1
|
Liu Y, Wang Z, Yang L, Zhang M, Li M, Zhang J, Tang L, Jiang Z, Li X, Deng J, Meng Q, Liu S, Wang K, Qi L. Identification of a rank-based radiomic signature with individualized prognostic value for lung adenocarcinoma in a multi-cohort study. Eur J Radiol 2024; 181:111782. [PMID: 39427495 DOI: 10.1016/j.ejrad.2024.111782] [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: 07/02/2024] [Revised: 09/29/2024] [Accepted: 10/08/2024] [Indexed: 10/22/2024]
Abstract
OBJECTIVES Radiomics provides an opportunity to evaluate cancer prognosis noninvasively. However, the susceptibility of the radiomic quantitative features to multicenter effects, leads to the clinical dilemma of the radiomic signatures. This study aimed to develop a radiomic signature to circumvent multicenter effects, achieving the individualized prognostic assessment of lung adenocarcinoma (LUAD). METHODS Using computed tomography (CT) imaging of 234 stage I-IIIA LUAD patients derived from three public multicenter cohorts, we proposed a rank-based method that utilized the relative rank patterns of quantitative values between radiomic feature pairs within individual patients and established a feature pair signature for LUAD prognosis. We collected a new clinical cohort with 162 LUAD patients for independent validation. RESULTS A rank-based radiomic signature, consisting of 12 feature pairs, was developed, and it could determine the mortality risk for an individual according to the rank patterns of 12 feature pairs within the patient's CT imaging. The prognostic performance of the rank-based signature was effectively validated in the new clinical cohort (log-rank P = 0.0051, C-index = 0.73), whereas other signatures lost their prognostic ability across centers. The novel proposed radiomic nomogram significantly improved the prognostic performance of clinicopathological factors. The further radiogenomic analyses revealed the underlying biological characteristics (e.g., Stemness, Ferroptosis, 'ECM') reflected by the rank-based radiomic signature. CONCLUSIONS This multicenter study illustrates the accuracy and stability of the rank-based radiomic signature for LUAD prognosis, and demonstrates a unique advantage of clinical individualized application. The biological characteristics underlying the rank-based radiomic signature would accelerate its clinical application.
Collapse
Affiliation(s)
- Yixin Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China; Modern Education Technology Center, Harbin Medical University, Harbin, China
| | - Zhihui Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Liping Yang
- PET-CT/MR Department, Harbin Medical University Cancer Hospital, Harbin, China
| | - Meng Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Mengyue Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Juxuan Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Lefan Tang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Zhiyun Jiang
- PET-CT/MR Department, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xin Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Jiaxing Deng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Qingwei Meng
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150040, China
| | - Shilong Liu
- Department of Thoracic Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin 150086, China.
| | - Kezheng Wang
- PET-CT/MR Department, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Lishuang Qi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China.
| |
Collapse
|
2
|
Miceli V, Gennarini M, Tomao F, Cupertino A, Lombardo D, Palaia I, Curti F, Riccardi S, Ninkova R, Maccioni F, Ricci P, Catalano C, Rizzo SMR, Manganaro L. Imaging of Peritoneal Carcinomatosis in Advanced Ovarian Cancer: CT, MRI, Radiomic Features and Resectability Criteria. Cancers (Basel) 2023; 15:5827. [PMID: 38136373 PMCID: PMC10741537 DOI: 10.3390/cancers15245827] [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: 10/27/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
PC represents the most striking picture of the loco-regional spread of ovarian cancer, configuring stage III. In the last few years, many papers have evaluated the role of imaging and therapeutic management in patients with ovarian cancer and PC. This paper summed up the literature on traditional approaches to the imaging of peritoneal carcinomatosis in advanced ovarian cancer, presenting classification systems, most frequent patterns, routes of spread and sites that are difficult to identify. The role of imaging in diagnosis was investigated, with particular attention to the reported sensitivity and specificity data-computed tomography (CT), magnetic resonance imaging (MRI), positron emission tomography-CT (PET-CT)-and to the peritoneal cancer index (PCI). In addition, we explored the therapeutic possibilities and radiomics applications that can impact management of patients with ovarian cancer. Careful staging is mandatory, and patient selection is one of the most important factors influencing complete cytoreduction (CCR) outcome: an accurate pre-operative imaging may allow selection of patients that may benefit most from primary cytoreductive surgery.
Collapse
Affiliation(s)
- Valentina Miceli
- Department of Radiological, Oncology and Patological Sciences, “Sapienza” University of Rome, 00185 Rome, Italy; (V.M.); (M.G.); (A.C.); (D.L.); (F.C.); (S.R.); (R.N.); (F.M.); (P.R.); (C.C.)
| | - Marco Gennarini
- Department of Radiological, Oncology and Patological Sciences, “Sapienza” University of Rome, 00185 Rome, Italy; (V.M.); (M.G.); (A.C.); (D.L.); (F.C.); (S.R.); (R.N.); (F.M.); (P.R.); (C.C.)
| | - Federica Tomao
- Department of Gynecological, Obstetrical and Urological Sciences, “Sapienza” University of Rome, 00185 Rome, Italy; (F.T.); (I.P.)
| | - Angelica Cupertino
- Department of Radiological, Oncology and Patological Sciences, “Sapienza” University of Rome, 00185 Rome, Italy; (V.M.); (M.G.); (A.C.); (D.L.); (F.C.); (S.R.); (R.N.); (F.M.); (P.R.); (C.C.)
| | - Dario Lombardo
- Department of Radiological, Oncology and Patological Sciences, “Sapienza” University of Rome, 00185 Rome, Italy; (V.M.); (M.G.); (A.C.); (D.L.); (F.C.); (S.R.); (R.N.); (F.M.); (P.R.); (C.C.)
| | - Innocenza Palaia
- Department of Gynecological, Obstetrical and Urological Sciences, “Sapienza” University of Rome, 00185 Rome, Italy; (F.T.); (I.P.)
| | - Federica Curti
- Department of Radiological, Oncology and Patological Sciences, “Sapienza” University of Rome, 00185 Rome, Italy; (V.M.); (M.G.); (A.C.); (D.L.); (F.C.); (S.R.); (R.N.); (F.M.); (P.R.); (C.C.)
| | - Sandrine Riccardi
- Department of Radiological, Oncology and Patological Sciences, “Sapienza” University of Rome, 00185 Rome, Italy; (V.M.); (M.G.); (A.C.); (D.L.); (F.C.); (S.R.); (R.N.); (F.M.); (P.R.); (C.C.)
| | - Roberta Ninkova
- Department of Radiological, Oncology and Patological Sciences, “Sapienza” University of Rome, 00185 Rome, Italy; (V.M.); (M.G.); (A.C.); (D.L.); (F.C.); (S.R.); (R.N.); (F.M.); (P.R.); (C.C.)
| | - Francesca Maccioni
- Department of Radiological, Oncology and Patological Sciences, “Sapienza” University of Rome, 00185 Rome, Italy; (V.M.); (M.G.); (A.C.); (D.L.); (F.C.); (S.R.); (R.N.); (F.M.); (P.R.); (C.C.)
| | - Paolo Ricci
- Department of Radiological, Oncology and Patological Sciences, “Sapienza” University of Rome, 00185 Rome, Italy; (V.M.); (M.G.); (A.C.); (D.L.); (F.C.); (S.R.); (R.N.); (F.M.); (P.R.); (C.C.)
| | - Carlo Catalano
- Department of Radiological, Oncology and Patological Sciences, “Sapienza” University of Rome, 00185 Rome, Italy; (V.M.); (M.G.); (A.C.); (D.L.); (F.C.); (S.R.); (R.N.); (F.M.); (P.R.); (C.C.)
| | - Stefania Maria Rita Rizzo
- Clinica di Radiologia EOC, Istituto Imaging della Svizzera Italiana (IIMSI), 6900 Lugano, Switzerland;
- Facoltà di Scienze Biomediche, Università della Svizzera Italiana (USI), 6900 Lugano, Switzerland
| | - Lucia Manganaro
- Department of Radiological, Oncology and Patological Sciences, “Sapienza” University of Rome, 00185 Rome, Italy; (V.M.); (M.G.); (A.C.); (D.L.); (F.C.); (S.R.); (R.N.); (F.M.); (P.R.); (C.C.)
| |
Collapse
|
3
|
Mumtaz H, Saqib M, Jabeen S, Muneeb M, Mughal W, Sohail H, Safdar M, Mehmood Q, Khan MA, Ismail SM. Exploring alternative approaches to precision medicine through genomics and artificial intelligence - a systematic review. Front Med (Lausanne) 2023; 10:1227168. [PMID: 37849490 PMCID: PMC10577305 DOI: 10.3389/fmed.2023.1227168] [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: 05/22/2023] [Accepted: 09/20/2023] [Indexed: 10/19/2023] Open
Abstract
The core idea behind precision medicine is to pinpoint the subpopulations that differ from one another in terms of disease risk, drug responsiveness, and treatment outcomes due to differences in biology and other traits. Biomarkers are found through genomic sequencing. Multi-dimensional clinical and biological data are created using these biomarkers. Better analytic methods are needed for these multidimensional data, which can be accomplished by using artificial intelligence (AI). An updated review of 80 latest original publications is presented on four main fronts-preventive medicine, medication development, treatment outcomes, and diagnostic medicine-All these studies effectively illustrated the significance of AI in precision medicine. Artificial intelligence (AI) has revolutionized precision medicine by swiftly analyzing vast amounts of data to provide tailored treatments and predictive diagnostics. Through machine learning algorithms and high-resolution imaging, AI assists in precise diagnoses and early disease detection. AI's ability to decode complex biological factors aids in identifying novel therapeutic targets, allowing personalized interventions and optimizing treatment outcomes. Furthermore, AI accelerates drug discovery by navigating chemical structures and predicting drug-target interactions, expediting the development of life-saving medications. With its unrivaled capacity to comprehend and interpret data, AI stands as an invaluable tool in the pursuit of enhanced patient care and improved health outcomes. It's evident that AI can open a new horizon for precision medicine by translating complex data into actionable information. To get better results in this regard and to fully exploit the great potential of AI, further research is required on this pressing subject.
Collapse
Affiliation(s)
| | | | | | - Muhammad Muneeb
- Department of Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Wajiha Mughal
- Department of Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Hassan Sohail
- Department of Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Myra Safdar
- Armed Forces Institute of Cardiology and National Institute of Heart Diseases (AFIC-NIHD), Rawalpindi, Pakistan
| | - Qasim Mehmood
- Department of Medicine, King Edward Medical University, Lahore, Pakistan
| | - Muhammad Ahsan Khan
- Department of Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | | |
Collapse
|
4
|
Raia G, Del Grande M, Colombo I, Nerone M, Manganaro L, Gasparri ML, Papadia A, Del Grande F, Rizzo S. Whole-Body Composition Features by Computed Tomography in Ovarian Cancer: Pilot Data on Survival Correlations. Cancers (Basel) 2023; 15:2602. [PMID: 37174067 PMCID: PMC10177066 DOI: 10.3390/cancers15092602] [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: 04/04/2023] [Revised: 04/28/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
BACKGROUND The primary objective of this study was to assess the associations of computed tomography (CT)-based whole-body composition values with overall survival (OS) and progression-free survival (PFS) in epithelial ovarian cancer (EOC) patients. The secondary objective was the association of body composition with chemotherapy-related toxicity. METHODS Thirty-four patients (median age 64.9 years; interquartile range 55.4-75.4) with EOC and thorax and abdomen CT scans were included. Clinical data recorded: age; weight; height; stage; chemotherapy-related toxicity; and date of last contact, progression and death. Automatic extraction of body composition values was performed by dedicated software. Sarcopenia was defined according to predefined cutoffs. Statistical analysis included univariate tests to investigate associations of sarcopenia and body composition with chemotoxicity. Association of body composition parameters and OS/PFS was evaluated by log-rank test and Cox proportional hazard model. Multivariate models were adjusted for FIGO stage and/or age at diagnosis. RESULTS We found significant associations of skeletal muscle volume with OS (p = 0.04) and PFS (p = 0.04); intramuscular fat volume with PFS (p = 0.03); and visceral adipose tissue, epicardial and paracardial fat with PFS (p = 0.04, 0.01 and 0.02, respectively). We found no significant associations between body composition parameters and chemotherapy-related toxicity. CONCLUSIONS In this exploratory study, we found significant associations of whole-body composition parameters with OS and PFS. These results open a window to the possibility to perform body composition profiling without approximate estimations.
Collapse
Affiliation(s)
- Giorgio Raia
- Istituto di Imaging della Svizzera Italiana (IIMSI), Ente Ospedaliero Cantonale, 6900 Lugano, Switzerland; (G.R.); (F.D.G.)
| | - Maria Del Grande
- Service of Medical Oncology, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland; (M.D.G.); (I.C.); (M.N.)
| | - Ilaria Colombo
- Service of Medical Oncology, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland; (M.D.G.); (I.C.); (M.N.)
| | - Marta Nerone
- Service of Medical Oncology, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale (EOC), 6500 Bellinzona, Switzerland; (M.D.G.); (I.C.); (M.N.)
| | - Lucia Manganaro
- Department of Radiological, Oncological and Pathological Sciences, University of Rome Sapienza (IT), 00185 Roma, Italy;
| | - Maria Luisa Gasparri
- Department of Gynecology and Obstetrics, Ente Ospedaliero Cantonale of Lugano (EOC), 6900 Lugano, Switzerland; (M.L.G.); (A.P.)
| | - Andrea Papadia
- Department of Gynecology and Obstetrics, Ente Ospedaliero Cantonale of Lugano (EOC), 6900 Lugano, Switzerland; (M.L.G.); (A.P.)
- Facoltà di Scienze Biomediche, Università della Svizzera Italiana, 6900 Lugano, Switzerland
| | - Filippo Del Grande
- Istituto di Imaging della Svizzera Italiana (IIMSI), Ente Ospedaliero Cantonale, 6900 Lugano, Switzerland; (G.R.); (F.D.G.)
- Facoltà di Scienze Biomediche, Università della Svizzera Italiana, 6900 Lugano, Switzerland
| | - Stefania Rizzo
- Istituto di Imaging della Svizzera Italiana (IIMSI), Ente Ospedaliero Cantonale, 6900 Lugano, Switzerland; (G.R.); (F.D.G.)
- Facoltà di Scienze Biomediche, Università della Svizzera Italiana, 6900 Lugano, Switzerland
| |
Collapse
|
5
|
Petrella F, Rizzo S, Attili I, Passaro A, Zilli T, Martucci F, Bonomo L, Del Grande F, Casiraghi M, De Marinis F, Spaggiari L. Stage III Non-Small-Cell Lung Cancer: An Overview of Treatment Options. Curr Oncol 2023; 30:3160-3175. [PMID: 36975452 PMCID: PMC10047909 DOI: 10.3390/curroncol30030239] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/27/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
Lung cancer is the second-most commonly diagnosed cancer and the leading cause of cancer death worldwide. The most common histological type is non-small-cell lung cancer, accounting for 85% of all lung cancer cases. About one out of three new cases of non-small-cell lung cancer are diagnosed at a locally advanced stage—mainly stage III—consisting of a widely heterogeneous group of patients presenting significant differences in terms of tumor volume, local diffusion, and lymph nodal involvement. Stage III NSCLC therapy is based on the pivotal role of multimodal treatment, including surgery, radiotherapy, and a wide-ranging option of systemic treatments. Radical surgery is indicated in the case of hilar lymphnodal involvement or single station mediastinal ipsilateral involvement, possibly after neoadjuvant chemotherapy; the best appropriate treatment for multistation mediastinal lymph node involvement still represents a matter of debate. Although the main scope of treatments in this setting is potentially curative, the overall survival rates are still poor, ranging from 36% to 26% and 13% in stages IIIA, IIIB, and IIIC, respectively. The aim of this article is to provide an up-to-date, comprehensive overview of the state-of-the-art treatments for stage III non-small-cell lung cancer.
Collapse
Affiliation(s)
- Francesco Petrella
- Department of Thoracic Surgery, European Institute of Oncology IRCCS, 20141 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy
- Correspondence: ; Tel.: +0039-0257489362
| | - Stefania Rizzo
- Service of Radiology, Imaging Institute of Southern Switzerland (IIMSI), EOC, Via Tesserete 46, 6900 Lugano, Switzerland
- Faculty of Biomedical Sciences, University of Italian Switzerland, Via Buffi 13, 6900 Lugano, Switzerland
| | - Ilaria Attili
- Division of Thoracic Oncology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Antonio Passaro
- Division of Thoracic Oncology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Thomas Zilli
- Faculty of Biomedical Sciences, University of Italian Switzerland, Via Buffi 13, 6900 Lugano, Switzerland
- Radiation Oncology, Oncological Institute of Southern Switzerland, EOC, 6500 Bellinzona, Switzerland
- Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Francesco Martucci
- Radiation Oncology, Oncological Institute of Southern Switzerland, EOC, 6500 Bellinzona, Switzerland
| | - Luca Bonomo
- Service of Radiology, Imaging Institute of Southern Switzerland (IIMSI), EOC, Via Tesserete 46, 6900 Lugano, Switzerland
| | - Filippo Del Grande
- Service of Radiology, Imaging Institute of Southern Switzerland (IIMSI), EOC, Via Tesserete 46, 6900 Lugano, Switzerland
- Faculty of Biomedical Sciences, University of Italian Switzerland, Via Buffi 13, 6900 Lugano, Switzerland
| | - Monica Casiraghi
- Department of Thoracic Surgery, European Institute of Oncology IRCCS, 20141 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy
| | - Filippo De Marinis
- Division of Thoracic Oncology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Lorenzo Spaggiari
- Department of Thoracic Surgery, European Institute of Oncology IRCCS, 20141 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy
| |
Collapse
|
6
|
Lin X, Wang H, Chen J, Lu T, Zheng D, Chen Y, Chen Y. The value of CT-based radiomics in early assessment of chemotherapeutic effect in patients with advanced lung adenocarcinoma: a preliminary study. Acta Radiol 2023; 64:524-532. [PMID: 35137628 DOI: 10.1177/02841851221078290] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Computed tomography (CT) is the preferred method for evaluating the therapeutic effect of lung cancer. Radiomics parameters can provide a lot of supplementary information for clinical diagnosis and treatment. PURPOSE To investigate the value of radiomics features of CT imaging to predict and evaluate the early efficacy of chemotherapy in patients with advanced lung adenocarcinoma. MATERIAL AND METHODS A total of 101 patients with advanced lung adenocarcinoma were enrolled. Patients were classified into a response group and non-response group according to RECIST 1.1 standard. All patients underwent chest CT examination before and after two cycles of chemotherapy. A total of 293 radiomics features were calculated. The features between response group and non-response group were compared before and after chemotherapy. The diagnostic efficacy was evaluated using the receiver operating characteristic curve. RESULTS The six pre-chemotherapy radiomics features were selected, with area under the curve (AUC), sensitivity, and specificity at 0.720, 68.3%, and 69.0% in the training group and 0.573, 50.0%, and 76.9% in the test group, respectively. The eleven post-chemotherapy radiomics features were selected, with AUC, sensitivity, specificity at 0.789, 75.6%, and 75.9% in the training group and 0.718, 61.1%, and 76.9% in the test group, respectively. The prognostic value of △f8, △f16, %f8, and %f16 were higher than the other features with AUCs of 0.787, 0.837, 0.763, and 0.877, respectively. CONCLUSION Radiomics is expected to provide more valuable information for evaluating the chemotherapy efficacy of lung adenocarcinoma.
Collapse
Affiliation(s)
- Xi Lin
- Department of Radiology, 66552Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fujian Provincial Clinical Research Center for Cancer Radiotherapy and Immunotherapy, Fujian Province, PR China
| | - Huaming Wang
- Department of Ultrasound, 117889The Second Affiliated Hospital of Fujian Medical University, Fujian Province, PR China
| | - Jiayou Chen
- Department of Radiology, 66552Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fujian Provincial Clinical Research Center for Cancer Radiotherapy and Immunotherapy, Fujian Province, PR China
| | - Tao Lu
- Department of Radiology, 66552Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fujian Provincial Clinical Research Center for Cancer Radiotherapy and Immunotherapy, Fujian Province, PR China
| | - Dechun Zheng
- Department of Radiology, 66552Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fujian Provincial Clinical Research Center for Cancer Radiotherapy and Immunotherapy, Fujian Province, PR China
| | - Ying Chen
- Department of Radiology, 66552Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fujian Provincial Clinical Research Center for Cancer Radiotherapy and Immunotherapy, Fujian Province, PR China
| | - Yunbin Chen
- Department of Radiology, 66552Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fujian Provincial Clinical Research Center for Cancer Radiotherapy and Immunotherapy, Fujian Province, PR China
| |
Collapse
|
7
|
Can quantitative peritumoral CT radiomics features predict the prognosis of patients with non-small cell lung cancer? A systematic review. Eur Radiol 2023; 33:2105-2117. [PMID: 36307554 PMCID: PMC9935659 DOI: 10.1007/s00330-022-09174-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/20/2022] [Accepted: 09/16/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVES To provide an overarching evaluation of the value of peritumoral CT radiomics features for predicting the prognosis of non-small cell lung cancer and to assess the quality of the available studies. METHODS The PubMed, Embase, Web of Science, and Cochrane Library databases were searched for studies predicting the prognosis in patients with non-small cell lung cancer (NSCLC) using CT-based peritumoral radiomics features. Information about the patient, CT-scanner, and radiomics analyses were all extracted for the included studies. Study quality was assessed using the Radiomics Quality Score (RQS) and the Prediction Model Risk of Bias Assessment Tool (PROBAST). RESULTS Thirteen studies were included with 2942 patients from 2017 to 2022. Only one study was prospective, and the others were all retrospectively designed. Manual segmentation and multicenter studies were performed by 69% and 46% of the included studies, respectively. 3D-Slicer and MATLAB software were most commonly used for the segmentation of lesions and extraction of features. The peritumoral region was most frequently defined as dilated from the tumor boundary of 15 mm, 20 mm, or 30 mm. The median RQS of the studies was 13 (range 4-19), while all of included studies were assessed as having a high risk of bias (ROB) overall. CONCLUSIONS Peritumoral radiomics features based on CT images showed promise in predicting the prognosis of NSCLC, although well-designed studies and further biological validation are still needed. KEY POINTS • Peritumoral radiomics features based on CT images are promising and encouraging for predicting the prognosis of non-small cell lung cancer. • The peritumoral region was often dilated from the tumor boundary of 15 mm or 20 mm because these were considered safe margins. • The median Radiomics Quality Score of the included studies was 13 (range 4-19), and all of studies were considered to have a high risk of bias overall.
Collapse
|
8
|
Ferrante M, Rinaldi L, Botta F, Hu X, Dolp A, Minotti M, De Piano F, Funicelli G, Volpe S, Bellerba F, De Marco P, Raimondi S, Rizzo S, Shi K, Cremonesi M, Jereczek-Fossa BA, Spaggiari L, De Marinis F, Orecchia R, Origgi D. Application of nnU-Net for Automatic Segmentation of Lung Lesions on CT Images and Its Implication for Radiomic Models. J Clin Med 2022; 11:7334. [PMID: 36555950 PMCID: PMC9784875 DOI: 10.3390/jcm11247334] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/05/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
Radiomics investigates the predictive role of quantitative parameters calculated from radiological images. In oncology, tumour segmentation constitutes a crucial step of the radiomic workflow. Manual segmentation is time-consuming and prone to inter-observer variability. In this study, a state-of-the-art deep-learning network for automatic segmentation (nnU-Net) was applied to computed tomography images of lung tumour patients, and its impact on the performance of survival radiomic models was assessed. In total, 899 patients were included, from two proprietary and one public datasets. Different network architectures (2D, 3D) were trained and tested on different combinations of the datasets. Automatic segmentations were compared to reference manual segmentations performed by physicians using the DICE similarity coefficient. Subsequently, the accuracy of radiomic models for survival classification based on either manual or automatic segmentations were compared, considering both hand-crafted and deep-learning features. The best agreement between automatic and manual contours (DICE = 0.78 ± 0.12) was achieved averaging 2D and 3D predictions and applying customised post-processing. The accuracy of the survival classifier (ranging between 0.65 and 0.78) was not statistically different when using manual versus automatic contours, both with hand-crafted and deep features. These results support the promising role nnU-Net can play in automatic segmentation, accelerating the radiomic workflow without impairing the models' accuracy. Further investigations on different clinical endpoints and populations are encouraged to confirm and generalise these findings.
Collapse
Affiliation(s)
- Matteo Ferrante
- Medical Physics Unit, IEO European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
| | - Lisa Rinaldi
- Radiation Research Unit, IEO European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
| | - Francesca Botta
- Medical Physics Unit, IEO European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
| | - Xiaobin Hu
- Department of Informatics, Technical University of Munich, Arcisstraße 21, 80333 Munich, Germany
| | - Andreas Dolp
- Department of Informatics, Technical University of Munich, Arcisstraße 21, 80333 Munich, Germany
| | - Marta Minotti
- Division of Radiology, IEO, European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
| | - Francesca De Piano
- Division of Radiology, IEO, European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
| | - Gianluigi Funicelli
- Division of Radiology, IEO, European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
| | - Stefania Volpe
- Division of Radiation Oncology, IEO European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, via Festa del Perdono 7, 20122 Milan, Italy
| | - Federica Bellerba
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
| | - Paolo De Marco
- Medical Physics Unit, IEO European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
| | - Sara Raimondi
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
| | - Stefania Rizzo
- Clinica di Radiologia EOC, Istituto Imaging della Svizzera Italiana (IIMSI), via Tesserete 46, 6900 Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), via G. Buffi 13, 6900 Lugano, Switzerland
| | - Kuangyu Shi
- Chair for Computer-Aided Medical Procedures, Department of Informatics, Technical University of Munich, Arcisstraße 21, 80333 Munich, Germany
- Department of Nuclear Medicine, Bern University Hospital, University of Bern, Freiburgstrasse 18, 3010 Bern, Switzerland
| | - Marta Cremonesi
- Radiation Research Unit, IEO European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
| | - Barbara A. Jereczek-Fossa
- Division of Radiation Oncology, IEO European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, via Festa del Perdono 7, 20122 Milan, Italy
| | - Lorenzo Spaggiari
- Department of Oncology and Hemato-Oncology, University of Milan, via Festa del Perdono 7, 20122 Milan, Italy
- Division of Thoracic Surgery, IEO, European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
| | - Filippo De Marinis
- Division of Thoracic Oncology, IEO, European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
| | - Roberto Orecchia
- Division of Radiology, IEO, European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
- Scientific Direction, IEO, European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
| | - Daniela Origgi
- Medical Physics Unit, IEO European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy
| |
Collapse
|
9
|
Petrella F, Manganaro L, Rizzo S. Editorial: State of the art body composition profiling: Advances in imaging modalities and patient outcomes. Front Oncol 2022; 12:1096671. [PMID: 36544701 PMCID: PMC9761766 DOI: 10.3389/fonc.2022.1096671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 11/22/2022] [Indexed: 12/09/2022] Open
Affiliation(s)
- Francesco Petrella
- Department of Thoracic Surgery, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) European Institute of Oncology, Milan, Italy,Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy,*Correspondence: Francesco Petrella, ;;
| | - Lucia Manganaro
- Department of Radiological, Oncological and Pathological Sciences, University of Rome Sapienza, Rome, Italy
| | - Stefania Rizzo
- Istituto di Imaging della Svizzera Italiana (IIMSI), Ente Ospedaliero Cantonale (EOC), Lugano, Switzerland,Facoltà di Scienze Biomediche, Università della Svizzera italiana (USI), Lugano, Switzerland
| |
Collapse
|
10
|
Chen Z, Yi L, Peng Z, Zhou J, Zhang Z, Tao Y, Lin Z, He A, Jin M, Zuo M. Development and validation of a radiomic nomogram based on pretherapy dual-energy CT for distinguishing adenocarcinoma from squamous cell carcinoma of the lung. Front Oncol 2022; 12:949111. [PMID: 36505773 PMCID: PMC9727167 DOI: 10.3389/fonc.2022.949111] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 10/26/2022] [Indexed: 11/24/2022] Open
Abstract
Objective Based on pretherapy dual-energy computed tomography (DECT) images, we developed and validated a nomogram combined with clinical parameters and radiomic features to predict the pathologic subtypes of non-small cell lung cancer (NSCLC) - adenocarcinoma (ADC) and squamous cell carcinoma (SCC). Methods A total of 129 pathologically confirmed NSCLC patients treated at the Second Affiliated Hospital of Nanchang University from October 2017 to October 2021 were retrospectively analyzed. Patients were randomly divided in a ratio of 7:3 (n=90) into training and validation cohorts (n=39). Patients' pretherapy clinical parameters were recorded. Radiomics features of the primary lesion were extracted from two sets of monoenergetic images (40 keV and 100 keV) in arterial phases (AP) and venous phases (VP). Features were selected successively through the intra-class correlation coefficient (ICC) and the least absolute shrinkage and selection operator (LASSO). Multivariate logistic regression analysis was then performed to establish predictive models. The prediction performance between models was evaluated and compared using the receiver operating characteristic (ROC) curve, DeLong test, and Akaike information criterion (AIC). A nomogram was developed based on the model with the best predictive performance to evaluate its calibration and clinical utility. Results A total of 87 ADC and 42 SCC patients were enrolled in this study. Among the five constructed models, the integrative model (AUC: Model 4 = 0.92, Model 5 = 0.93) combining clinical parameters and radiomic features had a higher AUC than the individual clinical models or radiomic models (AUC: Model 1 = 0.84, Model 2 = 0.79, Model 3 = 0.84). The combined clinical-venous phase radiomics model had the best predictive performance, goodness of fit, and parsimony; the area under the ROC curve (AUC) of the training and validation cohorts was 0.93 and 0.90, respectively, and the AIC value was 60.16. Then, this model was visualized as a nomogram. The calibration curves demonstrated it's good calibration, and decision curve analysis (DCA) proved its clinical utility. Conclusion The combined clinical-radiomics model based on pretherapy DECT showed good performance in distinguishing ADC and SCC of the lung. The nomogram constructed based on the best-performing combined clinical-venous phase radiomics model provides a relatively accurate, convenient and noninvasive method for predicting the pathological subtypes of ADC and SCC in NSCLC.
Collapse
Affiliation(s)
- Zhiyong Chen
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Li Yi
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhiwei Peng
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianzhong Zhou
- Department of Radiology, The Quzhou City People’s Hospital, Quzhou, Zhejiang, China
| | - Zhaotao Zhang
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yahong Tao
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ze Lin
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Anjing He
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Mengni Jin
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Minjing Zuo
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China,*Correspondence: Minjing Zuo,
| |
Collapse
|
11
|
Hao P, Deng BY, Huang CT, Xu J, Zhou F, Liu ZX, Zhou W, Xu YK. Predicting anaplastic lymphoma kinase rearrangement status in patients with non-small cell lung cancer using a machine learning algorithm that combines clinical features and CT images. Front Oncol 2022; 12:994285. [PMID: 36338735 PMCID: PMC9630325 DOI: 10.3389/fonc.2022.994285] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/26/2022] [Indexed: 12/01/2023] Open
Abstract
PURPOSE To develop an appropriate machine learning model for predicting anaplastic lymphoma kinase (ALK) rearrangement status in non-small cell lung cancer (NSCLC) patients using computed tomography (CT) images and clinical features. METHOD AND MATERIALS This study included 193 patients with NSCLC (154 in the training cohort, 39 in the validation cohort), 68 of whom tested positive for ALK rearrangements and 125 of whom tested negative. From the nonenhanced CT scans, 157 radiomic characteristics were extracted, and 8 clinical features were collected. Five machine learning (ML) models were assessed to find the best classification model for predicting ALK rearrangement status. A radiomic signature was developed using the least absolute shrinkage and selection operator (LASSO) algorithm. The predictive performance of the models based on radiomic features, clinical features, and their combination was assessed by receiver operating characteristic (ROC) curves. RESULTS The support vector machine (SVM) model had the highest AUC of 0.914 for classification. The clinical features model had an AUC=0.805 (95% CI 0.731-0.877) and an AUC=0.735 (95% CI 0.566-0.863) in the training and validation cohorts, respectively. The CT image-based ML model had an AUC=0.953 (95% CI 0.913-1.0) in the training cohort and an AUC=0.890 (95% CI 0.778-0.971) in the validation cohort. For predicting ALK rearrangement status, the ML model based on CT images and clinical features performed better than the model based on only clinical information or CT images, with an AUC of 0.965 (95% CI 0.826-0.882) in the primary cohort and an AUC of 0.914 (95% CI 0.804-0.893) in the validation cohort. CONCLUSION Our findings revealed that ALK rearrangement status could be accurately predicted using an ML-based classification model based on CT images and clinical data.
Collapse
Affiliation(s)
- Peng Hao
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Bo-Yu Deng
- School of Medical Information Engineering, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chan-Tao Huang
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jun Xu
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fang Zhou
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhe-Xing Liu
- School of Biomedical Engineering, Southern Medical Uinversity, Guangzhou, China
| | - Wu Zhou
- School of Medical Information Engineering, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yi-Kai Xu
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
12
|
Dang S, Guo Y, Han D, Ma G, Yu N, Yang Q, Duan X, Duan H, Ren J. MRI-based radiomics analysis in differentiating solid non-small-cell from small-cell lung carcinoma: a pilot study. Clin Radiol 2022; 77:e749-e757. [PMID: 35817610 DOI: 10.1016/j.crad.2022.06.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/29/2022] [Accepted: 06/01/2022] [Indexed: 12/24/2022]
Abstract
AIM To investigate the ability of a T2-weighted (W) magnetic resonance imaging (MRI)-based radiomics signature to differentiate solid non-small-cell lung carcinoma (NSCLC) from small-cell lung carcinoma (SCLC). MATERIALS AND METHODS The present retrospective study enrolled 152 eligible patients (NSCLC = 125, SCLC = 27). All patients underwent MRI using a 3 T scanner and radiomics features were extracted from T2W MRI. The least absolute shrinkage and selection operator (LASSO) logistic regression model was used to identify the optimal radiomics features for the construction of a radiomics model to differentiate solid NSCLC from SCLC. Threefold cross validation repeated 10 times was used for model training and evaluation. The conventional MRI morphology features of the lesions were also evaluated. The performance of the conventional MRI morphological features, and the radiomics signature model and nomogram model (combining radiomics signature with conventional MRI morphological features) was evaluated using receiver operating characteristic (ROC) curve analysis. RESULTS Five optimal features were chosen to build a radiomics signature. There was no significant difference in age, gender, and the largest diameter. The radiomics signature and conventional MRI morphological features (only pleural indentation and lymph node enlargement) were independent predictive factors for differentiating solid NSCLC from SCLC. The area under the ROC curves (AUCs) for MRI morphological features, and the radiomics model, and nomogram model was 0.69, 0.85, and 0.90 (ROC), respectively. CONCLUSIONS The T2W MRI-based radiomics signature is a potential non-invasive approach for distinguishing solid NSCLC from SCLC.
Collapse
Affiliation(s)
- S Dang
- Department of Radiology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang 712000, China
| | - Y Guo
- Department of Radiology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang 712000, China
| | - D Han
- Department of Radiology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang 712000, China
| | - G Ma
- Department of Radiology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang 712000, China
| | - N Yu
- Department of Radiology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang 712000, China; Shaanxi University of Chinese Medicine, Xianyang, China
| | - Q Yang
- Department of Radiology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang 712000, China
| | - X Duan
- Department of Radiology, The First Affiliated Hospital of Xi'an Jiaotong University, China
| | - H Duan
- Department of Radiology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang 712000, China; Shaanxi University of Chinese Medicine, Xianyang, China.
| | - J Ren
- GE Healthcare China, Daxing District, Beijing, China
| |
Collapse
|
13
|
Yang B, Liu C, Wu R, Zhong J, Li A, Ma L, Zhong J, Yin S, Zhou C, Ge Y, Tao X, Zhang L, Lu G. Development and Validation of a DeepSurv Nomogram to Predict Survival Outcomes and Guide Personalized Adjuvant Chemotherapy in Non-Small Cell Lung Cancer. Front Oncol 2022; 12:895014. [PMID: 35814402 PMCID: PMC9260694 DOI: 10.3389/fonc.2022.895014] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 05/02/2022] [Indexed: 11/22/2022] Open
Abstract
Objective To develop and validate a DeepSurv nomogram based on radiomic features extracted from computed tomography images and clinicopathological factors, to predict the overall survival and guide individualized adjuvant chemotherapy in patients with non-small cell lung cancer (NSCLC). Patients and Methods This retrospective study involved 976 consecutive patients with NSCLC (training cohort, n=683; validation cohort, n=293). DeepSurv was constructed based on 1,227 radiomic features, and the risk score was calculated for each patient as the output. A clinical multivariate Cox regression model was built with clinicopathological factors to determine the independent risk factors. Finally, a DeepSurv nomogram was constructed by integrating the risk score and independent clinicopathological factors. The discrimination capability, calibration, and clinical usefulness of the nomogram performance were assessed using concordance index evaluation, the Greenwood-Nam-D’Agostino test, and decision curve analysis, respectively. The treatment strategy was analyzed using a Kaplan–Meier curve and log-rank test for the high- and low-risk groups. Results The DeepSurv nomogram yielded a significantly better concordance index (training cohort, 0.821; validation cohort 0.768) with goodness-of-fit (P<0.05). The risk score, age, thyroid transcription factor-1, Ki-67, and disease stage were the independent risk factors for NSCLC.The Greenwood-Nam-D’Agostino test showed good calibration performance (P=0.39). Both high- and low-risk patients did not benefit from adjuvant chemotherapy, and chemotherapy in low-risk groups may lead to a poorer prognosis. Conclusions The DeepSurv nomogram, which is based on the risk score and independent risk factors, had good predictive performance for survival outcome. Further, it could be used to guide personalized adjuvant chemotherapy in patients with NSCLC.
Collapse
Affiliation(s)
- Bin Yang
- Medical Imaging Center, Calmette Hospital and The First Hospital of Kunming (Affiliated Calmette Hospital of Kunming Medical University), Kunming, China
- Department of Medical Imaging, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Chengxing Liu
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ren Wu
- Department of Medical Imaging, Jinling Hospital, Nanjing Medical University, Nanjing, China
| | - Jing Zhong
- Department of Medical Imaging, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Ang Li
- Department of Medical Imaging, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lu Ma
- Department of Medical Imaging, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jian Zhong
- Department of Medical Imaging, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Saisai Yin
- Department of Medical Imaging, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Changsheng Zhou
- Department of Medical Imaging, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | | | - Xinwei Tao
- Siemens Healthineers Ltd., Shanghai, China
| | - Longjiang Zhang
- Department of Medical Imaging, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
- *Correspondence: Guangming Lu, ; Longjiang Zhang,
| | - Guangming Lu
- Department of Medical Imaging, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
- Department of Medical Imaging, Jinling Hospital, Nanjing Medical University, Nanjing, China
- *Correspondence: Guangming Lu, ; Longjiang Zhang,
| |
Collapse
|
14
|
Popescu ER, Geantă M, Brand A. Mapping of clinical research on artificial intelligence in the treatment of cancer and the challenges and opportunities underpinning its integration in the European Union health sector. Eur J Public Health 2022; 32:443-449. [PMID: 35238918 PMCID: PMC9159319 DOI: 10.1093/eurpub/ckac016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Although current efforts are made to diminish the incidence and burden of disease, cancer is still widely identified late at stage. This study aims to conduct a systematic review mapping the existent and emerging clinical research on artificial intelligence (AI) in the treatment of cancer and to underpin its integration challenges and opportunities in the European Union (EU) health sector. METHODS A systematic literature review (SLR) evaluating global clinical trials (CTs; published between 2010 and 2020 or forthcoming) was concluded. Additionally, a horizon scanning (HS) exercise focusing on emerging trends (published between 2017 and 2020) was conducted. RESULTS Forty-four CTs were identified and analyzed. Selected CTs were divided into three research areas: (i) potential of AI combined with imaging techniques, (ii) AI's applicability in robotic surgery interventions and (iii) AI's potential in clinical decision making. Twenty-one studies presented an interventional nature, nine papers were observational and 14 articles did not explicitly mention the type of study performed. The papers presented an increased heterogeneity in sample size, type of tumour, type of study and reporting of results. In addition, a shift in research is observed and only a small fraction of studies were completed in the EU. These findings could be further linked to the current socio-economic, political, scientific, technological and environmental state of the EU in regard to AI innovation. CONCLUSION To overcome the challenges threatening the EU's integration of such technology in the healthcare field, new strategies taking into account the EU's socio-economic and political environment are deemed necessary.
Collapse
Affiliation(s)
| | - Marius Geantă
- Center for Innovation in Medicine, Bucharest, Romania
- KOL Medical Media, Bucharest, Romania
- United Nations University—Maastricht Economic and Social Research Institute on Innovation and Technology, Maastricht, The Netherlands
| | - Angela Brand
- United Nations University—Maastricht Economic and Social Research Institute on Innovation and Technology, Maastricht, The Netherlands
- Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
- Department of Public Health Genomics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
- Dr. TMA Pai Endowment Chair in Public Health Genomics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
15
|
Tang X, Huang H, Du P, Wang L, Yin H, Xu X. Intratumoral and peritumoral CT-based radiomics strategy reveals distinct subtypes of non-small-cell lung cancer. J Cancer Res Clin Oncol 2022; 148:2247-2260. [DOI: 10.1007/s00432-022-04015-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 04/04/2022] [Indexed: 12/24/2022]
|
16
|
Anagnostopoulos AK, Gaitanis A, Gkiozos I, Athanasiadis EI, Chatziioannou SN, Syrigos KN, Thanos D, Chatziioannou AN, Papanikolaou N. Radiomics/Radiogenomics in Lung Cancer: Basic Principles and Initial Clinical Results. Cancers (Basel) 2022; 14:cancers14071657. [PMID: 35406429 PMCID: PMC8997041 DOI: 10.3390/cancers14071657] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Radiogenomics is a promising new approach in cancer assessment, providing an evaluation of the molecular basis of imaging phenotypes after establishing associations between radiological features and molecular features at the genomic–transcriptomic–proteomic level. This review focuses on describing key aspects of radiogenomics while discussing limitations of translatability to the clinic and possible solutions to these challenges, providing the clinician with an up-to-date handbook on how to use this new tool. Abstract Lung cancer is the leading cause of cancer-related deaths worldwide, and elucidation of its complicated pathobiology has been traditionally targeted by studies incorporating genomic as well other high-throughput approaches. Recently, a collection of methods used for cancer imaging, supplemented by quantitative aspects leading towards imaging biomarker assessment termed “radiomics”, has introduced a novel dimension in cancer research. Integration of genomics and radiomics approaches, where identifying the biological basis of imaging phenotypes is feasible due to the establishment of associations between molecular features at the genomic–transcriptomic–proteomic level and radiological features, has recently emerged termed radiogenomics. This review article aims to briefly describe the main aspects of radiogenomics, while discussing its basic limitations related to lung cancer clinical applications for clinicians, researchers and patients.
Collapse
Affiliation(s)
- Athanasios K. Anagnostopoulos
- Division of Biotechnology, Center of Systems Biology, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11525 Athens, Greece
- Correspondence:
| | - Anastasios Gaitanis
- Clinical and Translational Research, Center of Experimental Surgery, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece;
| | - Ioannis Gkiozos
- Third Department of Internal Medicine, “Sotiria” Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.G.); (K.N.S.)
| | - Emmanouil I. Athanasiadis
- Greek Genome Centre, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece; (E.I.A.); (D.T.)
| | - Sofia N. Chatziioannou
- Nuclear Medicine Division, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece;
| | - Konstantinos N. Syrigos
- Third Department of Internal Medicine, “Sotiria” Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.G.); (K.N.S.)
| | - Dimitris Thanos
- Greek Genome Centre, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece; (E.I.A.); (D.T.)
| | - Achilles N. Chatziioannou
- First Department of Radiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Nikolaos Papanikolaou
- Computational Clinical Imaging Group, Centre for the Unknown, Champalimaud Foundation, 1400-038 Lisbon, Portugal;
- Machine Learning Group, The Royal Marsden, London SM2 5MG, UK
- The Institute of Cancer Research, London SM2 5MG, UK
- Karolinska Institutet, 14186 Stockholm, Sweden
- Institute of Computer Science, FORTH, 70013 Heraklion, Greece
| |
Collapse
|
17
|
Araujo-Filho JAB, Mayoral M, Horvat N, Santini F, Gibbs P, Ginsberg MS. Radiogenomics in personalized management of lung cancer patients: Where are we? Clin Imaging 2022; 84:54-60. [DOI: 10.1016/j.clinimag.2022.01.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 01/03/2022] [Accepted: 01/24/2022] [Indexed: 11/03/2022]
|
18
|
Yang B, Zhou L, Zhong J, Lv T, Li A, Ma L, Zhong J, Yin S, Huang L, Zhou C, Li X, Ge YQ, Tao X, Zhang L, Son Y, Lu G. Combination of computed tomography imaging-based radiomics and clinicopathological characteristics for predicting the clinical benefits of immune checkpoint inhibitors in lung cancer. Respir Res 2021; 22:189. [PMID: 34183009 PMCID: PMC8240400 DOI: 10.1186/s12931-021-01780-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 06/14/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND In this study, we tested whether a combination of radiomic features extracted from baseline pre-immunotherapy computed tomography (CT) images and clinicopathological characteristics could be used as novel noninvasive biomarkers for predicting the clinical benefits of non-small cell lung cancer (NSCLC) patients treated with immune checkpoint inhibitors (ICIs). METHODS The data from 92 consecutive patients with lung cancer who had been treated with ICIs were retrospectively analyzed. In total, 88 radiomic features were selected from the pretreatment CT images for the construction of a random forest model. Radiomics model 1 was constructed based on the Rad-score. Using multivariate logistic regression analysis, the Rad-score and significant predictors were integrated into a single predictive model (radiomics nomogram model 1) to predict the durable clinical benefit (DCB) of ICIs. Radiomics model 2 was developed based on the same Rad-score as radiomics model 1.Using multivariate Cox proportional hazards regression analysis, the Rad-score, and independent risk factors, radiomics nomogram model 2 was constructed to predict the progression-free survival (PFS). RESULTS The models successfully predicted the patients who would benefit from ICIs. For radiomics model 1, the area under the receiver operating characteristic curve values for the training and validation cohorts were 0.848 and 0.795, respectively, whereas for radiomics nomogram model 1, the values were 0.902 and 0.877, respectively. For the PFS prediction, the Harrell's concordance indexes for the training and validation cohorts were 0.717 and 0.760, respectively, using radiomics model 2, whereas they were 0.749 and 0.791, respectively, using radiomics nomogram model 2. CONCLUSIONS CT-based radiomic features and clinicopathological factors can be used prior to the initiation of immunotherapy for identifying NSCLC patients who are the most likely to benefit from the therapy. This could guide the individualized treatment strategy for advanced NSCLC.
Collapse
Affiliation(s)
- Bin Yang
- Department of Medical Imaging, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Li Zhou
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Sch Med, Nanjing, 210002, Jiangsu, China
| | - Jing Zhong
- Department of Medical Imaging, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Tangfeng Lv
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Sch Med, Nanjing, 210002, Jiangsu, China
| | - Ang Li
- Department of Medical Imaging, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Lu Ma
- Department of Medical Imaging, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Jian Zhong
- Department of Medical Imaging, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Saisai Yin
- Department of Medical Imaging, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Litang Huang
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Southeast University, Sch Med, Nanjing, 210002, Nanjing, China
| | - Changsheng Zhou
- Department of Medical Imaging, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Xinyu Li
- Department of Medical Imaging, Affiliated Jinling Hospital, Nanjing Medical University, Nanjing, 210002, China
| | - Ying Qian Ge
- Siemens Healthineers Ltd., Shanghai, 200000, China
| | - Xinwei Tao
- Siemens Healthineers Ltd., Shanghai, 200000, China
| | - Longjiang Zhang
- Department of Medical Imaging, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China.
| | - Yong Son
- Department of Respiratory and Critical Care Medicine, Affiliated Jinling Hospital, Medical School of Nanjing University, Sch Med, Nanjing, 210002, Jiangsu, China.
| | - Guangming Lu
- Department of Medical Imaging, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China.
| |
Collapse
|
19
|
Yu H, Lam KO, Green MD, Wu H, Yang L, Wang W, Jin J, Hu C, Wang Y, Jolly S, (Spring) Kong FM. Significance of radiation esophagitis: Conditional survival assessment in patients with non-small cell lung cancer. JOURNAL OF THE NATIONAL CANCER CENTER 2021; 1:31-38. [PMID: 39035770 PMCID: PMC11256695 DOI: 10.1016/j.jncc.2021.02.003] [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: 01/07/2021] [Revised: 02/07/2021] [Accepted: 02/12/2021] [Indexed: 11/29/2022] Open
Abstract
Purpose This study aimed to examine the effect of radiation esophagitis (RE) and the dynamics of RE on subsequent survival in non-small cell lung cancer (NSCLC) patients who underwent radiotherapy. Experimental Design Patients with NSCLC treated with fractionated thoracic radiotherapy enrolled in prospective trials were eligible. RE was graded prospectively according to Common Terminology Criteria for Adverse Events (CTCAE) v3.0 per protocol requirement weekly during-RT and 1 month after RT. This study applied conditional survival assessment which has advantage over traditional survival analysis as it assesses the survival from the event instead of from the baseline. P-value less than 0.05 was considered to be significant. The primary endpoint is overall survival. Results A total of 177 patients were eligible, with a median follow-up of 5 years. The presence of RE, the maximum RE grade, the evolution of RE and the onset timing of RE events were all correlated with subsequent survival. At all conditional time points, patients first presented with RE grade1 (initial RE1) had significant inferior subsequent survival (multivariable HRs median: 1.63, all P-values<0.05); meanwhile those with RE progressed had significant inferior subsequent survival than those never develop RE (multivariable HRs median: 2.08, all P-values<0.05). Multivariable Cox proportional-hazards analysis showed significantly higher C-indexes for models with inclusion of RE events than those without (all P-values<0.05). Conclusion This study comprehensively evaluated the impact of RE with conditional survival assessment and demonstrated that RE is associated with inferior survival in NSCLC patients treated with RT.
Collapse
Affiliation(s)
- Hao Yu
- Biomedical Engineering, Shenzhen Polytechnic, Shenzhen, China
- BioHealth Informatics, School of Informatics and Computing, IUPUI, Indianapolis, IN, USA
| | - Ka-On Lam
- Department of Clinical Oncology, LKS Faculty of Medicine, the University of Hong Kong, Hong Kong, China
- Clinical Oncology Center, the University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Michael D. Green
- Radiation Oncology, Ann Arbor VA Health Care, Ann Arbor, MI, USA
- Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Huanmei Wu
- BioHealth Informatics, School of Informatics and Computing, IUPUI, Indianapolis, IN, USA
| | - Li Yang
- Clinical Oncology Center, the University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Weili Wang
- University Hospitals/Cleveland Medical Center, Seidman Cancer Center and Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Jianyue Jin
- University Hospitals/Cleveland Medical Center, Seidman Cancer Center and Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Chen Hu
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yang Wang
- Biomedical Engineering, Shenzhen Polytechnic, Shenzhen, China
| | - Shruti Jolly
- Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Feng-Ming (Spring) Kong
- Department of Clinical Oncology, LKS Faculty of Medicine, the University of Hong Kong, Hong Kong, China
- Clinical Oncology Center, the University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- University Hospitals/Cleveland Medical Center, Seidman Cancer Center and Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
20
|
Lin T, Mai J, Yan M, Li Z, Quan X, Chen X. A Nomogram Based on CT Deep Learning Signature: A Potential Tool for the Prediction of Overall Survival in Resected Non-Small Cell Lung Cancer Patients. Cancer Manag Res 2021; 13:2897-2906. [PMID: 33833572 PMCID: PMC8019610 DOI: 10.2147/cmar.s299020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/02/2021] [Indexed: 12/29/2022] Open
Abstract
PURPOSE To develop and further validate a deep learning signature-based nomogram from computed tomography (CT) images for prediction of the overall survival (OS) in resected non-small cell lung cancer (NSCLC) patients. PATIENTS AND METHODS A total of 1792 deep learning features were extracted from non-enhanced and venous-phase CT images for each NSCLC patient in training cohort (n=231). Then, a deep learning signature was built with the least absolute shrinkage and selection operator (LASSO) Cox regression model for OS estimation. At last, a nomogram was constructed with the signature and other independent clinical risk factors. The performance of nomogram was assessed by discrimination, calibration and clinical usefulness. In addition, in order to quantify the improvement in performance added by deep learning signature, the net reclassification improvement (NRI) was calculated. The results were validated in external validation cohort (n=77). RESULTS A deep learning signature with 9 selected features was significantly associated with OS in both training cohort (hazard ratio [HR]=5.455, 95% CI: 3.393-8.769, P<0.001) and external validation cohort (HR=3.029, 95% CI: 1.673-5.485, P=0.004). The nomogram combining deep learning signature with clinical risk factors of TNM stage, lymphatic vessel invasion and differentiation grade showed favorable discriminative ability with C-index of 0.800 as well as a good calibration, which was validated in external validation cohort (C-index=0.723). Additional value of deep learning signature to the nomogram was statistically significant (NRI=0.093, P=0.027 for training cohort; NRI=0.106, P=0.040 for validation cohort). Decision curve analysis confirmed the clinical usefulness of this nomogram in predicting OS. CONCLUSION The deep learning signature-based nomogram is a robust tool for prognostic prediction in resected NSCLC patients.
Collapse
Affiliation(s)
- Ting Lin
- Department of Radiology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People’s Republic of China
| | - Jinhai Mai
- School of Automation Science and Engineering, South China University of Technology, Guangzhou, 510641, People’s Republic of China
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People’s Republic of China
| | - Meng Yan
- Department of Radiology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People’s Republic of China
| | - Zhenhui Li
- Department of Radiology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, People’s Republic of China
| | - Xianyue Quan
- Department of Radiology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, People’s Republic of China
| | - Xin Chen
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People’s Republic of China
- Department Of Radiology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, People’s Republic of China
| |
Collapse
|
21
|
Rizzo S, Manganaro L, Dolciami M, Gasparri ML, Papadia A, Del Grande F. Computed Tomography Based Radiomics as a Predictor of Survival in Ovarian Cancer Patients: A Systematic Review. Cancers (Basel) 2021; 13:cancers13030573. [PMID: 33540655 PMCID: PMC7867247 DOI: 10.3390/cancers13030573] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 01/27/2021] [Accepted: 01/29/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Ovarian cancer represents the most lethal gynecological malignancy. Since many new drugs have been recently introduced as adjunctive treatments for this pathology, an early prediction of outcome might be helpful to further improve outcomes. Radiomics represents a recent advancement, relying on extraction of quantitative features from imaging examinations. Indeed, clinical images, such as computed tomography images, may contain quantitative information, reflecting the underlying pathophysiology of a tumoral tissue. Radiomic analyses can be performed in tumor regions and metastatic lesions, as well as in normal tissues. The radiomic process relies on quantitative features, usually extracted by dedicated software, and then culminates in analysis and model building, according to a defined clinical question. This systematic review aims to evaluate association between radiomics based on computed tomography images and survival (in terms of overall survival and progression free survival) in ovarian cancer patients. Abstract The objective of this systematic review was to assess the results of radiomics for prediction of overall survival (OS) and progression free survival (PFS) in ovarian cancer (OC) patients. A secondary objective was to evaluate the findings of papers that based their analyses on inter-site heterogeneity. This systematic review was conducted according to the PRISMA statement. After the initial retrieval of 145 articles, the final systematic review comprised six articles. Association between radiomic features and OS was evaluated in 3/6 studies (50%); all articles showed a significant association between radiomic features and OS. Association with PFS was evaluated in 5/6 (83%) articles; the period of follow-up ranged between six and 36 months. All the articles showed significant association between radiomic models and PFS. Inter-site textural features were used for analysis in 2/6 (33%) articles. They demonstrated that high levels of inter-site textural heterogeneity were significantly associated with incomplete surgical resection in breast cancer gene-negative patients, and that lower heterogeneity was associated with complete resectability. There were some differences among papers in methodology; for example, only 3/6 (50%) articles included validation cohorts. In conclusion, radiomic models have demonstrated promising results as predictors of survival in OC patients, although larger studies are needed to allow clinical applicability.
Collapse
Affiliation(s)
- Stefania Rizzo
- Istituto di Imaging della Svizzera Italiana (IIMSI), Ente Ospedaliero Cantonale (EOC), Via Tesserete 46, 6900 Lugano, Switzerland;
- Facoltà di Scienze Biomediche, Università della Svizzera Italiana, Via Buffi 13, 6900 Lugano, Switzerland; (M.L.G.); (A.P.)
- Correspondence: ; Tel.: +41-91-811-6676
| | - Lucia Manganaro
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, 00185 Rome, Italy; (L.M.); (M.D.)
| | - Miriam Dolciami
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, 00185 Rome, Italy; (L.M.); (M.D.)
| | - Maria Luisa Gasparri
- Facoltà di Scienze Biomediche, Università della Svizzera Italiana, Via Buffi 13, 6900 Lugano, Switzerland; (M.L.G.); (A.P.)
- Department of Gynecology and Obstetrics, Ente Ospedaliero Cantonale (EOC), Via Tesserete 46, 6900 Lugano, Switzerland
| | - Andrea Papadia
- Facoltà di Scienze Biomediche, Università della Svizzera Italiana, Via Buffi 13, 6900 Lugano, Switzerland; (M.L.G.); (A.P.)
- Department of Gynecology and Obstetrics, Ente Ospedaliero Cantonale (EOC), Via Tesserete 46, 6900 Lugano, Switzerland
| | - Filippo Del Grande
- Istituto di Imaging della Svizzera Italiana (IIMSI), Ente Ospedaliero Cantonale (EOC), Via Tesserete 46, 6900 Lugano, Switzerland;
- Facoltà di Scienze Biomediche, Università della Svizzera Italiana, Via Buffi 13, 6900 Lugano, Switzerland; (M.L.G.); (A.P.)
| |
Collapse
|
22
|
Bortolotto C, Lancia A, Stelitano C, Montesano M, Merizzoli E, Agustoni F, Stella G, Preda L, Filippi AR. Radiomics features as predictive and prognostic biomarkers in NSCLC. Expert Rev Anticancer Ther 2020; 21:257-266. [PMID: 33216651 DOI: 10.1080/14737140.2021.1852935] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Introduction: Radiomics extracts a large amount of quantitative information from medical images using specific data characterization algorithms. This information, called radiomic features, can be combined with clinical data to build prediction models for prognostic evaluation and treatment selection.Areas covered: We outlined a series of studies investigating the correlation between radiomics features and outcome (prognostic) as well as response to therapy (predictive) in non-small cell lung cancer (NSCLC). We performed our analysis both in the setting of early and advanced stage of disease, with a focus on the different therapies and imaging modalities adopted.Expert opinion: The prognostic and predictive potential of the radiomic approach, combined with clinical models, could help decision-making process and guide toward the creation of an optimal and 'tailored' therapeutic strategy for lung cancer patients. However, due to the low reproducibility of most of the conducted studies and the lack of validated results, such a desirable scenario has not yet been translated to routine clinical practice.
Collapse
Affiliation(s)
| | - Andrea Lancia
- Radiation Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Chiara Stelitano
- Radiology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Marianna Montesano
- Radiation Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Elisa Merizzoli
- Radiation Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | | | - Giulia Stella
- Respiratory Disease Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Lorenzo Preda
- Radiology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | | |
Collapse
|
23
|
Li J, Li X, Chen X, Ma S. [Research Advances and Obstacles of CT-based Radiomics in Diagnosis and Treatment of Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2020; 23:904-908. [PMID: 32798440 PMCID: PMC7583873 DOI: 10.3779/j.issn.1009-3419.2020.101.36] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
影像组学是一种基于多模态医学影像处理分析的技术,该技术能够基于高性能计算机及算法从目前普遍使用的计算机断层扫描(computed tomography, CT)、磁共振图像(magnetic resonance imaging, MRI)和正电子发射/断层图像(positron emission tomography/computed tomography, PET/CT)中自动提取海量数据进行分析,对疾病的早期诊断、良恶性肿瘤鉴别、疾病治疗全程管理,个体化精准治疗等需求提供更多有价值信息。近年来,许多研究表明基于CT的影像组学技术在肺癌的早期诊断、基因表型预测、疗效预测及预后评估均有良好的应用价值,且影像学检查具有无创、经济、可重复等优势。其对临床的指导价值已有所展露,在肺癌的个体化、精准化治疗和研究方面具有较大价值,但是,影像组学特征的重复性和一致性问题以及在肺部肿瘤图像提取中的特征筛选还需进一步研究。
Collapse
Affiliation(s)
- Jiawei Li
- Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xiadong Li
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou 310002, China
| | - Xueqin Chen
- Department of Oncology, Hangzhou First People's Hospital, Hangzhou 310006, China
| | - Shenglin Ma
- Department of Oncology, Hangzhou First People's Hospital, Hangzhou 310006, China
| |
Collapse
|
24
|
Beyond tissue biopsy: a diagnostic framework to address tumor heterogeneity in lung cancer. Curr Opin Oncol 2020; 32:68-77. [PMID: 31714259 DOI: 10.1097/cco.0000000000000598] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW The objective of this review is to discuss the strength and limitations of tissue and liquid biopsy and functional imaging to capture spatial and temporal tumor heterogeneity either alone or as part of a diagnostic framework in non-small cell lung cancer (NSCLC). RECENT FINDINGS NSCLC displays genetic and phenotypic heterogeneity - a detailed knowledge of which is crucial to personalize treatment. Tissue biopsy often lacks spatial and temporal resolution. Thus, NSCLC needs to be characterized by complementary diagnostic methods to resolve heterogeneity. Liquid biopsy offers detection of tumor biomarkers and for example, the classification and monitoring of EGFR mutations in NSCLC. It allows repeated sampling, and therefore, appears promising to address temporal aspects of tumor heterogeneity. Functional imaging methods and emerging image analytic tools, such as radiomics capture temporal and spatial heterogeneity. Further standardization of radiomics is required to allow introduction into clinical routine. SUMMARY To augment the potential of precision therapy, improved diagnostic characterization of tumors is pivotal. We suggest a comprehensive diagnostic framework combining tissue and liquid biopsy and functional imaging to address the known aspects of spatial and temporal tumor heterogeneity on the example of NSCLC. We envision how this framework might be implemented in clinical practice.
Collapse
|
25
|
Sheng K. Artificial intelligence in radiotherapy: a technological review. Front Med 2020; 14:431-449. [PMID: 32728877 DOI: 10.1007/s11684-020-0761-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 02/14/2020] [Indexed: 12/19/2022]
Abstract
Radiation therapy (RT) is widely used to treat cancer. Technological advances in RT have occurred in the past 30 years. These advances, such as three-dimensional image guidance, intensity modulation, and robotics, created challenges and opportunities for the next breakthrough, in which artificial intelligence (AI) will possibly play important roles. AI will replace certain repetitive and labor-intensive tasks and improve the accuracy and consistency of others, particularly those with increased complexity because of technological advances. The improvement in efficiency and consistency is important to manage the increasing cancer patient burden to the society. Furthermore, AI may provide new functionalities that facilitate satisfactory RT. The functionalities include superior images for real-time intervention and adaptive and personalized RT. AI may effectively synthesize and analyze big data for such purposes. This review describes the RT workflow and identifies areas, including imaging, treatment planning, quality assurance, and outcome prediction, that benefit from AI. This review primarily focuses on deep-learning techniques, although conventional machine-learning techniques are also mentioned.
Collapse
Affiliation(s)
- Ke Sheng
- Department of Radiation Oncology, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
26
|
Fornacon-Wood I, Faivre-Finn C, O'Connor JPB, Price GJ. Radiomics as a personalized medicine tool in lung cancer: Separating the hope from the hype. Lung Cancer 2020; 146:197-208. [PMID: 32563015 PMCID: PMC7383235 DOI: 10.1016/j.lungcan.2020.05.028] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 05/18/2020] [Accepted: 05/23/2020] [Indexed: 12/24/2022]
Abstract
Radiomics has become a popular image analysis method in the last few years. Its key hypothesis is that medical images harbor biological, prognostic and predictive information that is not revealed upon visual inspection. In contrast to previous work with a priori defined imaging biomarkers, radiomics instead calculates image features at scale and uses statistical methods to identify those most strongly associated to outcome. This builds on years of research into computer aided diagnosis and pattern recognition. While the potential of radiomics to aid personalized medicine is widely recognized, several technical limitations exist which hinder biomarker translation. Aspects of the radiomic workflow lack repeatability or reproducibility under particular circumstances, which is a key requirement for the translation of imaging biomarkers into clinical practice. One of the most commonly studied uses of radiomics is for personalized medicine applications in Non-Small Cell Lung Cancer (NSCLC). In this review, we summarize reported methodological limitations in CT based radiomic analyses together with suggested solutions. We then evaluate the current NSCLC radiomics literature to assess the risk associated with accepting the published conclusions with respect to these limitations. We review different complementary scoring systems and initiatives that can be used to critically appraise data from radiomics studies. Wider awareness should improve the quality of ongoing and future radiomics studies and advance their potential as clinically relevant biomarkers for personalized medicine in patients with NSCLC.
Collapse
Affiliation(s)
| | - Corinne Faivre-Finn
- Division of Cancer Sciences, University of Manchester, Manchester, UK; Department of Radiation Oncology, The Christie Hospital NHS Foundation Trust, Manchester, UK
| | - James P B O'Connor
- Division of Cancer Sciences, University of Manchester, Manchester, UK; Department of Radiology, The Christie Hospital NHS Foundation Trust, Manchester, UK
| | - Gareth J Price
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
27
|
Recent developments and advances in secondary prevention of lung cancer. Eur J Cancer Prev 2020; 29:321-328. [PMID: 32452945 DOI: 10.1097/cej.0000000000000586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Lung cancer prevention may include primary prevention strategies, such as corrections of working conditions and life style - primarily smoking cessation - as well as secondary prevention strategies, aiming at early detection that allows better survival rates and limited resections. This review summarizes recent developments and advances in secondary prevention, focusing on recent technological tools for an effective early diagnosis.
Collapse
|
28
|
Mazzaschi G, Milanese G, Pagano P, Madeddu D, Gnetti L, Trentini F, Falco A, Frati C, Lorusso B, Lagrasta C, Minari R, Ampollini L, Silva M, Sverzellati N, Quaini F, Roti G, Tiseo M. Integrated CT imaging and tissue immune features disclose a radio-immune signature with high prognostic impact on surgically resected NSCLC. Lung Cancer 2020; 144:30-39. [PMID: 32361033 DOI: 10.1016/j.lungcan.2020.04.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 03/06/2020] [Accepted: 04/07/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Qualitative and quantitative CT imaging features might intercept the multifaceted tumor immune microenvironment (TIME), providing a non-invasive approach to design new prognostic models in NSCLC patients. MATERIALS AND METHODS Our study population consisted of 100 surgically resected NSCLC patients among which 31 served as a validation cohort for quantitative image analysis. TIME was classified according to PD-L1 expression and the magnitude of Tumor Infiltrating Lymphocytes (TILs) and further defined as hot or cold by the tissue analysis of effector (CD8-to-CD3high/PD-1-to-CD8low) or inert (CD8-to-CD3low/PD-1-to-CD8high) phenotypes. CT datasets acted as source for qualitative (semantic, CT-SFs) and quantitative (radiomic, CT-RFs) features which were correlated with clinico-pathological and TIME profiles to determine their impact on survival outcome. RESULTS Specific CT-SFs (texture [TXT], effect [EFC] and margins [MRG]) strongly correlated to PD-L1 and TILs status and showed significant impact on survival outcome (TXT, HR:3.39, 95 % CI 1.12-10-27, P < 0.05; EFC, HR:0.41, 95 % CI 0.18-0.93, P < 0.05; MRG, HR:1.93, 95 % CI 0.88-4.25, P = 0.09). Seven CT derived radiomic features were able to sharply discriminate cases with hot (inflamed) vs cold (desert) TIME, which also exhibited opposite OS (long vs short, HR:0.09, 95 % CI 0.04-0.23, P < 0.001) and DFS (long vs short, HR:0.31, 95 % CI 0.16-0.58, P < 0.001). Moreover, we identified 6 prognostic radiomic features among which ClusterProminence displayed the highest statistical significance (HR:0.13, 95 % CI 0.06-0.31, P < 0.001). These findings were independently validated in an additional cohort of NSCLC (HR:0.11, 95 % CI 0.03-0.40, P = 0.001). Finally, in our training cohort we developed a multiparametric prognostic model, interlacing TIME and clinico-pathological characteristics with CT-SFs (ROC curve AUC:0.83, 95 % CI 0.71-0.92, P < 0.001) or CT-RFs (AUC: 0.91, 95 % CI 0.83-0.99, P < 0.001), which appeared to outperform pTNM staging (AUC: 0.66, 95 % CI 0.51-0.80, P < 0.05) in the risk assessment of NSCLC. CONCLUSION Higher order CT extracted features associated with specific TIME profiles may reveal a radio-immune signature with prognostic impact on resected NSCLC.
Collapse
Affiliation(s)
- Giulia Mazzaschi
- Department of Medicine and Surgery, University of Parma, Medical Oncology Unit, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Gianluca Milanese
- Department of Medicine and Surgery, University of Parma, Institute of Radiologic Science, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Paolo Pagano
- Department of Medicine and Surgery, University of Parma, Institute of Radiologic Science, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Denise Madeddu
- Department of Medicine and Surgery, University of Parma, Pathology Unit, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Letizia Gnetti
- Department of Medicine and Surgery, University of Parma, Pathology Unit, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Francesca Trentini
- Department of Medicine and Surgery, University of Parma, Medical Oncology Unit, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Angela Falco
- Department of Medicine and Surgery, University of Parma, Pathology Unit, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Caterina Frati
- Department of Medicine and Surgery, University of Parma, Pathology Unit, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Bruno Lorusso
- Department of Medicine and Surgery, University of Parma, Pathology Unit, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Costanza Lagrasta
- Department of Medicine and Surgery, University of Parma, Pathology Unit, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Roberta Minari
- Department of Medicine and Surgery, University of Parma, Medical Oncology Unit, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Luca Ampollini
- Department of Medicine and Surgery, University of Parma, Thoracic Surgery, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Mario Silva
- Department of Medicine and Surgery, University of Parma, Institute of Radiologic Science, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Nicola Sverzellati
- Department of Medicine and Surgery, University of Parma, Institute of Radiologic Science, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Federico Quaini
- Department of Medicine and Surgery, Hematology and Bone Marrow Transplantation, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Giovanni Roti
- Department of Medicine and Surgery, Hematology and Bone Marrow Transplantation, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Marcello Tiseo
- Department of Medicine and Surgery, University of Parma, Medical Oncology Unit, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| |
Collapse
|
29
|
Song L, Zhu Z, Mao L, Li X, Han W, Du H, Wu H, Song W, Jin Z. Clinical, Conventional CT and Radiomic Feature-Based Machine Learning Models for Predicting ALK Rearrangement Status in Lung Adenocarcinoma Patients. Front Oncol 2020; 10:369. [PMID: 32266148 PMCID: PMC7099003 DOI: 10.3389/fonc.2020.00369] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 03/03/2020] [Indexed: 12/25/2022] Open
Abstract
Objectives: To predict the anaplastic lymphoma kinase (ALK) mutations in lung adenocarcinoma patients non-invasively with machine learning models that combine clinical, conventional CT and radiomic features. Methods: This retrospective study included 335 lung adenocarcinoma patients who were randomly divided into a primary cohort (268 patients; 90 ALK-rearranged; and 178 ALK wild-type) and a test cohort (67 patients; 22 ALK-rearranged; and 45 ALK wild-type). One thousand two hundred and eighteen quantitative radiomic features were extracted from the semi-automatically delineated volume of interest (VOI) of the entire tumor using both the original and the pre-processed non-enhanced CT images. Twelve conventional CT features and seven clinical features were also collected. Normalized features were selected using a sequential of the F-test-based method, the density-based spatial clustering of applications with noise (DBSCAN) method, and the recursive feature elimination (RFE) method. Selected features were then used to build three predictive models (radiomic, radiological, and integrated models) for the ALK-rearranged phenotype by a soft voting classifier. Models were evaluated in the test cohort using the area under the receiver operating characteristic curve (AUC), accuracy, sensitivity, and specificity, and the performances of three models were compared using the DeLong test. Results: Our results showed that the addition of clinical information and conventional CT features significantly enhanced the validation performance of the radiomic model in the primary cohort (AUC = 0.83–0.88, P = 0.01), but not in the test cohort (AUC = 0.80–0.88, P = 0.29). The majority of radiomic features associated with ALK mutations reflected information around and within the high-intensity voxels of lesions. The presence of the cavity and left lower lobe location were new imaging phenotypic patterns in association with ALK-rearranged tumors. Current smoking was strongly correlated with non-ALK-mutated lung adenocarcinoma. Conclusions: Our study demonstrates that radiomics-derived machine learning models can potentially serve as a non-invasive tool to identify ALK mutation of lung adenocarcinoma.
Collapse
Affiliation(s)
- Lan Song
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhenchen Zhu
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,4+4 MD Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li Mao
- Deepwise AI Lab, Deepwise Inc., Beijing, China
| | - Xiuli Li
- Deepwise AI Lab, Deepwise Inc., Beijing, China
| | - Wei Han
- Department of Epidemiology and Biostatistics, Institute of Basic Medicine Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Huayang Du
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huanwen Wu
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Song
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhengyu Jin
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
30
|
Kakino R, Nakamura M, Mitsuyoshi T, Shintani T, Hirashima H, Matsuo Y, Mizowaki T. Comparison of radiomic features in diagnostic CT images with and without contrast enhancement in the delayed phase for NSCLC patients. Phys Med 2020; 69:176-182. [PMID: 31918370 DOI: 10.1016/j.ejmp.2019.12.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 11/18/2019] [Accepted: 12/18/2019] [Indexed: 12/18/2022] Open
Abstract
PURPOSE To compare radiomic features extracted from diagnostic computed tomography (CT) images with and without contrast enhancement in delayed phase for non-small cell lung cancer (NSCLC) patients. METHODS Diagnostic CT images from 269 tumors [non-contrast CT, 188 (dataset NE); contrast-enhanced CT, 81 (dataset CE)] were enrolled in this study. Eighteen first-order and seventy-five texture features were extracted by setting five bin width levels for CT values. Reproducible features were selected by the intraclass correlation coefficient (ICC). Radiomic features were compared between datasets NE and CE. Subgroup analyses were performed based on the CT acquisition period, exposure value, and patient characteristics. RESULTS Eighty features were considered reproducible (0.5 ≤ ICC). Twelve of the sixteen first-order features, independent of the bin width levels, were statistically different between datasets NE and CE (p < 0.05), and the p-values of two first-order features depending on the bin width levels were reduced with narrower bin widths. Sixteen out of sixty-two features showed a significant difference, regardless of the bin width (p < 0.05). There were significant differences between datasets NE and CE with older age, lighter body weight, better performance status, being a smoker, larger gross tumor volume, and tumor location at central region. CONCLUSIONS Contrast enhancement in the delayed phase of CT images for NSCLC patients affected some of the radiomic features and the variability of radiomic features due to contrast uptake may depend largely on the patient characteristics.
Collapse
Affiliation(s)
- Ryo Kakino
- Division of Medical Physics, Department of Information Technology and Medical Engineering, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan; Department of Radiation Oncology and Image-applied Therapy, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan; Research Fellow at Japan Society for the Promotion of Science, Tokyo, Japan
| | - Mitsuhiro Nakamura
- Division of Medical Physics, Department of Information Technology and Medical Engineering, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan; Department of Radiation Oncology and Image-applied Therapy, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan.
| | - Takamasa Mitsuyoshi
- Department of Radiation Oncology and Image-applied Therapy, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Takashi Shintani
- Department of Radiation Oncology and Image-applied Therapy, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Hideaki Hirashima
- Department of Radiation Oncology and Image-applied Therapy, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Yukinori Matsuo
- Department of Radiation Oncology and Image-applied Therapy, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Takashi Mizowaki
- Department of Radiation Oncology and Image-applied Therapy, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
31
|
Is tumour sphericity an important prognostic factor in patients with lung cancer? Radiother Oncol 2019; 143:73-80. [PMID: 31472998 DOI: 10.1016/j.radonc.2019.08.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 07/05/2019] [Accepted: 08/05/2019] [Indexed: 01/05/2023]
Abstract
BACKGROUND AND PURPOSE Quantitative tumour shape features extracted from radiotherapy planning scans have shown potential as prognostic markers. In this study, we investigated if sphericity of the gross tumour volume (GTV) on planning computed tomography (CT) is an independent predictor of overall survival (OS) in lung cancer patients treated with standard radiotherapy. In the analysis, we considered whether tumour sphericity is correlated with clinical prognostic factors or influenced by the inclusion of lymph nodes in the GTV. MATERIALS AND METHODS Sphericity of single GTV delineation was extracted for 457 lung cancer patients. Relationships between sphericity, and common patient and tumour characteristics were investigated via correlation analysis and multivariate Cox regression to assess prognostic value of GTV sphericity. A subset analysis was performed for 290 nodal stage N0 patients to determine prognostic value of primary tumour sphericity. RESULTS Sphericity is correlated with clinical variables: tumour volume, mean lung dose, N stage, and T stage. Sphericity is strongly associated with OS (p < 0.001, hazard ratio (HR) (95% confidence interval (CI)) = 0.13 (0.04-0.41)) in univariate analysis. However, this association did not remain significant in multivariate analysis (p = 0.826, HR (95% CI) = 0.83 (0.16-4.31), and inclusion of sphericity to a clinical model did not improve model performance. In addition, no significant relationship between sphericity and OS was detected in univariate (p = 0.072) or multivariate (p = 0.920) analysis of N0 subset. CONCLUSION Sphericity correlates clearly with clinical prognostic factors, which are often unaccounted for in radiomic studies. Sphericity is also influenced by the presence of nodal involvement within the GTV contour.
Collapse
|
32
|
van Timmeren JE, van Elmpt W, Leijenaar RTH, Reymen B, Monshouwer R, Bussink J, Paelinck L, Bogaert E, De Wagter C, Elhaseen E, Lievens Y, Hansen O, Brink C, Lambin P. Longitudinal radiomics of cone-beam CT images from non-small cell lung cancer patients: Evaluation of the added prognostic value for overall survival and locoregional recurrence. Radiother Oncol 2019; 136:78-85. [PMID: 31015133 PMCID: PMC6598851 DOI: 10.1016/j.radonc.2019.03.032] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 03/07/2019] [Accepted: 03/29/2019] [Indexed: 11/23/2022]
Abstract
Longitudinal CBCT radiomics does not show added prognostic information for NSCLC. A CT-radiomics model could be validated in an external validation dataset. Dataset heterogeneity and small cohort sizes could cause poor validation performance.
Background and purpose The prognostic value of radiomics for non-small cell lung cancer (NSCLC) patients has been investigated for images acquired prior to treatment, but no prognostic model has been developed that includes the change of radiomic features during treatment. Therefore, the aim of this study was to investigate the potential added prognostic value of a longitudinal radiomics approach using cone-beam computed tomography (CBCT) for NSCLC patients. Materials and methods This retrospective study includes a training dataset of 141 stage I–IV NSCLC patients and three external validation datasets of 94, 61 and 41 patients, all treated with curative intended (chemo)radiotherapy. The change of radiomic features extracted from CBCT images was summarized as the slope of a linear regression. The CBCT slope-features and CT-extracted features were used as input for a Cox proportional hazards model. Moreover, prognostic performance of clinical parameters was investigated for overall survival and locoregional recurrence. Model performances were assessed using the Kaplan–Meier curves and c-index. Results The radiomics model contained only CT-derived features and reached a c-index of 0.63 for overall survival and could be validated on the first validation dataset. No model for locoregional recurrence could be developed that validated on the validation datasets. The clinical parameters model could not be validated for either overall survival or locoregional recurrence. Conclusion In this study we could not confirm our hypothesis that longitudinal CBCT-extracted radiomic features contribute to improved prognostic information. Moreover, performance of baseline radiomic features or clinical parameters was poor, probably affected by heterogeneity within and between datasets.
Collapse
Affiliation(s)
- Janna E van Timmeren
- The D-Lab: Decision Support for Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, the Netherlands.
| | - Wouter van Elmpt
- Department of Radiation Oncology (MAASTRO), GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre (MUMC), Maastricht, the Netherlands
| | - Ralph T H Leijenaar
- The D-Lab: Decision Support for Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Bart Reymen
- Department of Radiation Oncology (MAASTRO), GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre (MUMC), Maastricht, the Netherlands
| | - René Monshouwer
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Johan Bussink
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Leen Paelinck
- Ghent University Hospital and Ghent University, Ghent, Belgium
| | - Evelien Bogaert
- Ghent University Hospital and Ghent University, Ghent, Belgium
| | | | - Elamin Elhaseen
- Ghent University Hospital and Ghent University, Ghent, Belgium
| | - Yolande Lievens
- Ghent University Hospital and Ghent University, Ghent, Belgium
| | - Olfred Hansen
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark; Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Carsten Brink
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark; Laboratory of Radiation Physics, Odense University Hospital, Odense, Denmark
| | - Philippe Lambin
- The D-Lab: Decision Support for Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, the Netherlands
| |
Collapse
|
33
|
Astaraki M, Wang C, Buizza G, Toma-Dasu I, Lazzeroni M, Smedby Ö. Early survival prediction in non-small cell lung cancer from PET/CT images using an intra-tumor partitioning method. Phys Med 2019; 60:58-65. [DOI: 10.1016/j.ejmp.2019.03.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 02/12/2019] [Accepted: 03/21/2019] [Indexed: 12/26/2022] Open
|