1
|
Yang Z, Wang S, Yin W, Wang Y, Liu F, Xu J, Han L, Liu C. Radiomics-clinical nomogram for preoperative tumor-node-metastasis staging prediction in breast cancer patients using dynamic enhanced magnetic resonance imaging. Transl Cancer Res 2025; 14:1836-1848. [PMID: 40225004 PMCID: PMC11985186 DOI: 10.21037/tcr-24-1559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 01/09/2025] [Indexed: 04/15/2025]
Abstract
Background Breast cancer is one of the most commonly diagnosed malignancies in women worldwide, and the disease burden continues to aggravate. The tumor-node-metastasis (TNM) staging information is crucial for oncology physicians to develop appropriate clinical strategies. This study aimed to investigate the value of a radiomics-clinical model for predicting TNM stage in breast cancer patients using dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI). Methods DCE-MRI images from 166 patients with pathologically confirmed breast cancer were retrospectively collected, including early stage (TNM0-TNM2) and locally advanced or advanced stage (TNM3-TNM4). Included patients were divided into a training cohort (n=116) and a test cohort (n=50). The radiomics, clinical and integrated models were constructed and a nomogram was established to distinguish the TNM0-TNM2 stage from the TNM3-TNM4 stage. Receiver operating characteristic (ROC) curves, calibration curves and decision curve analysis (DCA) were employed to assess the predictability of the models. Results Eighty-five patients were at the early stages, while 81 patients were at the other stages. In the training and test cohorts, the area under the curve (AUC) values for distinguishing early and advanced breast cancer were 0.870 and 0.818 for the nomogram, respectively. The nomogram calibration curves showed good agreement between the predicted and observed TNM stages in the training and test cohorts. The Hosmer-Lemeshow test showed that the nomogram fit perfectly in the two cohorts. DCA indicated that the nomogram displayed clear superiority in forecasting TNM staging over clinical and radiomic signatures. Conclusions Compared to traditional imaging methods, the clinical-radiomics nomogram acquired by DCE-MRI could potentially be utilized to preoperatively evaluate the TNM stage of breast cancer with relatively high accuracy. It can be an effective method to guide clinical decisions.
Collapse
Affiliation(s)
- Zhe Yang
- Department of Radiology, the Second Affiliated Hospital of Shandong First Medical University, Tai’an, China
| | - Shouen Wang
- Department of Pathology, the Second Affiliated Hospital of Shandong First Medical University, Tai’an, China
| | - Wei Yin
- Department of Radiology, Beijing Friendship Hospital of Capital Medical University, Beijing, China
| | - Ying Wang
- Department of Radiology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Fanghua Liu
- Department of Radiology, the Second Affiliated Hospital of Shandong First Medical University, Tai’an, China
| | - Jianshu Xu
- Department of Radiology, the Second Affiliated Hospital of Shandong First Medical University, Tai’an, China
| | - Long Han
- Department of Radiology, the Second Affiliated Hospital of Shandong First Medical University, Tai’an, China
| | - Chenglong Liu
- Department of Radiology, the Second Affiliated Hospital of Shandong First Medical University, Tai’an, China
| |
Collapse
|
2
|
Kotsifa E, Saffioti F, Mavroeidis VK. Cholangiocarcinoma: The era of liquid biopsy. World J Gastroenterol 2025; 31:104170. [PMID: 40124277 PMCID: PMC11924015 DOI: 10.3748/wjg.v31.i11.104170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/28/2025] [Accepted: 02/14/2025] [Indexed: 03/13/2025] Open
Abstract
Cholangiocarcinoma (CCA) is a highly aggressive and heterogeneous malignancy arising from the epithelial cells of the biliary tract. The limitations of the current methods in the diagnosis of CCA highlight the urgent need for new, accurate tools for early cancer detection, better prognostication and patient monitoring. Liquid biopsy (LB) is a modern and non-invasive technique comprising a diverse group of methodologies aiming to detect tumour biomarkers from body fluids. These biomarkers include circulating tumour cells, cell-free DNA, circulating tumour DNA, RNA and extracellular vesicles. The aim of this review is to explore the current and potential future applications of LB in CCA management, with a focus on diagnosis, prognostication and monitoring. We examine both its significant potential and the inevitable limitations associated with this technology. We conclude that LB holds considerable promise, but further research is necessary to fully integrate it into precision oncology for CCA.
Collapse
Affiliation(s)
- Evgenia Kotsifa
- The Second Propaedeutic Department of Surgery, National and Kapodistrian University of Athens, General Hospital of Athens “Laiko”, Athens 11527, Greece
| | - Francesca Saffioti
- Department of Gastroenterology and Hepatology, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, United Kingdom
- University College London Institute for Liver and Digestive Health and Sheila Sherlock Liver Unit, Royal Free Hospital and University College London, London NW3 2QG, United Kingdom
- Division of Clinical and Molecular Hepatology, Department of Clinical and Experimental Medicine, University Hospital of Messina, Messina 98124, Italy
| | - Vasileios K Mavroeidis
- Department of Transplant Surgery, North Bristol NHS Trust, Southmead Hospital, Bristol BS10 5NB, United Kingdom
- Department of Gastrointestinal Surgery, North Bristol NHS Trust, Southmead Hospital, Bristol BS10 5NB, United Kingdom
- Department of HPB Surgery, Bristol Royal Infirmary, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol BS2 8HW, United Kingdom
| |
Collapse
|
3
|
Zwanenburg A, Price G, Löck S. Artificial intelligence for response prediction and personalisation in radiation oncology. Strahlenther Onkol 2025; 201:266-273. [PMID: 39212687 PMCID: PMC11839704 DOI: 10.1007/s00066-024-02281-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/14/2024] [Indexed: 09/04/2024]
Abstract
Artificial intelligence (AI) systems may personalise radiotherapy by assessing complex and multifaceted patient data and predicting tumour and normal tissue responses to radiotherapy. Here we describe three distinct generations of AI systems, namely personalised radiotherapy based on pretreatment data, response-driven radiotherapy and dynamically optimised radiotherapy. Finally, we discuss the main challenges in clinical translation of AI systems for radiotherapy personalisation.
Collapse
Affiliation(s)
- Alex Zwanenburg
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden-Rossendorf, Fetscherstr. 74, PF 41, 01307, Dresden, Germany.
- National Center for Tumor Diseases Dresden (NCT/UCC), Germany:, German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.
- German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany.
| | - Gareth Price
- Division of Cancer Sciences, University of Manchester, Manchester, UK
- The Christie NHS Foundation Trust, Manchester, UK
| | - Steffen Löck
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden-Rossendorf, Fetscherstr. 74, PF 41, 01307, Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| |
Collapse
|
4
|
Guo Y, Liao J, Li S, Shang Y, Wang Y, Wu Q, Wu Y, Wang M, Yan F, Tan H. Preoperative Prediction of Breast Cancer Histological Grade Using Intratumoral and Peritumoral Radiomics Features from T2WI and DWI MR Sequences. BREAST CANCER (DOVE MEDICAL PRESS) 2024; 16:981-991. [PMID: 39720357 PMCID: PMC11668253 DOI: 10.2147/bctt.s487988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 12/03/2024] [Indexed: 12/26/2024]
Abstract
Background Histological grade is an acknowledged prognostic factor for breast cancer, essential for determining clinical treatment strategies and prognosis assessment. Our study aims to establish intra- and peritumoral radiomics models using T2WI and DWI MR sequences for predicting the histological grade of breast cancer. Methods 700 breast cancer cases who had MRI scans before surgery were included. The intratumoral region (ITR) of interest was manually delineated, while the peritumoral region (PTR-3 mm) was automatically obtained by expanding the ITR by 3 mm. Radiomics features were extracted using the intra- and peritumoral images from T2WI and DWI sequences on breast MRI. Then, the key features with the strongest predictivity of histological grade were selected. Finally, 9 predictive radiomics models were established based on T2WI-ITR, T2WI-3mmPTR, DWI-ITR, DWI-3mmPTR, T2WI-ITR + 3mmPTR, DWI-ITR + 3mmPTR, (T2WI + DWI)-ITR, (T2WI + DWI)-3mmPTR and (T2WI + DWI)-ITR + 3mmPTR. Results The (T2WI + DWI)-ITR + 3mmPTR contained 13 DWI features which included a shape feature, a texture feature, and 11 filtered features, as well as 10 T2WI features, all of which were filtered features. Among the 9 models, the combined models showed better performance than the single models in both the training and test sets, especially for the (T2WI + DWI)-ITR + 3mmPTR radiomics model. The (T2WI + DWI)-ITR + 3mmPTR radiomics model achieved a sensitivity, specificity, accuracy, and AUC of 80.4%, 72.4%, 75.0%, and 0.860 in the training set, and 68.9%, 70.5%, 70.0%, and 0.781 in the test set. Decision curve analysis (DCA) showed that the (T2WI + DWI)-ITR + 3mmPTR model had the greatest net clinical benefit compared to the other models. Conclusion The intra- and peritumoral radiomics methodologies using T2WI and DWI MR sequences could be utilized to assess histological grade for breast cancer, particularly with the (T2WI + DWI)-ITR + 3mmPTR radiomics model demonstrating significant potential for clinical application.
Collapse
Affiliation(s)
- Yaxin Guo
- Department of Radiology, People’s Hospital of Zhengzhou University & Henan Provincial People’s Hospital, Zhengzhou, People’s Republic of China
| | - Jun Liao
- Department of Radiology, People’s Hospital of Zhengzhou University & Henan Provincial People’s Hospital, Zhengzhou, People’s Republic of China
| | - Shunian Li
- Department of Radiology, People’s Hospital of Zhengzhou University & Henan Provincial People’s Hospital, Zhengzhou, People’s Republic of China
| | - Yiyan Shang
- Department of Radiology, People’s Hospital of Henan University, Zhengzhou, People’s Republic of China
| | - Yunxia Wang
- Department of Radiology, People’s Hospital of Henan University, Zhengzhou, People’s Republic of China
| | - Qingxia Wu
- Beijing United Imaging Research Institute of Intelligent Imaging & United Imaging Intelligence (Beijing) Co., Ltd, Beijing, People’s Republic of China
| | - Yaping Wu
- Department of Radiology, People’s Hospital of Zhengzhou University & Henan Provincial People’s Hospital, Zhengzhou, People’s Republic of China
| | - Meiyun Wang
- Department of Radiology, People’s Hospital of Zhengzhou University & Henan Provincial People’s Hospital, Zhengzhou, People’s Republic of China
| | - Fengshan Yan
- Department of Radiology, People’s Hospital of Zhengzhou University & Henan Provincial People’s Hospital, Zhengzhou, People’s Republic of China
| | - Hongna Tan
- Department of Radiology, People’s Hospital of Zhengzhou University & Henan Provincial People’s Hospital, Zhengzhou, People’s Republic of China
| |
Collapse
|
5
|
Shahzadi M, Rafique H, Waheed A, Naz H, Waheed A, Zokirova FR, Khan H. Artificial intelligence for chimeric antigen receptor-based therapies: a comprehensive review of current applications and future perspectives. Ther Adv Vaccines Immunother 2024; 12:25151355241305856. [PMID: 39691280 PMCID: PMC11650588 DOI: 10.1177/25151355241305856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/18/2024] [Indexed: 12/19/2024] Open
Abstract
Using artificial intelligence (AI) to enhance chimeric antigen receptor (CAR)-based therapies' design, production, and delivery is a novel and promising approach. This review provides an overview of the current applications and challenges of AI for CAR-based therapies and suggests some directions for future research and development. This paper examines some of the recent advances of AI for CAR-based therapies, for example, using deep learning (DL) to design CARs that target multiple antigens and avoid antigen escape; using natural language processing to extract relevant information from clinical reports and literature; using computer vision to analyze the morphology and phenotype of CAR cells; using reinforcement learning to optimize the dose and schedule of CAR infusion; and using AI to predict the efficacy and toxicity of CAR-based therapies. These applications demonstrate the potential of AI to improve the quality and efficiency of CAR-based therapies and to provide personalized and precise treatments for cancer patients. However, there are also some challenges and limitations of using AI for CAR-based therapies, for example, the lack of high-quality and standardized data; the need for validation and verification of AI models; the risk of bias and error in AI outputs; the ethical, legal, and social issues of using AI for health care; and the possible impact of AI on the human role and responsibility in cancer immunotherapy. It is important to establish a multidisciplinary collaboration among researchers, clinicians, regulators, and patients to address these challenges and to ensure the safe and responsible use of AI for CAR-based therapies.
Collapse
Affiliation(s)
- Muqadas Shahzadi
- Department of Zoology, Faculty of Life Sciences, University of Okara, Okara, Pakistan
| | - Hamad Rafique
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi’an, Shaanxi, China
| | - Ahmad Waheed
- Department of Zoology, Faculty of Life Sciences, University of Okara, 2 KM Lahore Road, Renala Khurd, Okara 56130, Punjab, Pakistan
| | - Hina Naz
- Department of Zoology, Faculty of Life Sciences, University of Okara, Okara, Pakistan
| | - Atifa Waheed
- Department of Biology, Faculty of Life Sciences, University of Okara, Okara, Pakistan
| | | | - Humera Khan
- Department of Biochemistry, Sahiwal Medical College, Sahiwal, Pakistan
| |
Collapse
|
6
|
Hasanabadi S, Aghamiri SMR, Abin AA, Abdollahi H, Arabi H, Zaidi H. Enhancing Lymphoma Diagnosis, Treatment, and Follow-Up Using 18F-FDG PET/CT Imaging: Contribution of Artificial Intelligence and Radiomics Analysis. Cancers (Basel) 2024; 16:3511. [PMID: 39456604 PMCID: PMC11505665 DOI: 10.3390/cancers16203511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/11/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Lymphoma, encompassing a wide spectrum of immune system malignancies, presents significant complexities in its early detection, management, and prognosis assessment since it can mimic post-infectious/inflammatory diseases. The heterogeneous nature of lymphoma makes it challenging to definitively pinpoint valuable biomarkers for predicting tumor biology and selecting the most effective treatment strategies. Although molecular imaging modalities, such as positron emission tomography/computed tomography (PET/CT), specifically 18F-FDG PET/CT, hold significant importance in the diagnosis of lymphoma, prognostication, and assessment of treatment response, they still face significant challenges. Over the past few years, radiomics and artificial intelligence (AI) have surfaced as valuable tools for detecting subtle features within medical images that may not be easily discerned by visual assessment. The rapid expansion of AI and its application in medicine/radiomics is opening up new opportunities in the nuclear medicine field. Radiomics and AI capabilities seem to hold promise across various clinical scenarios related to lymphoma. Nevertheless, the need for more extensive prospective trials is evident to substantiate their reliability and standardize their applications. This review aims to provide a comprehensive perspective on the current literature regarding the application of AI and radiomics applied/extracted on/from 18F-FDG PET/CT in the management of lymphoma patients.
Collapse
Affiliation(s)
- Setareh Hasanabadi
- Department of Medical Radiation Engineering, Shahid Beheshti University, Tehran 1983969411, Iran; (S.H.); (S.M.R.A.)
| | - Seyed Mahmud Reza Aghamiri
- Department of Medical Radiation Engineering, Shahid Beheshti University, Tehran 1983969411, Iran; (S.H.); (S.M.R.A.)
| | - Ahmad Ali Abin
- Faculty of Computer Science and Engineering, Shahid Beheshti University, Tehran 1983969411, Iran;
| | - Hamid Abdollahi
- Department of Radiology, University of British Columbia, Vancouver, BC V5Z 1M9, Canada;
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
| | - Hossein Arabi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211 Geneva, Switzerland;
| | - Habib Zaidi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211 Geneva, Switzerland;
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
- Department of Nuclear Medicine, University of Southern Denmark, 500 Odense, Denmark
- University Research and Innovation Center, Óbuda University, 1034 Budapest, Hungary
| |
Collapse
|
7
|
Yang Z, Liu C. Research on the application of radiomics in breast cancer: A bibliometrics and visualization analysis. Medicine (Baltimore) 2024; 103:e39463. [PMID: 39213225 PMCID: PMC11365679 DOI: 10.1097/md.0000000000039463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/24/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
Breast cancer is the most prevalent form of cancer worldwide. Therefore, improved disease detection has emerged as a focal point in clinical studies. At the forefront of innovation, radiomics has the capability to extract comprehensive insights from medical images, ultimately enhancing the accuracy of diagnostic procedures. There has been rapid growth in the field of radiomics research on breast cancer in the past few years. We explored pertinent research articles in the Web of Science Core Collection database to gain a thorough understanding of breast cancer radiomics. We used CiteSpace to conduct a bibliometric analysis of the annual distribution of different nations, institutions, journals, authors, keywords, and references in the field of breast cancer radiomics. GraphPad Prism software was used to examine and graph yearly and country-specific trends and the proportions of publications. The tools utilized for the visualization of science mapping included CiteSpace and VOSviewer. Of the 891 publications, most were original articles (731, 91.09%) and a few were reviews (160, 8.91%). Most academic research has been published in China and the United States. The study centers predominantly consisted of major academic institutions, such as Fudan University and the Chinese Academy of Sciences, with some of their members being prominent figures in the field. Pinker, Katja has published the largest number of research papers. The majority of these studies have been published in medical journals focusing on radiology and oncology in recent years. In the realm of cutting-edge medical research, the top two keywords, magnetic resonance imaging and machine learning stand at the forefront as current areas of intense focus. Breast cancer radiomics is advancing rapidly, presenting numerous opportunities and obstacles. Our study of the literature in this academic area aimed to pinpoint the primary themes addressed in the studies and anticipate prospective avenues for research.
Collapse
Affiliation(s)
- Zhe Yang
- Department of Radiology, the Second Affiliated Hospital of Shandong First Medical University, Tai’an, Shandong, China
| | - Chenglong Liu
- Department of Radiology, the Second Affiliated Hospital of Shandong First Medical University, Tai’an, Shandong, China
| |
Collapse
|
8
|
Arif WM. Radiologic Technology Students' Perceptions on Adoption of Artificial Intelligence Technology in Radiology. Int J Gen Med 2024; 17:3129-3136. [PMID: 39049835 PMCID: PMC11268710 DOI: 10.2147/ijgm.s465944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 07/11/2024] [Indexed: 07/27/2024] Open
Abstract
Study Purpose This study aims to analyze radiologic technology student's perceptions of artificial intelligence (AI) and its applications in radiology. Methods A quantitative cross-sectional survey was conducted. A pre-validated survey questionnaire with 17 items related to students perceptions of AI and its applications was used. The sample included radiologic technology students from three universities in Saudi Arabia. The survey was conducted online for several weeks, resulting in a sample of 280 radiologic technology students. Results Of the participants, 63.9% were aware of AI and its applications. T-tests revealed a statistically significant difference (p = 0.0471) between genders with male participants reflecting slightly higher AI awareness than female participants. Regarding the choice of radiology as specialization, 35% of the participants stated that they would not choose radiology, whereas 65% preferred it. Approximately 56% of the participants expressed concerns about the potential replacement of radiology technologists with AI, and 62.1% strongly agreed on the necessity of incorporating known ethical principles into AI. Conclusion The findings reflect a positive evaluation of the applications of this technology, which is attributed to its essential support role. However, tailored education and training programs are necessary to prepare future healthcare professionals for the increasing role of AI in medical sciences.
Collapse
Affiliation(s)
- Wejdan M Arif
- King Saud University, College of Applied Medical Sciences, Department of Radiological Sciences, Riyadh, Saudi Arabia
| |
Collapse
|
9
|
Huang W, Tao Z, Younis MH, Cai W, Kang L. Nuclear medicine radiomics in digestive system tumors: Concept, applications, challenges, and future perspectives. VIEW 2023; 4:20230032. [PMID: 38179181 PMCID: PMC10766416 DOI: 10.1002/viw.20230032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 07/20/2023] [Indexed: 01/06/2024] Open
Abstract
Radiomics aims to develop novel biomarkers and provide relevant deeper subvisual information about pathology, immunophenotype, and tumor microenvironment. It uses automated or semiautomated quantitative analysis of high-dimensional images to improve characterization, diagnosis, and prognosis. Recent years have seen a rapid increase in radiomics applications in nuclear medicine, leading to some promising research results in digestive system oncology, which have been driven by big data analysis and the development of artificial intelligence. Although radiomics advances one step further toward the non-invasive precision medical analysis, it is still a step away from clinical application and faces many challenges. This review article summarizes the available literature on digestive system tumors regarding radiomics in nuclear medicine. First, we describe the workflow and steps involved in radiomics analysis. Subsequently, we discuss the progress in clinical application regarding the utilization of radiomics for distinguishing between various diseases and evaluating their prognosis, and demonstrate how radiomics advances this field. Finally, we offer our viewpoint on how the field can progress by addressing the challenges facing clinical implementation.
Collapse
Affiliation(s)
- Wenpeng Huang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, China
| | - Zihao Tao
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, China
| | - Muhsin H. Younis
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Lei Kang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, China
| |
Collapse
|
10
|
De Muzio F, Pellegrino F, Fusco R, Tafuto S, Scaglione M, Ottaiano A, Petrillo A, Izzo F, Granata V. Prognostic Assessment of Gastropancreatic Neuroendocrine Neoplasm: Prospects and Limits of Radiomics. Diagnostics (Basel) 2023; 13:2877. [PMID: 37761243 PMCID: PMC10529975 DOI: 10.3390/diagnostics13182877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Neuroendocrine neoplasms (NENs) are a group of lesions originating from cells of the diffuse neuroendocrine system. NENs may involve different sites, including the gastrointestinal tract (GEP-NENs). The incidence and prevalence of GEP-NENs has been constantly rising thanks to the increased diagnostic power of imaging and immuno-histochemistry. Despite the plethora of biochemical markers and imaging techniques, the prognosis and therapeutic choice in GEP-NENs still represents a challenge, mainly due to the great heterogeneity in terms of tumor lesions and clinical behavior. The concept that biomedical images contain information about tissue heterogeneity and pathological processes invisible to the human eye is now well established. From this substrate comes the idea of radiomics. Computational analysis has achieved promising results in several oncological settings, and the use of radiomics in different types of GEP-NENs is growing in the field of research, yet with conflicting results. The aim of this narrative review is to provide a comprehensive update on the role of radiomics on GEP-NEN management, focusing on the main clinical aspects analyzed by most existing reports: predicting tumor grade, distinguishing NET from other tumors, and prognosis assessment.
Collapse
Affiliation(s)
- Federica De Muzio
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy;
| | | | - Roberta Fusco
- Medical Oncology Division, Igea SpA, 80013 Napoli, Italy;
| | - Salvatore Tafuto
- Unit of Sarcomi e Tumori Rari, Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, 80131 Naples, Italy;
| | - Mariano Scaglione
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy
| | - Alessandro Ottaiano
- Unit for Innovative Therapies of Abdominal Metastastes, Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, 80131 Naples, Italy;
| | - Antonella Petrillo
- Division of Radiology, Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, 80131 Naples, Italy;
| | - Francesco Izzo
- Division of Hepatobiliary Surgery, Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, 80131 Naples, Italy
| | - Vincenza Granata
- Division of Radiology, Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, 80131 Naples, Italy;
| |
Collapse
|
11
|
Yan W, Quan C, Mourad WF, Yuan J, Shi Z, Yang J, Lu Q, Zhang J. Application of radiomics in lung immuno-oncology. PRECISION RADIATION ONCOLOGY 2023; 7:128-136. [PMID: 40337267 PMCID: PMC11935008 DOI: 10.1002/pro6.1191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 02/22/2023] [Accepted: 02/26/2023] [Indexed: 04/08/2023] Open
Abstract
Radiomics is a rapidly evolving field of research that extracts and analyzes quantitative features within medical images. Those features are termed as radiomic features that can characterize a tumor in a comprehensive and quantitative manner with regard to its internal structure and heterogeneity. Radiomic features can be used, alone or in combination with demographic, histological, genomic, or proteomic data, for predicting prognosis or treatment response. Immunotherapy, or immune-oncology, is the study of cancer treatment by taking advantage of the body's immune system to prevent, control, and eliminate cancer. In this review, we first provide a brief introduction to both radiomics and immune-oncology in lung cancer. Then, we discuss the need for developing immune-oncology biomarkers, and the advantages of radiomics in identifying biomarkers related to immunotherapy. We also discuss potential areas in and out of tumors, such as the intra-tumoral hypoxic region and tumor microenvironment, where radiomic markers might be extracted, as well as a potential application of radiomic biomarkers in clinical lung cancer management. Finally, we present radiation and immune modulation in non-small cell lung cancer, clinical trials and their design to incorporate radiomic biomarkers, and radiomics-guided precision radiation therapy.
Collapse
Affiliation(s)
- Weisi Yan
- Baptist Health SystemLexingtonKentuckyUSA
| | - Chen Quan
- City of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Waleed F. Mourad
- Department of Radiation MedicineUniversity of KentuckyLexingtonKentuckyUSA
| | - Jianda Yuan
- Translational Oncology at Merck & CoKenilworthNew JerseyUSA
| | | | - Jun Yang
- Foshan Chancheng HospitalFoshanGuangdongChina
| | - Qiuxia Lu
- Foshan Chancheng HospitalFoshanGuangdongChina
| | - Jie Zhang
- Department of RadiologyUniversity of KentuckyLexingtonKentuckyUSA
| |
Collapse
|
12
|
Liu Y, Wei X, Feng X, Liu Y, Feng G, Du Y. Repeatability of radiomics studies in colorectal cancer: a systematic review. BMC Gastroenterol 2023; 23:125. [PMID: 37059990 PMCID: PMC10105401 DOI: 10.1186/s12876-023-02743-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 03/22/2023] [Indexed: 04/16/2023] Open
Abstract
BACKGROUND Recently, radiomics has been widely used in colorectal cancer, but many variable factors affect the repeatability of radiomics research. This review aims to analyze the repeatability of radiomics studies in colorectal cancer and to evaluate the current status of radiomics in the field of colorectal cancer. METHODS The included studies in this review by searching from the PubMed and Embase databases. Then each study in our review was evaluated using the Radiomics Quality Score (RQS). We analyzed the factors that may affect the repeatability in the radiomics workflow and discussed the repeatability of the included studies. RESULTS A total of 188 studies was included in this review, of which only two (2/188, 1.06%) studies controlled the influence of individual factors. In addition, the median score of RQS was 11 (out of 36), range-1 to 27. CONCLUSIONS The RQS score was moderately low, and most studies did not consider the repeatability of radiomics features, especially in terms of Intra-individual, scanners, and scanning parameters. To improve the generalization of the radiomics model, it is necessary to further control the variable factors of repeatability.
Collapse
Affiliation(s)
- Ying Liu
- School of Medical Imaging, North Sichuan Medical College, Sichuan Province, Nanchong City, 637000, China
| | - Xiaoqin Wei
- School of Medical Imaging, North Sichuan Medical College, Sichuan Province, Nanchong City, 637000, China
| | | | - Yan Liu
- Department of Radiology, the Affiliated Hospital of North Sichuan Medical College, 1 Maoyuannan Road, Sichuan Province, 637000, Nanchong City, China
| | - Guiling Feng
- Department of Radiology, the Affiliated Hospital of North Sichuan Medical College, 1 Maoyuannan Road, Sichuan Province, 637000, Nanchong City, China
| | - Yong Du
- Department of Radiology, the Affiliated Hospital of North Sichuan Medical College, 1 Maoyuannan Road, Sichuan Province, 637000, Nanchong City, China.
| |
Collapse
|
13
|
Granata V, Fusco R, Setola SV, Galdiero R, Maggialetti N, Patrone R, Ottaiano A, Nasti G, Silvestro L, Cassata A, Grassi F, Avallone A, Izzo F, Petrillo A. Colorectal liver metastases patients prognostic assessment: prospects and limits of radiomics and radiogenomics. Infect Agent Cancer 2023; 18:18. [PMID: 36927442 PMCID: PMC10018963 DOI: 10.1186/s13027-023-00495-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/07/2023] [Indexed: 03/18/2023] Open
Abstract
In this narrative review, we reported un up-to-date on the role of radiomics to assess prognostic features, which can impact on the liver metastases patient treatment choice. In the liver metastases patients, the possibility to assess mutational status (RAS or MSI), the tumor growth pattern and the histological subtype (NOS or mucinous) allows a better treatment selection to avoid unnecessary therapies. However, today, the detection of these features require an invasive approach. Recently, radiomics analysis application has improved rapidly, with a consequent growing interest in the oncological field. Radiomics analysis allows the textural characteristics assessment, which are correlated to biological data. This approach is captivating since it should allow to extract biological data from the radiological images, without invasive approach, so that to reduce costs and time, avoiding any risk for the patients. Several studies showed the ability of Radiomics to identify mutational status, tumor growth pattern and histological type in colorectal liver metastases. Although, radiomics analysis in a non-invasive and repeatable way, however features as the poor standardization and generalization of clinical studies results limit the translation of this analysis into clinical practice. Clear limits are data-quality control, reproducibility, repeatability, generalizability of results, and issues related to model overfitting.
Collapse
Affiliation(s)
- Vincenza Granata
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli", Naples, Italy.
| | - Roberta Fusco
- Medical Oncology Division, Igea SpA, Napoli, Italy.,Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Via della Signora 2, Milan, 20122, Italy
| | - Sergio Venanzio Setola
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli", Naples, Italy
| | - Roberta Galdiero
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli", Naples, Italy
| | - Nicola Maggialetti
- Department of Medical Science, Neuroscience and Sensory Organs (DSMBNOS), University of Bari "Aldo Moro", Bari, 70124, Italy
| | - Renato Patrone
- Division of Epatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale-IRCCS di Napoli, Naples, 80131, Italy
| | - Alessandro Ottaiano
- Clinical Sperimental Abdominal Oncology Unit, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, Napoli, 80131, Italy
| | - Guglielmo Nasti
- Clinical Sperimental Abdominal Oncology Unit, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, Napoli, 80131, Italy
| | - Lucrezia Silvestro
- Clinical Sperimental Abdominal Oncology Unit, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, Napoli, 80131, Italy
| | - Antonio Cassata
- Clinical Sperimental Abdominal Oncology Unit, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, Napoli, 80131, Italy
| | - Francesca Grassi
- Division of Radiology, "Università degli Studi della Campania Luigi Vanvitelli", Naples, 80138, Italy
| | - Antonio Avallone
- Clinical Sperimental Abdominal Oncology Unit, Istituto Nazionale Tumori, IRCCS Fondazione G. Pascale, Napoli, 80131, Italy
| | - Francesco Izzo
- Division of Epatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale-IRCCS di Napoli, Naples, 80131, Italy
| | - Antonella Petrillo
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli", Naples, Italy
| |
Collapse
|
14
|
Barreiro-Ares A, Morales-Santiago A, Sendra-Portero F, Souto-Bayarri M. Impact of the Rise of Artificial Intelligence in Radiology: What Do Students Think? INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:1589. [PMID: 36674348 PMCID: PMC9867061 DOI: 10.3390/ijerph20021589] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 06/17/2023]
Abstract
The rise of artificial intelligence (AI) in medicine, and particularly in radiology, is becoming increasingly prominent. Its impact will transform the way the specialty is practiced and the current and future education model. The aim of this study is to analyze the perception that undergraduate medical students have about the current situation of AI in medicine, especially in radiology. A survey with 17 items was distributed to medical students between 3 January to 31 March 2022. Two hundred and eighty-one students correctly responded the questionnaire; 79.3% of them claimed that they knew what AI is. However, their objective knowledge about AI was low but acceptable. Only 24.9% would choose radiology as a specialty, and only 40% of them as one of their first three options. The applications of this technology were valued positively by most students, who give it an important Support Role, without fear that the radiologist will be replaced by AI (79.7%). The majority (95.7%) agreed with the need to implement well-established ethical principles in AI, and 80% valued academic training in AI positively. Surveyed medical students have a basic understanding of AI and perceive it as a useful tool that will transform radiology.
Collapse
Affiliation(s)
- Andrés Barreiro-Ares
- Department of Radiology, School of Medicine, University of Santiago de Compostela/CHUS/IDIS (Instituto de Investigación Sanitaria de Santiago), 15782 Santiago de Compostela, Spain
| | - Annia Morales-Santiago
- Department of Radiology, School of Medicine, University of Santiago de Compostela/CHUS/IDIS (Instituto de Investigación Sanitaria de Santiago), 15782 Santiago de Compostela, Spain
| | - Francisco Sendra-Portero
- Department of Radiology and Physical Medicine, School of Medicine, University of Malaga, 29010 Málaga, Spain
| | - Miguel Souto-Bayarri
- Department of Radiology, School of Medicine, University of Santiago de Compostela/CHUS/IDIS (Instituto de Investigación Sanitaria de Santiago), 15782 Santiago de Compostela, Spain
| |
Collapse
|
15
|
Ghani M, Liau J, Eskander R, Mell L, Yusufaly T, Obrzut S. Imaging Biomarkers and Liquid Biopsy in Assessment of Cervical Cancer. J Comput Assist Tomogr 2022; 46:707-715. [PMID: 35995483 PMCID: PMC9474655 DOI: 10.1097/rct.0000000000001358] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT The role of imaging has been increasing in pretherapy planning and response assessment in cervical cancer, particularly in high-resource settings that provide access to computed tomography (CT), magnetic resonance imaging (MRI), and positron emission tomography (PET). In 2018, imaging was incorporated into the International Federation of Gynecology and Obstetrics staging system for cervical cancer. Magnetic resonance imaging is advantageous over CT for evaluation of the primary cervical cancer size and extent, because of superior contrast resolution. Furthermore, quantitative methods, including diffusion-weighted and dynamic contrast-enhanced MRI, show promise in improving treatment response and prognosis evaluation. Molecular imaging with fluorodeoxyglucose-PET/CT and PET/MRI can be particularly helpful in the detection of nodal disease and distant metastases. Semiautomated delineation of 3-dimensional tumor regions of interest has facilitated the development of novel PET-derived biomarkers that include metabolic volume and radiomics textural analysis features for prediction of outcomes. However, posttreatment inflammatory changes can be a confounder and lymph node evaluation is challenging, even with the use of PET/CT. Liquid biopsy has emerged as a promising tool that may be able to overcome some of the drawbacks inherent with imaging, such as limited ability to detect microscopic metastases or to distinguish between postchemoradiotherapy changes and residual tumor. Preliminary evidence suggests that liquid biopsy may be able to identify cervical cancer treatment response and resistance earlier than traditional methods. Future work should prioritize how to best synergize imaging and liquid biopsy as an integrated approach for optimal cervical cancer management.
Collapse
Affiliation(s)
- Mansur Ghani
- Department of Radiology, University of California San Diego, CA, USA
| | - Joy Liau
- Department of Radiology, University of California San Diego, CA, USA
| | - Ramez Eskander
- Division of Hematology/Oncology, University of California San Diego, CA, USA
| | - Loren Mell
- Department of Radiation Oncology, University of California San Diego, CA, USA
| | - Tahir Yusufaly
- Department of Radiology, Johns Hopkins University, MD, USA
| | - Sebastian Obrzut
- Department of Radiology, University of California San Diego, CA, USA
| |
Collapse
|
16
|
Update on quantitative radiomics of pancreatic tumors. Abdom Radiol (NY) 2022; 47:3118-3160. [PMID: 34292365 DOI: 10.1007/s00261-021-03216-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 02/07/2023]
Abstract
Radiomics is a newer approach for analyzing radiological images obtained from conventional imaging modalities such as computed tomography, magnetic resonance imaging, endoscopic ultrasonography, and positron emission tomography. Radiomics involves extracting quantitative data from the images and assessing them to identify diagnostic or prognostic features such as tumor grade, resectability, tumor response to neoadjuvant therapy, and survival. The purpose of this review is to discuss the basic principles of radiomics and provide an overview of the current clinical applications of radiomics in the field of pancreatic tumors.
Collapse
|
17
|
Kondylakis H, Ciarrocchi E, Cerda-Alberich L, Chouvarda I, Fromont LA, Garcia-Aznar JM, Kalokyri V, Kosvyra A, Walker D, Yang G, Neri E, the AI4HealthImaging Working Group on metadata models**. Position of the AI for Health Imaging (AI4HI) network on metadata models for imaging biobanks. Eur Radiol Exp 2022; 6:29. [PMID: 35773546 PMCID: PMC9247122 DOI: 10.1186/s41747-022-00281-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 04/20/2022] [Indexed: 11/10/2022] Open
Abstract
A huge amount of imaging data is becoming available worldwide and an incredible range of possible improvements can be provided by artificial intelligence algorithms in clinical care for diagnosis and decision support. In this context, it has become essential to properly manage and handle these medical images and to define which metadata have to be considered, in order for the images to provide their full potential. Metadata are additional data associated with the images, which provide a complete description of the image acquisition, curation, analysis, and of the relevant clinical variables associated with the images. Currently, several data models are available to describe one or more subcategories of metadata, but a unique, common, and standard data model capable of fully representing the heterogeneity of medical metadata has not been yet developed. This paper reports the state of the art on metadata models for medical imaging, the current limitations and further developments, and describes the strategy adopted by the Horizon 2020 "AI for Health Imaging" projects, which are all dedicated to the creation of imaging biobanks.
Collapse
Affiliation(s)
| | - Esther Ciarrocchi
- grid.5395.a0000 0004 1757 3729Department of Translational Research, University of Pisa, Pisa, Italy
| | | | - Ioanna Chouvarda
- grid.4793.90000000109457005Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Lauren A. Fromont
- grid.11478.3b0000 0004 1766 3695Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | | | - Varvara Kalokyri
- grid.5395.a0000 0004 1757 3729Department of Translational Research, University of Pisa, Pisa, Italy
| | - Alexandra Kosvyra
- grid.4793.90000000109457005Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dawn Walker
- grid.11835.3e0000 0004 1936 9262Department of Computer Science and Insigneo Institute of in silico Medicine, University of Sheffield, Sheffield, UK
| | - Guang Yang
- grid.7445.20000 0001 2113 8111National Heart and Lung Institute, Imperial College London, London, UK
| | - Emanuele Neri
- grid.5395.a0000 0004 1757 3729Department of Translational Research, University of Pisa, Pisa, Italy
| | | |
Collapse
|
18
|
Saba L, Antignani PL, Gupta A, Cau R, Paraskevas KI, Poredos P, Wasserman B, Kamel H, Avgerinos ED, Salgado R, Caobelli F, Aluigi L, Savastano L, Brown M, Hatsukami T, Hussein E, Suri JS, Mansilha A, Wintermark M, Staub D, Montequin JF, Rodriguez RTT, Balu N, Pitha J, Kooi ME, Lal BK, Spence JD, Lanzino G, Marcus HS, Mancini M, Chaturvedi S, Blinc A. International Union of Angiology (IUA) consensus paper on imaging strategies in atherosclerotic carotid artery imaging: From basic strategies to advanced approaches. Atherosclerosis 2022; 354:23-40. [DOI: 10.1016/j.atherosclerosis.2022.06.1014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/10/2022] [Accepted: 06/14/2022] [Indexed: 12/24/2022]
|
19
|
Valladares A, Oberoi G, Berg A, Beyer T, Unger E, Rausch I. Additively manufactured, solid object structures for adjustable image contrast in Magnetic Resonance Imaging. Z Med Phys 2022; 32:466-476. [PMID: 35597743 PMCID: PMC9948875 DOI: 10.1016/j.zemedi.2022.03.003] [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: 11/26/2021] [Revised: 02/08/2022] [Accepted: 03/15/2022] [Indexed: 11/28/2022]
Abstract
The choice of materials challenges the development of Magnetic Resonance Imaging (MRI) phantoms and, to date, is mainly limited to water-filled compartments or gel-based components. Recently, solid materials have been introduced through additive manufacturing (AM) to mimic complex geometrical structures. Nonetheless, no such manufactured solid materials are available with controllable MRI contrast to mimic organ substructures or lesion heterogeneities. Here, we present a novel AM design that allows MRI contrast manipulation by varying the partial volume contribution to a ROI/voxel of MRI-visible material within an imaging object. Two sets of 11 cubes and three replicates of a spherical tumour model were designed and printed using AM. Most samples presented varying MRI-contrast in standard MRI sequences, based mainly on spin density and partial volume signal variation. A smooth and continuous MRI-contrast gradient could be generated in a single-compartment tumour model. This concept supports the development of more complex MRI phantoms that mimic the appearance of heterogeneous tumour tissues.
Collapse
Affiliation(s)
- Alejandra Valladares
- QIMP Team, Centre for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Gunpreet Oberoi
- Centre for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Andreas Berg
- Centre for Medical Physics and Biomedical Engineering, MR-Physics, Medical University of Vienna, Vienna, Austria,High-field MR-Center, Medical University of Vienna, Vienna, Austria
| | - Thomas Beyer
- QIMP Team, Centre for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Ewald Unger
- Centre for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Ivo Rausch
- QIMP Team, Centre for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
20
|
Tagliafico AS, Campi C, Bianca B, Bortolotto C, Buccicardi D, Francesca C, Prost R, Rengo M, Faggioni L. Blockchain in radiology research and clinical practice: current trends and future directions. LA RADIOLOGIA MEDICA 2022; 127:391-397. [PMID: 35194720 PMCID: PMC8863512 DOI: 10.1007/s11547-022-01460-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 01/21/2022] [Indexed: 12/31/2022]
Abstract
Blockchain usage in healthcare, in radiology, in particular, is at its very early infancy. Only a few research applications have been tested, however, blockchain technology is widely known outside healthcare and widely adopted, especially in Finance, since 2009 at least. Learning by history, radiology is a potential ideal scenario to apply this technology. Blockchain could have the potential to increase radiological data value in both clinical and research settings for the patient digital record, radiological reports, privacy control, quantitative image analysis, cybersecurity, radiomics and artificial intelligence.Up-to-date experiences using blockchain in radiology are still limited, but radiologists should be aware of the emergence of this technology and follow its next developments. We present here the potentials of some applications of blockchain in radiology.
Collapse
Affiliation(s)
- Alberto Stefano Tagliafico
- IRCCS Ospedale Policlinico San Martino, Genova, Genoa, Italy
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| | - Cristina Campi
- Dipartimento Di Matematica, Università Di Genova, via Dodecaneso 35, 16146 Genova, Italy
| | - Bignotti Bianca
- IRCCS Ospedale Policlinico San Martino, Genova, Genoa, Italy
| | - Chandra Bortolotto
- Dipartimento Di Radiologia, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | | | - Coppola Francesca
- Department of Radiology, IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Roberto Prost
- Azienda Ospedaliera Brotzu, Cagliari, Sardegna Italy
| | - Marco Rengo
- Department of Radiological Sciences, Oncology and Pathology, Sapienza University of Rome - I.C.O.T. Hospital, Via Franco Faggiana, 1668, 04100 Latina, Italy
| | - Lorenzo Faggioni
- Diagnostic and Interventional Radiology, University Hospital of Pisa, Via Paradisa 2, 56100 Pisa, Italy
| |
Collapse
|
21
|
Bonmatí LM, Miguel A, Suárez A, Aznar M, Beregi JP, Fournier L, Neri E, Laghi A, França M, Sardanelli F, Penzkofer T, Lambin P, Blanquer I, Menzel M, Seymour K, Figueiras S, Krischak K, Martínez R, Mirsky Y, Yang G, Alberich-Bayarri Á. CHAIMELEON Project: Creation of a Pan-European Repository of Health Imaging Data for the Development of AI-Powered Cancer Management Tools. Front Oncol 2022; 12:742701. [PMID: 35280732 PMCID: PMC8913333 DOI: 10.3389/fonc.2022.742701] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 01/28/2022] [Indexed: 12/13/2022] Open
Abstract
The CHAIMELEON project aims to set up a pan-European repository of health imaging data, tools and methodologies, with the ambition to set a standard and provide resources for future AI experimentation for cancer management. The project is a 4 year long, EU-funded project tackling some of the most ambitious research in the fields of biomedical imaging, artificial intelligence and cancer treatment, addressing the four types of cancer that currently have the highest prevalence worldwide: lung, breast, prostate and colorectal. To allow this, clinical partners and external collaborators will populate the repository with multimodality (MR, CT, PET/CT) imaging and related clinical data. Subsequently, AI developers will enable a multimodal analytical data engine facilitating the interpretation, extraction and exploitation of the information stored at the repository. The development and implementation of AI-powered pipelines will enable advancement towards automating data deidentification, curation, annotation, integrity securing and image harmonization. By the end of the project, the usability and performance of the repository as a tool fostering AI experimentation will be technically validated, including a validation subphase by world-class European AI developers, participating in Open Challenges to the AI Community. Upon successful validation of the repository, a set of selected AI tools will undergo early in-silico validation in observational clinical studies coordinated by leading experts in the partner hospitals. Tool performance will be assessed, including external independent validation on hallmark clinical decisions in response to some of the currently most important clinical end points in cancer. The project brings together a consortium of 18 European partners including hospitals, universities, R&D centers and private research companies, constituting an ecosystem of infrastructures, biobanks, AI/in-silico experimentation and cloud computing technologies in oncology.
Collapse
Affiliation(s)
- Luis Martí Bonmatí
- Medical Imaging Department, La Fe University and Polytechnic Hospital & Biomedical Imaging Research Group Grupo de Investigación Biomédica en Imagen (GIBI2) at La Fe University and Polytechnic Hospital and Health Research Institute, Valencia, Spain,*Correspondence: Luis Martí Bonmatí,
| | - Ana Miguel
- Medical Imaging Department, La Fe University and Polytechnic Hospital & Biomedical Imaging Research Group Grupo de Investigación Biomédica en Imagen (GIBI2) at La Fe University and Polytechnic Hospital and Health Research Institute, Valencia, Spain
| | | | | | | | - Laure Fournier
- Collège des enseignants en radiologie de France, Paris, France
| | - Emanuele Neri
- Diagnostic Radiology 3, Department of Translational Research, University of Pisa, Pisa, Italy
| | - Andrea Laghi
- Medicina Traslazionale e Oncologia, Sant Andrea Sapienza Rome, Rome, Italy
| | - Manuela França
- Department of Radiology, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Francesco Sardanelli
- Servizio di Diagnostica per Immagini, “Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Donato, Milanese, Italy
| | - Tobias Penzkofer
- Department of Radiology, CHARITÉ-Universitätsmedizin Berlin, Berlin, Germany
| | - Phillipe Lambin
- Department of Precision Medicine, Maastricht University, Maastricht, Netherlands
| | - Ignacio Blanquer
- Computing Science Department, Universitat Politècnica de València, València, Spain
| | - Marion I. Menzel
- GE Healthcare, München, Germany,Department of Physics, Technical University of Munich, Garching, Germany
| | | | | | - Katharina Krischak
- European Institute for Biomedical Imaging Research, EIBIR gemeinnützige GmbH, Vienna, Austria
| | - Ricard Martínez
- Departamento de Derecho Constitucional, Ciencia Política y Administración, Universitat de València, València, Spain
| | - Yisroel Mirsky
- Software and Information Systems Engineering, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Guang Yang
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | | |
Collapse
|
22
|
Treatment-driven tumour heterogeneity and drug resistance: lessons from solid tumours. Cancer Treat Rev 2022; 104:102340. [DOI: 10.1016/j.ctrv.2022.102340] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/16/2022] [Accepted: 01/17/2022] [Indexed: 02/07/2023]
|
23
|
Bridging gaps between images and data: a systematic update on imaging biobanks. Eur Radiol 2022; 32:3173-3186. [PMID: 35001159 DOI: 10.1007/s00330-021-08431-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 10/01/2021] [Accepted: 10/22/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND OBJECTIVE The systematic collection of medical images combined with imaging biomarkers and patient non-imaging data is the core concept of imaging biobanks, a key element for fuelling the development of modern precision medicine. Our purpose is to review the existing image repositories fulfilling the criteria for imaging biobanks. METHODS Pubmed, Scopus and Web of Science were searched for articles published in English from January 2010 to July 2021 using a combination of the terms: "imaging" AND "biobanks" and "imaging" AND "repository". Moreover, the Community Research and Development Information Service (CORDIS) database ( https://cordis.europa.eu/projects ) was searched using the terms: "imaging" AND "biobanks", also including collections, projects, project deliverables, project publications and programmes. RESULTS Of 9272 articles retrieved, only 54 related to biobanks containing imaging data were finally selected, of which 33 were disease-oriented (61.1%) and 21 population-based (38.9%). Most imaging biobanks were European (26/54, 48.1%), followed by American biobanks (20/54, 37.0%). Among disease-oriented biobanks, the majority were focused on neurodegenerative (9/33, 27.3%) and oncological diseases (9/33, 27.3%). The number of patients enrolled ranged from 240 to 3,370,929, and the imaging modality most frequently involved was MRI (40/54, 74.1%), followed by CT (20/54, 37.0%), PET (13/54, 24.1%), and ultrasound (12/54, 22.2%). Most biobanks (38/54, 70.4%) were accessible under request. CONCLUSIONS Imaging biobanks can serve as a platform for collecting, sharing and analysing medical images integrated with imaging biomarkers, biological and clinical data. A relatively small proportion of current biobanks also contain images and can thus be classified as imaging biobanks. KEY POINTS • Imaging biobanks are a powerful tool for large-scale collection and processing of medical images integrated with imaging biomarkers and patient non-imaging data. • Most imaging biobanks retrieved were European, disease-oriented and accessible under user request. • While many biobanks have been developed so far, only a relatively small proportion of them are imaging biobanks.
Collapse
|
24
|
Scapicchio C, Gabelloni M, Barucci A, Cioni D, Saba L, Neri E. A deep look into radiomics. LA RADIOLOGIA MEDICA 2021; 126:1296-1311. [PMID: 34213702 PMCID: PMC8520512 DOI: 10.1007/s11547-021-01389-x] [Citation(s) in RCA: 209] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 06/15/2021] [Indexed: 11/29/2022]
Abstract
Radiomics is a process that allows the extraction and analysis of quantitative data from medical images. It is an evolving field of research with many potential applications in medical imaging. The purpose of this review is to offer a deep look into radiomics, from the basis, deeply discussed from a technical point of view, through the main applications, to the challenges that have to be addressed to translate this process in clinical practice. A detailed description of the main techniques used in the various steps of radiomics workflow, which includes image acquisition, reconstruction, pre-processing, segmentation, features extraction and analysis, is here proposed, as well as an overview of the main promising results achieved in various applications, focusing on the limitations and possible solutions for clinical implementation. Only an in-depth and comprehensive description of current methods and applications can suggest the potential power of radiomics in fostering precision medicine and thus the care of patients, especially in cancer detection, diagnosis, prognosis and treatment evaluation.
Collapse
Affiliation(s)
- Camilla Scapicchio
- Academic Radiology, Department of Translational Research, University of Pisa, Via Roma 67, 56126, Pisa, Italy.
| | - Michela Gabelloni
- Academic Radiology, Department of Translational Research, University of Pisa, Via Roma 67, 56126, Pisa, Italy
| | - Andrea Barucci
- CNR-IFAC Institute of Applied Physics "N. Carrara", 50019, Sesto Fiorentino, Italy
| | - Dania Cioni
- Academic Radiology, Department of Surgical, Medical, Molecular Pathology and Emergency Medicine, University of Pisa, Via Roma 67, 56126, Pisa, Italy
| | - Luca Saba
- Department of Radiology, Azienda Ospedaliero Universitaria (A.O.U.), Monserrato (Cagliari),Cagliari, Italy
| | - Emanuele Neri
- Academic Radiology, Department of Translational Research, University of Pisa, Via Roma 67, 56126, Pisa, Italy
- Italian Society of Medical and Interventional Radiology, SIRM Foundation, Via della Signora 2, 20122, Milano, Italy
| |
Collapse
|
25
|
Cucchiara F, Petrini I, Romei C, Crucitta S, Lucchesi M, Valleggi S, Scavone C, Capuano A, De Liperi A, Chella A, Danesi R, Del Re M. Combining liquid biopsy and radiomics for personalized treatment of lung cancer patients. State of the art and new perspectives. Pharmacol Res 2021; 169:105643. [PMID: 33940185 DOI: 10.1016/j.phrs.2021.105643] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/22/2021] [Accepted: 04/22/2021] [Indexed: 12/11/2022]
Abstract
Lung cancer has become a paradigm for precision medicine in oncology, and liquid biopsy (LB) together with radiomics may have a great potential in this scenario. They are both minimally invasive, easy to perform, and can be repeated during patient's follow-up. Also, increasing evidence suggest that LB and radiomics may provide an efficient way to screen and diagnose tumors at an early stage, including the monitoring of any change in the tumor molecular profile. This could allow treatment optimization, improvement of patients' quality of life, and healthcare-related costs reduction. Latest reports on lung cancer patients suggest a combination of these two strategies, along with cutting-edge data analysis, to decode valuable information regarding tumor type, aggressiveness, progression, and response to treatment. The approach seems more compatible with clinical practice than the current standard, and provides new diagnostic companions being able to suggest the best treatment strategy compared to conventional methods. To implement radiomics and liquid biopsy directly into clinical practice, an artificial intelligence (AI)-based system could help to link patients' clinical data together with tumor molecular profiles and imaging characteristics. AI could also solve problems and limitations related to LB and radiomics methodologies. Further work is needed, including new health policies and the access to large amounts of high-quality and well-organized data, allowing a complementary and synergistic combination of LB and imaging, to provide an attractive choice e in the personalized treatment of lung cancer.
Collapse
Affiliation(s)
- Federico Cucchiara
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Iacopo Petrini
- Unit of Pneumology, Department of Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Chiara Romei
- Unit II of Radio-diagnostics, Department of Diagnostic and Imaging, University Hospital of Pisa, Pisa, Italy
| | - Stefania Crucitta
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Maurizio Lucchesi
- Unit of Pneumology, Department of Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Simona Valleggi
- Unit of Pneumology, Department of Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Cristina Scavone
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Annalisa Capuano
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Annalisa De Liperi
- Unit II of Radio-diagnostics, Department of Diagnostic and Imaging, University Hospital of Pisa, Pisa, Italy
| | - Antonio Chella
- Unit of Pneumology, Department of Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Romano Danesi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy.
| | - Marzia Del Re
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| |
Collapse
|
26
|
Badic B, Tixier F, Cheze Le Rest C, Hatt M, Visvikis D. Radiogenomics in Colorectal Cancer. Cancers (Basel) 2021; 13:cancers13050973. [PMID: 33652647 PMCID: PMC7956421 DOI: 10.3390/cancers13050973] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 02/07/2021] [Accepted: 02/20/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Colorectal carcinoma is characterized by intratumoral heterogeneity that can be assessed by radiogenomics. Radiomics, high-throughput quantitative data extracted from medical imaging, combined with molecular analysis, through genomic and transcriptomic data, is expected to lead to significant advances in personalized medicine. However, a radiogenomics approach in colorectal cancer is still in its early stages and many problems remain to be solved. Here we review the progress and challenges in this field at its current stage, as well as future developments. Abstract The steady improvement of high-throughput technologies greatly facilitates the implementation of personalized precision medicine. Characterization of tumor heterogeneity through image-derived features—radiomics and genetic profile modifications—genomics, is a rapidly evolving field known as radiogenomics. Various radiogenomics studies have been dedicated to colorectal cancer so far, highlighting the potential of these approaches to enhance clinical decision-making. In this review, a general outline of colorectal radiogenomics literature is provided, discussing the current limitations and suggested further developments.
Collapse
Affiliation(s)
- Bogdan Badic
- National Institute of Health and Medical Research, LaTIM—Laboratory of Medical Information Processing (INSERM LaTIM), UMR 1101, Université Bretagne Occidentale, 29238 Brest, France; (F.T.); (C.C.L.R.); (M.H.); (D.V.)
- Correspondence: ; Tel.: +33-298-347-215
| | - Florent Tixier
- National Institute of Health and Medical Research, LaTIM—Laboratory of Medical Information Processing (INSERM LaTIM), UMR 1101, Université Bretagne Occidentale, 29238 Brest, France; (F.T.); (C.C.L.R.); (M.H.); (D.V.)
| | - Catherine Cheze Le Rest
- National Institute of Health and Medical Research, LaTIM—Laboratory of Medical Information Processing (INSERM LaTIM), UMR 1101, Université Bretagne Occidentale, 29238 Brest, France; (F.T.); (C.C.L.R.); (M.H.); (D.V.)
- Department of Nuclear Medicine, University Hospital of Poitiers, 86021 Poitiers, France
| | - Mathieu Hatt
- National Institute of Health and Medical Research, LaTIM—Laboratory of Medical Information Processing (INSERM LaTIM), UMR 1101, Université Bretagne Occidentale, 29238 Brest, France; (F.T.); (C.C.L.R.); (M.H.); (D.V.)
| | - Dimitris Visvikis
- National Institute of Health and Medical Research, LaTIM—Laboratory of Medical Information Processing (INSERM LaTIM), UMR 1101, Université Bretagne Occidentale, 29238 Brest, France; (F.T.); (C.C.L.R.); (M.H.); (D.V.)
| |
Collapse
|
27
|
Del Re M, Cucchiara F, Rofi E, Fontanelli L, Petrini I, Gri N, Pasquini G, Rizzo M, Gabelloni M, Belluomini L, Crucitta S, Ciampi R, Frassoldati A, Neri E, Porta C, Danesi R. A multiparametric approach to improve the prediction of response to immunotherapy in patients with metastatic NSCLC. Cancer Immunol Immunother 2021; 70:1667-1678. [PMID: 33315149 PMCID: PMC8139911 DOI: 10.1007/s00262-020-02810-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 11/23/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND It is still unclear how to combine biomarkers to identify patients who will truly benefit from anti-PD-1 agents in NSCLC. This study investigates exosomal mRNA expression of PD-L1 and IFN-γ, PD-L1 polymorphisms, tumor mutational load (TML) in circulating cell-free DNA (cfDNA) and radiomic features as possible predictive markers of response to nivolumab and pembrolizumab in metastatic NSCLC patients. METHODS Patients were enrolled and blood (12 ml) was collected at baseline before receiving anti-PD-1 therapy. Exosome-derived mRNA and cfDNA were extracted to analyse PD-L1 and IFN-γ expression and tumor mutational load (TML) by digital droplet PCR (ddPCR) and next-generation sequencing (NGS), respectively. The PD-L1 single nucleotide polymorphisms (SNPs) c.-14-368 T > C and c.*395G > C, were analysed on genomic DNA by Real-Time PCR. A radiomic analysis was performed on the QUIBIM Precision® V3.0 platform. RESULTS Thirty-eight patients were enrolled. High baseline IFN-γ was independently associated with shorter median PFS (5.6 months vs. not reached p = 0.0057), and levels of PD-L1 showed an increase at 3 months vs. baseline in patients who progressed (p = 0.01). PD-L1 baseline levels showed significant direct and inverse relationships with radiomic features. Radiomic features also inversely correlated with PD-L1 expression in tumor tissue. In subjects receiving nivolumab, median PFS was shorter in carriers of c.*395GG vs. c.*395GC/CC genotype (2.3 months vs. not reached, p = 0.041). Lastly, responders had higher non-synonymous mutations and more links between co-occurring genetic somatic mutations and ARID1A alterations as well. CONCLUSIONS A combined multiparametric approach may provide a better understanding of the molecular determinants of response to immunotherapy.
Collapse
Affiliation(s)
- Marzia Del Re
- grid.5395.a0000 0004 1757 3729Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Federico Cucchiara
- grid.5395.a0000 0004 1757 3729Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Eleonora Rofi
- grid.5395.a0000 0004 1757 3729Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Lorenzo Fontanelli
- grid.5395.a0000 0004 1757 3729Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Iacopo Petrini
- grid.5395.a0000 0004 1757 3729General Pathology, Department of Translational Research and New Technologies in Medicine, University of Pisa, Pisa, Italy
| | - Nicole Gri
- Division of Translational Oncology, IRCCS Istituti Clinici Scientifici Maugeri, Pavia, Italy
| | - Giulia Pasquini
- grid.5395.a0000 0004 1757 3729General Pathology, Department of Translational Research and New Technologies in Medicine, University of Pisa, Pisa, Italy
| | - Mimma Rizzo
- Division of Translational Oncology, IRCCS Istituti Clinici Scientifici Maugeri, Pavia, Italy
| | - Michela Gabelloni
- grid.5395.a0000 0004 1757 3729Diagnostic and Interventional Radiology, Department of Translational Research and New Technologies in Medicine, University of Pisa, Pisa, Italy
| | - Lorenzo Belluomini
- grid.416315.4Unit of Clinical Oncology, Specialist Medical Department, S. Anna University Hospital, Ferrara, Italy
| | - Stefania Crucitta
- grid.5395.a0000 0004 1757 3729Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Raffaele Ciampi
- grid.5395.a0000 0004 1757 3729Endocrinology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Antonio Frassoldati
- grid.416315.4Unit of Clinical Oncology, Specialist Medical Department, S. Anna University Hospital, Ferrara, Italy
| | - Emanuele Neri
- grid.5395.a0000 0004 1757 3729Diagnostic and Interventional Radiology, Department of Translational Research and New Technologies in Medicine, University of Pisa, Pisa, Italy
| | - Camillo Porta
- Division of Translational Oncology, IRCCS Istituti Clinici Scientifici Maugeri, Pavia, Italy ,grid.8982.b0000 0004 1762 5736Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy ,grid.7644.10000 0001 0120 3326Present Address: Unit of Medical Oncology, Department of Biomedical Sciences and Human Oncology, University of Bari ‘A. Moro’, Bari, Italy
| | - Romano Danesi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.
| |
Collapse
|
28
|
Litvin A, Burkin D, Kropinov A, Paramzin F. Radiomics and Digital Image Texture Analysis in Oncology (Review). Sovrem Tekhnologii Med 2021; 13:97-104. [PMID: 34513082 PMCID: PMC8353717 DOI: 10.17691/stm2021.13.2.11] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Indexed: 12/12/2022] Open
Abstract
One of the most promising areas of diagnosis and prognosis of diseases is radiomics, a science combining radiology, mathematical modeling, and deep machine learning. The main concept of radiomics is image biomarkers (IBMs), the parameters characterizing various pathological changes and calculated based on the analysis of digital image texture. IBMs are used for quantitative assessment of digital imaging results (CT, MRI, ultrasound, PET). The use of IBMs in the form of "virtual biopsy" is of particular relevance in oncology. The article provides the basic concepts of radiomics identifying the main stages of obtaining IBMs: data collection and preprocessing, tumor segmentation, data detection and extraction, modeling, statistical processing, and data validation. The authors have analyzed the possibilities of using IBMs in oncology, describing the currently known features and advantages of using radiomics and image texture analysis in the diagnosis and prognosis of cancer. The limitations and problems associated with the use of radiomics data are considered. Although the novel effective tool for performing virtual biopsy of human tissue is at the development stage, quite a few projects have already been implemented, and medical software packages for radiomics analysis of digital images have been created.
Collapse
Affiliation(s)
- A.A. Litvin
- Professor, Department of Surgical Disciplines, Immanuel Kant Baltic Federal University, 14 A. Nevskogo St., Kaliningrad, 236016, Russia; Deputy Head Physician for Medical Aspects, Regional Clinical Hospital of the Kaliningrad Region, 74 Klinicheskaya St., Kaliningrad, 236016, Russia
| | - D.A. Burkin
- PhD Student in Information Science and Computer Engineering, Immanuel Kant Baltic Federal University, 14 A. Nevskogo St., Kaliningrad, 236016, Russia
| | - A.A. Kropinov
- Therapeutist, Central City Clinical Hospital, 3 Letnyaya St., Kaliningrad, 236005, Russia
| | - F.N. Paramzin
- Oncologist, Central City Clinical Hospital, 3 Letnyaya St., Kaliningrad, 236005, Russia
| |
Collapse
|
29
|
Cucchiara F, Del Re M, Valleggi S, Romei C, Petrini I, Lucchesi M, Crucitta S, Rofi E, De Liperi A, Chella A, Russo A, Danesi R. Integrating Liquid Biopsy and Radiomics to Monitor Clonal Heterogeneity of EGFR-Positive Non-Small Cell Lung Cancer. Front Oncol 2020; 10:593831. [PMID: 33489892 PMCID: PMC7819134 DOI: 10.3389/fonc.2020.593831] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 10/30/2020] [Indexed: 12/19/2022] Open
Abstract
Background EGFR-positive Non-small Cell Lung Cancer (NSCLC) is a dynamic entity and tumor progression and resistance to tyrosine kinase inhibitors (TKIs) arise from the accumulation, over time and across different disease sites, of subclonal genetic mutations. For instance, the occurrence of EGFR T790M is associated with resistance to gefitinib, erlotinib, and afatinib, while EGFR C797S causes osimertinib to lose activity. Sensitive technologies as radiomics and liquid biopsy have great potential to monitor tumor heterogeneity since they are both minimally invasive, easy to perform, and can be repeated over patient’s follow-up, enabling the extraction of valuable information. Yet, to date, there are no reported cases associating liquid biopsy and radiomics during treatment. Case presentation In this case series, seven patients with metastatic EGFR-positive NSCLC have been monitored during target therapy. Plasma-derived cell free DNA (cfDNA) was analyzed by a digital droplet PCR (ddPCR), while radiomic analyses were performed using the validated LifeX® software on computed tomography (CT)-images. The dynamics of EGFR mutations in cfDNA was compared with that of radiomic features. Then, for each EGFR mutation, a radiomic signature was defines as the sum of the most predictive features, weighted by their corresponding regression coefficients for the least absolute shrinkage and selection operator (LASSO) model. The receiver operating characteristic (ROC) curves were computed to estimate their diagnostic performance. The signatures achieved promising performance on predicting the presence of EGFR mutations (R2 = 0.447, p <0.001 EGFR activating mutations R2 = 0.301, p = 0.003 for T790M; and R2 = 0.354, p = 0.001 for activating plus resistance mutations), confirmed by ROC analysis. Conclusion To our knowledge, these are the first cases to highlight a potentially promising strategy to detect clonal heterogeneity and ultimately identify patients at risk of progression during treatment. Together, radiomics and liquid biopsy could detect the appearance of new mutations and therefore suggest new therapeutic management.
Collapse
Affiliation(s)
- Federico Cucchiara
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Marzia Del Re
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Simona Valleggi
- Pneumology Unit, Cardiovascular and Thoracic Department, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Chiara Romei
- Radiology Unit 2, Department of Diagnostics and Imaging, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Iacopo Petrini
- Pneumology Unit, Cardiovascular and Thoracic Department, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy.,Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Maurizio Lucchesi
- Pneumology Unit, Cardiovascular and Thoracic Department, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Stefania Crucitta
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Eleonora Rofi
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Annalisa De Liperi
- Radiology Unit 2, Department of Diagnostics and Imaging, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Antonio Chella
- Pneumology Unit, Cardiovascular and Thoracic Department, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Antonio Russo
- Section of Medical Oncology, Department of Surgical, Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy
| | - Romano Danesi
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
30
|
|
31
|
Gill AB, Rundo L, Wan JCM, Lau D, Zawaideh JP, Woitek R, Zaccagna F, Beer L, Gale D, Sala E, Couturier DL, Corrie PG, Rosenfeld N, Gallagher FA. Correlating Radiomic Features of Heterogeneity on CT with Circulating Tumor DNA in Metastatic Melanoma. Cancers (Basel) 2020; 12:E3493. [PMID: 33255267 PMCID: PMC7759931 DOI: 10.3390/cancers12123493] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 11/17/2020] [Indexed: 12/18/2022] Open
Abstract
Clinical imaging methods, such as computed tomography (CT), are used for routine tumor response monitoring. Imaging can also reveal intratumoral, intermetastatic, and interpatient heterogeneity, which can be quantified using radiomics. Circulating tumor DNA (ctDNA) in the plasma is a sensitive and specific biomarker for response monitoring. Here we evaluated the interrelationship between circulating tumor DNA mutant allele fraction (ctDNAmaf), obtained by targeted amplicon sequencing and shallow whole genome sequencing, and radiomic measurements of CT heterogeneity in patients with stage IV melanoma. ctDNAmaf and radiomic observations were obtained from 15 patients with a total of 70 CT examinations acquired as part of a prospective trial. 26 of 39 radiomic features showed a significant relationship with log(ctDNAmaf). Principal component analysis was used to define a radiomics signature that predicted ctDNAmaf independent of lesion volume. This radiomics signature and serum lactate dehydrogenase were independent predictors of ctDNAmaf. Together, these results suggest that radiomic features and ctDNAmaf may serve as complementary clinical tools for treatment monitoring.
Collapse
Affiliation(s)
- Andrew B. Gill
- Department of Radiology, University of Cambridge, Cambridge CB2 0QQ, UK; (L.R.); (D.L.); (J.P.Z.); (R.W.); (F.Z.); (L.B.); (E.S.); (F.A.G.)
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge CB2 0RE, UK; (D.G.); (N.R.)
- Imaging Department, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Leonardo Rundo
- Department of Radiology, University of Cambridge, Cambridge CB2 0QQ, UK; (L.R.); (D.L.); (J.P.Z.); (R.W.); (F.Z.); (L.B.); (E.S.); (F.A.G.)
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge CB2 0RE, UK; (D.G.); (N.R.)
| | - Jonathan C. M. Wan
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK; (J.C.M.W.); (D.-L.C.)
| | - Doreen Lau
- Department of Radiology, University of Cambridge, Cambridge CB2 0QQ, UK; (L.R.); (D.L.); (J.P.Z.); (R.W.); (F.Z.); (L.B.); (E.S.); (F.A.G.)
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge CB2 0RE, UK; (D.G.); (N.R.)
| | - Jeries P. Zawaideh
- Department of Radiology, University of Cambridge, Cambridge CB2 0QQ, UK; (L.R.); (D.L.); (J.P.Z.); (R.W.); (F.Z.); (L.B.); (E.S.); (F.A.G.)
| | - Ramona Woitek
- Department of Radiology, University of Cambridge, Cambridge CB2 0QQ, UK; (L.R.); (D.L.); (J.P.Z.); (R.W.); (F.Z.); (L.B.); (E.S.); (F.A.G.)
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge CB2 0RE, UK; (D.G.); (N.R.)
| | - Fulvio Zaccagna
- Department of Radiology, University of Cambridge, Cambridge CB2 0QQ, UK; (L.R.); (D.L.); (J.P.Z.); (R.W.); (F.Z.); (L.B.); (E.S.); (F.A.G.)
| | - Lucian Beer
- Department of Radiology, University of Cambridge, Cambridge CB2 0QQ, UK; (L.R.); (D.L.); (J.P.Z.); (R.W.); (F.Z.); (L.B.); (E.S.); (F.A.G.)
| | - Davina Gale
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge CB2 0RE, UK; (D.G.); (N.R.)
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK; (J.C.M.W.); (D.-L.C.)
| | - Evis Sala
- Department of Radiology, University of Cambridge, Cambridge CB2 0QQ, UK; (L.R.); (D.L.); (J.P.Z.); (R.W.); (F.Z.); (L.B.); (E.S.); (F.A.G.)
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge CB2 0RE, UK; (D.G.); (N.R.)
| | - Dominique-Laurent Couturier
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK; (J.C.M.W.); (D.-L.C.)
| | - Pippa G. Corrie
- Department of Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK;
| | - Nitzan Rosenfeld
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge CB2 0RE, UK; (D.G.); (N.R.)
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK; (J.C.M.W.); (D.-L.C.)
| | - Ferdia A. Gallagher
- Department of Radiology, University of Cambridge, Cambridge CB2 0QQ, UK; (L.R.); (D.L.); (J.P.Z.); (R.W.); (F.Z.); (L.B.); (E.S.); (F.A.G.)
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge CB2 0RE, UK; (D.G.); (N.R.)
- Imaging Department, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| |
Collapse
|
32
|
Gabelloni M, Faggioni L, Attanasio S, Vani V, Goddi A, Colantonio S, Germanese D, Caudai C, Bruschini L, Scarano M, Seccia V, Neri E. Can Magnetic Resonance Radiomics Analysis Discriminate Parotid Gland Tumors? A Pilot Study. Diagnostics (Basel) 2020; 10:diagnostics10110900. [PMID: 33153140 PMCID: PMC7692594 DOI: 10.3390/diagnostics10110900] [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] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 10/28/2020] [Accepted: 11/01/2020] [Indexed: 12/29/2022] Open
Abstract
Our purpose is to evaluate the performance of magnetic resonance (MR) radiomics analysis for differentiating between malignant and benign parotid neoplasms and, among the latter, between pleomorphic adenomas and Warthin tumors. We retrospectively evaluated 75 T2-weighted images of parotid gland lesions, of which 61 were benign tumors (32 pleomorphic adenomas, 23 Warthin tumors and 6 oncocytomas) and 14 were malignant tumors. A receiver operating characteristics (ROC) curve analysis was performed to find the threshold values for the most discriminative features and determine their sensitivity, specificity and area under the ROC curve (AUROC). The most discriminative features were used to train a support vector machine classifier. The best classification performance was obtained by comparing a pleomorphic adenoma with a Warthin tumor (yielding sensitivity, specificity and a diagnostic accuracy as high as 0.8695, 0.9062 and 0.8909, respectively) and a pleomorphic adenoma with malignant tumors (sensitivity, specificity and a diagnostic accuracy of 0.6666, 0.8709 and 0.8043, respectively). Radiomics analysis of parotid tumors on conventional T2-weighted MR images allows the discrimination of pleomorphic adenomas from Warthin tumors and malignant tumors with a high sensitivity, specificity and diagnostic accuracy.
Collapse
Affiliation(s)
- Michela Gabelloni
- Master in Oncologic Imaging, Diagnostic and Interventional Radiology, Department of Translational Research, University of Pisa, Via Roma, 67, 56126 Pisa, Italy; (M.G.); (E.N.)
| | - Lorenzo Faggioni
- Master in Oncologic Imaging, Diagnostic and Interventional Radiology, Department of Translational Research, University of Pisa, Via Roma, 67, 56126 Pisa, Italy; (M.G.); (E.N.)
- Correspondence: ; Tel.: +39-050-995835
| | - Simona Attanasio
- Diagnostic and Interventional Radiology, Department of Translational Research, University of Pisa, Via Roma, 67, 56126 Pisa, Italy; (S.A.); (V.V.); (A.G.)
| | - Vanina Vani
- Diagnostic and Interventional Radiology, Department of Translational Research, University of Pisa, Via Roma, 67, 56126 Pisa, Italy; (S.A.); (V.V.); (A.G.)
| | - Antonio Goddi
- Diagnostic and Interventional Radiology, Department of Translational Research, University of Pisa, Via Roma, 67, 56126 Pisa, Italy; (S.A.); (V.V.); (A.G.)
| | - Sara Colantonio
- Institute of Information Science and Technologies “A. Faedo” of the National Research Council of Italy (ISTI-CNR), 56124 Pisa, Italy; (S.C.); (D.G.); (C.C.)
| | - Danila Germanese
- Institute of Information Science and Technologies “A. Faedo” of the National Research Council of Italy (ISTI-CNR), 56124 Pisa, Italy; (S.C.); (D.G.); (C.C.)
| | - Claudia Caudai
- Institute of Information Science and Technologies “A. Faedo” of the National Research Council of Italy (ISTI-CNR), 56124 Pisa, Italy; (S.C.); (D.G.); (C.C.)
| | - Luca Bruschini
- Otolaryngology, Audiology, and Phoniatric Operative Unit, Department of Surgical, Medical, Molecular Pathology, and Critical Care Medicine, Azienda Ospedaliero Universitaria Pisana, University of Pisa, 56124 Pisa, Italy; (L.B.); (M.S.); (V.S.)
| | - Mariella Scarano
- Otolaryngology, Audiology, and Phoniatric Operative Unit, Department of Surgical, Medical, Molecular Pathology, and Critical Care Medicine, Azienda Ospedaliero Universitaria Pisana, University of Pisa, 56124 Pisa, Italy; (L.B.); (M.S.); (V.S.)
| | - Veronica Seccia
- Otolaryngology, Audiology, and Phoniatric Operative Unit, Department of Surgical, Medical, Molecular Pathology, and Critical Care Medicine, Azienda Ospedaliero Universitaria Pisana, University of Pisa, 56124 Pisa, Italy; (L.B.); (M.S.); (V.S.)
| | - Emanuele Neri
- Master in Oncologic Imaging, Diagnostic and Interventional Radiology, Department of Translational Research, University of Pisa, Via Roma, 67, 56126 Pisa, Italy; (M.G.); (E.N.)
| |
Collapse
|
33
|
Trimboli RM, Giorgi Rossi P, Battisti NML, Cozzi A, Magni V, Zanardo M, Sardanelli F. Do we still need breast cancer screening in the era of targeted therapies and precision medicine? Insights Imaging 2020; 11:105. [PMID: 32975658 PMCID: PMC7519022 DOI: 10.1186/s13244-020-00905-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 08/20/2020] [Indexed: 12/27/2022] Open
Abstract
Breast cancer (BC) is the most common female cancer and the second cause of death among women worldwide. The 5-year relative survival rate recently improved up to 90% due to increased population coverage and women's attendance to organised mammography screening as well as to advances in therapies, especially systemic treatments. Screening attendance is associated with a mortality reduction of at least 30% and a 40% lower risk of advanced disease. The stage at diagnosis remains the strongest predictor of recurrences. Systemic treatments evolved dramatically over the last 20 years: aromatase inhibitors improved the treatment of early-stage luminal BC; targeted monoclonal antibodies changed the natural history of anti-human epidermal growth factor receptor 2-positive (HER2) disease; immunotherapy is currently investigated in patients with triple-negative BC; gene expression profiling is now used with the aim of personalising systemic treatments. In the era of precision medicine, it is a challenging task to define the relative contribution of early diagnosis by screening mammography and systemic treatments in determining BC survival. Estimated contributions before 2000 were 46% for screening and 54% for treatment advances and after 2000, 37% and 63%, respectively. A model showed that the 10-year recurrence rate would be 30% and 25% using respectively chemotherapy or novel treatments in the absence of screening, but would drop to 19% and 15% respectively if associated with mammography screening. Early detection per se has not a curative intent and systemic treatment has limited benefit on advanced stages. Both screening mammography and systemic therapies continue to positively contribute to BC prognosis.
Collapse
Affiliation(s)
- Rubina Manuela Trimboli
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Via Mangiagalli 31, 20133 Milan, Italy
| | - Paolo Giorgi Rossi
- Epidemiology Unit, Azienda USL–IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Nicolò Matteo Luca Battisti
- Breast Unit–Department of Medicine, The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, London, SM2 5PT UK
- Breast Cancer Research Division, The Institute of Cancer Research, 15 Cotswold Road, Sutton, London, SM2 5NG UK
| | - Andrea Cozzi
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Via Mangiagalli 31, 20133 Milan, Italy
| | - Veronica Magni
- Medical School, Università degli Studi di Milano, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Moreno Zanardo
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Via Mangiagalli 31, 20133 Milan, Italy
| | - Francesco Sardanelli
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Via Mangiagalli 31, 20133 Milan, Italy
- Unit of Radiology, IRCCS Policlinico San Donato, Via Morandi 30, 20097 San Donato Milanese, Italy
| |
Collapse
|
34
|
Del Re M, Cucchiara F, Petrini I, Fogli S, Passaro A, Crucitta S, Attili I, De Marinis F, Chella A, Danesi R. erbB in NSCLC as a molecular target: current evidences and future directions. ESMO Open 2020; 5:e000724. [PMID: 32820012 PMCID: PMC7443272 DOI: 10.1136/esmoopen-2020-000724] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/06/2020] [Accepted: 06/08/2020] [Indexed: 12/13/2022] Open
Abstract
A number of treatments have been developed for HER1, 2 and 3-driven non-small cell lung cancer (NSCLC), of which the most successful have been the epidermal growth factor receptor-tyrosine kinase inhibitors in HER1-mutant tumours resulting in highly improved progression-free survival. Human epidermal growth factor (HER)2 and 3-driven tumours represent the minority of NSCLC, and effective therapies in these patients still represent an unmet medical need. The encouraging results seen with anti-HER2 and anti-HER3 monoclonal antibodies need to be validated in larger studies, even if the greatest obstacle is represented by the exiguous number of patients bearing deregulated HER2/3 system and abnormalities of signal transduction pathway. Considering NSCLC tumour heterogeneity, which affects response and resistance to treatment, combined multiparametric approaches, such as liquid biopsy together with radiomics, may provide a better understanding of the tumour dynamics and clonal selection during the treatments.
Collapse
Affiliation(s)
- Marzia Del Re
- Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Federico Cucchiara
- Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Iacopo Petrini
- Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Stefano Fogli
- Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Antonio Passaro
- Division of Thoracic Oncology, European Institute of Oncology - IRCCS, Milan, Italy
| | - Stefania Crucitta
- Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Ilaria Attili
- Division of Thoracic Oncology, European Institute of Oncology - IRCCS, Milan, Italy
| | - Filippo De Marinis
- Division of Thoracic Oncology, European Institute of Oncology - IRCCS, Milan, Italy
| | - Antonio Chella
- Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
| | - Romano Danesi
- Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| |
Collapse
|
35
|
Ninatti G, Kirienko M, Neri E, Sollini M, Chiti A. Imaging-Based Prediction of Molecular Therapy Targets in NSCLC by Radiogenomics and AI Approaches: A Systematic Review. Diagnostics (Basel) 2020; 10:E359. [PMID: 32486314 PMCID: PMC7345054 DOI: 10.3390/diagnostics10060359] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/28/2020] [Accepted: 05/29/2020] [Indexed: 12/11/2022] Open
Abstract
The objective of this systematic review was to analyze the current state of the art of imaging-derived biomarkers predictive of genetic alterations and immunotherapy targets in lung cancer. We included original research studies reporting the development and validation of imaging feature-based models. The overall quality, the standard of reporting and the advancements towards clinical practice were assessed. Eighteen out of the 24 selected articles were classified as "high-quality" studies according to the Quality Assessment of Diagnostic Accuracy Studies 2 (QUADAS-2). The 18 "high-quality papers" adhered to Transparent Reporting of a multivariable prediction model for Individual Prognosis or Diagnosis (TRIPOD) with a mean of 62.9%. The majority of "high-quality" studies (16/18) were classified as phase II. The most commonly used imaging predictors were radiomic features, followed by visual qualitative computed tomography (CT) features, convolutional neural network-based approaches and positron emission tomography (PET) parameters, all used alone or combined with clinicopathologic features. The majority (14/18) were focused on the prediction of epidermal growth factor receptor (EGFR) mutation. Thirty-five imaging-based models were built to predict the EGFR status. The model's performances ranged from weak (n = 5) to acceptable (n = 11), to excellent (n = 18) and outstanding (n = 1) in the validation set. Positive outcomes were also reported for the prediction of ALK rearrangement, ALK/ROS1/RET fusions and programmed cell death ligand 1 (PD-L1) expression. Despite the promising results in terms of predictive performance, image-based models, suffering from methodological bias, require further validation before replacing traditional molecular pathology testing.
Collapse
Affiliation(s)
- Gaia Ninatti
- Humanitas University, Pieve Emanuele, 20090 Milan, Italy; (G.N.); (A.C.)
| | | | - Emanuele Neri
- Department of Translational Research, Diagnostic Radiology 3, University of Pisa, 56126 Pisa, Italy;
| | - Martina Sollini
- Humanitas University, Pieve Emanuele, 20090 Milan, Italy; (G.N.); (A.C.)
- Humanitas Clinical and Research Center-IRCCS, Rozzano, 20089 Milan, Italy
| | - Arturo Chiti
- Humanitas University, Pieve Emanuele, 20090 Milan, Italy; (G.N.); (A.C.)
- Humanitas Clinical and Research Center-IRCCS, Rozzano, 20089 Milan, Italy
| |
Collapse
|
36
|
Adversarial radiomics: the rising of potential risks in medical imaging from adversarial learning. Eur J Nucl Med Mol Imaging 2020; 47:2941-2943. [DOI: 10.1007/s00259-020-04879-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
37
|
The texture analysis as a predictive method in the assessment of the cytological specimen of CT-guided FNAC of the lung cancer. Med Oncol 2020; 37:54. [PMID: 32424733 DOI: 10.1007/s12032-020-01375-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 04/13/2020] [Indexed: 02/07/2023]
Abstract
The lung cancer is the principle cause of the worldwide deaths and its prognosis is poor with a 5-year overall survival rate. Computed tomography (CT) gives many information about the prognosis, but the problem is the subject interpretation of the findings. Thanks to the computer-aided diagnosis/detection (CAD), it is possible to reduce the second opinion. "Radiomics" is an extension of CAD and overlaps the quantitative imaging data of the CT texture analysis (CTTA) with the clinical information, increasing the power and precision of the decision going through the personalized medicine. The aim of this study is to describe the role of the radiomics in the characterization of the pulmonary nodule. For this study, we retrospectively analyzed the images of the 87 NSCLC patients with a waiver of informed consent from the Institutional Review Board (IRB) at the Campania University "Luigi Vanvitelli" of Naples. All tumors were semiautomatically segmented by a radiologist with 10 years of experience using three diameters (AW Server 3.2). The examinations were acquired using 128 MDCT (GSI CT, GE) with a peak tube voltage of 120 kVp, tube current of 100 or 200 mA, and rotation times of 0.5 or 0.8 s. To confirm the imaging results, the FNAC was performed and for every nodule the following parameters were extracted: the presence of the solid component (named = 1), papillary component (named = 2), and mixed component (named = 3). Feature calculation was performed using the HealthMyne software and Integrated Platform That Enables Better Patient Management Decisions For Oncology. The radiologist uses the Rapid Precise Metrics (RPM)™ functionality to identify a lesion with the algorithm and these methods are put to work. The correlation between each feature and the tumor volume was calculated using a two-step cluster statistical analysis. In this retrospective study, in one year from 2018 to 2019 20 patients with lung adenocarcinoma confirmed with FNAC were enrolled. The pathologic results were subdivided into three categories: the solid architecture (group 1), papillary architecture (group 2), and mixed architecture (group 3). Nine lesions resulted with component 1, seven patients with component 2, and 3 patients with component 3. Eight females and 12 males with a median age 61 and 15 years (mean ± SD = 67.4 ± 9.7 years, range 39-73 years) were enrolled. The two results suggest, with p < 0.05, that the GGO variable is a good discriminating estimator of the kurtosis variable: GGO = "no" implies a high kurtosis value, while GGO = "yes" implies a low value. The numerous data obtained from the automatic analysis allow to have a fertile ground on which to develop a new concept of medicine which is precision medicine. The limit of this study is the poor sample. In the future, in order to have a more mature and consolidated discipline, it is necessary to increase the large scale of observations with further studies to establish the rigorous evaluation criteria. In order for radiomics to mature as a discipline in the future, it will be necessary to develop studies that consolidate its role to standardize the collected data.
Collapse
|
38
|
Valladares A, Beyer T, Rausch I. Physical imaging phantoms for simulation of tumor heterogeneity in PET, CT, and MRI: An overview of existing designs. Med Phys 2020; 47:2023-2037. [PMID: 31981214 PMCID: PMC7216968 DOI: 10.1002/mp.14045] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 01/09/2020] [Accepted: 01/10/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND In oncology, lesion characterization is essential for tumor grading, treatment planning, and follow-up of cancer patients. Hybrid imaging systems, such as Single Photon Emission Computed Tomography (SPECT)/CT, Positron Emission Tomography (PET)/CT, or PET/magnetic resonance imaging (MRI), play an essential role for the noninvasive quantification of tumor characteristics. However, most of the existing approaches are challenged by intra- and intertumor heterogeneity. Novel quantitative imaging parameters that can be derived from textural feature analysis (as part of radiomics) are promising complements for improved characterization of tumor heterogeneity, thus, supporting clinically relevant implementations of personalized medicine concepts. Nevertheless, establishing new quantitative parameters for tumor characterization requires the use of standardized imaging objects to test the reliability of results prior to their implementation in patient studies. METHODS In this review, we summarize existing reports on heterogeneous phantoms with a focus on simulating tumor heterogeneity. We discuss the techniques, materials, advantages, and limitations of the existing phantoms for PET, CT, and MR imaging modalities. CONCLUSIONS Finally, we outline the future directions and requirements for the design of cross modality imaging phantoms.
Collapse
Affiliation(s)
- Alejandra Valladares
- QIMP TeamCentre for Medical Physics and Biomedical EngineeringMedical University of ViennaVienna1090Austria
| | - Thomas Beyer
- QIMP TeamCentre for Medical Physics and Biomedical EngineeringMedical University of ViennaVienna1090Austria
| | - Ivo Rausch
- QIMP TeamCentre for Medical Physics and Biomedical EngineeringMedical University of ViennaVienna1090Austria
| |
Collapse
|
39
|
Hu HT, Shan QY, Chen SL, Li B, Feng ST, Xu EJ, Li X, Long JY, Xie XY, Lu MD, Kuang M, Shen JX, Wang W. CT-based radiomics for preoperative prediction of early recurrent hepatocellular carcinoma: technical reproducibility of acquisition and scanners. Radiol Med 2020; 125:697-705. [PMID: 32200455 DOI: 10.1007/s11547-020-01174-2] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 03/11/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE To test the technical reproducibility of acquisition and scanners of CT image-based radiomics model for early recurrent hepatocellular carcinoma (HCC). METHODS We included primary HCC patient undergone curative therapies, using early recurrence as endpoint. Four datasets were constructed: 109 images from hospital #1 for training (set 1: 1-mm image slice thickness), 47 images from hospital #1 for internal validation (sets 2 and 3: 1-mm and 10-mm image slice thicknesses, respectively), and 47 images from hospital #2 for external validation (set 4: vastly different from training dataset). A radiomics model was constructed. Radiomics technical reproducibility was measured by overfitting and calibration deviation in external validation dataset. The influence of slice thickness on reproducibility was evaluated in two internal validation datasets. RESULTS Compared with set 1, the model in set 2 indicated favorable prediction efficiency (the area under the curve 0.79 vs. 0.80, P = 0.47) and good calibration (unreliability statistic U: P = 0.33). However, in set 4, significant overfitting (0.63 vs. 0.80, P < 0.01) and calibration deviation (U: P < 0.01) were observed. Similar poor performance was also observed in set 3 (0.56 vs. 0.80, P = 0.02; U: P < 0.01). CONCLUSIONS CT-based radiomics has poor reproducibility between centers. Image heterogeneity, such as slice thickness, can be a significant influencing factor.
Collapse
Affiliation(s)
- Hang-Tong Hu
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, Ultrasomics Artificial Intelligence X-Lab, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhong Shan Road 2, Guangzhou, 510080, China
| | - Quan-Yuan Shan
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, Ultrasomics Artificial Intelligence X-Lab, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhong Shan Road 2, Guangzhou, 510080, China
| | - Shu-Ling Chen
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, Ultrasomics Artificial Intelligence X-Lab, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhong Shan Road 2, Guangzhou, 510080, China
| | - Bin Li
- Clinical trials Unit, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Shi-Ting Feng
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhong Shan Road 2, Guangzhou, 510080, China
| | - Er-Jiao Xu
- Department of Medical Ultrasonics, The Third Affiliated Hospital of Sun Yat-Sen University, Guangdong Key Laboratory of Liver Disease Research, Guangzhou, 510630, Guangdong Province, China
| | - Xin Li
- GE Healthcare, Shanghai, 200030, China
| | - Jian-Yan Long
- Clinical trials Unit, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiao-Yan Xie
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, Ultrasomics Artificial Intelligence X-Lab, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhong Shan Road 2, Guangzhou, 510080, China
| | - Ming-de Lu
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, Ultrasomics Artificial Intelligence X-Lab, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhong Shan Road 2, Guangzhou, 510080, China.,Department of Liver Surgery, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhong Shan Road 2, Guangzhou, 510080, China
| | - Ming Kuang
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, Ultrasomics Artificial Intelligence X-Lab, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhong Shan Road 2, Guangzhou, 510080, China.,Department of Liver Surgery, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhong Shan Road 2, Guangzhou, 510080, China
| | - Jing-Xian Shen
- Department of Radiology, State Key Laboratory of Oncology in South China, The Cancer Center, Sun Yat-sen University, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, China.
| | - Wei Wang
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, Ultrasomics Artificial Intelligence X-Lab, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhong Shan Road 2, Guangzhou, 510080, China.
| |
Collapse
|
40
|
Calandri M, Siravegna G, Yevich SM, Stranieri G, Gazzera C, Kopetz S, Fonio P, Gupta S, Bardelli A, Veltri A, Odisio BC. Liquid biopsy, a paradigm shift in oncology: what interventional radiologists should know. Eur Radiol 2020; 30:4496-4503. [PMID: 32193642 DOI: 10.1007/s00330-020-06700-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/21/2020] [Accepted: 01/31/2020] [Indexed: 02/08/2023]
Abstract
The acquisition of adequate tumor sample is required to verify primary tumor type and specific biomarkers and to assess response to therapy. Historically, invasive surgical procedures were the standard methods to acquire tumor samples until advancements in imaging and minimally invasive equipment facilitated the paradigm shift image-guided biopsy. Image-guided biopsy has improved sampling yield and minimized risk to the patient; however, there are still limitations, such as its invasive nature and its consequent limitations to longitudinal tumor monitoring. The next paradigm shift in sampling technique will need to address these issues to provide a more reliable and less invasive technique. Recently, liquid biopsy (LB) has emerged as a non-invasive alternative to tissue sampling. This technique relies on direct sampling of blood or other bodily fluids in contact with the tumor in order to collect circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), and circulating RNAs-in particular microRNA (miRNAs). Clinical applications of LB involve different steps of cancer patient management including screening, detection of disease recurrence, and evaluation of acquired resistance. With any paradigm shift, old techniques are often relegated to a secondary option. Although image-guided biopsies may appear as a passive spectator on the rapid advancement of LB, the two techniques may well be codependent. Interventional radiology may be integral to directly sample the liquid surrounding or draining from the tumor. In addition, LB may help to correctly select the patients for image-guided loco-regional treatments, to determine its treatment endpoint, and to early detect recurrence. KEY POINTS: • Liquid biopsy is a novel technology with potential high impact in the management of patients undergoing image-guided procedures. • Interventional radiology procedures may increase liquid biopsy sensitivity through direct fluid sampling. • Liquid biopsy techniques may provide a venue for improving patients' selection and enhance outcomes of interventional loco-regional therapies performed by interventional radiologists.
Collapse
Affiliation(s)
- Marco Calandri
- Radiology Unit, A.O.U. San Luigi Gonzaga - Orbassano (To), Orbassano, TO, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | - Giulia Siravegna
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo (To), Candiolo, TO, Italy.,Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Steven M Yevich
- Department of Interventional Radiology, MD Anderson Cancer Center, The University of Texas, Houston, TX, USA
| | - Giuseppe Stranieri
- Radiology Unit, A.O.U. San Luigi Gonzaga - Orbassano (To), Orbassano, TO, Italy
| | - Carlo Gazzera
- Radiology Institute, Città della Salute e della Scienza - Torino Department of Surgical Sciences, University of Turin, Turin, Italy
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, MD Anderson Cancer Center, The University of Texas, Houston, TX, USA
| | - Paolo Fonio
- Radiology Institute, Città della Salute e della Scienza - Torino Department of Surgical Sciences, University of Turin, Turin, Italy
| | - Sanjay Gupta
- Department of Interventional Radiology, MD Anderson Cancer Center, The University of Texas, Houston, TX, USA
| | - Alberto Bardelli
- Department of Oncology, University of Torino, Turin, Italy.,Candiolo Cancer Institute-FPO, IRCCS, Candiolo (To), Candiolo, TO, Italy
| | - Andrea Veltri
- Radiology Unit, A.O.U. San Luigi Gonzaga - Orbassano (To), Orbassano, TO, Italy.,Department of Oncology, University of Torino, Turin, Italy
| | - Bruno C Odisio
- Department of Interventional Radiology, MD Anderson Cancer Center, The University of Texas, Houston, TX, USA.
| |
Collapse
|
41
|
Borazanci E, Korn R, Liang WS, Guarnieri C, Haag S, Snyder C, Hendrickson K, Caldwell L, Von Hoff D, Jameson G. An Analysis of Patients with DNA Repair Pathway Mutations Treated with a PARP Inhibitor. Oncologist 2020; 25:e60-e67. [PMID: 31391296 PMCID: PMC6964119 DOI: 10.1634/theoncologist.2018-0905] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 07/05/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Molecular analysis has revealed four subtypes of pancreatic ductal adenocarcinoma (PDAC). One subtype identified for the presence of DNA damage repair deficiency can be targeted therapeutically with the poly (ADP-ribose) polymerase (PARP) inhibitor olaparib. We performed a single institution retrospective analysis of treatment response in patients with PDAC treated with olaparib who have DNA damage repair deficiency mutations. SUBJECTS, MATERIALS, AND METHODS Patients with germline or somatic mutations involving the DNA repair pathway were identified and treated with olaparib. The primary objective was to examine the objective response rate (ORR). The secondary objectives were assessing tolerability, overall survival, and change in cancer antigen 19-9. Quantitative texture analysis (QTA) was evaluated from CT scans to explore imaging biomarkers. RESULTS Thirteen individuals with metastatic PDAC were treated with Olaparib. The ORR to Olaparib was 23%. Median overall survival (OS) was 16.47 months. Four of seven patients with BRCA mutations had an effect on RAD51 binding, with a median OS of 24.60 months. Exploratory analysis of index lesions using QTA revealed correlations between lesion texture and OS (hepatic lesion tumor texture correlation coefficient [CC], 0.683, p = .042) and time on olaparib (primary pancreatic lesion tumor texture CC, 0.778, p = .023). CONCLUSION In individuals with metastatic PDAC who have mutations involved in DNA repair, Olaparib may provide clinical benefit. BRCA mutations affecting RAD51 binding domains translated to improved median OS. QTA of individual tumors may allow for additional information that predicts outcomes to treatment with PARP inhibitors. IMPLICATIONS FOR PRACTICE Pursuing germline and somatic DNA sequencing in individuals with pancreatic ductal adenocarcinoma may yield abnormalities in DNA repair pathways. These individuals may receive benefit with poly (ADP-ribose) polymerase (PARP) inhibition. Radiomics and deep sequencing analysis may yet uncover additional information that may predict outcome to treatment with PARP inhibitors.
Collapse
Affiliation(s)
- Erkut Borazanci
- HonorHealth Research InstituteScottsdaleArizonaUSA
- Translational Genomics Research InstitutePhoenixArizonaUSA
| | | | | | | | - Susan Haag
- HonorHealth Research InstituteScottsdaleArizonaUSA
| | | | | | | | - Dan Von Hoff
- HonorHealth Research InstituteScottsdaleArizonaUSA
- Translational Genomics Research InstitutePhoenixArizonaUSA
| | - Gayle Jameson
- HonorHealth Research InstituteScottsdaleArizonaUSA
- Translational Genomics Research InstitutePhoenixArizonaUSA
| |
Collapse
|
42
|
Dalal V, Carmicheal J, Dhaliwal A, Jain M, Kaur S, Batra SK. Radiomics in stratification of pancreatic cystic lesions: Machine learning in action. Cancer Lett 2019; 469:228-237. [PMID: 31629933 DOI: 10.1016/j.canlet.2019.10.023] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/03/2019] [Accepted: 10/15/2019] [Indexed: 12/15/2022]
Abstract
Pancreatic cystic lesions (PCLs) are well-known precursors of pancreatic cancer. Their diagnosis can be challenging as their behavior varies from benign to malignant disease. Precise and timely management of malignant pancreatic cysts might prevent transformation to pancreatic cancer. However, the current consensus guidelines, which rely on standard imaging features to predict cyst malignancy potential, are conflicting and unclear. This has led to an increased interest in radiomics, a high-throughput extraction of comprehensible data from standard of care images. Radiomics can be used as a diagnostic and prognostic tool in personalized medicine. It utilizes quantitative image analysis to extract features in conjunction with machine learning and artificial intelligence (AI) methods like support vector machines, random forest, and convolutional neural network for feature selection and classification. Selected features can then serve as imaging biomarkers to predict high-risk PCLs. Radiomics studies conducted heretofore on PCLs have shown promising results. This cost-effective approach would help us to differentiate benign PCLs from malignant ones and potentially guide clinical decision-making leading to better utilization of healthcare resources. In this review, we discuss the process of radiomics, its myriad applications such as diagnosis, prognosis, and prediction of therapy response. We also discuss the outcomes of studies involving radiomic analysis of PCLs and pancreatic cancer, and challenges associated with this novel field along with possible solutions. Although these studies highlight the potential benefit of radiomics in the prevention and optimal treatment of pancreatic cancer, further studies are warranted before incorporating radiomics into the clinical decision support system.
Collapse
Affiliation(s)
- Vipin Dalal
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Joseph Carmicheal
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Amaninder Dhaliwal
- Department of Gastroenterology and Hepatology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA; The Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sukhwinder Kaur
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA; The Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
43
|
Du R, Lee VH, Yuan H, Lam KO, Pang HH, Chen Y, Lam EY, Khong PL, Lee AW, Kwong DL, Vardhanabhuti V. Radiomics Model to Predict Early Progression of Nonmetastatic Nasopharyngeal Carcinoma after Intensity Modulation Radiation Therapy: A Multicenter Study. Radiol Artif Intell 2019; 1:e180075. [PMID: 33937796 DOI: 10.1148/ryai.2019180075] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 04/04/2019] [Accepted: 05/07/2019] [Indexed: 12/23/2022]
Abstract
Purpose To examine the prognostic value of a machine learning model trained with pretreatment MRI radiomic features in the assessment of patients with nonmetastatic nasopharyngeal carcinoma (NPC) who are at risk for 3-year disease progression after intensity-modulated radiation therapy and to explain the radiomics features in the model. Materials and Methods A total of 277 patients with nonmetastatic NPC admitted between March 2008 and December 2014 at two imaging centers were retrospectively reviewed. Patients were allocated to a discovery or validation cohort based on where they underwent MRI (discovery cohort, n = 217; validation cohort, n = 60). A total of 525 radiomics features extracted from contrast material-enhanced T1- or T2-weighted MRI studies and five clinical features were subjected to radiomic machine learning modeling to predict 3-year disease progression. Feature selection was performed by analyzing robustness to resampling, reproducibility between observers, and redundancy. Features for the final model were selected with Kaplan-Meier analysis and the log-rank test. A support vector machine was used as the classifier for the model. To interpret the pattern learned from the model, Shapley additive explanations (SHAP) was applied. Results The final model yielded an area under the receiver operating characteristic curve of 0.80 in both the discovery (95% bootstrap confidence interval: 0.80, 0.81) and independent validation (95% bootstrap confidence interval: 0.73, 0.89) cohorts. Analysis with SHAP revealed that tumor shape sphericity, first-order mean absolute deviation, T stage, and overall stage were important factors in 3-year disease progression. Conclusion These results add to the growing evidence of the role of radiomics in the assessment of NPC. By using explanatory techniques, such as SHAP, the complex interaction of features learned by the model may be understood.© RSNA, 2019Supplemental material is available for this article.
Collapse
Affiliation(s)
- Richard Du
- Departments of Diagnostic Radiology (R.D., H.Y., P.L.K., V.V.) and Clinical Oncology (V.H.L., K.O.L., A.W.L., D.L.K.) and the School of Public Health (H.H.P.), Li Ka Shing Faculty of Medicine, The University of Hong Kong, Room 406, Block K, Queen Mary Hospital, Pok Fu Lam Road, Hong Kong SAR; Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China (Y.C.); and Department of Electrical and Electronic Engineering, Faculty of Engineering, The University of Hong Kong, Hong Kong SAR (E.Y.L.)
| | - Victor H Lee
- Departments of Diagnostic Radiology (R.D., H.Y., P.L.K., V.V.) and Clinical Oncology (V.H.L., K.O.L., A.W.L., D.L.K.) and the School of Public Health (H.H.P.), Li Ka Shing Faculty of Medicine, The University of Hong Kong, Room 406, Block K, Queen Mary Hospital, Pok Fu Lam Road, Hong Kong SAR; Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China (Y.C.); and Department of Electrical and Electronic Engineering, Faculty of Engineering, The University of Hong Kong, Hong Kong SAR (E.Y.L.)
| | - Hui Yuan
- Departments of Diagnostic Radiology (R.D., H.Y., P.L.K., V.V.) and Clinical Oncology (V.H.L., K.O.L., A.W.L., D.L.K.) and the School of Public Health (H.H.P.), Li Ka Shing Faculty of Medicine, The University of Hong Kong, Room 406, Block K, Queen Mary Hospital, Pok Fu Lam Road, Hong Kong SAR; Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China (Y.C.); and Department of Electrical and Electronic Engineering, Faculty of Engineering, The University of Hong Kong, Hong Kong SAR (E.Y.L.)
| | - Ka-On Lam
- Departments of Diagnostic Radiology (R.D., H.Y., P.L.K., V.V.) and Clinical Oncology (V.H.L., K.O.L., A.W.L., D.L.K.) and the School of Public Health (H.H.P.), Li Ka Shing Faculty of Medicine, The University of Hong Kong, Room 406, Block K, Queen Mary Hospital, Pok Fu Lam Road, Hong Kong SAR; Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China (Y.C.); and Department of Electrical and Electronic Engineering, Faculty of Engineering, The University of Hong Kong, Hong Kong SAR (E.Y.L.)
| | - Herbert H Pang
- Departments of Diagnostic Radiology (R.D., H.Y., P.L.K., V.V.) and Clinical Oncology (V.H.L., K.O.L., A.W.L., D.L.K.) and the School of Public Health (H.H.P.), Li Ka Shing Faculty of Medicine, The University of Hong Kong, Room 406, Block K, Queen Mary Hospital, Pok Fu Lam Road, Hong Kong SAR; Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China (Y.C.); and Department of Electrical and Electronic Engineering, Faculty of Engineering, The University of Hong Kong, Hong Kong SAR (E.Y.L.)
| | - Yu Chen
- Departments of Diagnostic Radiology (R.D., H.Y., P.L.K., V.V.) and Clinical Oncology (V.H.L., K.O.L., A.W.L., D.L.K.) and the School of Public Health (H.H.P.), Li Ka Shing Faculty of Medicine, The University of Hong Kong, Room 406, Block K, Queen Mary Hospital, Pok Fu Lam Road, Hong Kong SAR; Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China (Y.C.); and Department of Electrical and Electronic Engineering, Faculty of Engineering, The University of Hong Kong, Hong Kong SAR (E.Y.L.)
| | - Edmund Y Lam
- Departments of Diagnostic Radiology (R.D., H.Y., P.L.K., V.V.) and Clinical Oncology (V.H.L., K.O.L., A.W.L., D.L.K.) and the School of Public Health (H.H.P.), Li Ka Shing Faculty of Medicine, The University of Hong Kong, Room 406, Block K, Queen Mary Hospital, Pok Fu Lam Road, Hong Kong SAR; Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China (Y.C.); and Department of Electrical and Electronic Engineering, Faculty of Engineering, The University of Hong Kong, Hong Kong SAR (E.Y.L.)
| | - Pek-Lan Khong
- Departments of Diagnostic Radiology (R.D., H.Y., P.L.K., V.V.) and Clinical Oncology (V.H.L., K.O.L., A.W.L., D.L.K.) and the School of Public Health (H.H.P.), Li Ka Shing Faculty of Medicine, The University of Hong Kong, Room 406, Block K, Queen Mary Hospital, Pok Fu Lam Road, Hong Kong SAR; Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China (Y.C.); and Department of Electrical and Electronic Engineering, Faculty of Engineering, The University of Hong Kong, Hong Kong SAR (E.Y.L.)
| | - Anne W Lee
- Departments of Diagnostic Radiology (R.D., H.Y., P.L.K., V.V.) and Clinical Oncology (V.H.L., K.O.L., A.W.L., D.L.K.) and the School of Public Health (H.H.P.), Li Ka Shing Faculty of Medicine, The University of Hong Kong, Room 406, Block K, Queen Mary Hospital, Pok Fu Lam Road, Hong Kong SAR; Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China (Y.C.); and Department of Electrical and Electronic Engineering, Faculty of Engineering, The University of Hong Kong, Hong Kong SAR (E.Y.L.)
| | - Dora L Kwong
- Departments of Diagnostic Radiology (R.D., H.Y., P.L.K., V.V.) and Clinical Oncology (V.H.L., K.O.L., A.W.L., D.L.K.) and the School of Public Health (H.H.P.), Li Ka Shing Faculty of Medicine, The University of Hong Kong, Room 406, Block K, Queen Mary Hospital, Pok Fu Lam Road, Hong Kong SAR; Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China (Y.C.); and Department of Electrical and Electronic Engineering, Faculty of Engineering, The University of Hong Kong, Hong Kong SAR (E.Y.L.)
| | - Varut Vardhanabhuti
- Departments of Diagnostic Radiology (R.D., H.Y., P.L.K., V.V.) and Clinical Oncology (V.H.L., K.O.L., A.W.L., D.L.K.) and the School of Public Health (H.H.P.), Li Ka Shing Faculty of Medicine, The University of Hong Kong, Room 406, Block K, Queen Mary Hospital, Pok Fu Lam Road, Hong Kong SAR; Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China (Y.C.); and Department of Electrical and Electronic Engineering, Faculty of Engineering, The University of Hong Kong, Hong Kong SAR (E.Y.L.)
| |
Collapse
|
44
|
Gabelloni M, Faggioni L, Neri E. Imaging biomarkers in upper gastrointestinal cancers. BJR Open 2019; 1:20190001. [PMID: 33178936 PMCID: PMC7592483 DOI: 10.1259/bjro.20190001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 02/23/2019] [Accepted: 03/29/2019] [Indexed: 12/02/2022] Open
Abstract
In parallel with the increasingly widespread availability of high performance imaging platforms and recent progresses in pathobiological characterisation and treatment of gastrointestinal malignancies, imaging biomarkers have become a major research topic due to their potential to provide additional quantitative information to conventional imaging modalities that can improve accuracy at staging and follow-up, predict outcome, and guide treatment planning in an individualised manner. The aim of this review is to briefly examine the status of current knowledge about imaging biomarkers in the field of upper gastrointestinal cancers, highlighting their potential applications and future perspectives in patient management from diagnosis onwards.
Collapse
Affiliation(s)
- Michela Gabelloni
- Department of Translational Research, Diagnostic and Interventional Radiology, University of Pisa, Pisa, Italy
| | - Lorenzo Faggioni
- Department of Translational Research, Diagnostic and Interventional Radiology, University of Pisa, Pisa, Italy
| | - Emanuele Neri
- Department of Translational Research, Diagnostic and Interventional Radiology, University of Pisa, Pisa, Italy
| |
Collapse
|
45
|
Zhang P, Zhou H, Lu K, Wang Y, Feng T. Circulating tumor cells in the clinical cancer diagnosis. Clin Transl Oncol 2019; 22:279-282. [DOI: 10.1007/s12094-019-02139-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Accepted: 05/21/2019] [Indexed: 12/24/2022]
|
46
|
European Society of Radiology (ESR). What the radiologist should know about artificial intelligence - an ESR white paper. Insights Imaging 2019; 10:44. [PMID: 30949865 PMCID: PMC6449411 DOI: 10.1186/s13244-019-0738-2] [Citation(s) in RCA: 177] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 03/20/2019] [Indexed: 02/08/2023] Open
Abstract
This paper aims to provide a review of the basis for application of AI in radiology, to discuss the immediate ethical and professional impact in radiology, and to consider possible future evolution.Even if AI does add significant value to image interpretation, there are implications outside the traditional radiology activities of lesion detection and characterisation. In radiomics, AI can foster the analysis of the features and help in the correlation with other omics data. Imaging biobanks would become a necessary infrastructure to organise and share the image data from which AI models can be trained. AI can be used as an optimising tool to assist the technologist and radiologist in choosing a personalised patient's protocol, tracking the patient's dose parameters, providing an estimate of the radiation risks. AI can also aid the reporting workflow and help the linking between words, images, and quantitative data. Finally, AI coupled with CDS can improve the decision process and thereby optimise clinical and radiological workflow.
Collapse
|
47
|
Lucignani G, Neri E. Integration of imaging biomarkers into systems biomedicine: a renaissance for medical imaging. Clin Transl Imaging 2019. [DOI: 10.1007/s40336-019-00320-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
48
|
Artificial intelligence: a challenge for third millennium radiologist. Radiol Med 2019; 124:241-242. [PMID: 30707375 DOI: 10.1007/s11547-019-00990-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 01/14/2019] [Indexed: 12/24/2022]
|