlation (ICC) analysis. RESULTS Fractional extravascular, extracellular volume (Ve) of all models were significantly smaller in cervix carcinoma than normal cervix tissue, and were associated with large values of area under ROC curve (AUC 0.884-0.961). Capillary permeability PS derived from the ATH, CC, and DP models also yielded large AUC values (0.730, 0.860, and 0.797). Transfer constant Ktrans derived from TOFTS and EXTOFTS models yielded smaller AUC (0.587 and 0.701). Repeatability of parameters derived from all models was robust to AIF sampling, with ICC coefficients typically larger than 0.80. CONCLUSIONS With the use of high-temporal resolution DCE-MRI, all tracer kinetic models could reflect pathophysiological differences between cervix carcinoma and normal tissue (with significant differences in Ve and PS) and potentially yield radiomic features with diagnostic value.
Collapse
Affiliation(s)
- Yi Lu
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Wenwen Peng
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Jiao Song
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Tao Chen
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Xue Wang
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Zujun Hou
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Zhihan Yan
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Tong San Koh
- Department of Oncologic Imaging, National Cancer Centre, 247969, Singapore, Singapore
| |
Collapse
|
28
|
Vuong L, Kotecha RR, Voss MH, Hakimi AA. Tumor Microenvironment Dynamics in Clear-Cell Renal Cell Carcinoma. Cancer Discov 2019; 9:1349-1357. [PMID: 31527133 DOI: 10.1158/2159-8290.cd-19-0499] [Citation(s) in RCA: 277] [Impact Index Per Article: 46.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/02/2019] [Accepted: 08/02/2019] [Indexed: 12/30/2022]
Abstract
Renal cell carcinoma stands out as one of the most immune-infiltrated tumors in pan-cancer comparisons. Features of the tumor microenvironment heavily affect disease biology and may affect responses to systemic therapy. With evolving frontline options in the metastatic setting, several immune checkpoint blockade regimens have emerged as efficacious, and there is growing interest in characterizing features of tumor biology that can reproducibly prognosticate patients and/or predict the likelihood of their deriving therapeutic benefit. Herein, we review pertinent characteristics of the tumor microenvironment with dedicated attention to candidate prognostic and predictive signatures as well as possible targets for future drug development. SIGNIFICANCE: Tumor microenvironment features broadly characterizing angiogenesis and inflammatory signatures have shown striking differences in response to immune checkpoint blockade and antiangiogenic agents. Integration of stromal and immune biomarkers may hence produce predictive and prognostic signatures to guide management with existing regimens as well as future drug development.
Collapse
Affiliation(s)
- Lynda Vuong
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ritesh R Kotecha
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Martin H Voss
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - A Ari Hakimi
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York. .,Department of Urology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
29
|
Woolf DK, Li SP, Detre S, Liu A, Gogbashian A, Simcock IC, Stirling J, Kosmin M, Cook GJ, Siddique M, Dowsett M, Makris A, Goh V. Assessment of the Spatial Heterogeneity of Breast Cancers: Associations Between Computed Tomography and Immunohistochemistry. BIOMARKERS IN CANCER 2019; 11:1179299X19851513. [PMID: 31210736 PMCID: PMC6552350 DOI: 10.1177/1179299x19851513] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 04/23/2019] [Indexed: 11/17/2022]
Abstract
BACKGROUND Tumour heterogeneity is considered an important mechanism of treatment failure. Imaging-based assessment of tumour heterogeneity is showing promise but the relationship between these mathematically derived measures and accepted 'gold standards' of tumour biology such as immunohistochemical measures is not established. METHODS A total of 20 women with primary breast cancer underwent a research dynamic contrast-enhanced computed tomography prior to treatment with data being available for 15 of these. Texture analysis was performed of the primary tumours to extract 13 locoregional and global parameters. Immunohistochemical analysis associations were assessed by the Spearman rank correlation. RESULTS Hypoxia-inducible factor-1α was correlated with first-order kurtosis (r = -0.533, P = .041) and higher order neighbourhood grey-tone difference matrix coarseness (r = 0.54, P = .038). Vascular maturity-related smooth muscle actin was correlated with higher order grey-level run-length long-run emphasis (r = -0.52, P = .047), fractal dimension (r = 0.613, P = .015), and lacunarity (r = -0.634, P = .011). Micro-vessel density, reflecting angiogenesis, was also associated with lacunarity (r = 0.547, P = .035). CONCLUSIONS The associations suggest a biological basis for these image-based heterogeneity features and support the use of imaging, already part of standard care, for assessing intratumoural heterogeneity.
Collapse
Affiliation(s)
- David K Woolf
- Breast Cancer Research Unit, Mount Vernon Cancer Centre, Northwood, UK
- Department of Clinical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Sonia P Li
- Breast Cancer Research Unit, Mount Vernon Cancer Centre, Northwood, UK
| | - Simone Detre
- Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital, London, UK
| | - Alison Liu
- Division of Imaging Sciences, King’s College London, St Thomas’ Hospital, London, UK
| | - Andrew Gogbashian
- Paul Strickland Scanner Centre, Mount Vernon Cancer Centre, Northwood, UK
| | - Ian C Simcock
- Paul Strickland Scanner Centre, Mount Vernon Cancer Centre, Northwood, UK
| | - James Stirling
- Paul Strickland Scanner Centre, Mount Vernon Cancer Centre, Northwood, UK
| | - Michael Kosmin
- Breast Cancer Research Unit, Mount Vernon Cancer Centre, Northwood, UK
| | - Gary J Cook
- Division of Imaging Sciences, King’s College London, St Thomas’ Hospital, London, UK
| | - Muhammad Siddique
- Division of Imaging Sciences, King’s College London, St Thomas’ Hospital, London, UK
| | - Mitch Dowsett
- Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital, London, UK
| | - Andreas Makris
- Breast Cancer Research Unit, Mount Vernon Cancer Centre, Northwood, UK
| | - Vicky Goh
- Division of Imaging Sciences, King’s College London, St Thomas’ Hospital, London, UK
- Paul Strickland Scanner Centre, Mount Vernon Cancer Centre, Northwood, UK
| |
Collapse
|
30
|
Alessandrino F, Shinagare AB, Bossé D, Choueiri TK, Krajewski KM. Radiogenomics in renal cell carcinoma. Abdom Radiol (NY) 2019; 44:1990-1998. [PMID: 29713740 DOI: 10.1007/s00261-018-1624-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Radiogenomics, a field of radiology investigating the association between the imaging features of a disease and its gene expression pattern, has expanded considerably in the last few years. Recent advances in whole-genome sequencing of clear cell renal cell carcinoma (ccRCC) and the identification of mutations with prognostic significance have led to increased interest in the relationship between imaging and genomic data. ccRCC is particularly suitable for radiogenomic analysis as the relative paucity of mutated genes allows for more straightforward genomic-imaging associations. The ultimate aim of radiogenomics of ccRCC is to retrieve additional data for accurate diagnosis, prognostic stratification, and optimization of therapy. In this review article, we will present the state-of-the-art of radiogenomics of ccRCC, and after briefly reviewing updates in genomics, we will discuss imaging-genomic associations for diagnosis and staging, prognosis, and for assessment of optimal therapy in ccRCC.
Collapse
Affiliation(s)
- Francesco Alessandrino
- Department of Imaging, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02215, USA.
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA.
| | - Atul B Shinagare
- Department of Imaging, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02215, USA
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Dominick Bossé
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Dana 1230, Boston, MA, 02215, USA
| | - Toni K Choueiri
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Dana 1230, Boston, MA, 02215, USA
| | - Katherine M Krajewski
- Department of Imaging, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02215, USA
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| |
Collapse
|
31
|
Deep learning and radiomics: the utility of Google TensorFlow™ Inception in classifying clear cell renal cell carcinoma and oncocytoma on multiphasic CT. Abdom Radiol (NY) 2019; 44:2009-2020. [PMID: 30778739 DOI: 10.1007/s00261-019-01929-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE Currently, all solid enhancing renal masses without microscopic fat are considered malignant until proven otherwise and there is substantial overlap in the imaging findings of benign and malignant renal masses, particularly between clear cell RCC (ccRCC) and benign oncocytoma (ONC). Radiomics has attracted increased attention for its utility in pre-operative work-up on routine clinical images. Radiomics based approaches have converted medical images into mineable data and identified prognostic imaging signatures that machine learning algorithms can use to construct predictive models by learning the decision boundaries of the underlying data distribution. The TensorFlow™ framework from Google is a state-of-the-art open-source software library that can be used for training deep learning neural networks for performing machine learning tasks. The purpose of this study was to investigate the diagnostic value and feasibility of a deep learning-based renal lesion classifier using open-source Google TensorFlow™ Inception in differentiating ccRCC from ONC on routine four-phase MDCT in patients with pathologically confirmed renal masses. METHODS With institutional review board approval for this 1996 Health Insurance Portability and Accountability Act compliant retrospective study and a waiver of informed consent, we queried our institution's pathology, clinical, and radiology databases for histologically proven cases of ccRCC and ONC obtained between January 2000 and January 2016 scanned with a an intravenous contrast-enhanced four-phase renal mass protocol (unenhanced (UN), corticomedullary (CM), nephrographic (NP), and excretory (EX) phases). To extract features to be used for the machine learning model, the entire renal mass was contoured in the axial plane in each of the four phases, resulting in a 3D volume of interest (VOI) representative of the entire renal mass. We investigated thirteen different approaches to convert the acquired VOI data into a set of images that adequately represented each tumor which was used to train the final layer of the neural network model. Training was performed over 4000 iterations. In each iteration, 90% of the data were designated as training data and the remaining 10% served as validation data and a leave-one-out cross-validation scheme was implemented. Accuracy, sensitivity, specificity, positive (PPV) and negative predictive (NPV) values, and CIs were calculated for the classification of the thirteen processing modes. RESULTS We analyzed 179 consecutive patients with 179 lesions (128 ccRCC and 51 ONC). The ccRCC cohort had a mean size of 3.8 cm (range 0.8-14.6 cm) and the ONC cohort had a mean lesion size of 3.9 cm (range 1.0-13.1 cm). The highest specificity and PPV (52.9% and 80.3%, respectively) were achieved in the EX phase when we analyzed the single mid-slice of the tumor in the axial, coronal and sagittal plane, and when we increased the number of mid-slices of the tumor to three, with an accuracy of 75.4%, which also increased the sensitivity to 88.3% and the PPV to 79.6%. Using the entire tumor volume also showed that classification performance was best in the EX phase with an accuracy of 74.4%, a sensitivity of 85.8% and a PPV of 80.1%. When the entire tumor volume, plus mid-slices from all phases and all planes presented as tiled images, were submitted to the final layer of the neural network we achieved a PPV of 82.5%. CONCLUSIONS The best classification result was obtained in the EX phase among the thirteen classification methods tested. Our proof of concept study is the first step towards understanding the utility of machine learning in the differentiation of ccRCC from ONC on routine CT images. We hope this could lead to future investigation into the development of a multivariate machine learning model which may augment our ability to accurately predict renal lesion histology on imaging.
Collapse
|
32
|
Branchini M, Zorz A, Zucchetta P, Bettinelli A, De Monte F, Cecchin D, Paiusco M. Impact of acquisition count statistics reduction and SUV discretization on PET radiomic features in pediatric 18F-FDG-PET/MRI examinations. Phys Med 2019; 59:117-126. [PMID: 30928060 DOI: 10.1016/j.ejmp.2019.03.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 03/02/2019] [Accepted: 03/07/2019] [Indexed: 01/09/2023] Open
Abstract
PURPOSE The evaluation of features robustness with respect to acquisition and post-processing parameter changes is fundamental for the reliability of radiomics studies. The aim of this study was to investigate the sensitivity of PET radiomic features to acquisition statistics reduction and standardized-uptake-volume (SUV) discretization in PET/MRI pediatric examinations. METHODS Twenty-seven lesions were detected from the analysis of twenty-one 18F-FDG-PET/MRI pediatric examinations. By decreasing the count-statistics of the original list-mode data (3 MBq/kg), injected activity reduction was simulated. Two SUV discretization approaches were applied: 1) resampling lesion SUV range into fixed bins numbers (FBN); 2) rounding lesion SUV into fixed bin size (FBS). One hundred and six radiomic features were extracted. Intraclass Correlation Coefficient (ICC), Spearman correlation coefficient and coefficient-of-variation (COV) were calculated to assess feature reproducibility between low tracer activities and full tracer activity feature values. RESULTS More than 70% of Shape and first order features, and around 70% and 40% of textural features, when using FBS and FBN methods respectively, resulted robust till 1.2 MBk/kg. Differences in median features reproducibility (ICC) between FBS and FBN datasets were statistically significant for every activity level independently from bin number/size, with higher values for FBS. Differences in median Spearman coefficient (i.e. patient ranking according to feature values) were not statistically significant, varying the intensity resolution (i.e. bin number/size) for either FBS and FBN methods. CONCLUSIONS For each simulated count-statistic level, robust PET radiomic features were determined for pediatric PET/MRI examinations. A larger number of robust features were detected when using FBS methods.
Collapse
Affiliation(s)
- Marco Branchini
- Medical Physics Department, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy.
| | - Alessandra Zorz
- Medical Physics Department, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Pietro Zucchetta
- Nuclear Medicine Unit, Department of Medicine DIMED, University Hospital of Padua, Padova, Italy
| | - Andrea Bettinelli
- Medical Physics Department, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Francesca De Monte
- Medical Physics Department, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Diego Cecchin
- Nuclear Medicine Unit, Department of Medicine DIMED, University Hospital of Padua, Padova, Italy
| | - Marta Paiusco
- Medical Physics Department, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| |
Collapse
|
33
|
Coy H, Young JR, Douek ML, Pantuck A, Brown MS, Sayre J, Raman SS. Association of qualitative and quantitative imaging features on multiphasic multidetector CT with tumor grade in clear cell renal cell carcinoma. Abdom Radiol (NY) 2019; 44:180-189. [PMID: 29987358 DOI: 10.1007/s00261-018-1688-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE The purpose of the study was to determine if enhancement features and qualitative imaging features on multiphasic multidetector computed tomography (MDCT) were associated with tumor grade in patients with clear cell renal cell carcinoma (ccRCC). METHODS In this retrospective, IRB approved, HIPAA-compliant, institutional review board-approved study with waiver of informed consent, 127 consecutive patients with 89 low grade (LG) and 43 high grade (HG) ccRCCs underwent preoperative four-phase MDCT in unenhanced (UN), corticomedullary (CM), nephrographic (NP), and excretory (EX) phases. Previously published quantitative (absolute peak lesion enhancement, absolute peak lesion enhancement relative to normal enhancing renal cortex, 3D whole lesion enhancement and the wash-in/wash-out of enhancement within the 3D whole lesion ROI) and qualitative (enhancement pattern; presence of necrosis; pattern of; tumor margin; tumor-parenchymal interface, tumor-parenchymal interaction; intratumoral vascularity; collecting system infiltration; renal vein invasion; and calcification) assessments were obtained for each lesion independently by two fellowship-trained genitourinary radiologists. Comparisons between variables included χ2, ANOVA, and student t test. p values less than 0.05 were considered to be significant. Inter-reader agreement was obtained with the Gwet agreement coefficient (AC1) and standard error (SE) was reported. RESULTS No significant differences were observed between the LG and HG ccRCC cohorts with respect to absolute peak lesion enhancement and relative lesion enhancement ratio. There was a significant inverse correlation between low and high grade ccRCC and tumor enhancement the NP (71 HU vs. 54 HU, p < 0.001) and EX (52 HU vs. 39 HU, p < 0.001) phases using the 3D whole lesion ROI method. The percent wash-in of 3D enhancement from the UN to the CM phase was also significantly different between LG and HG ccRCCs (352% vs. 255%, p = 0.003). HG lesions showed significantly more calcification, necrosis, collecting system infiltration and ill-defined tumor margins (p < 0.05). Overall agreement between the two readers had a mean AC1 of 0.8172 (SE 0.0235). CONCLUSIONS Quantitatively, high grade ccRCC had significantly lower whole lesion enhancement in the NP and EX phases on MDCT. Qualitatively, high grade ccRCC were significantly more likely to be associated with calcifications, necrosis, collecting system infiltration, and an ill-defined tumor margin.
Collapse
Affiliation(s)
- Heidi Coy
- Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Ronald Reagan-UCLA Medical Center, 924 Westwood Boulevard, Suite 650, Los Angeles, CA, 90024, USA.
| | - Jonathan R Young
- Department of Radiology, University of California, Davis, 4860 Y Street, Suite 3100, Sacramento, CA, 95817, USA
| | - Michael L Douek
- Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Ronald Reagan-UCLA Medical Center, 924 Westwood Boulevard, Suite 650, Los Angeles, CA, 90024, USA
| | - Alan Pantuck
- Department of Urology, David Geffen School of Medicine at UCLA, Ronald Reagan-UCLA Medical Center, Clark Urology Center-Westwood, 200 Medical Plaza, Suite 140, Los Angeles, CA, 90095, USA
| | - Matthew S Brown
- Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Ronald Reagan-UCLA Medical Center, 924 Westwood Boulevard, Suite 650, Los Angeles, CA, 90024, USA
| | - James Sayre
- Department of Biostatistics, UCLA School of Public Heath, Room 51-253A, Los Angeles, CA, 90095, USA
| | - Steven S Raman
- UCLA Department of Radiological Sciences, Ronald Reagan UCLA Medical Center, 757 Westwood Plaza, RRUMC 1621H, Box 957437, Los Angeles, CA, 90095, USA
| |
Collapse
|
34
|
Arimura H, Soufi M, Kamezawa H, Ninomiya K, Yamada M. Radiomics with artificial intelligence for precision medicine in radiation therapy. JOURNAL OF RADIATION RESEARCH 2019; 60:150-157. [PMID: 30247662 PMCID: PMC6373667 DOI: 10.1093/jrr/rry077] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 07/21/2018] [Indexed: 05/27/2023]
Abstract
Recently, the concept of radiomics has emerged from radiation oncology. It is a novel approach for solving the issues of precision medicine and how it can be performed, based on multimodality medical images that are non-invasive, fast and low in cost. Radiomics is the comprehensive analysis of massive numbers of medical images in order to extract a large number of phenotypic features (radiomic biomarkers) reflecting cancer traits, and it explores the associations between the features and patients' prognoses in order to improve decision-making in precision medicine. Individual patients can be stratified into subtypes based on radiomic biomarkers that contain information about cancer traits that determine the patient's prognosis. Machine-learning algorithms of AI are boosting the powers of radiomics for prediction of prognoses or factors associated with treatment strategies, such as survival time, recurrence, adverse events, and subtypes. Therefore, radiomic approaches, in combination with AI, may potentially enable practical use of precision medicine in radiation therapy by predicting outcomes and toxicity for individual patients.
Collapse
Affiliation(s)
- Hidetaka Arimura
- Division of Medical Quantum Science, Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, Japan
| | - Mazen Soufi
- Division of Information Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5, Takayama-cho, Ikoma, Nara, Japan
| | - Hidemi Kamezawa
- Department of Radiological Technology, Faculty of Fukuoka Medical Technology, Teikyo University, 6-22, Misaki-machi, Omuta, Fukuoka, Japan
| | - Kenta Ninomiya
- Division of Medical Quantum Science, Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, Japan
| | - Masahiro Yamada
- Division of Medical Quantum Science, Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, Japan
| |
Collapse
|
35
|
Ajith TA. Current insights and future perspectives of hypoxia-inducible factor-targeted therapy in cancer. J Basic Clin Physiol Pharmacol 2018; 30:11-18. [PMID: 30260792 DOI: 10.1515/jbcpp-2017-0167] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 07/31/2018] [Indexed: 06/08/2023]
Abstract
Hypoxia-inducible factors (HIFs) are transcription factors that are expressed in the hypoxic tumor microenvironment. They are involved in the cellular adaptations by improving the metabolism of glucose and enhance the expression of vascular endothelial growth factor, platelet-derived growth factor and angiopoietin, thereby they play a pivotal role in the angiogenesis. Hypoxia can increase the expression of nuclear factor-kappa B which promotes the pro-inflammatory status. Abnormally high angiogenesis, inflammation, antiapoptosis and anaerobic glycolysis can augment the progression and metastasis of tumor. Hence, HIFs remain one of the promising antiangiogenic agents as well as a direct target for interfering with the energetic of cancer cells in order to regulate the tumor growth. Previous studies found agents like topotecan, acriflavine and benzophenone-1B etc. to block the HIF-α mediated angiogenesis. The effect is mediated through interfering any one of the processes in the activation of HIF such as nuclear translocation of HIF-1α; dimerization of HIF-1α with β in the nucleus; HIF-1α/HIF-2α mediated induction of VEGF or translation of HIF-1α mRNA. Despite the experimental studies on the inhibitory molecules of HIFs, none of them are available for the clinical use. This review article discusses the recent update on the HIF-targeted therapy in cancer.
Collapse
Affiliation(s)
- Thekkuttuparambil A Ajith
- Professor Biochemistry, Department of Biochemistry, Amala Institute of Medical Sciences, Amala Nagar, Thrissur-680 555, Kerala, India
| |
Collapse
|
36
|
Meyer HJ, Höhn AK, Hamerla G, Surov A. Histogram parameters derived from T2 weighted images are associated with histopathological findings in rectal cancer - a preliminary study. Am J Transl Res 2018; 10:3790-3796. [PMID: 30662629 PMCID: PMC6291705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 09/01/2018] [Indexed: 06/09/2023]
Abstract
Histogram analysis can better reflect tumor heterogeneity than conventional imaging analysis. The present study analyzed possible correlations between histogram parameters derived from T2 weighted images and histopathological features in rectal cancer. Seventeen patients with histopathological proven rectal adenocarcinoma were retrospectively acquired with prebioptic 3 T MRI and available histopathological specimens. Histogram analysis was performed using an in-house matlab tool conducting a whole lesion measurement. Histopathology was investigated using Ki67 specimens with calculation of Ki67-index as well as cellularity and nucleic areas and CD31 specimens, with estimation of microvessel density. Several histogram parameters correlated with average nucleic area. Skewness showed a moderate correlation with microvessel density (P = 0.54, P = 0.02). None of the parameters correlated with Ki67-index. Skewness derived from T2 weighted images might be used as a surrogate parameter for average nucleic area and microvessel density. However, none of the parameters were associated with proliferation index.
Collapse
Affiliation(s)
- Hans-Jonas Meyer
- Department of Diagnostic and Interventional Radiology, University of LeipzigLeipzig 04103, Germany
| | - Anne Kathrin Höhn
- Department of Pathology, University of LeipzigLeipzig 04103, Germany
| | - Gordian Hamerla
- Department of Diagnostic and Interventional Radiology, University of LeipzigLeipzig 04103, Germany
| | - Alexey Surov
- Department of Diagnostic and Interventional Radiology, University of LeipzigLeipzig 04103, Germany
| |
Collapse
|
37
|
Shu J, Tang Y, Cui J, Yang R, Meng X, Cai Z, Zhang J, Xu W, Wen D, Yin H. Clear cell renal cell carcinoma: CT-based radiomics features for the prediction of Fuhrman grade. Eur J Radiol 2018; 109:8-12. [PMID: 30527316 DOI: 10.1016/j.ejrad.2018.10.005] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 08/22/2018] [Accepted: 10/04/2018] [Indexed: 01/08/2023]
Abstract
OBJECTIVES To discriminate low grade (Fuhrman I/II) and high grade (Fuhrman III/IV) clear cell renal cell carcinoma (CCRCC) by using CT-based radiomic features. METHODS 161 and 99 patients diagnosed with low and high grade CCRCCs from January 2011 to May 2018 were enrolled in this study. 1029 radiomic features were extracted from corticomedullary (CMP), and nephrographic phase (NP) CT images of all patients. We used interclass correlation coefficient (ICC) and the least absolute shrinkage and selection operator (LASSO) regression method to select features, then the selected features were constructed three classification models (CMP, NP and with their combination) to discriminate high and low grades CCRCC. These three models were built by logistic regression method using 5-fold cross validation strategy, evaluated with receiver operating characteristics curve (ROC) and compared using DeLong test. RESULTS We found 11 and 24 CMP and NP features were independently significantly associated with the Fuhrman grades. The model of CMP, NP and Combined model using radiomic feature set showed diagnostic accuracy of 0.719 (AUC [area under the curve], 0.766; 95% CI [confidence interval]: 0.709-0.816; sensitivity, 0.602; specificity, 0.838), 0.738 (AUC, 0.818; 95% CI:0.765-0.838; sensitivity, 0.693; specificity, 0.838), 0.777(AUC, 0.822; 95% CI: 0.769-0.866; sensitivity, 0.677; specificity, 0.839). There were significant differences in AUC between CMP model and Combined model (P = 0.0208), meanwhile, the differences between CMP model and NP model, NP model and Combined model reached no significant (P = 0.0844, 0.7915). CONCLUSIONS Radiomic features could be used as biomarker for the preoperative evaluation of the CCRCC Fuhrman grades.
Collapse
Affiliation(s)
- Jun Shu
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Changle West Road 127#, Xi'an City, 710032, People's Republic of China
| | - Yongqiang Tang
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Changle West Road 127#, Xi'an City, 710032, People's Republic of China
| | - Jingjing Cui
- Huiying Medical Technology Co., Ltd, Room C103, B2, Dongsheng Science and Technology Park, HaiDian District, Beijing City, 100192, People's Republic of China
| | - Ruwu Yang
- Department of Radiology, Xi'an XD Group Hospital, Shaanxi University of Chinese Medicine, FengDeng Road 97#, Xi'an City, 710077, People's Republic of China
| | - Xiaoli Meng
- Department of Radiology, Xi'an XD Group Hospital, Shaanxi University of Chinese Medicine, FengDeng Road 97#, Xi'an City, 710077, People's Republic of China
| | - Zhengting Cai
- Huiying Medical Technology Co., Ltd, Room C103, B2, Dongsheng Science and Technology Park, HaiDian District, Beijing City, 100192, People's Republic of China
| | - Jingsong Zhang
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Changle West Road 127#, Xi'an City, 710032, People's Republic of China
| | - Wanni Xu
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Changle West Road 127#, Xi'an City 710032, People's Republic of China
| | - Didi Wen
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Changle West Road 127#, Xi'an City, 710032, People's Republic of China
| | - Hong Yin
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Changle West Road 127#, Xi'an City, 710032, People's Republic of China.
| |
Collapse
|
38
|
Differentiation of Papillary Renal Cell Carcinoma Subtypes on MRI: Qualitative and Texture Analysis. AJR Am J Roentgenol 2018; 211:1234-1245. [PMID: 30240294 DOI: 10.2214/ajr.17.19213] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE The objective of this study was to determine whether quantitative texture analysis of MR images would improve the ability to distinguish papillary renal cell carcinoma (RCC) subtypes, compared with analysis of qualitative MRI features alone. MATERIALS AND METHODS A total of 47 pathologically proven papillary RCC tumors were retrospectively evaluated, with 31 (66%) classified as type 1 tumors and 16 (34%) classified as type 2 tumors. MR images were reviewed by two readers to determine tumor size, signal intensity, heterogeneity, enhancement pattern, margins, perilesional stranding, vein thrombosis, and metastasis. Quantitative texture analysis of gray-scale images was performed. A logistic regression was derived from qualitative and quantitative features. Model performance was compared with and without texture features. RESULTS The significant qualitative MR features noted were necrosis, enhancement appearance, perilesional stranding, and metastasis. A multivariable model based on qualitative features did not identify any factor as an independent predictor of a type 2 tumor. The logistic regression model for predicting papillary RCCs on the basis of qualitative and quantitative analysis identified probability of the 2D volumetric interpolated breath-hold examination (VIBE) sequence (AUC value, 0.87; 95% CI, 0.77-0.98) as an independent predictor of a type 2 tumor. No difference in the model AUC value was noted when texture features were included in the analysis; however, the model had increased sensitivity and an improved predictive value without loss of specificity. CONCLUSION The addition of texture analysis to analysis of conventional qualitative MRI features increased the probability of predicting a type 2 papillary RCC tumor, which may be clinically important.
Collapse
|
39
|
Wu Q, Shi D, Dou S, Shi L, Liu M, Dong L, Chang X, Wang M. Radiomics Analysis of Multiparametric MRI Evaluates the Pathological Features of Cervical Squamous Cell Carcinoma. J Magn Reson Imaging 2018; 49:1141-1148. [PMID: 30230114 DOI: 10.1002/jmri.26301] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 07/31/2018] [Accepted: 07/31/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Robust parameters to evaluate pathological aggressiveness are needed to provide individualized therapy for cervical cancer patients. PURPOSE To investigate the radiomics analysis of multiparametric MRI to evaluate tumor grade, lymphovascular space invasion (LVSI), and lymph node (LN) metastasis of cervical squamous cell carcinoma (CSCC). STUDY TYPE Retrospective. SUBJECTS Fifty-six patients with histopathologically confirmed CSCC. FIELD STRENGTH/SEQUENCE 3T, axial T2 and T2 with fat suppression (FS), diffusion-weighted imaging (DWI) (multi-b values), axial dynamic contrast enhanced (DCE) MRI (8 sec temporal resolution). ASSESSMENT Regions of interest were drawn around the tumor on each axial slice and fused to generate the whole tumor volume. Sixty-six radiomics features were derived from each image sequence, including axial T2 and T2 FS, ADC maps, and Ktrans , Ve , and Vp maps from DCE MRI. STATISTICAL TESTS A univariate analysis was performed to assess each parameter's association with tumor grade and the presence of lymphovascular space invasion (LVSI) and lymph node (LN) metastasis. A principal component analysis was employed for dimension reduction and to generate new discriminative valuables. Using logistic regression, a discriminative model of each parameter was built and a receiver operating characteristic curve (ROC) was generated. RESULTS The area under the ROC curve (AUC) of anatomical, diffusion, and permeability parameters in discriminating the presence of LVSI ranged from 0.659 to 0.814, with Ve showing the best discriminative value. The AUC in discriminating the presence of LN metastasis and distinguishing tumor grade ranged from 0.747 to 0.850, 0.668 to 0.757, with ADC and Ve showing the best discriminative value, respectively. DATA CONCLUSION Functional maps exhibit better discriminative values than anatomical images for discriminating the pathological features of CSCC, with ADC maps showing the best discrimination performance for LN metastasis and Ve maps showing the best discriminative value for LVSI and tumor grade. LEVEL OF EVIDENCE 3 Technical Efficacy: Stage 2 J. Magn. Reson. Imaging 2019;49:1141-1148.
Collapse
Affiliation(s)
- Qingxia Wu
- Radiological Department of Henan Provincial People's Hospital, Zhengzhou, Henan, P.R. China
| | - Dapeng Shi
- Radiological Department of Henan Provincial People's Hospital, Zhengzhou, Henan, P.R. China
| | - Shewei Dou
- Radiological Department of Henan Provincial People's Hospital, Zhengzhou, Henan, P.R. China
| | - Ligang Shi
- Pathological Department of Henan Provincial People's Hospital, Zhengzhou, Henan, P.R. China
| | - Mingbo Liu
- Radiotherapeutical Department of Henan Provincial People's Hospital, Zhengzhou, Henan, P.R. China
| | - Li Dong
- Obstetrics and Gynecology Department of Henan Provincial People's Hospital, Zhengzhou, Henan, P.R. China
| | - Xiaowan Chang
- Operation and Management Department of Henan Provincial People's Hospital, Zhengzhou, Henan, P.R. China
| | - Meiyun Wang
- Radiological Department of Henan Provincial People's Hospital, Zhengzhou, Henan, P.R. China
| |
Collapse
|
40
|
Abstract
Precision medicine is increasingly pushed forward, also with respect to upcoming new targeted therapies. Individual characterization of diseases on the basis of biomarkers is a prerequisite for this development. So far, biomarkers are characterized clinically, histologically or on a molecular level. The implementation of broad screening methods (“Omics”) and the analysis of big data – in addition to single markers – allow to define biomarker signatures. Next to “Genomics”, “Proteomics”, and “Metabolicis”, “Radiomics” gained increasing interest during the last years. Based on radiologic imaging, multiple radiomic markers are extracted with the help of specific algorithms. These are correlated with clinical, (immuno-) histopathological, or genomic data. Underlying structural differences are based on the imaging metadata and are often not visible and therefore not detectable without specific software. Radiomics are depicted numerically or by graphs. The fact that radiomic information can be extracted from routinely performed imaging adds a specific appeal to this method. Radiomics could potentially replace biopsies and additional investigations. Alternatively, radiomics could complement other biomarkers and thus lead to a more precise, multimodal prediction. Until now, radiomics are primarily used to investigate solid tumors. Some promising studies in head and neck cancer have already been published.
Collapse
|
41
|
Salo T, Dourado MR, Sundquist E, Apu EH, Alahuhta I, Tuomainen K, Vasara J, Al-Samadi A. Organotypic three-dimensional assays based on human leiomyoma-derived matrices. Philos Trans R Soc Lond B Biol Sci 2018; 373:rstb.2016.0482. [PMID: 29158312 PMCID: PMC5717437 DOI: 10.1098/rstb.2016.0482] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2017] [Indexed: 12/19/2022] Open
Abstract
Alongside cancer cells, tumours exhibit a complex stroma containing a repertoire of cells, matrix molecules and soluble factors that actively crosstalk between each other. Recognition of this multifaceted concept of the tumour microenvironment (TME) calls for authentic TME mimetics to study cancer in vitro. Traditionally, tumourigenesis has been investigated in non-human, three-dimensional rat type I collagen containing organotypic discs or by means of mouse sarcoma-derived gel, such as Matrigel®. However, the molecular compositions of these simplified assays do not properly simulate human TME. Here, we review the main properties and benefits of using human leiomyoma discs and their matrix Myogel for in vitro assays. Myoma discs are practical for investigating the invasion of cancer cells, as are cocultures of cancer and stromal cells in a stiff, hypoxic TME mimetic. Myoma discs contain soluble factors and matrix molecules commonly present in neoplastic stroma. In Transwell, IncuCyte, spheroid and sandwich assays, cancer cells move faster and form larger colonies in Myogel than in Matrigel®. Additionally, Myogel can replace Matrigel® in hanging-drop and tube-formation assays. Myogel also suits three-dimensional drug testing and extracellular vesicle interactions. To conclude, we describe the application of our myoma-derived matrices in 3D in vitro cancer assays. This article is part of the discussion meeting issue ‘Extracellular vesicles and the tumour microenvironment’.
Collapse
Affiliation(s)
- Tuula Salo
- Cancer and Translational Medicine Research Unit, University of Oulu, Oulu 90014, Finland .,Medical Research Centre, Oulu University Hospital, Oulu, Finland.,Department of Oral and Maxillofacial Diseases, University of Helsinki, Helsinki 0014, Finland.,Helsinki University Hospital, Helsinki 0014, Finland.,Department of Oral Diagnosis, Oral Pathology Division, Piracicaba Dental School, University of Campinas, Campinas 13414-903, Brazil
| | - Mauricio Rocha Dourado
- Cancer and Translational Medicine Research Unit, University of Oulu, Oulu 90014, Finland.,Medical Research Centre, Oulu University Hospital, Oulu, Finland.,Department of Oral Diagnosis, Oral Pathology Division, Piracicaba Dental School, University of Campinas, Campinas 13414-903, Brazil
| | - Elias Sundquist
- Cancer and Translational Medicine Research Unit, University of Oulu, Oulu 90014, Finland.,Medical Research Centre, Oulu University Hospital, Oulu, Finland
| | - Ehsanul Hoque Apu
- Cancer and Translational Medicine Research Unit, University of Oulu, Oulu 90014, Finland.,Medical Research Centre, Oulu University Hospital, Oulu, Finland
| | - Ilkka Alahuhta
- Cancer and Translational Medicine Research Unit, University of Oulu, Oulu 90014, Finland.,Medical Research Centre, Oulu University Hospital, Oulu, Finland
| | - Katja Tuomainen
- Department of Oral and Maxillofacial Diseases, University of Helsinki, Helsinki 0014, Finland
| | - Jenni Vasara
- Department of Oral and Maxillofacial Diseases, University of Helsinki, Helsinki 0014, Finland
| | - Ahmed Al-Samadi
- Department of Oral and Maxillofacial Diseases, University of Helsinki, Helsinki 0014, Finland
| |
Collapse
|
42
|
Sanduleanu S, Woodruff HC, de Jong EE, van Timmeren JE, Jochems A, Dubois L, Lambin P. Tracking tumor biology with radiomics: A systematic review utilizing a radiomics quality score. Radiother Oncol 2018; 127:349-360. [DOI: 10.1016/j.radonc.2018.03.033] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 03/02/2018] [Accepted: 03/29/2018] [Indexed: 02/07/2023]
|
43
|
Yin Q, Hung SC, Rathmell WK, Shen L, Wang L, Lin W, Fielding JR, Khandani AH, Woods ME, Milowsky MI, Brooks SA, Wallen EM, Shen D. Integrative radiomics expression predicts molecular subtypes of primary clear cell renal cell carcinoma. Clin Radiol 2018; 73:782-791. [PMID: 29801658 DOI: 10.1016/j.crad.2018.04.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 04/17/2018] [Indexed: 02/06/2023]
Abstract
AIM To identify combined positron-emission tomography (PET)/magnetic resonance imaging (MRI)-based radiomics as a surrogate biomarker of intratumour disease risk for molecular subtype ccA and ccB in patients with primary clear cell renal cell carcinoma (ccRCC). MATERIALS AND METHODS PET/MRI data were analysed retrospectively from eight patients. One hundred and sixty-eight radiomics features for each tumour sampling based on the regionally sampled tumours with 23 specimens were extracted. Sparse partial least squares discriminant analysis (SPLS-DA) was applied to feature screening on high-throughput radiomics features and project the selected features to low-dimensional intrinsic latent components as radiomics signatures. In addition, multilevel omics datasets were leveraged to explore the complementing information and elevate the discriminative ability. RESULTS The correct classification rate (CCR) for molecular subtype classification by SPLS-DA using only radiomics features was 86.96% with permutation test p=7×10-4. When multi-omics datasets including mRNA, microvascular density, and clinical parameters from each specimen were combined with radiomics features to refine the model of SPLS-DA, the best CCR was 95.65% with permutation test, p<10-4; however, even in the case of generating the classification based on transcription features, which is the reference standard, there is roughly 10% classification ambiguity. Thus, this classification level (86.96-95.65%) of the proposed method represents the discriminating level that is consistent with reality. CONCLUSION Featured with high accuracy, an integrated multi-omics model of PET/MRI-based radiomics could be the first non-invasive investigation for disease risk stratification and guidance of treatment in patients with primary ccRCC.
Collapse
Affiliation(s)
- Q Yin
- Information Science and Technology College, Dalian Maritime University, Dalian, 116026, China; Department of Radiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - S-C Hung
- Department of Radiology, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Radiology, Taipei Veterans General Hospital, Taipei 11217, Taiwan; School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan; Department of Biomedical Imaging and Radiological Sciences, School of Biomedical Science of Engineering, National Yang-Ming University, Taipei 11221, Taiwan
| | - W K Rathmell
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN 37232, USA
| | - L Shen
- Information Science and Technology College, Dalian Maritime University, Dalian, 116026, China
| | - L Wang
- Department of Radiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - W Lin
- Department of Radiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - J R Fielding
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - A H Khandani
- Department of Radiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - M E Woods
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Urology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - M I Milowsky
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Urology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - S A Brooks
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - E M Wallen
- Department of Urology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - D Shen
- Department of Radiology, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Brain and Cognitive Engineering, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
44
|
Mannheim JG, Schmid AM, Schwenck J, Katiyar P, Herfert K, Pichler BJ, Disselhorst JA. PET/MRI Hybrid Systems. Semin Nucl Med 2018; 48:332-347. [PMID: 29852943 DOI: 10.1053/j.semnuclmed.2018.02.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Over the last decade, the combination of PET and MRI in one system has proven to be highly successful in basic preclinical research, as well as in clinical research. Nowadays, PET/MRI systems are well established in preclinical imaging and are progressing into clinical applications to provide further insights into specific diseases, therapeutic assessments, and biological pathways. Certain challenges in terms of hardware had to be resolved concurrently with the development of new techniques to be able to reach the full potential of both combined techniques. This review provides an overview of these challenges and describes the opportunities that simultaneous PET/MRI systems can exploit in comparison with stand-alone or other combined hybrid systems. New approaches were developed for simultaneous PET/MRI systems to correct for attenuation of 511 keV photons because MRI does not provide direct information on gamma photon attenuation properties. Furthermore, new algorithms to correct for motion were developed, because MRI can accurately detect motion with high temporal resolution. The additional information gained by the MRI can be employed to correct for partial volume effects as well. The development of new detector designs in combination with fast-decaying scintillator crystal materials enabled time-of-flight detection and incorporation in the reconstruction algorithms. Furthermore, this review lists the currently commercially available systems both for preclinical and clinical imaging and provides an overview of applications in both fields. In this regard, special emphasis has been placed on data analysis and the potential for both modalities to evolve with advanced image analysis tools, such as cluster analysis and machine learning.
Collapse
Affiliation(s)
- Julia G Mannheim
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Andreas M Schmid
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Johannes Schwenck
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany; Department of Nuclear Medicine and Clinical Molecular Imaging, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Prateek Katiyar
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Kristina Herfert
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Bernd J Pichler
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany.
| | - Jonathan A Disselhorst
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University Tuebingen, Tuebingen, Germany
| |
Collapse
|
45
|
Abstract
Radiomics, the high-throughput mining of quantitative image features from standard-of-care medical imaging that enables data to be extracted and applied within clinical-decision support systems to improve diagnostic, prognostic, and predictive accuracy, is gaining importance in cancer research. Radiomic analysis exploits sophisticated image analysis tools and the rapid development and validation of medical imaging data that uses image-based signatures for precision diagnosis and treatment, providing a powerful tool in modern medicine. Herein, we describe the process of radiomics, its pitfalls, challenges, opportunities, and its capacity to improve clinical decision making, emphasizing the utility for patients with cancer. Currently, the field of radiomics lacks standardized evaluation of both the scientific integrity and the clinical relevance of the numerous published radiomics investigations resulting from the rapid growth of this area. Rigorous evaluation criteria and reporting guidelines need to be established in order for radiomics to mature as a discipline. Herein, we provide guidance for investigations to meet this urgent need in the field of radiomics.
Collapse
|
46
|
Xie T, Chen X, Fang J, Kang H, Xue W, Tong H, Cao P, Wang S, Yang Y, Zhang W. Textural features of dynamic contrast-enhanced MRI derived model-free and model-based parameter maps in glioma grading. J Magn Reson Imaging 2017; 47:1099-1111. [PMID: 28845594 DOI: 10.1002/jmri.25835] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 07/25/2017] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Presurgical glioma grading by dynamic contrast-enhanced MRI (DCE-MRI) has unresolved issues. PURPOSE The aim of this study was to investigate the ability of textural features derived from pharmacokinetic model-based or model-free parameter maps of DCE-MRI in discriminating between different grades of gliomas, and their correlation with pathological index. STUDY TYPE Retrospective. SUBJECTS Forty-two adults with brain gliomas. FIELD STRENGTH/SEQUENCE 3.0T, including conventional anatomic sequences and DCE-MRI sequences (variable flip angle T1-weighted imaging and three-dimensional gradient echo volumetric imaging). ASSESSMENT Regions of interest on the cross-sectional images with maximal tumor lesion. Five commonly used textural features, including Energy, Entropy, Inertia, Correlation, and Inverse Difference Moment (IDM), were generated. RESULTS All textural features of model-free parameters (initial area under curve [IAUC], maximal signal intensity [Max SI], maximal up-slope [Max Slope]) could effectively differentiate between grade II (n = 15), grade III (n = 13), and grade IV (n = 14) gliomas (P < 0.05). Two textural features, Entropy and IDM, of four DCE-MRI parameters, including Max SI, Max Slope (model-free parameters), vp (Extended Tofts), and vp (Patlak) could differentiate grade III and IV gliomas (P < 0.01) in four measurements. Both Entropy and IDM of Patlak-based Ktrans and vp could differentiate grade II (n = 15) from III (n = 13) gliomas (P < 0.01) in four measurements. No textural features of any DCE-MRI parameter maps could discriminate between subtypes of grade II and III gliomas (P < 0.05). Both Entropy and IDM of Extended Tofts- and Patlak-based vp showed highest area under curve in discriminating between grade III and IV gliomas. However, intraclass correlation coefficient (ICC) of these features revealed relatively lower inter-observer agreement. No significant correlation was found between microvascular density and textural features, compared with a moderate correlation found between cellular proliferation index and those features. DATA CONCLUSION Textural features of DCE-MRI parameter maps displayed a good ability in glioma grading. LEVEL OF EVIDENCE 3 Technical Efficacy: Stage 2 J. Magn. Reson. Imaging 2018;47:1099-1111.
Collapse
Affiliation(s)
- Tian Xie
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Xiao Chen
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Jingqin Fang
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Houyi Kang
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Wei Xue
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Haipeng Tong
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Peng Cao
- GE HealthCare (China), Pudong, Shanghai, China
| | - Sumei Wang
- Department of Radiology, Division of Neuroradiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yizeng Yang
- Department of Medicine, Gastroenterology Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Weiguo Zhang
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China.,Chongqing Clinical Research Center of Imaging and Nuclear Medicine, Chongqing, China
| |
Collapse
|
47
|
PET-MRI of the Pancreas and Kidneys. CURRENT RADIOLOGY REPORTS 2017. [DOI: 10.1007/s40134-017-0229-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|