51
|
Bao J, Feng X, Ma Y, Wang Y, Qi J, Qin C, Tan X, Tian Y. The latest application progress of radiomics in prediction and diagnosis of liver diseases. Expert Rev Gastroenterol Hepatol 2022; 16:707-719. [PMID: 35880549 DOI: 10.1080/17474124.2022.2104711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Early detection and individualized treatment of patients with liver disease is the key to survival. Radiomics can extract high-throughput quantitative features by multimode imaging, which has good application prospects for the diagnosis, staging and prognosis of benign and malignant liver diseases. Therefore, this paper summarizes the current research status in the field of liver disease, in order to help these patients achieve personalized and precision medical care. AREAS COVERED This paper uses several keywords on the PubMed database to search the references, and reviews the workflow of traditional radiomics, as well as the characteristics and influencing factors of different imaging modes. At the same time, the references on the application of imaging in different benign and malignant liver diseases were also summarized. EXPERT OPINION For patients with liver disease, the traditional imaging evaluation can only provide limited information. Radiomics exploits the characteristics of high-throughput and high-dimensional extraction, enabling liver imaging capabilities far beyond the scope of traditional visual image analysis. Recent studies have demonstrated the prospect of this technology in personalized diagnosis and treatment decision in various fields of the liver. However, further clinical validation is needed in its application and practice.
Collapse
Affiliation(s)
- Jiaying Bao
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, P.R. China
| | - Xiao Feng
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, P.R. China
| | - Yan Ma
- Department of Ultrasound, Zibo Central Hospital, Zibo, P.R. China
| | - Yanyan Wang
- Departments of Emergency Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| | - Jianni Qi
- Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| | - Chengyong Qin
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, P.R. China
| | - Xu Tan
- Department of Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| | - Yongmei Tian
- Department of Geriatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| |
Collapse
|
52
|
Moskowitz CS, Welch ML, Jacobs MA, Kurland BF, Simpson AL. Radiomic Analysis: Study Design, Statistical Analysis, and Other Bias Mitigation Strategies. Radiology 2022; 304:265-273. [PMID: 35579522 PMCID: PMC9340236 DOI: 10.1148/radiol.211597] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 01/30/2022] [Accepted: 02/02/2022] [Indexed: 12/13/2022]
Abstract
Rapid advances in automated methods for extracting large numbers of quantitative features from medical images have led to tremendous growth of publications reporting on radiomic analyses. Translation of these research studies into clinical practice can be hindered by biases introduced during the design, analysis, or reporting of the studies. Herein, the authors review biases, sources of variability, and pitfalls that frequently arise in radiomic research, with an emphasis on study design and statistical analysis considerations. Drawing on existing work in the statistical, radiologic, and machine learning literature, approaches for avoiding these pitfalls are described.
Collapse
Affiliation(s)
| | | | - Michael A. Jacobs
- From the Department of Epidemiology and Biostatistics, Memorial Sloan
Kettering Cancer Center, 485 Lexington Ave, 2nd Floor, New York, NY, NY 10017
(C.S.M.); Cancer Digital Intelligence Program, University Health Network,
Toronto, ON, Canada (M.L.W.); The Russell H. Morgan Department of Radiology and
Radiological Science and Sidney Kimmel Comprehensive Cancer Center, The Johns
Hopkins School of Medicine, Baltimore, Md (M.A.J.); ERT, Pittsburgh, Pa
(B.F.K.); and School of Computing, Department of Biomedical and Molecular
Sciences, Queen’s University, Kingston, ON, Canada (A.L.S.)
| | - Brenda F. Kurland
- From the Department of Epidemiology and Biostatistics, Memorial Sloan
Kettering Cancer Center, 485 Lexington Ave, 2nd Floor, New York, NY, NY 10017
(C.S.M.); Cancer Digital Intelligence Program, University Health Network,
Toronto, ON, Canada (M.L.W.); The Russell H. Morgan Department of Radiology and
Radiological Science and Sidney Kimmel Comprehensive Cancer Center, The Johns
Hopkins School of Medicine, Baltimore, Md (M.A.J.); ERT, Pittsburgh, Pa
(B.F.K.); and School of Computing, Department of Biomedical and Molecular
Sciences, Queen’s University, Kingston, ON, Canada (A.L.S.)
| | - Amber L. Simpson
- From the Department of Epidemiology and Biostatistics, Memorial Sloan
Kettering Cancer Center, 485 Lexington Ave, 2nd Floor, New York, NY, NY 10017
(C.S.M.); Cancer Digital Intelligence Program, University Health Network,
Toronto, ON, Canada (M.L.W.); The Russell H. Morgan Department of Radiology and
Radiological Science and Sidney Kimmel Comprehensive Cancer Center, The Johns
Hopkins School of Medicine, Baltimore, Md (M.A.J.); ERT, Pittsburgh, Pa
(B.F.K.); and School of Computing, Department of Biomedical and Molecular
Sciences, Queen’s University, Kingston, ON, Canada (A.L.S.)
| |
Collapse
|
53
|
Quraishi MI. Radiomics-Guided Precision Medicine Approaches for Colorectal Cancer. Front Oncol 2022; 12:872656. [PMID: 35756680 PMCID: PMC9218262 DOI: 10.3389/fonc.2022.872656] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 05/06/2022] [Indexed: 11/13/2022] Open
Abstract
The concept of precision oncology entails molecular profiling of tumors to guide therapeutic interventions. Genomic testing through next-generation sequencing (NGS) molecular analysis provides the basis of such highly targeted therapeutics in oncology. As radiomic analysis delivers an array of structural and functional imaging-based biomarkers that depict these molecular mechanisms and correlate with key genetic alterations related to cancers. There is an opportunity to synergize these two big-data approaches to determine the molecular guidance for precision therapeutics. Colorectal cancer is one such disease whose therapeutic management is being guided by genetic and genomic analyses. We review the rationale and utility of radiomics as a combinative strategy for these approaches in the management of colorectal cancer.
Collapse
Affiliation(s)
- Mohammed I Quraishi
- Department of Radiology, University of Tennessee Medical Center, Knoxville, TN, United States
| |
Collapse
|
54
|
Granata V, Fusco R, De Muzio F, Cutolo C, Setola SV, Dell'Aversana F, Grassi F, Belli A, Silvestro L, Ottaiano A, Nasti G, Avallone A, Flammia F, Miele V, Tatangelo F, Izzo F, Petrillo A. Radiomics and machine learning analysis based on magnetic resonance imaging in the assessment of liver mucinous colorectal metastases. Radiol Med 2022; 127:763-772. [PMID: 35653011 DOI: 10.1007/s11547-022-01501-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 04/27/2022] [Indexed: 12/11/2022]
Abstract
PURPOSE The purpose of this study is to evaluate the Radiomics and Machine Learning Analysis based on MRI in the assessment of Liver Mucinous Colorectal Metastases.Query METHODS: The cohort of patients included a training set (121 cases) and an external validation set (30 cases) with colorectal liver metastases with pathological proof and MRI study enrolled in this approved study retrospectively. About 851 radiomics features were extracted as median values by means of the PyRadiomics tool on volume on interest segmented manually by two expert radiologists. Univariate analysis, linear regression modelling and pattern recognition methods were used as statistical and classification procedures. RESULTS The best results at univariate analysis were reached by the wavelet_LLH_glcm_JointEntropy extracted by T2W SPACE sequence with accuracy of 92%. Linear regression model increased the performance obtained respect to the univariate analysis. The best results were obtained by a linear regression model of 15 significant features extracted by the T2W SPACE sequence with accuracy of 94%, a sensitivity of 92% and a specificity of 95%. The best classifier among the tested pattern recognition approaches was k-nearest neighbours (KNN); however, KNN achieved lower precision than the best linear regression model. CONCLUSIONS Radiomics metrics allow the mucinous subtype lesion characterization, in order to obtain a more personalized approach. We demonstrated that the best performance was obtained by T2-W extracted textural metrics.
Collapse
Affiliation(s)
- Vincenza Granata
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale-IRCCS Di Napoli, Naples, Italy
| | | | - Federica De Muzio
- Department of Medicine and Health Sciences V. Tiberio, University of Molise, 86100, Campobasso, Italy
| | - Carmen Cutolo
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84084, Fisciano, Italy
| | - Sergio Venanzio Setola
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale-IRCCS Di Napoli, Naples, Italy
| | - Federica Dell'Aversana
- Division of Radiology, Università Degli Studi Della Campania Luigi Vanvitelli, Naples, Italy
| | - Francesca Grassi
- Division of Radiology, Università Degli Studi Della Campania Luigi Vanvitelli, Naples, Italy
| | - Andrea Belli
- Division of Hepatobiliary Surgery, Istituto Nazionale Tumori IRCCS Fondazione Pascale-IRCCS Di Napoli, Naples, Italy
| | - Lucrezia Silvestro
- Division of Abdominal Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Naples, Italy
| | - Alessandro Ottaiano
- Division of Abdominal Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Naples, Italy
| | - Guglielmo Nasti
- Division of Abdominal Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Naples, Italy
| | - Antonio Avallone
- Division of Abdominal Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Naples, Italy
| | - Federica Flammia
- Division of Radiology, Azienda Ospedaliera Universitaria Careggi, 50134, Florence, Italy
| | - Vittorio Miele
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, via della Signora 2, 20122, Milan, Italy.,Division of Radiology, Azienda Ospedaliera Universitaria Careggi, 50134, Florence, Italy
| | - Fabiana Tatangelo
- Division of Pathology, Istituto Nazionale Tumori IRCCS Fondazione Pascale-IRCCS Di Napoli, 80131, Naples, Italy
| | - Francesco Izzo
- Division of Hepatobiliary Surgery, Istituto Nazionale Tumori IRCCS Fondazione Pascale-IRCCS Di Napoli, Naples, Italy
| | - Antonella Petrillo
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale-IRCCS Di Napoli, Naples, Italy
| |
Collapse
|
55
|
Maniar A, Wei AZ, Dercle L, Bien HH, Fojo T, Bates SE, Schwartz LH. Assessing Outcomes in NSCLC: Radiomic analysis, kinetic analysis and circulating tumor DNA. Semin Oncol 2022; 49:298-305. [PMID: 35914982 DOI: 10.1053/j.seminoncol.2022.06.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 06/06/2022] [Indexed: 11/11/2022]
Abstract
Current radiographic methods of measuring treatment response for patients with nonsmall cell lung cancer have significant limitations. Recently, new modalities using standard of care images or minimally invasive blood-based DNA tests have gained interest as methods of evaluating treatment response. This article highlights three emerging modalities: radiomic analysis, kinetic analysis and serum-based measurement of circulating tumor DNA, with a focus on the clinical evidence supporting these methods. Additionally, we discuss the possibility of combining these modalities to develop a robust biomarker with strong correlation to clinically meaningful outcomes that could impact clinical trial design and patient care. At Last, we focus on how these methods specifically apply to a Veteran population.
Collapse
Affiliation(s)
- Ashray Maniar
- Columbia University Irving Medical Center, Division of Hematology and Oncology, New York, NY
| | - Alexander Z Wei
- Columbia University Irving Medical Center, Division of Hematology and Oncology, New York, NY
| | - Laurent Dercle
- Columbia University Irving Medical Center, Division of Radiology, New York, NY
| | - Harold H Bien
- Northport VA Medical Center, Division of Hematology and Oncology, Northport, NY
| | - Tito Fojo
- Columbia University Irving Medical Center, Division of Hematology and Oncology, New York, NY; James J. Peters Bronx VA Medical Center, Division of Hematology and Oncology, Bronx, NY
| | - Susan E Bates
- Columbia University Irving Medical Center, Division of Hematology and Oncology, New York, NY; Northport VA Medical Center, Division of Hematology and Oncology, Northport, NY.
| | - Lawrence H Schwartz
- Columbia University Irving Medical Center, Division of Radiology, New York, NY
| |
Collapse
|
56
|
Radiomics and Machine Learning Analysis Based on Magnetic Resonance Imaging in the Assessment of Colorectal Liver Metastases Growth Pattern. Diagnostics (Basel) 2022; 12:diagnostics12051115. [PMID: 35626271 PMCID: PMC9140199 DOI: 10.3390/diagnostics12051115] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/11/2022] [Accepted: 04/27/2022] [Indexed: 02/07/2023] Open
Abstract
To assess Radiomics and Machine Learning Analysis in Liver Colon and Rectal Cancer Metastases (CRLM) Growth Pattern, we evaluated, retrospectively, a training set of 51 patients with 121 liver metastases and an external validation set of 30 patients with a single lesion. All patients were subjected to MRI studies in pre-surgical setting. For each segmented volume of interest (VOI), 851 radiomics features were extracted using PyRadiomics package. Nonparametric test, univariate, linear regression analysis and patter recognition approaches were performed. The best results to discriminate expansive versus infiltrative front of tumor growth with the highest accuracy and AUC at univariate analysis were obtained by the wavelet_LHH_glrlm_ShortRunLowGray Level Emphasis from portal phase of contrast study. With regard to linear regression model, this increased the performance obtained respect to the univariate analysis for each sequence except that for EOB-phase sequence. The best results were obtained by a linear regression model of 15 significant features extracted by the T2-W SPACE sequence. Furthermore, using pattern recognition approaches, the diagnostic performance to discriminate the expansive versus infiltrative front of tumor growth increased again and the best classifier was a weighted KNN trained with the 9 significant metrics extracted from the portal phase of contrast study, with an accuracy of 92% on training set and of 91% on validation set. In the present study, we have demonstrated as Radiomics and Machine Learning Analysis, based on EOB-MRI study, allow to identify several biomarkers that permit to recognise the different Growth Patterns in CRLM.
Collapse
|
57
|
Deantonio L, Garo ML, Paone G, Valli MC, Cappio S, La Regina D, Cefali M, Palmarocchi MC, Vannelli A, De Dosso S. 18F-FDG PET Radiomics as Predictor of Treatment Response in Oesophageal Cancer: A Systematic Review and Meta-Analysis. Front Oncol 2022; 12:861638. [PMID: 35371989 PMCID: PMC8965232 DOI: 10.3389/fonc.2022.861638] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 02/16/2022] [Indexed: 12/22/2022] Open
Abstract
The best treatment strategy for oesophageal cancer patients achieving a complete clinical response after neoadjuvant chemoradiation is a burning topic. The available diagnostic tools, such as 18F-FDG PET/CT performed routinely, cannot accurately evaluate the presence or absence of the residual tumour. The emerging field of radiomics may encounter the critical challenge of personalised treatment. Radiomics is based on medical image analysis, executed by extracting information from many image features; it has been shown to provide valuable information for predicting treatment responses in oesophageal cancer. This systematic review with a meta-analysis aims to provide current evidence of 18F-FDG PET-based radiomics in predicting response treatments following neoadjuvant chemoradiotherapy in oesophageal cancer. A comprehensive literature review identified 1160 studies, of which five were finally included in the study. Our findings provided that pooled Area Under the Curve (AUC) of the five selected studies was relatively high at 0.821 (95% CI: 0.737–0.904) and not influenced by the sample size of the studies. Radiomics models exhibited a good performance in predicting pathological complete responses (pCRs). This review further strengthens the great potential of 18F-FDG PET-based radiomics to predict pCRs in oesophageal cancer patients who underwent neoadjuvant chemoradiotherapy. Additionally, our review imparts additional support to prospective studies on 18F-FDG PET radiomics for a tailored treatment strategy of oesophageal cancer patients.
Collapse
Affiliation(s)
- Letizia Deantonio
- Radiation Oncology Clinic, Oncology Institute of Southern Switzerland (IOSI), Bellinzona, Switzerland.,University of Southern Switzerland, Faculty of Biomedical Sciences, Lugano, Switzerland
| | | | - Gaetano Paone
- Clinic for Nuclear Medicine and Molecular Imaging, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Maria Carla Valli
- Radiation Oncology Clinic, Oncology Institute of Southern Switzerland (IOSI), Bellinzona, Switzerland
| | - Stefano Cappio
- Clinic for Radiology, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Davide La Regina
- Department of Surgery, Ente Ospedaliero Cantonale, Bellinzona, Switzerland.,University of Southern Switzerland, Faculty of Biomedical Sciences, Lugano, Switzerland
| | - Marco Cefali
- Department of Medical Oncology, Oncology Institute of Southern Switzerland (IOSI), Bellinzona, Switzerland
| | - Maria Celeste Palmarocchi
- Department of Medical Oncology, Oncology Institute of Southern Switzerland (IOSI), Bellinzona, Switzerland
| | | | - Sara De Dosso
- University of Southern Switzerland, Faculty of Biomedical Sciences, Lugano, Switzerland.,Department of Medical Oncology, Oncology Institute of Southern Switzerland (IOSI), Bellinzona, Switzerland
| |
Collapse
|
58
|
Lu J, Xue Z, Xu BB, Wu D, Zheng HL, Xie JW, Wang JB, Lin JX, Chen QY, Li P, Huang CM, Zheng CH. Application of an artificial neural network for predicting the potential chemotherapy benefit of patients with gastric cancer after radical surgery. Surgery 2022; 171:955-965. [PMID: 34756492 DOI: 10.1016/j.surg.2021.08.055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 08/19/2021] [Accepted: 08/31/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND Artificial neural network models have a strong self-learning ability and can deal with complex biological information, but there is no artificial neural network model for predicting the benefits of adjuvant chemotherapy in patients with gastric cancer. METHODS The clinicopathological data of patients who underwent radical resection of gastric cancer from January 2010 to September 2014 were analyzed retrospectively. Patients who underwent surgery combined with adjuvant chemotherapy were randomly divided into a training cohort (70%) and a validation cohort (30%). An artificial neural network model (potential-CT-benefit-ANN) was established, and its ability to predict the potential benefit of chemotherapy was evaluated by the C-index. The prognostic prediction and stratification ability of potential-CT-benefit-ANN and the eighth American Joint Committee on Cancer staging system were compared by receiver operating characteristic curves and Kaplan-Meier curves. RESULTS In both the training and validation cohort, potential-CT-benefit-ANN shows good prediction accuracy for potential adjuvant chemotherapy benefit. The receiver operating characteristic curve showed that the prediction accuracy of potential-CT-benefit-ANN was better than that of the eighth American Joint Committee on Cancer staging system in all groups. The calibration plots showed that the predicted prognosis of potential-CT-benefit-ANN was highly consistent with the actual value. The survival curves showed that potential-CT-benefit-ANN could stratify prognosis well for all groups and performed significantly better than the eighth AJCC staging system. CONCLUSION The potential-CT-benefit-ANN model developed in this study can accurately predict the potential benefits of adjuvant chemotherapy in patients with stage II/III gastric cancer. The benefit score based on potential-CT-benefit-ANN can predict the long-term prognosis of patients with adjuvant chemotherapy and has good prognostic stratification ability.
Collapse
Affiliation(s)
- Jun Lu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Zhen Xue
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Bin-Bin Xu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Dong Wu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Hua-Long Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Jia-Bin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China.
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
59
|
Granata V, Fusco R, De Muzio F, Cutolo C, Setola SV, Grassi R, Grassi F, Ottaiano A, Nasti G, Tatangelo F, Pilone V, Miele V, Brunese MC, Izzo F, Petrillo A. Radiomics textural features by MR imaging to assess clinical outcomes following liver resection in colorectal liver metastases. Radiol Med 2022; 127:461-470. [PMID: 35347583 DOI: 10.1007/s11547-022-01477-6] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/25/2022] [Indexed: 01/18/2023]
Abstract
PURPOSE To assess the efficacy of radiomics features obtained by T2-weighted sequences to predict clinical outcomes following liver resection in colorectal liver metastases patients. METHODS This retrospective analysis was approved by the local Ethical Committee board and radiological databases were interrogated, from January 2018 to May 2021, to select patients with liver metastases with pathological proof and MRI study in pre-surgical setting. The cohort of patients included a training set and an external validation set. The internal training set included 51 patients with 61 years of median age and 121 liver metastases. The validation cohort consisted a total of 30 patients with single lesion with 60 years of median age. For each volume of interest, 851 radiomics features were extracted as median values using PyRadiomics. Nonparametric test, intraclass correlation, receiver operating characteristic (ROC) analysis, linear regression modelling and pattern recognition methods (support vector machine (SVM), k-nearest neighbours (KNN), artificial neural network (NNET) and decision tree (DT)) were considered. RESULTS The best predictor to discriminate expansive versus infiltrative front of tumour growth was obtained by wavelet_LHL_gldm_DependenceNonUniformityNormalized with an accuracy of 82%; to discriminate high grade versus low grade or absent was the wavelet_LLH_glcm_Imc1 with accuracy of 88%; to differentiate the mucinous type of tumour was the wavelet_LLH_glcm_JointEntropy with accuracy of 92% while to identify tumour recurrence was the wavelet_LLL_glcm_Correlation with accuracy of 85%. Linear regression model increased the performance obtained with respect to the univariate analysis exclusively in the discrimination of expansive versus infiltrative front of tumour growth reaching an accuracy of 90%, a sensitivity of 95% and a specificity of 80%. Considering significant texture metrics tested with pattern recognition approaches, the best performance was reached by the KNN in the discrimination of the tumour budding considering the four textural predictors obtaining an accuracy of 93%, a sensitivity of 81% and a specificity of 97%. CONCLUSIONS Ours results confirmed the capacity of radiomics to identify as biomarkers, several prognostic features that could affect the treatment choice in patients with liver metastases, in order to obtain a more personalized approach.
Collapse
Affiliation(s)
- Vincenza Granata
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli, Naples, Italy.
| | | | - Federica De Muzio
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, 86100, Campobasso, Italy
| | - Carmen Cutolo
- Department of Medicine, Surgery and Dentistry, University of Salerno, Salerno, Italy
| | - Sergio Venanzio Setola
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli, Naples, Italy
| | - Roberta Grassi
- Division of Radiology, Università Degli Studi Della Campania Luigi Vanvitelli, Naples, Italy
| | - Francesca Grassi
- Division of Radiology, Università Degli Studi Della Campania Luigi Vanvitelli, Naples, Italy
| | - Alessandro Ottaiano
- Division of Abdominal Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli, Naples, Italy
| | - Guglielmo Nasti
- Division of Abdominal Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli, Naples, Italy
| | - Fabiana Tatangelo
- Division of Pathology, Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli, Naples, Italy
| | - Vincenzo Pilone
- Department of Medicine, Surgery and Dentistry, University of Salerno, Salerno, Italy
| | - Vittorio Miele
- Division of Radiology, Azienda Ospedaliera Universitaria Careggi, Florence, Italy.,Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, via della Signora 2, 20122, Milan, Italy
| | - Maria Chiara Brunese
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, 86100, Campobasso, Italy
| | - Francesco Izzo
- Division of Epatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli, Naples, Italy
| | - Antonella Petrillo
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli, Naples, Italy
| |
Collapse
|
60
|
Tharmaseelan H, Hertel A, Tollens F, Rink J, Woźnicki P, Haselmann V, Ayx I, Nörenberg D, Schoenberg SO, Froelich MF. Identification of CT Imaging Phenotypes of Colorectal Liver Metastases from Radiomics Signatures-Towards Assessment of Interlesional Tumor Heterogeneity. Cancers (Basel) 2022; 14:cancers14071646. [PMID: 35406418 PMCID: PMC8997087 DOI: 10.3390/cancers14071646] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 02/06/2023] Open
Abstract
(1) Background: Tumoral heterogeneity (TH) is a major challenge in the treatment of metastatic colorectal cancer (mCRC) and is associated with inferior response. Therefore, the identification of TH would be beneficial for treatment planning. TH can be assessed by identifying genetic alterations. In this work, a radiomics-based approach for assessment of TH in colorectal liver metastases (CRLM) in CT scans is demonstrated. (2) Methods: In this retrospective study, CRLM of mCRC were segmented and radiomics features extracted using pyradiomics. Unsupervised k-means clustering was applied to features and lesions. Feature redundancy was evaluated by principal component analysis and reduced by Pearson correlation coefficient cutoff. Feature selection was conducted by LASSO regression and visual analysis of the clusters by radiologists. (3) Results: A total of 47 patients’ (36% female, median age 64) CTs with 261 lesions were included. Five clusters were identified, and the categories small disseminated (n = 31), heterogeneous (n = 105), homogeneous (n = 64), mixed (n = 59), and very large type (n = 2) were assigned based on visual characteristics. Further statistical analysis showed correlation (p < 0.01) of clusters with sex, primary location, T- and N-status, and mutational status. Feature reduction and selection resulted in the identification of four features as a final set for cluster definition. (4) Conclusions: Radiomics features can characterize TH in liver metastases of mCRC in CT scans, and may be suitable for a better pretherapeutic classification of liver lesion phenotypes.
Collapse
Affiliation(s)
- Hishan Tharmaseelan
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
| | - Alexander Hertel
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
| | - Fabian Tollens
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
| | - Johann Rink
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
| | - Piotr Woźnicki
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
| | - Verena Haselmann
- Institute of Clinical Chemistry, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany;
| | - Isabelle Ayx
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
| | - Dominik Nörenberg
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
| | - Stefan O. Schoenberg
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
| | - Matthias F. Froelich
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim of the University of Heidelberg, 68167 Mannheim, Germany; (H.T.); (A.H.); (F.T.); (J.R.); (P.W.); (I.A.); (D.N.); (S.O.S.)
- Correspondence:
| |
Collapse
|
61
|
CT-Based Radiomics Analysis to Predict Histopathological Outcomes Following Liver Resection in Colorectal Liver Metastases. Cancers (Basel) 2022; 14:cancers14071648. [PMID: 35406419 PMCID: PMC8996874 DOI: 10.3390/cancers14071648] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary The objective of the study was to assess the radiomic features obtained by computed tomography (CT) examination as prognostic biomarkers in patients with colorectal liver metastases, in order to predict histopathological outcomes following liver resection. We obtained good performance considering the single significant textural metric in the identification of the front of tumor growth (expansive versus infiltrative) and tumor budding (high grade versus low grade or absent), in the recognition of mucinous type, and in the detection of recurrences. Abstract Purpose: We aimed to assess the efficacy of radiomic features extracted by computed tomography (CT) in predicting histopathological outcomes following liver resection in colorectal liver metastases patients, evaluating recurrence, mutational status, histopathological characteristics (mucinous), and surgical resection margin. Methods: This retrospectively approved study included a training set and an external validation set. The internal training set included 49 patients with a median age of 60 years and 119 liver colorectal metastases. The validation cohort consisted of 28 patients with single liver colorectal metastasis and a median age of 61 years. Radiomic features were extracted using PyRadiomics on CT portal phase. Nonparametric Kruskal–Wallis tests, intraclass correlation, receiver operating characteristic (ROC) analyses, linear regression modeling, and pattern recognition methods (support vector machine (SVM), k-nearest neighbors (KNN), artificial neural network (NNET), and decision tree (DT)) were considered. Results: The median value of intraclass correlation coefficients for the features was 0.92 (range 0.87–0.96). The best performance in discriminating expansive versus infiltrative front of tumor growth was wavelet_HHL_glcm_Imc2, with an accuracy of 79%, a sensitivity of 84%, and a specificity of 67%. The best performance in discriminating expansive versus tumor budding was wavelet_LLL_firstorder_Mean, with an accuracy of 86%, a sensitivity of 91%, and a specificity of 65%. The best performance in differentiating the mucinous type of tumor was original_firstorder_RobustMeanAbsoluteDeviation, with an accuracy of 88%, a sensitivity of 42%, and a specificity of 100%. The best performance in identifying tumor recurrence was the wavelet_HLH_glcm_Idmn, with an accuracy of 85%, a sensitivity of 81%, and a specificity of 88%. The best linear regression model was obtained with the identification of recurrence considering the linear combination of the 16 significant textural metrics (accuracy of 97%, sensitivity of 94%, and specificity of 98%). The best performance for each outcome was reached using KNN as a classifier with an accuracy greater than 86% in the training and validation sets for each classification problem; the best results were obtained with the identification of tumor front growth considering the seven significant textural features (accuracy of 97%, sensitivity of 90%, and specificity of 100%). Conclusions: This study confirmed the capacity of radiomics data to identify several prognostic features that may affect the treatment choice in patients with liver metastases, in order to obtain a more personalized approach.
Collapse
|
62
|
Dercle L, Zhao B, Gönen M, Moskowitz CS, Firas A, Beylergil V, Connors DE, Yang H, Lu L, Fojo T, Carvajal R, Karovic S, Maitland ML, Goldmacher GV, Oxnard GR, Postow MA, Schwartz LH. Early Readout on Overall Survival of Patients With Melanoma Treated With Immunotherapy Using a Novel Imaging Analysis. JAMA Oncol 2022; 8:385-392. [PMID: 35050320 PMCID: PMC8778619 DOI: 10.1001/jamaoncol.2021.6818] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
IMPORTANCE Existing criteria to estimate the benefit of a therapy in patients with cancer rely almost exclusively on tumor size, an approach that was not designed to estimate survival benefit and is challenged by the unique properties of immunotherapy. More accurate prediction of survival by treatment could enhance treatment decisions. OBJECTIVE To validate, using radiomics and machine learning, the performance of a signature of quantitative computed tomography (CT) imaging features for estimating overall survival (OS) in patients with advanced melanoma treated with immunotherapy. DESIGN, SETTING, AND PARTICIPANTS This prognostic study used radiomics and machine learning to retrospectively analyze CT images obtained at baseline and first follow-up and their associated clinical metadata. Data were prospectively collected in the KEYNOTE-002 (Study of Pembrolizumab [MK-3475] Versus Chemotherapy in Participants With Advanced Melanoma; 2017 analysis) and KEYNOTE-006 (Study to Evaluate the Safety and Efficacy of Two Different Dosing Schedules of Pembrolizumab [MK-3475] Compared to Ipilimumab in Participants With Advanced Melanoma; 2016 analysis) multicenter clinical trials. Participants included 575 patients with a diagnosis of advanced melanoma who were randomly assigned to training and validation sets. Data for the present study were collected from November 20, 2012, to June 3, 2019, and analyzed from July 1, 2019, to September 15, 2021. INTERVENTIONS KEYNOTE-002 featured trial groups testing intravenous pembrolizumab, 2 mg/kg or 10 mg/kg every 2 or every 3 weeks based on randomization, or investigator-choice chemotherapy; KEYNOTE-006 featured trial groups testing intravenous ipilimumab, 3 mg/kg every 3 weeks and intravenous pembrolizumab, 10 mg/kg every 2 or 3 weeks based on randomization. MAIN OUTCOMES AND MEASURES The performance of the signature CT imaging features for estimating OS at the month 6 posttreatment landmark in patients who received pembrolizumab was measured using an area under the time-dependent receiver operating characteristics curve (AUC). RESULTS A random forest model combined 25 imaging features extracted from tumors segmented on CT images to identify the combination (signature) that best estimated OS with pembrolizumab in 575 patients. The signature combined 4 imaging features, 2 related to tumor size and 2 reflecting changes in tumor imaging phenotype. In the validation set (287 patients treated with pembrolizumab), the signature reached an AUC for estimation of OS status of 0.92 (95% CI, 0.89-0.95). The standard method, Response Evaluation Criteria in Solid Tumors 1.1, achieved an AUC of 0.80 (95% CI, 0.75-0.84) and classified tumor outcomes as partial or complete response (93 of 287 [32.4%]), stable disease (90 of 287 [31.3%]), or progressive disease (104 of 287 [36.2%]). CONCLUSIONS AND RELEVANCE The findings of this prognostic study suggest that the radiomic signature discerned from conventional CT images at baseline and on first follow-up may be used in clinical settings to provide an accurate early readout of future OS probability in patients with melanoma treated with single-agent programmed cell death 1 blockade.
Collapse
Affiliation(s)
- Laurent Dercle
- Department of Radiology, Columbia University Medical Center, New York, New York,Department of Radiology, New York Presbyterian Hospital, New York, New York
| | - Binsheng Zhao
- Department of Radiology, Columbia University Medical Center, New York, New York,Department of Radiology, New York Presbyterian Hospital, New York, New York
| | - Mithat Gönen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Chaya S. Moskowitz
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ahmed Firas
- Department of Radiology, Columbia University Medical Center, New York, New York,Department of Radiology, New York Presbyterian Hospital, New York, New York
| | - Volkan Beylergil
- Department of Radiology, Columbia University Medical Center, New York, New York,Department of Radiology, New York Presbyterian Hospital, New York, New York
| | - Dana E. Connors
- Foundation for the National Institutes of Health, North Bethesda, Maryland
| | - Hao Yang
- Department of Radiology, Columbia University Medical Center, New York, New York,Department of Radiology, New York Presbyterian Hospital, New York, New York
| | - Lin Lu
- Department of Radiology, Columbia University Medical Center, New York, New York,Department of Radiology, New York Presbyterian Hospital, New York, New York
| | - Tito Fojo
- Columbia University Herbert Irving Comprehensive Cancer Center, New York, New York
| | - Richard Carvajal
- Columbia University Herbert Irving Comprehensive Cancer Center, New York, New York
| | - Sanja Karovic
- Inova Center for Personalized Health and Schar Cancer Institute, Fairfax, Virginia
| | - Michael L. Maitland
- Inova Center for Personalized Health and Schar Cancer Institute, Fairfax, Virginia,University of Virginia Cancer Center, Charlottesville
| | | | - Geoffrey R. Oxnard
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Michael A. Postow
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York,Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Lawrence H. Schwartz
- Department of Radiology, Columbia University Medical Center, New York, New York,Department of Radiology, New York Presbyterian Hospital, New York, New York
| |
Collapse
|
63
|
EOB-MR Based Radiomics Analysis to Assess Clinical Outcomes following Liver Resection in Colorectal Liver Metastases. Cancers (Basel) 2022; 14:cancers14051239. [PMID: 35267544 PMCID: PMC8909637 DOI: 10.3390/cancers14051239] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 02/07/2023] Open
Abstract
Simple Summary The aim of this study was to assess the efficacy of radiomics features obtained by EOB-MRI phase in order to predict clinical outcomes following liver resection in Colorectal Liver Metastases Patients, and evaluate recurrence, mutational status, pathological characteristic (mucinous) and surgical resection margin. Ours results confirmed the capacity of radiomics to identify, as biomarkers, several prognostic features that could affect the treatment choice in patients with liver metastases, in order to obtain a more personalized approach. These results were confirmed by external validation dataset. We obtained a good performance considering the single textural significant metric in the identification of front of tumor growth (expansive versus infiltrative) and tumor budding (high grade versus low grade or absent), in the recognition of mucinous type and in the detection of recurrences. Abstract The aim of this study was to assess the efficacy of radiomics features obtained by EOB-MRI phase in order to predict clinical outcomes following liver resection in Colorectal Liver Metastases Patients, and evaluate recurrence, mutational status, pathological characteristic (mucinous) and surgical resection margin. This retrospective analysis was approved by the local Ethical Committee board of National Cancer of Naples, IRCCS “Fondazione Pascale”. Radiological databases were interrogated from January 2018 to May 2021 in order to select patients with liver metastases with pathological proof and EOB-MRI study in pre-surgical setting. The cohort of patients included a training set (51 patients with 61 years of median age and 121 liver metastases) and an external validation set (30 patients with single lesion with 60 years of median age). For each segmented volume of interest by 2 expert radiologists, 851 radiomics features were extracted as median values using PyRadiomics. non-parametric test, intraclass correlation, receiver operating characteristic (ROC) analysis, linear regression modelling and pattern recognition methods (support vector machine (SVM), k-nearest neighbors (KNN), artificial neural network (NNET), and decision tree (DT)) were considered. The best predictor to discriminate expansive versus infiltrative front of tumor growth was HLH_glcm_MaximumProbability extraxted on VIBE_FA30 with an accuracy of 84%, a sensitivity of 83%, and a specificity of 82%. The best predictor to discriminate tumor budding was Inverse Variance obtained by the original GLCM matrix extraxted on VIBE_FA30 with an accuracy of 89%, a sensitivity of 96% and a specificity of 65%. The best predictor to differentiate the mucinous type of tumor was the HHL_glszm_ZoneVariance extraxted on VIBE_FA30 with an accuracy of 85%, a sensitivity of 46% and a specificity of 95%. The best predictor to identify tumor recurrence was the LHL_glcm_Correlation extraxted on VIBE_FA30 with an accuracy of 86%, a sensitivity of 52% and a specificity of 97%. The best linear regression model was obtained in the identification of the tumor growth front considering the height textural significant metrics by VIBE_FA10 (an accuracy of 89%; sensitivity of 93% and a specificity of 82%). Considering significant texture metrics tested with pattern recognition approaches, the best performance for each outcome was reached by a KNN in the identification of recurrence with the 3 textural significant features extracted by VIBE_FA10 (AUC of 91%, an accuracy of 93%; sensitivity of 99% and a specificity of 77%). Ours results confirmed the capacity of radiomics to identify as biomarkers, several prognostic features that could affect the treatment choice in patients with liver metastases, in order to obtain a more personalized approach.
Collapse
|
64
|
Granata V, Fusco R, De Muzio F, Cutolo C, Setola SV, dell’ Aversana F, Ottaiano A, Avallone A, Nasti G, Grassi F, Pilone V, Miele V, Brunese L, Izzo F, Petrillo A. Contrast MR-Based Radiomics and Machine Learning Analysis to Assess Clinical Outcomes following Liver Resection in Colorectal Liver Metastases: A Preliminary Study. Cancers (Basel) 2022; 14:cancers14051110. [PMID: 35267418 PMCID: PMC8909569 DOI: 10.3390/cancers14051110] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/17/2022] [Accepted: 02/21/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary The objective of the study was to evaluate the radiomics features obtained by contrast MRI studies as prognostic biomarkers in colorectal liver metastases patients to predict clinical outcomes following liver resection. We demonstrated a good performance considering the single textural significant metric in the identification of front of tumor growth (expansive versus infiltrative) and tumor budding (high grade versus low grade or absent), in the recognition of mucinous type and in the detection of recurrences. Moreover, considering linear regression models or neural network classifiers in a multivariate approach was possible to increase the performance in terms of accuracy, sensitivity, and specificity. Abstract Purpose: To assess radiomics features efficacy obtained by arterial and portal MRI phase in the prediction of clinical outcomes in the colorectal liver metastases patients, evaluating recurrence, mutational status, pathological characteristic (mucinous and tumor budding) and surgical resection margin. Methods: This retrospective analysis was approved by the local Ethical Committee board, and radiological databases were used to select patients with colorectal liver metastases with pathological proof and MRI study in a pre-surgical setting after neoadjuvant chemotherapy. The cohort of patients included a training set (51 patients with 61 years of median age and 121 liver metastases) and an external validation set (30 patients with single lesion with 60 years of median age). For each segmented volume of interest on MRI by two expert radiologists, 851 radiomics features were extracted as median values using the PyRadiomics tool. Non-parametric Kruskal-Wallis test, intraclass correlation, receiver operating characteristic (ROC) analysis, linear regression modelling and pattern recognition methods (support vector machine (SVM), k-nearest neighbors (KNN), artificial neural network (NNET), and decision tree (DT)) were considered. Results: The best predictor to discriminate expansive versus infiltrative tumor growth front was wavelet_LHH_glrlm_ShortRunLowGrayLevelEmphasis extracted on portal phase with accuracy of 82%, sensitivity of 84%, and specificity of 77%. The best predictor to discriminate tumor budding was wavelet_LLH_firstorder_10Percentile extracted on portal phase with accuracy of 92%, a sensitivity of 96%, and a specificity of 81%. The best predictor to differentiate the mucinous type of tumor was the wavelet_LLL_glcm_ClusterTendency extracted on portal phase with accuracy of 88%, a sensitivity of 38%, and a specificity of 100%. The best predictor to identify the recurrence was the wavelet_HLH_ngtdm_Complexity extracted on arterial phase with accuracy of 90%, a sensitivity of 71%, and a specificity of 95%. The best linear regression model was obtained in the identification of mucinous type considering the 13 textural significant metrics extracted by arterial phase (accuracy of 94%, sensitivity of 77% and a specificity of 99%). The best results were obtained in the identification of tumor budding with the eleven textural significant features extracted by arterial phase using a KNN (accuracy of 95%, sensitivity of 84%, and a specificity of 99%). Conclusions: Our results confirmed the capacity of radiomics to identify as biomarkers and several prognostic features that could affect the treatment choice in patients with liver metastases in order to obtain a more personalized approach.
Collapse
Affiliation(s)
- Vincenza Granata
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale–IRCCS di Napoli, 80131 Naples, Italy; (S.V.S.); (A.P.)
- Correspondence:
| | - Roberta Fusco
- Medical Oncology Division, Igea SpA, 80013 Napoli, Italy;
| | - Federica De Muzio
- Department of Medicine and Health Sciences V. Tiberio, University of Molise, 86100 Campobasso, Italy; (F.D.M.); (L.B.)
| | - Carmen Cutolo
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84084 Salerno, Italy; (C.C.); (V.P.)
| | - Sergio Venanzio Setola
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale–IRCCS di Napoli, 80131 Naples, Italy; (S.V.S.); (A.P.)
| | - Federica dell’ Aversana
- Division of Radiology, Università Degli Studi Della Campania Luigi Vanvitelli, 80138 Naples, Italy; (F.d.A.); (F.G.)
| | - Alessandro Ottaiano
- Division of Abdominal Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale–IRCCS di Napoli, 80131 Naples, Italy; (A.O.); (A.A.); (G.N.)
| | - Antonio Avallone
- Division of Abdominal Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale–IRCCS di Napoli, 80131 Naples, Italy; (A.O.); (A.A.); (G.N.)
| | - Guglielmo Nasti
- Division of Abdominal Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale–IRCCS di Napoli, 80131 Naples, Italy; (A.O.); (A.A.); (G.N.)
| | - Francesca Grassi
- Division of Radiology, Università Degli Studi Della Campania Luigi Vanvitelli, 80138 Naples, Italy; (F.d.A.); (F.G.)
| | - Vincenzo Pilone
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84084 Salerno, Italy; (C.C.); (V.P.)
| | - Vittorio Miele
- Division of Radiology, Azienda Ospedaliera Universitaria Careggi, 50134 Florence, Italy;
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Via della Signora 2, 20122 Milan, Italy
| | - Luca Brunese
- Department of Medicine and Health Sciences V. Tiberio, University of Molise, 86100 Campobasso, Italy; (F.D.M.); (L.B.)
| | - Francesco Izzo
- Division of Epatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale–IRCCS di Napoli, 80131 Naples, Italy;
| | - Antonella Petrillo
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale–IRCCS di Napoli, 80131 Naples, Italy; (S.V.S.); (A.P.)
| |
Collapse
|
65
|
Viganò L, Jayakody Arachchige VS, Fiz F. Is precision medicine for colorectal liver metastases still a utopia? New perspectives by modern biomarkers, radiomics, and artificial intelligence. World J Gastroenterol 2022; 28:608-623. [PMID: 35317421 PMCID: PMC8900542 DOI: 10.3748/wjg.v28.i6.608] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/29/2021] [Accepted: 01/20/2022] [Indexed: 02/06/2023] Open
Abstract
The management of patients with liver metastases from colorectal cancer is still debated. Several therapeutic options and treatment strategies are available for an extremely heterogeneous clinical scenario. Adequate prediction of patients’ outcomes and of the effectiveness of chemotherapy and loco-regional treatments are crucial to reach a precision medicine approach. This has been an unmet need for a long time, but recent studies have opened new perspectives. New morphological biomarkers have been identified. The dynamic evaluation of the metastases across a time interval, with or without chemotherapy, provided a reliable assessment of the tumor biology. Genetics have been explored and, thanks to their strong association with prognosis, have the potential to drive treatment planning. The liver-tumor interface has been identified as one of the main determinants of tumor progression, and its components, in particular the immune infiltrate, are the focus of major research. Image mining and analyses provided new insights on tumor biology and are expected to have a relevant impact on clinical practice. Artificial intelligence is a further step forward. The present paper depicts the evolution of clinical decision-making for patients affected by colorectal liver metastases, facing modern biomarkers and innovative opportunities that will characterize the evolution of clinical research and practice in the next few years.
Collapse
Affiliation(s)
- Luca Viganò
- Department of Hepatobiliary and General Surgery, IRCCS Humanitas Research Hospital, Rozzano 20089, MI, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele 20072, MI, Italy
| | - Visala S Jayakody Arachchige
- Department of Hepatobiliary and General Surgery, IRCCS Humanitas Research Hospital, Rozzano 20089, MI, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele 20072, MI, Italy
| | - Francesco Fiz
- Nuclear Medicine, IRCCS Humanitas Research Hospital, Rozzano 20089, MI, Italy
| |
Collapse
|
66
|
Rompianesi G, Pegoraro F, Ceresa CDL, Montalti R, Troisi RI. Artificial intelligence in the diagnosis and management of colorectal cancer liver metastases. World J Gastroenterol 2022; 28:108-122. [PMID: 35125822 PMCID: PMC8793013 DOI: 10.3748/wjg.v28.i1.108] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 10/12/2021] [Accepted: 12/25/2021] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common malignancy worldwide, with approximately 50% of patients developing colorectal cancer liver metastasis (CRLM) during the follow-up period. Management of CRLM is best achieved via a multidisciplinary approach and the diagnostic and therapeutic decision-making process is complex. In order to optimize patients' survival and quality of life, there are several unsolved challenges which must be overcome. These primarily include a timely diagnosis and the identification of reliable prognostic factors. Furthermore, to allow optimal treatment options, a precision-medicine, personalized approach is required. The widespread digitalization of healthcare generates a vast amount of data and together with accessible high-performance computing, artificial intelligence (AI) technologies can be applied. By increasing diagnostic accuracy, reducing timings and costs, the application of AI could help mitigate the current shortcomings in CRLM management. In this review we explore the available evidence of the possible role of AI in all phases of the CRLM natural history. Radiomics analysis and convolutional neural networks (CNN) which combine computed tomography (CT) images with clinical data have been developed to predict CRLM development in CRC patients. AI models have also proven themselves to perform similarly or better than expert radiologists in detecting CRLM on CT and magnetic resonance scans or identifying them from the noninvasive analysis of patients' exhaled air. The application of AI and machine learning (ML) in diagnosing CRLM has also been extended to histopathological examination in order to rapidly and accurately identify CRLM tissue and its different histopathological growth patterns. ML and CNN have shown good accuracy in predicting response to chemotherapy, early local tumor progression after ablation treatment, and patient survival after surgical treatment or chemotherapy. Despite the initial enthusiasm and the accumulating evidence, AI technologies' role in healthcare and CRLM management is not yet fully established. Its limitations mainly concern safety and the lack of regulation and ethical considerations. AI is unlikely to fully replace any human role but could be actively integrated to facilitate physicians in their everyday practice. Moving towards a personalized and evidence-based patient approach and management, further larger, prospective and rigorous studies evaluating AI technologies in patients at risk or affected by CRLM are needed.
Collapse
Affiliation(s)
- Gianluca Rompianesi
- Division of Hepato-Bilio-Pancreatic, Minimally Invasive and Robotic Surgery, Department of Clinical Medicine and Surgery, Federico II University Hospital, Naples 80125, Italy
| | - Francesca Pegoraro
- Division of Hepato-Bilio-Pancreatic, Minimally Invasive and Robotic Surgery, Department of Clinical Medicine and Surgery, Federico II University Hospital, Naples 80125, Italy
| | - Carlo DL Ceresa
- Department of Hepato-Pancreato-Biliary Surgery, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9ES, United Kingdom
| | - Roberto Montalti
- Division of Hepato-Bilio-Pancreatic, Minimally Invasive and Robotic Surgery, Department of Public Health, Federico II University Hospital, Naples 80125, Italy
| | - Roberto Ivan Troisi
- Division of Hepato-Bilio-Pancreatic, Minimally Invasive and Robotic Surgery, Department of Clinical Medicine and Surgery, Federico II University Hospital, Naples 80125, Italy
| |
Collapse
|
67
|
Wang Y, Ma LY, Yin XP, Gao BL. Radiomics and Radiogenomics in Evaluation of Colorectal Cancer Liver Metastasis. Front Oncol 2022; 11:689509. [PMID: 35070948 PMCID: PMC8776634 DOI: 10.3389/fonc.2021.689509] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 12/03/2021] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer is one common digestive malignancy, and the most common approach of blood metastasis of colorectal cancer is through the portal vein system to the liver. Early detection and treatment of liver metastasis is the key to improving the prognosis of the patients. Radiomics and radiogenomics use non-invasive methods to evaluate the biological properties of tumors by deeply mining the texture features of images and quantifying the heterogeneity of metastatic tumors. Radiomics and radiogenomics have been applied widely in the detection, treatment, and prognostic evaluation of colorectal cancer liver metastases. Based on the imaging features of the liver, this paper reviews the current application of radiomics and radiogenomics in the diagnosis, treatment, monitor of disease progression, and prognosis of patients with colorectal cancer liver metastases.
Collapse
Affiliation(s)
| | | | - Xiao-Ping Yin
- CT-MRI Room, Affiliated Hospital of Hebei University, Baoding, China
| | | |
Collapse
|
68
|
Giannini V, Pusceddu L, Defeudis A, Nicoletti G, Cappello G, Mazzetti S, Sartore-Bianchi A, Siena S, Vanzulli A, Rizzetto F, Fenocchio E, Lazzari L, Bardelli A, Marsoni S, Regge D. Delta-Radiomics Predicts Response to First-Line Oxaliplatin-Based Chemotherapy in Colorectal Cancer Patients with Liver Metastases. Cancers (Basel) 2022; 14:cancers14010241. [PMID: 35008405 PMCID: PMC8750408 DOI: 10.3390/cancers14010241] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 12/29/2021] [Accepted: 12/30/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Oxaliplatin-based chemotherapy remains the mainstay of first-line therapy in patients with metastatic colorectal cancer (mCRC). Unfortunately, only approximately 60% of treated patients achieve response, and half of responders will experience an early onset of disease progression. Furthermore, some individuals will develop a mixed response due to the emergence of resistant tumor subclones. The ability to predicting which patients will acquire resistance could help them avoid the unnecessary toxicity of oxaliplatin therapies. Furthermore, sorting out lesions that do not respond, in the context of an overall good response, could trigger further investigation into their mutational landscape, providing mechanistic insight towards the planning of a more comprehensive treatment. In this study, we validated a delta-radiomics signature capable of predicting response to oxaliplatin-based first-line treatment of individual liver colorectal cancer metastases. Findings could pave the way to a more personalized treatment of patients with mCRC. Abstract The purpose of this paper is to develop and validate a delta-radiomics score to predict the response of individual colorectal cancer liver metastases (lmCRC) to first-line FOLFOX chemotherapy. Three hundred one lmCRC were manually segmented on both CT performed at baseline and after the first cycle of first-line FOLFOX, and 107 radiomics features were computed by subtracting textural features of CT at baseline from those at timepoint 1 (TP1). LmCRC were classified as nonresponders (R−) if they showed progression of disease (PD), according to RECIST1.1, before 8 months, and as responders (R+), otherwise. After feature selection, we developed a decision tree statistical model trained using all lmCRC coming from one hospital. The final output was a delta-radiomics signature subsequently validated on an external dataset. Sensitivity, specificity, positive (PPV), and negative (NPV) predictive values in correctly classifying individual lesions were assessed on both datasets. Per-lesion sensitivity, specificity, PPV, and NPV were 99%, 94%, 95%, 99%, 85%, 92%, 90%, and 87%, respectively, in the training and validation datasets. The delta-radiomics signature was able to reliably predict R− lmCRC, which were wrongly classified by lesion RECIST as R+ at TP1, (93%, averaging training and validation set, versus 67% of RECIST). The delta-radiomics signature developed in this study can reliably predict the response of individual lmCRC to oxaliplatin-based chemotherapy. Lesions forecasted as poor or nonresponders by the signature could be further investigated, potentially paving the way to lesion-specific therapies.
Collapse
Affiliation(s)
- Valentina Giannini
- Department of Surgical Sciences, University of Turin, 10124 Turin, Italy; (A.D.); (G.N.); (S.M.); (D.R.)
- Radiology Unit, Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy; (L.P.); (G.C.)
- Correspondence:
| | - Laura Pusceddu
- Radiology Unit, Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy; (L.P.); (G.C.)
| | - Arianna Defeudis
- Department of Surgical Sciences, University of Turin, 10124 Turin, Italy; (A.D.); (G.N.); (S.M.); (D.R.)
- Radiology Unit, Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy; (L.P.); (G.C.)
| | - Giulia Nicoletti
- Department of Surgical Sciences, University of Turin, 10124 Turin, Italy; (A.D.); (G.N.); (S.M.); (D.R.)
| | - Giovanni Cappello
- Radiology Unit, Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy; (L.P.); (G.C.)
| | - Simone Mazzetti
- Department of Surgical Sciences, University of Turin, 10124 Turin, Italy; (A.D.); (G.N.); (S.M.); (D.R.)
- Radiology Unit, Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy; (L.P.); (G.C.)
| | - Andrea Sartore-Bianchi
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, 20122 Milan, Italy; (A.S.-B.); (S.S.); (A.V.)
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy
| | - Salvatore Siena
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, 20122 Milan, Italy; (A.S.-B.); (S.S.); (A.V.)
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy
| | - Angelo Vanzulli
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, 20122 Milan, Italy; (A.S.-B.); (S.S.); (A.V.)
- Department of Radiology, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy;
| | - Francesco Rizzetto
- Department of Radiology, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy;
| | - Elisabetta Fenocchio
- Multidisciplinary Outpatient Oncology Clinic, Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy;
| | - Luca Lazzari
- Precision Oncology, IFOM-The FIRC Institute of Molecular Oncology, 20139 Milan, Italy; (L.L.); (S.M.)
| | - Alberto Bardelli
- Candiolo Cancer Institute-FPO, IRCCS, 10060 Candiolo, Italy;
- Department of Oncology, University of Torino, 10060 Candiolo, Italy
| | - Silvia Marsoni
- Precision Oncology, IFOM-The FIRC Institute of Molecular Oncology, 20139 Milan, Italy; (L.L.); (S.M.)
| | - Daniele Regge
- Department of Surgical Sciences, University of Turin, 10124 Turin, Italy; (A.D.); (G.N.); (S.M.); (D.R.)
- Radiology Unit, Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy; (L.P.); (G.C.)
| |
Collapse
|
69
|
Dercle L, Zhao B, Gönen M, Moskowitz CS, Connors DE, Yang H, Lu L, Reidy-Lagunes D, Fojo T, Karovic S, Maitland ML, Oxnard GR, Schwartz LH. An imaging signature to predict outcome in metastatic colorectal cancer using routine computed tomography scans. Eur J Cancer 2022; 161:138-147. [PMID: 34916122 PMCID: PMC10018811 DOI: 10.1016/j.ejca.2021.10.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/10/2021] [Accepted: 10/24/2021] [Indexed: 01/25/2023]
Abstract
BACKGROUND & AIMS Quantitative analysis of computed tomography (CT) scans of patients with metastatic colorectal cancer (mCRC) can identify imaging signatures that predict overall survival (OS). METHODS We retrospectively analysed CT images from 1584 mCRC patients on two phase III trials evaluating FOLFOX ± panitumumab (n = 331, 350) and FOLFIRI ± aflibercept (n = 437, 466). In the training set (n = 720), an algorithm was trained to predict OS landmarked from month 2; the output was a signature value on a scale from 0 to 1 (most to least favourable predicted OS). In the validation set (n = 864), hazard ratios (HRs) evaluated the association of the signature with OS using RECIST1.1 as a benchmark of comparison. RESULTS In the training set, the selected signature combined three features - change in tumour volume, change in tumour spatial heterogeneity, and tumour volume - to predict OS. In the validation set, RECIST1.1 classified patients in three categories: response (n = 166, 19.2%), stable disease (n = 636, 73.6%), and progression (n = 62, 7.2%). The HR was 3.93 (2.79-5.54). Using the same distribution for the signature, the HR was 21.04 (14.88-30.58), showing an incremental prognostic separation. Stable disease by RECIST1.1 was reclassified by the signature along a continuum where patients belonging to the most and least favourable signature quartiles had a median OS of 40.73 (28.49 to NA) months (n = 94) and 7.03 (5.66-7.89) months (n = 166), respectively. CONCLUSIONS A signature combining three imaging features provides early prognostic information that can improve treatment decisions for individual patients and clinical trial analyses.
Collapse
Affiliation(s)
- Laurent Dercle
- Department of Radiology, Columbia University Medical Center and New York Presbyterian Hospital, 710 West 168th St., New York, NY 10032, USA.
| | - Binsheng Zhao
- Department of Radiology, Columbia University Medical Center and New York Presbyterian Hospital, 710 West 168th St., New York, NY 10032, USA
| | - Mithat Gönen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Chaya S Moskowitz
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Dana E Connors
- Foundation for the National Institutes of Health (FNIH), 11400 Rockville Pike, Suite 600, North Bethesda, MD 20852, USA
| | - Hao Yang
- Department of Radiology, Columbia University Medical Center and New York Presbyterian Hospital, 710 West 168th St., New York, NY 10032, USA
| | - Lin Lu
- Department of Radiology, Columbia University Medical Center and New York Presbyterian Hospital, 710 West 168th St., New York, NY 10032, USA
| | - Diane Reidy-Lagunes
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Tito Fojo
- Columbia University Herbert Irving Comprehensive Cancer Center, 161 Fort Washington Ave., New York, NY 10032, USA
| | - Sanja Karovic
- Inova Center for Personalized Health and Schar Cancer Institute, 8100 Innovation Park Dr, Fairfax, VA 22031, USA
| | - Michael L Maitland
- Inova Center for Personalized Health and Schar Cancer Institute, 8100 Innovation Park Dr, Fairfax, VA 22031, USA; University of Virginia Cancer Center, 1240 Lee St., Charlottesville, VA 22903, USA
| | - Geoffrey R Oxnard
- Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave., Boston, MA 02215, USA
| | - Lawrence H Schwartz
- Department of Radiology, Columbia University Medical Center and New York Presbyterian Hospital, 710 West 168th St., New York, NY 10032, USA
| |
Collapse
|
70
|
Lu L, Dercle L, Zhao B, Schwartz LH. Deep learning for the prediction of early on-treatment response in metastatic colorectal cancer from serial medical imaging. Nat Commun 2021; 12:6654. [PMID: 34789774 PMCID: PMC8599694 DOI: 10.1038/s41467-021-26990-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 10/21/2021] [Indexed: 12/23/2022] Open
Abstract
In current clinical practice, tumor response assessment is usually based on tumor size change on serial computerized tomography (CT) scan images. However, evaluation of tumor response to anti-vascular endothelial growth factor therapies in metastatic colorectal cancer (mCRC) is limited because morphological change in tumor may occur earlier than tumor size change. Here we present an analysis utilizing a deep learning (DL) network to characterize tumor morphological change for response assessment in mCRC patients. We retrospectively analyzed 1,028 mCRC patients who were prospectively included in the VELOUR trial (NCT00561470). We found that DL network was able to predict early on-treatment response in mCRC and showed better performance than its size-based counterpart with C-Index: 0.649 (95% CI: 0.619,0.679) vs. 0.627 (95% CI: 0.567,0.638), p = 0.009, z-test. The integration of DL network with size-based methodology could further improve the prediction performance to C-Index: 0.694 (95% CI: 0.661,0.720), which was superior to size/DL-based-only models (all p < 0.001, z-test). Our study suggests that DL network could provide a noninvasive mean for quantitative and comprehensive characterization of tumor morphological change, which may potentially benefit personalized early on-treatment decision making.
Collapse
Affiliation(s)
- Lin Lu
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Laurent Dercle
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Binsheng Zhao
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Lawrence H Schwartz
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
71
|
de la Pinta C, Castillo ME, Collado M, Galindo-Pumariño C, Peña C. Radiogenomics: Hunting Down Liver Metastasis in Colorectal Cancer Patients. Cancers (Basel) 2021; 13:5547. [PMID: 34771709 PMCID: PMC8582778 DOI: 10.3390/cancers13215547] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 02/07/2023] Open
Abstract
Radiomics is a developing new discipline that analyzes conventional medical images to extract quantifiable data that can be mined for new biomarkers that show the biology of pathological processes at microscopic levels. These data can be converted into image-based signatures to improve diagnostic, prognostic and predictive accuracy in cancer patients. The combination of radiomics and molecular data, called radiogenomics, has clear implications for cancer patients' management. Though some studies have focused on radiogenomics signatures in hepatocellular carcinoma patients, only a few have examined colorectal cancer metastatic lesions in the liver. Moreover, the need to differentiate between liver lesions is fundamental for accurate diagnosis and treatment. In this review, we summarize the knowledge gained from radiomics and radiogenomics studies in hepatic metastatic colorectal cancer patients and their use in early diagnosis, response assessment and treatment decisions. We also investigate their value as possible prognostic biomarkers. In addition, the great potential of image mining to provide a comprehensive view of liver niche formation is examined thoroughly. Finally, new challenges and current limitations for the early detection of the liver premetastatic niche, based on radiomics and radiogenomics, are also discussed.
Collapse
Affiliation(s)
- Carolina de la Pinta
- Radiation Oncology Department, Ramón y Cajal University Hospital, IRYCIS, Alcalá University, 28034 Madrid, Spain
| | - María E. Castillo
- Medical Oncology Department, Ramón y Cajal University Hospital, IRYCIS, Alcalá University, 28034 Madrid, Spain; (M.E.C.); (M.C.); (C.G.-P.)
- Centro de Investigación Biomédica en Red de Cancer (CIBERONC), 28029 Madrid, Spain
| | - Manuel Collado
- Medical Oncology Department, Ramón y Cajal University Hospital, IRYCIS, Alcalá University, 28034 Madrid, Spain; (M.E.C.); (M.C.); (C.G.-P.)
| | - Cristina Galindo-Pumariño
- Medical Oncology Department, Ramón y Cajal University Hospital, IRYCIS, Alcalá University, 28034 Madrid, Spain; (M.E.C.); (M.C.); (C.G.-P.)
- Centro de Investigación Biomédica en Red de Cancer (CIBERONC), 28029 Madrid, Spain
| | - Cristina Peña
- Medical Oncology Department, Ramón y Cajal University Hospital, IRYCIS, Alcalá University, 28034 Madrid, Spain; (M.E.C.); (M.C.); (C.G.-P.)
- Centro de Investigación Biomédica en Red de Cancer (CIBERONC), 28029 Madrid, Spain
| |
Collapse
|
72
|
Machine Learning-Based Radiomics of the Optic Chiasm Predict Visual Outcome Following Pituitary Adenoma Surgery. J Pers Med 2021; 11:jpm11100991. [PMID: 34683132 PMCID: PMC8541242 DOI: 10.3390/jpm11100991] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/25/2021] [Accepted: 09/27/2021] [Indexed: 02/05/2023] Open
Abstract
Preoperative prediction of visual recovery after pituitary adenoma surgery remains a challenge. We aimed to investigate the value of MRI-based radiomics of the optic chiasm in predicting postoperative visual field outcome using machine learning technology. A total of 131 pituitary adenoma patients were retrospectively enrolled and divided into the recovery group (N = 79) and the non-recovery group (N = 52) according to visual field outcome following surgical chiasmal decompression. Radiomic features were extracted from the optic chiasm on preoperative coronal T2-weighted imaging. Least absolute shrinkage and selection operator regression were first used to select optimal features. Then, three machine learning algorithms were employed to develop radiomic models to predict visual recovery, including support vector machine (SVM), random forest and linear discriminant analysis. The prognostic performances of models were evaluated via five-fold cross-validation. The results showed that radiomic models using different machine learning algorithms all achieved area under the curve (AUC) over 0.750. The SVM-based model represented the best predictive performance for visual field recovery, with the highest AUC of 0.824. In conclusion, machine learning-based radiomics of the optic chiasm on routine MR imaging could potentially serve as a novel approach to preoperatively predict visual recovery and allow personalized counseling for individual pituitary adenoma patients.
Collapse
|
73
|
Chang YC, Chan MH, Li CH, Fang CY, Hsiao M, Chen CL. Exosomal Components and Modulators in Colorectal Cancer: Novel Diagnosis and Prognosis Biomarkers. Biomedicines 2021; 9:biomedicines9080931. [PMID: 34440135 PMCID: PMC8391321 DOI: 10.3390/biomedicines9080931] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 07/23/2021] [Accepted: 07/29/2021] [Indexed: 02/07/2023] Open
Abstract
The relatively high incidence and mortality rates for colorectal carcinoma (CRC) make it a formidable malignant tumor. Comprehensive strategies have been applied to predict patient survival and diagnosis. Various clinical regimens have also been developed to improve the therapeutic outcome. Extracellular vesicles (EVs) are recently proposed cellular structures that can be produced by natural or artificial methods and have been extensively studied. In addition to their innate functions, EVs can be manipulated to be drug carriers and exert many biological functions. The composition of EVs, their intravesicular components, and the surrounding tumor microenvironment are closely related to the development of colorectal cancer. Determining the expression profiles of exocytosis samples and using them as indicators for selecting effective combination therapy is an indispensable direction for EV study and should be regarded as a novel prediction platform in addition to cancer stage, prognosis, and other clinical assessments. In this review, we summarize the function, regulation, and application of EVs in the colon cancer research field. We provide an update on and discuss potential values for clinical applications of EVs. Moreover, we illustrate the specific markers, mediators, and genetic alterations of EVs in colorectal carcinogenesis. Furthermore, we outline the vital markers present in the EVs and discuss their plausible uses in colon cancer patient therapy in combination with the currently used clinical strategies. The development and application of these EVs will significantly improve the accuracy of diagnosis, lead to more precise prognoses, and may lead to the improved treatment of colorectal cancer.
Collapse
Affiliation(s)
- Yu-Chan Chang
- Department of Biomedical Imaging and Radiological Science, National Yang-Ming University, Taipei 112, Taiwan;
- Department of Biomedical Imaging and Radiological Science, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Ming-Hsien Chan
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan; (M.-H.C.); (C.-H.L.)
| | - Chien-Hsiu Li
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan; (M.-H.C.); (C.-H.L.)
| | - Chih-Yeu Fang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 350, Taiwan;
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan; (M.-H.C.); (C.-H.L.)
- Department of Biochemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Correspondence: (M.H.); (C.-L.C.); Tel.: +886-2-2787-1243 (M.H.); +886-2-2736-1661 (ext. 3139) (C.-L.C.); Fax: +886-2-2789-9931 (M.H.)
| | - Chi-Long Chen
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Department of Pathology, Taipei Medical University Hospital, Taipei 110, Taiwan
- Correspondence: (M.H.); (C.-L.C.); Tel.: +886-2-2787-1243 (M.H.); +886-2-2736-1661 (ext. 3139) (C.-L.C.); Fax: +886-2-2789-9931 (M.H.)
| |
Collapse
|
74
|
Furtado FS, Suarez-Weiss KE, Vangel M, Clark JW, Cusack JC, Hong T, Blaszkowsky L, Wo J, Striar R, Umutlu L, Daldrup-Link HE, Groshar D, Rocco R, Bordeianou L, Anderson MA, Mojtahed A, Qadan M, Ferrone C, Catalano OA. Clinical impact of PET/MRI in oligometastatic colorectal cancer. Br J Cancer 2021; 125:975-982. [PMID: 34282295 DOI: 10.1038/s41416-021-01494-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/08/2021] [Accepted: 07/08/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Oligometastatic colorectal cancer (CRC) is potentially curable and demands individualised strategies. METHODS This single-centre retrospective study investigated if positron emission tomography (PET)/magnetic resonance imaging (MR) had a clinical impact on oligometastatic CRC relative to the standard of care imaging (SCI). Adult patients with oligometastatic CRC on SCI who also underwent PET/MR between 3/2016 and 3/2019 were included. The exclusion criterion was lack of confirmatory standard of reference, either surgical pathology, intraoperative gross confirmation or imaging follow-up. SCI consisted of contrast-enhanced (CE) computed tomography (CT) of the chest/abdomen/pelvis, abdominal/pelvic CE-MR, and/or CE whole-body PET/CT with diagnostic quality (i.e. standard radiation dose) CT. Follow-up was evaluated until 3/2020. RESULTS Thirty-one patients constituted the cohort, 16 (52%) male, median patient age was 53 years (interquartile range: 49-65 years). PET/MR and SCI results were divergent in 19% (95% CI 9-37%) of the cases, with PET/MR leading to management changes in all of them. The diagnostic accuracy of PET/MR was 90 ± 5%, versus 71 ± 8% for SCI. In a pairwise analysis, PET/MR outperformed SCI when compared to the reference standard (p = 0.0412). CONCLUSIONS These findings suggest the potential usefulness of PET/MR in the management of oligometastatic CRC.
Collapse
Affiliation(s)
- Felipe S Furtado
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Martinos Center for Biomedical Imaging, Harvard Medical School, Boston, MA, USA
| | | | - Mark Vangel
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Martinos Center for Biomedical Imaging, Harvard Medical School, Boston, MA, USA.,Biostatistics Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jeffrey W Clark
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - James C Cusack
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Theodore Hong
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lawrence Blaszkowsky
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Newton-Wellesley Hospital, Newton, MA, USA
| | - Jennifer Wo
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Robin Striar
- Martinos Center for Biomedical Imaging, Harvard Medical School, Boston, MA, USA
| | | | | | - David Groshar
- Assuta Medical Centers, Tel Aviv, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ricciardi Rocco
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Mark A Anderson
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Motaz Qadan
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Cristina Ferrone
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Onofrio A Catalano
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA. .,Martinos Center for Biomedical Imaging, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
75
|
Pei Q, Yi X, Chen C, Pang P, Fu Y, Lei G, Chen C, Tan F, Gong G, Li Q, Zai H, Chen BT. Pre-treatment CT-based radiomics nomogram for predicting microsatellite instability status in colorectal cancer. Eur Radiol 2021; 32:714-724. [PMID: 34258636 DOI: 10.1007/s00330-021-08167-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 05/23/2021] [Accepted: 06/08/2021] [Indexed: 12/19/2022]
Abstract
OBJECTIVES Stratification of microsatellite instability (MSI) status in patients with colorectal cancer (CRC) improves clinical decision-making for cancer treatment. The present study aimed to develop a radiomics nomogram to predict the pre-treatment MSI status in patients with CRC. METHODS A total of 762 patients with CRC confirmed by surgical pathology and MSI status determined with polymerase chain reaction (PCR) method were retrospectively recruited between January 2013 and May 2019. Radiomics features were extracted from routine pre-treatment abdominal pelvic computed tomography (CT) scans acquired as part of the patients' clinical care. A radiomics nomogram was constructed using multivariate logistic regression. The performance of the nomogram was evaluated using discrimination, calibration, and decision curves. RESULTS The radiomics nomogram incorporating radiomics signatures, tumor location, patient age, high-density lipoprotein expression, and platelet counts showed good discrimination between patients with non-MSI-H and MSI-H, with an area under the curve (AUC) of 0.74 [95% CI, 0.68-0.80] in the training cohort and 0.77 [95% CI, 0.68-0.85] in the validation cohort. Favorable clinical application was observed using decision curve analysis. The addition of pathological characteristics to the nomogram failed to show incremental prognostic value. CONCLUSIONS We developed a radiomics nomogram incorporating radiomics signatures and clinical indicators, which could potentially be used to facilitate the individualized prediction of MSI status in patients with CRC. KEY POINTS • There is an unmet need to non-invasively determine MSI status prior to treatment. However, the traditional radiological evaluation of CT is limited for evaluating MSI status. • Our non-invasive CT imaging-based radiomics method could efficiently distinguish patients with high MSI disease from those with low MSI disease. • Our radiomics approach demonstrated promising diagnostic efficiency for MSI status, similar to the commonly used IHC method.
Collapse
Affiliation(s)
- Qian Pei
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Xiaoping Yi
- Department of Radiology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, 410008, People's Republic of China. .,National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Central South University, Changsha, 410008, People's Republic of China.
| | - Chen Chen
- Department of Radiology, 331 Hospital of Zhuzhou City, Zhuzhou, People's Republic of China
| | - Peipei Pang
- GE Healthcare, Hangzhou, 310000, People's Republic of China
| | - Yan Fu
- Department of Radiology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, 410008, People's Republic of China.,National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Central South University, Changsha, 410008, People's Republic of China
| | - Guangwu Lei
- Department of Radiology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, 410008, People's Republic of China
| | - Changyong Chen
- Department of Radiology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, 410008, People's Republic of China
| | - Fengbo Tan
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Guanghui Gong
- Department of Pathology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, 410008, People's Republic of China
| | - Qingling Li
- Department of Pathology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, 410008, People's Republic of China.
| | - Hongyan Zai
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Bihong T Chen
- Department of Diagnostic Radiology, City of Hope National Medical Center, Duarte, CA, USA
| |
Collapse
|
76
|
Mbugua SN, Njenga LW, Odhiambo RA, Wandiga SO, Onani MO. Beyond DNA-targeting in Cancer Chemotherapy. Emerging Frontiers - A Review. Curr Top Med Chem 2021; 21:28-47. [PMID: 32814532 DOI: 10.2174/1568026620666200819160213] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 12/14/2022]
Abstract
Modern anti-cancer drugs target DNA specifically for rapid division of malignant cells. One downside of this approach is that they also target other rapidly dividing healthy cells, such as those involved in hair growth leading to serious toxic side effects and hair loss. Therefore, it would be better to develop novel agents that address cellular signaling mechanisms unique to cancerous cells, and new research is now focussing on such approaches. Although the classical chemotherapy area involving DNA as the set target continues to produce important findings, nevertheless, a distinctly discernible emerging trend is the divergence from the cisplatin operation model that uses the metal as the primary active center of the drug. Many successful anti-cancer drugs present are associated with elevated toxicity levels. Cancers also develop immunity against most therapies and the area of cancer research can, therefore, be seen as an area with a high unaddressed need. Hence, ongoing work into cancer pathogenesis is important to create accurate preclinical tests that can contribute to the development of innovative drugs to manage and treat cancer. Some of the emergent frontiers utilizing different approaches include nanoparticles delivery, use of quantum dots, metal complexes, tumor ablation, magnetic hypothermia and hyperthermia by use of Superparamagnetic Iron oxide Nanostructures, pathomics and radiomics, laser surgery and exosomes. This review summarizes these new approaches in good detail, giving critical views with necessary comparisons. It also delves into what they carry for the future, including their advantages and disadvantages.
Collapse
Affiliation(s)
- Simon N Mbugua
- Department of Chemistry, University of Nairobi, P.O. Box 30197-00100, Nairobi, Kenya
| | - Lydia W Njenga
- Department of Chemistry, University of Nairobi, P.O. Box 30197-00100, Nairobi, Kenya
| | - Ruth A Odhiambo
- Department of Chemistry, University of Nairobi, P.O. Box 30197-00100, Nairobi, Kenya
| | - Shem O Wandiga
- Department of Chemistry, University of Nairobi, P.O. Box 30197-00100, Nairobi, Kenya
| | - Martin O Onani
- Organometallics and Nanomaterials, Department of Chemistry, University of the Western Cape, Private Bag X17, Bellville, 7535, South Africa
| |
Collapse
|
77
|
Fiz F, Costa G, Gennaro N, la Bella L, Boichuk A, Sollini M, Politi LS, Balzarini L, Torzilli G, Chiti A, Viganò L. Contrast Administration Impacts CT-Based Radiomics of Colorectal Liver Metastases and Non-Tumoral Liver Parenchyma Revealing the "Radiological" Tumour Microenvironment. Diagnostics (Basel) 2021; 11:diagnostics11071162. [PMID: 34202253 PMCID: PMC8305553 DOI: 10.3390/diagnostics11071162] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/11/2021] [Accepted: 06/22/2021] [Indexed: 12/29/2022] Open
Abstract
The impact of the contrast medium on the radiomic textural features (TF) extracted from the CT scan is unclear. We investigated the modification of TFs of colorectal liver metastases (CLM), peritumoral tissue, and liver parenchyma. One hundred and sixty-two patients with 409 CLMs undergoing resection (2017–2020) into a single institution were considered. We analyzed the following volumes of interest (VOIs): The CLM (Tumor-VOI); a 5-mm parenchyma rim around the CLM (Margin-VOI); and a 2-mL sample of parenchyma distant from CLM (Liver-VOI). Forty-five TFs were extracted from each VOI (LIFEx®®). Contrast enhancement affected most TFs of the Tumor-VOI (71%) and Margin-VOI (62%), and part of those of the Liver-VOI (44%, p = 0.010). After contrast administration, entropy increased and energy decreased in the Tumor-VOI (0.93 ± 0.10 vs. 0.85 ± 0.14 in pre-contrast; 0.14 ± 0.03 vs. 0.18 ± 0.04, p < 0.001) and Margin-VOI (0.89 ± 0.11 vs. 0.85 ± 0.12; 0.16 ± 0.04 vs. 0.18 ± 0.04, p < 0.001), while remaining stable in the Liver-VOI. Comparing the VOIs, pre-contrast Tumor and Margin-VOI had similar entropy and energy (0.85/0.18 for both), while Liver-VOI had lower values (0.76/0.21, p < 0.001). In the portal phase, a gradient was observed (entropy: Tumor > Margin > Liver; energy: Tumor < Margin < Liver, p < 0.001). Contrast enhancement affected TFs of CLM, while it did not modify entropy and energy of parenchyma. TFs of the peritumoral tissue had modifications similar to the Tumor-VOI despite its radiological aspect being equal to non-tumoral parenchyma.
Collapse
Affiliation(s)
- Francesco Fiz
- Nuclear Medicine Unit, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (M.S.); (A.C.)
- Correspondence: (F.F.); (L.V.); Tel.: +39-02-8224-7361 (L.V.)
| | - Guido Costa
- Division of Hepatobiliary and General Surgery, Department of Surgery, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (G.C.); (G.T.)
| | - Nicolò Gennaro
- Department of Diagnostic Imaging, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (N.G.); (L.S.P.); (L.B.)
| | - Ludovico la Bella
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20089 Milan, Italy; (L.l.B.); (A.B.)
| | - Alexandra Boichuk
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20089 Milan, Italy; (L.l.B.); (A.B.)
| | - Martina Sollini
- Nuclear Medicine Unit, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (M.S.); (A.C.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20089 Milan, Italy; (L.l.B.); (A.B.)
| | - Letterio S. Politi
- Department of Diagnostic Imaging, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (N.G.); (L.S.P.); (L.B.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20089 Milan, Italy; (L.l.B.); (A.B.)
| | - Luca Balzarini
- Department of Diagnostic Imaging, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (N.G.); (L.S.P.); (L.B.)
| | - Guido Torzilli
- Division of Hepatobiliary and General Surgery, Department of Surgery, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (G.C.); (G.T.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20089 Milan, Italy; (L.l.B.); (A.B.)
| | - Arturo Chiti
- Nuclear Medicine Unit, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (M.S.); (A.C.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20089 Milan, Italy; (L.l.B.); (A.B.)
| | - Luca Viganò
- Division of Hepatobiliary and General Surgery, Department of Surgery, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (G.C.); (G.T.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20089 Milan, Italy; (L.l.B.); (A.B.)
- Correspondence: (F.F.); (L.V.); Tel.: +39-02-8224-7361 (L.V.)
| |
Collapse
|
78
|
Prediction of recurrence after surgery in colorectal cancer patients using radiomics from diagnostic contrast-enhanced computed tomography: a two-center study. Eur Radiol 2021; 32:405-414. [PMID: 34170367 DOI: 10.1007/s00330-021-08104-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/11/2021] [Accepted: 05/27/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVES To assess the value of contrast-enhanced (CE) diagnostic CT scans characterized through radiomics as predictors of recurrence for patients with stage II and III colorectal cancer in a two-center context. MATERIALS AND METHODS This study included 193 patients diagnosed with stage II and III colorectal adenocarcinoma from 1 July 2008 to 15 March 2017 in two different French University Hospitals. To compensate for the variability in two-center data, a statistical harmonization method Bootstrapped ComBat (B-ComBat) was used. Models predicting disease-free survival (DFS) were built using 3 different machine learning (ML): (1) multivariate regression (MR) with 10-fold cross-validation after feature selection based on least absolute shrinkage and selection operator (LASSO), (2) random forest (RF), and (3) support vector machine (SVM), both with embedded feature selection. RESULTS The performance for both balanced and 95% sensitivity models was systematically higher after our proposed B-ComBat harmonization compared to the use of the original untransformed data. The most clinically relevant performance was achieved by the multivariate regression model combining a clinical variable (postoperative chemotherapy) with two radiomics shape descriptors (compactness and least axis length) with a BAcc of 0.78 and an MCC of 0.6 associated with a required sensitivity of 95%. The resulting stratification in terms of DFS was significant (p = 0.00021), especially compared to the use of unharmonized original data (p = 0.17). CONCLUSIONS Radiomics models derived from contrast-enhanced CT could be trained and validated in a two-center cohort with a good predictive performance of recurrence in stage II et III colorectal cancer patients. KEY POINTS • Adjuvant therapy decision in colorectal cancer can be a challenge in medical oncology. • Radiomics models, derived from diagnostic CT, trained and validated in a two-center cohort, could predict recurrence in stage II and III colorectal cancer patients. • Identifying patients with a low risk of recurrence, these models could facilitate treatment optimization and avoid unnecessary treatment.
Collapse
|
79
|
Osman MH, Mohamed RH, Sarhan HM, Park EJ, Baik SH, Lee KY, Kang J. Machine Learning Model for Predicting Postoperative Survival of Patients with Colorectal Cancer. Cancer Res Treat 2021; 54:517-524. [PMID: 34126702 PMCID: PMC9016295 DOI: 10.4143/crt.2021.206] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 06/13/2021] [Indexed: 11/24/2022] Open
Abstract
Purpose Machine learning (ML) is a strong candidate for making accurate predictions, as we can use large amount of data with powerful computational algorithms. We developed a ML based model to predict survival of patients with colorectal cancer (CRC) using data from two independent datasets. Materials and Methods A total of 364,316 and 1,572 CRC patients were included from the Surveillance, Epidemiology, and End Results (SEER) and a Korean dataset, respectively. As SEER combines data from 18 cancer registries, internal validation was done using 18-Fold-Cross-Validation then external validation was performed by testing the trained model on the Korean dataset. Performance was evaluated using area under the receiver operating characteristic curve (AUROC), sensitivity and positive predictive values. Results Clinicopathological characteristics were significantly different between the two datasets and the SEER showed a significant lower 5-year survival rate compared to the Korean dataset (60.1% vs. 75.3%, p < 0.001). The ML-based model using the Light gradient boosting algorithm achieved a better performance in predicting 5-year-survival compared to American Joint Committee on Cancer stage (AUROC, 0.804 vs. 0.736; p < 0.001). The most important features which influenced model performance were age, number of examined lymph nodes, and tumor size. Sensitivity and positive predictive values of predicting 5-year-survival for classes including dead or alive were reported as 68.14%, 77.51% and 49.88%, 88.1% respectively in the validation set. Survival probability can be checked using the web-based survival predictor (http://colorectalcancer.pythonanywhere.com). Conclusion ML-based model achieved a much better performance compared to staging in individualized estimation of survival of patients with CRC.
Collapse
Affiliation(s)
| | | | | | - Eun Jung Park
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Seung Hyuk Baik
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Kang Young Lee
- Department of Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jeonghyun Kang
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
80
|
Granata V, Fusco R, Barretta ML, Picone C, Avallone A, Belli A, Patrone R, Ferrante M, Cozzi D, Grassi R, Grassi R, Izzo F, Petrillo A. Radiomics in hepatic metastasis by colorectal cancer. Infect Agent Cancer 2021; 16:39. [PMID: 34078424 PMCID: PMC8173908 DOI: 10.1186/s13027-021-00379-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/12/2021] [Indexed: 02/06/2023] Open
Abstract
Background Radiomics is an emerging field and has a keen interest, especially in the oncology field. The process of a radiomics study consists of lesion segmentation, feature extraction, consistency analysis of features, feature selection, and model building. Manual segmentation is one of the most critical parts of radiomics. It can be time-consuming and suffers from variability in tumor delineation, which leads to the reproducibility problem of calculating parameters and assessing spatial tumor heterogeneity, particularly in large or multiple tumors. Radiomic features provides data on tumor phenotype as well as cancer microenvironment. Radiomics derived parameters, when associated with other pertinent data and correlated with outcomes data, can produce accurate robust evidence based clinical decision support systems. The principal challenge is the optimal collection and integration of diverse multimodal data sources in a quantitative manner that delivers unambiguous clinical predictions that accurately and robustly enable outcome prediction as a function of the impending decisions. Methods The search covered the years from January 2010 to January 2021. The inclusion criterion was: clinical study evaluating radiomics of liver colorectal metastases. Exclusion criteria were studies with no sufficient reported data, case report, review or editorial letter. Results We recognized 38 studies that assessed radiomics in mCRC from January 2010 to January 2021. Twenty were on different tpics, 5 corresponded to most criteria; 3 are review, or letter to editors; so 10 articles were included. Conclusions In colorectal liver metastases radiomics should be a valid tool for the characterization of lesions, in the stratification of patients based on the risk of relapse after surgical treatment and in the prediction of response to chemotherapy treatment.
Collapse
Affiliation(s)
- Vincenza Granata
- Radiology Division, "ISTITUTO NAZIONALE TUMORI - IRCCS - FONDAZIONE G. PASCALE, Napoli, Italy", Via Mariano Semmola, Naples, Italy
| | - Roberta Fusco
- Radiology Division, "ISTITUTO NAZIONALE TUMORI - IRCCS - FONDAZIONE G. PASCALE, Napoli, Italy", Via Mariano Semmola, Naples, Italy.
| | - Maria Luisa Barretta
- Radiology Division, "ISTITUTO NAZIONALE TUMORI - IRCCS - FONDAZIONE G. PASCALE, Napoli, Italy", Via Mariano Semmola, Naples, Italy
| | - Carmine Picone
- Radiology Division, "ISTITUTO NAZIONALE TUMORI - IRCCS - FONDAZIONE G. PASCALE, Napoli, Italy", Via Mariano Semmola, Naples, Italy
| | - Antonio Avallone
- Abdominal Oncology Division, "ISTITUTO NAZIONALE TUMORI - IRCCS - FONDAZIONE G. PASCALE, NAPOLI, ITALIA", Via Mariano Semmola, Naples, Italy
| | - Andrea Belli
- Hepatobiliary Surgical Oncology Division, "ISTITUTO NAZIONALE TUMORI - IRCCS - FONDAZIONE G. PASCALE, NAPOLI, ITALIA", Via Mariano Semmola, Naples, Italy
| | - Renato Patrone
- Hepatobiliary Surgical Oncology Division, "ISTITUTO NAZIONALE TUMORI - IRCCS - FONDAZIONE G. PASCALE, NAPOLI, ITALIA", Via Mariano Semmola, Naples, Italy
| | - Marilina Ferrante
- Division of Radiology, "Università degli Studi della Campania Luigi Vanvitelli", Naples, Italy
| | - Diletta Cozzi
- Division of Radiology, "Azienda Ospedaliera Universitaria Careggi", Florence, Italy
| | - Roberta Grassi
- Division of Radiology, "Università degli Studi della Campania Luigi Vanvitelli", Naples, Italy
| | - Roberto Grassi
- Division of Radiology, "Università degli Studi della Campania Luigi Vanvitelli", Naples, Italy.,Italian Society of Medical and Interventional Radiology SIRM, SIRM Foundation, Via della Signora 2, 20122, Milan, Italy
| | - Francesco Izzo
- Hepatobiliary Surgical Oncology Division, "ISTITUTO NAZIONALE TUMORI - IRCCS - FONDAZIONE G. PASCALE, NAPOLI, ITALIA", Via Mariano Semmola, Naples, Italy
| | - Antonella Petrillo
- Radiology Division, "ISTITUTO NAZIONALE TUMORI - IRCCS - FONDAZIONE G. PASCALE, Napoli, Italy", Via Mariano Semmola, Naples, Italy
| |
Collapse
|
81
|
Vercellino L, de Jong D, di Blasi R, Kanoun S, Reshef R, Schwartz LH, Dercle L. Current and Future Role of Medical Imaging in Guiding the Management of Patients With Relapsed and Refractory Non-Hodgkin Lymphoma Treated With CAR T-Cell Therapy. Front Oncol 2021; 11:664688. [PMID: 34123825 PMCID: PMC8195284 DOI: 10.3389/fonc.2021.664688] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 05/05/2021] [Indexed: 12/21/2022] Open
Abstract
Chimeric antigen receptor (CAR) T-cells are a novel immunotherapy available for patients with refractory/relapsed non-Hodgkin lymphoma. In this indication, clinical trials have demonstrated that CAR T-cells achieve high rates of response, complete response, and long-term response (up to 80%, 60%, and 40%, respectively). Nonetheless, the majority of patients ultimately relapsed. This review provides an overview about the current and future role of medical imaging in guiding the management of non-Hodgkin lymphoma patients treated with CAR T-cells. It discusses the value of predictive and prognostic biomarkers to better stratify the risk of relapse, and provide a patient-tailored therapeutic strategy. At baseline, high tumor volume (assessed on CT-scan or on [18F]-FDG PET/CT) is a prognostic factor associated with treatment failure. Response assessment has not been studied extensively yet. Available data suggests that current response assessment developed on CT-scan or on [18F]-FDG PET/CT for cytotoxic systemic therapies remains relevant to estimate lymphoma response to CAR T-cell therapy. Nonetheless, atypical patterns of response and progression have been observed and should be further analyzed. The potential advantages as well as limitations of artificial intelligence and radiomics as tools providing high throughput quantitative imaging features is described.
Collapse
Affiliation(s)
- Laetitia Vercellino
- Nuclear Medicine Department Saint Louis Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Dorine de Jong
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Roberta di Blasi
- Onco-Hematology Department Saint Louis Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Salim Kanoun
- Cancer Research Center of Toulouse (CRCT), Team 9, INSERM UMR 1037, Toulouse, France
| | - Ran Reshef
- Blood and Marrow Transplantation and Cell Therapy Program, Division of Hematology/Oncology and Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York City, NY, United States
| | - Lawrence H. Schwartz
- Department of Radiology, New York Presbyterian, Columbia University Irving Medical Center, New York City, NY, United States
| | - Laurent Dercle
- Department of Radiology, New York Presbyterian, Columbia University Irving Medical Center, New York City, NY, United States
| |
Collapse
|
82
|
Lu L, Ahmed FS, Akin O, Luk L, Guo X, Yang H, Yoon J, Hakimi AA, Schwartz LH, Zhao B. Uncontrolled Confounders May Lead to False or Overvalued Radiomics Signature: A Proof of Concept Using Survival Analysis in a Multicenter Cohort of Kidney Cancer. Front Oncol 2021; 11:638185. [PMID: 34123789 PMCID: PMC8191735 DOI: 10.3389/fonc.2021.638185] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 04/06/2021] [Indexed: 01/06/2023] Open
Abstract
Purpose We aimed to explore potential confounders of prognostic radiomics signature predicting survival outcomes in clear cell renal cell carcinoma (ccRCC) patients and demonstrate how to control for them. Materials and Methods Preoperative contrast enhanced abdominal CT scan of ccRCC patients along with pathological grade/stage, gene mutation status, and survival outcomes were retrieved from The Cancer Imaging Archive (TCIA)/The Cancer Genome Atlas-Kidney Renal Clear Cell Carcinoma (TCGA-KIRC) database, a publicly available dataset. A semi-automatic segmentation method was applied to segment ccRCC tumors, and 1,160 radiomics features were extracted from each segmented tumor on the CT images. Non-parametric principal component decomposition (PCD) and unsupervised hierarchical clustering were applied to build the radiomics signature models. The factors confounding the radiomics signature were investigated and controlled sequentially. Kaplan-Meier curves and Cox regression analyses were performed to test the association between radiomics signatures and survival outcomes. Results 183 patients of TCGA-KIRC cohort with available imaging, pathological, and clinical outcomes were included in this study. All 1,160 radiomics features were included in the first radiomics signature. Three additional radiomics signatures were then modelled in successive steps removing redundant radiomics features first, removing radiomics features biased by CT slice thickness second, and removing radiomics features dependent on tumor size third. The final radiomics signature model was the most parsimonious, unbiased by CT slice thickness, and independent of tumor size. This final radiomics signature stratified the cohort into radiomics phenotypes that are different by cancer-specific and recurrence-free survival; HR (95% CI) = 3.0 (1.5-5.7), p <0.05 and HR (95% CI) = 6.6 (3.1-14.1), p <0.05, respectively. Conclusion Radiomics signature can be confounded by multiple factors, including feature redundancy, image acquisition parameters like slice thickness, and tumor size. Attention to and proper control for these potential confounders are necessary for a reliable and clinically valuable radiomics signature.
Collapse
Affiliation(s)
- Lin Lu
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, United States
| | - Firas S Ahmed
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, United States
| | - Oguz Akin
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Lyndon Luk
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, United States
| | - Xiaotao Guo
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, United States
| | - Hao Yang
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, United States
| | - Jin Yoon
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, United States
| | - A Aari Hakimi
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Lawrence H Schwartz
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, United States
| | - Binsheng Zhao
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, United States
| |
Collapse
|
83
|
Terranova N, Venkatakrishnan K, Benincosa LJ. Application of Machine Learning in Translational Medicine: Current Status and Future Opportunities. AAPS JOURNAL 2021; 23:74. [PMID: 34008139 PMCID: PMC8130984 DOI: 10.1208/s12248-021-00593-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 04/08/2021] [Indexed: 02/06/2023]
Abstract
The exponential increase in our ability to harness multi-dimensional biological and clinical data from experimental to real-world settings has transformed pharmaceutical research and development in recent years, with increasing applications of artificial intelligence (AI) and machine learning (ML). Patient-centered iterative forward and reverse translation is at the heart of precision medicine discovery and development across the continuum from target validation to optimization of pharmacotherapy. Integration of advanced analytics into the practice of Translational Medicine is now a fundamental enabler to fully exploit information contained in diverse sources of big data sets such as “omics” data, as illustrated by deep characterizations of the genome, transcriptome, proteome, metabolome, microbiome, and exposome. In this commentary, we provide an overview of ML applications in drug discovery and development, aligned with the three strategic pillars of Translational Medicine (target, patient, dose) and offer perspectives on their potential to transform the science and practice of the discipline. Opportunities for integrating ML approaches into the discipline of Pharmacometrics are discussed and will revolutionize the practice of model-informed drug discovery and development. Finally, we posit that joint efforts of Clinical Pharmacology, Bioinformatics, and Biomarker Technology experts are vital in cross-functional team settings to realize the promise of AI/ML-enabled Translational and Precision Medicine.
Collapse
Affiliation(s)
- Nadia Terranova
- Translational Medicine, Merck Institute for Pharmacometrics, Merck Serono S.A., Lausanne, Switzerland
| | - Karthik Venkatakrishnan
- Translational Medicine, EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts, USA
| | - Lisa J Benincosa
- Translational Medicine, EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts, USA.
| |
Collapse
|
84
|
Chang E, Joel MZ, Chang HY, Du J, Khanna O, Omuro A, Chiang V, Aneja S. Comparison of radiomic feature aggregation methods for patients with multiple tumors. Sci Rep 2021; 11:9758. [PMID: 33963236 PMCID: PMC8105371 DOI: 10.1038/s41598-021-89114-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 04/21/2021] [Indexed: 12/28/2022] Open
Abstract
Radiomic feature analysis has been shown to be effective at analyzing diagnostic images to model cancer outcomes. It has not yet been established how to best combine radiomic features in cancer patients with multifocal tumors. As the number of patients with multifocal metastatic cancer continues to rise, there is a need for improving personalized patient-level prognosis to better inform treatment. We compared six mathematical methods of combining radiomic features of 3,596 tumors in 831 patients with multiple brain metastases and evaluated the performance of these aggregation methods using three survival models: a standard Cox proportional hazards model, a Cox proportional hazards model with LASSO regression, and a random survival forest. Across all three survival models, the weighted average of the largest three metastases had the highest concordance index (95% confidence interval) of 0.627 (0.595–0.661) for the Cox proportional hazards model, 0.628 (0.591–0.666) for the Cox proportional hazards model with LASSO regression, and 0.652 (0.565–0.727) for the random survival forest model. This finding was consistent when evaluating patients with different numbers of brain metastases and different tumor volumes. Radiomic features can be effectively combined to estimate patient-level outcomes in patients with multifocal brain metastases. Future studies are needed to confirm that the volume-weighted average of the largest three tumors is an effective method for combining radiomic features across other imaging modalities and tumor types.
Collapse
Affiliation(s)
- Enoch Chang
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, USA
| | - Marina Z Joel
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, USA
| | | | | | - Omaditya Khanna
- Department of Neurosurgery, Thomas Jefferson University, Philadelphia, USA
| | | | - Veronica Chiang
- Department of Neurosurgery, Yale School of Medicine, New Haven, USA
| | - Sanjay Aneja
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, USA. .,Yale Brain Tumor Center, New Haven, USA. .,Center for Outcomes Research and Evaluation, Yale School of Medicine, New Haven, USA.
| |
Collapse
|
85
|
Banegas-Luna AJ, Peña-García J, Iftene A, Guadagni F, Ferroni P, Scarpato N, Zanzotto FM, Bueno-Crespo A, Pérez-Sánchez H. Towards the Interpretability of Machine Learning Predictions for Medical Applications Targeting Personalised Therapies: A Cancer Case Survey. Int J Mol Sci 2021; 22:4394. [PMID: 33922356 PMCID: PMC8122817 DOI: 10.3390/ijms22094394] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 12/18/2022] Open
Abstract
Artificial Intelligence is providing astonishing results, with medicine being one of its favourite playgrounds. Machine Learning and, in particular, Deep Neural Networks are behind this revolution. Among the most challenging targets of interest in medicine are cancer diagnosis and therapies but, to start this revolution, software tools need to be adapted to cover the new requirements. In this sense, learning tools are becoming a commodity but, to be able to assist doctors on a daily basis, it is essential to fully understand how models can be interpreted. In this survey, we analyse current machine learning models and other in-silico tools as applied to medicine-specifically, to cancer research-and we discuss their interpretability, performance and the input data they are fed with. Artificial neural networks (ANN), logistic regression (LR) and support vector machines (SVM) have been observed to be the preferred models. In addition, convolutional neural networks (CNNs), supported by the rapid development of graphic processing units (GPUs) and high-performance computing (HPC) infrastructures, are gaining importance when image processing is feasible. However, the interpretability of machine learning predictions so that doctors can understand them, trust them and gain useful insights for the clinical practice is still rarely considered, which is a factor that needs to be improved to enhance doctors' predictive capacity and achieve individualised therapies in the near future.
Collapse
Affiliation(s)
- Antonio Jesús Banegas-Luna
- Structural Bioinformatics and High-Performance Computing Research Group (BIO-HPC), Universidad Católica de Murcia (UCAM), 30107 Murcia, Spain; (J.P.-G.); (A.B.-C.)
| | - Jorge Peña-García
- Structural Bioinformatics and High-Performance Computing Research Group (BIO-HPC), Universidad Católica de Murcia (UCAM), 30107 Murcia, Spain; (J.P.-G.); (A.B.-C.)
| | - Adrian Iftene
- Faculty of Computer Science, Universitatea Alexandru Ioan Cuza (UAIC), 700505 Jashi, Romania;
| | - Fiorella Guadagni
- Interinstitutional Multidisciplinary Biobank (BioBIM), IRCCS San Raffaele Roma, 00166 Rome, Italy; (F.G.); (P.F.)
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy;
| | - Patrizia Ferroni
- Interinstitutional Multidisciplinary Biobank (BioBIM), IRCCS San Raffaele Roma, 00166 Rome, Italy; (F.G.); (P.F.)
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy;
| | - Noemi Scarpato
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy;
| | - Fabio Massimo Zanzotto
- Dipartimento di Ingegneria dell’Impresa “Mario Lucertini”, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Andrés Bueno-Crespo
- Structural Bioinformatics and High-Performance Computing Research Group (BIO-HPC), Universidad Católica de Murcia (UCAM), 30107 Murcia, Spain; (J.P.-G.); (A.B.-C.)
| | - Horacio Pérez-Sánchez
- Structural Bioinformatics and High-Performance Computing Research Group (BIO-HPC), Universidad Católica de Murcia (UCAM), 30107 Murcia, Spain; (J.P.-G.); (A.B.-C.)
| |
Collapse
|
86
|
Wesdorp NJ, van Goor VJ, Kemna R, Jansma EP, van Waesberghe JHTM, Swijnenburg RJ, Punt CJA, Huiskens J, Kazemier G. Advanced image analytics predicting clinical outcomes in patients with colorectal liver metastases: A systematic review of the literature. Surg Oncol 2021; 38:101578. [PMID: 33866191 DOI: 10.1016/j.suronc.2021.101578] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/30/2021] [Accepted: 04/01/2021] [Indexed: 12/26/2022]
Abstract
BACKGROUND To better select patients with colorectal liver metastases (CRLM) for an optimal selection of treatment strategy (i.e. local, systemic or combined treatment) new prognostic models are warranted. In the last decade, radiomics has emerged as a field to create predictive models based on imaging features. This systematic review aims to investigate the current state and potential of radiomics to predict clinical outcomes in patients with CRLM. METHODS A comprehensive literature search was conducted in the electronic databases of PubMed, Embase, and Cochrane Library, according to PRISMA guidelines. Original studies reporting on radiomics predicting clinical outcome in patients diagnosed with CRLM were included. Clinical outcomes were defined as response to systemic treatment, recurrence of disease, and survival (overall, progression-free, disease-free). Primary outcome was the predictive performance of radiomics. A narrative synthesis of the results was made. Methodological quality was assessed using the radiomics quality score. RESULTS In 11 out of 14 included studies, radiomics was predictive for response to treatment, recurrence of disease, survival, or a combination of outcomes. Combining clinical parameters and radiomic features in multivariate modelling often improved the predictive performance. Different types of individual features were found prognostic. Noticeable were the contrary levels of heterogeneous and homogeneous features in patients with good response. The methodological quality as assessed by the radiomics quality score varied considerably between studies. CONCLUSION Radiomics appears a promising non-invasive method to predict clinical outcome and improve personalized decision-making in patients with CRLM. However, results were contradictory and difficult to compare. Standardized prospective studies are warranted to establish the added value of radiomics in patients with CRLM.
Collapse
Affiliation(s)
- N J Wesdorp
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Surgery, Cancer Center Amsterdam, De Boelelaan, 1117, Amsterdam, Netherlands.
| | - V J van Goor
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Surgery, Cancer Center Amsterdam, De Boelelaan, 1117, Amsterdam, Netherlands
| | - R Kemna
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Surgery, Cancer Center Amsterdam, De Boelelaan, 1117, Amsterdam, Netherlands
| | - E P Jansma
- Amsterdam UMC, University of Amsterdam, Department of Epidemiology and Biostatistics, Cancer Center Amsterdam, De Boelelaan, 1117, Amsterdam, Netherlands
| | - J H T M van Waesberghe
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Radiology and Molecular Imaging, Cancer Center Amsterdam, De Boelelaan, 1117, Amsterdam, Netherlands
| | - R J Swijnenburg
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Surgery, Cancer Center Amsterdam, Meibergdreef 9, Amsterdam, Netherlands
| | - C J A Punt
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Department of Epidemiology, Utrecht, the Netherlands
| | - J Huiskens
- SAS Institute B.V., Flevolaan 69, Huizen, the Netherlands
| | - G Kazemier
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Surgery, Cancer Center Amsterdam, De Boelelaan, 1117, Amsterdam, Netherlands
| |
Collapse
|
87
|
Alderson PO, Summers RM. The Evolving Status of Radiomics. J Natl Cancer Inst 2021; 112:869-870. [PMID: 32016420 DOI: 10.1093/jnci/djaa018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 01/24/2020] [Indexed: 12/30/2022] Open
Affiliation(s)
| | - Ronald M Summers
- Imaging Biomarkers and Computer-Aided, Diagnosis Laboratory NIH Clinical Center, Bethesda, MD, USA
| |
Collapse
|
88
|
Zhao B. Understanding Sources of Variation to Improve the Reproducibility of Radiomics. Front Oncol 2021; 11:633176. [PMID: 33854969 PMCID: PMC8039446 DOI: 10.3389/fonc.2021.633176] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 03/01/2021] [Indexed: 12/12/2022] Open
Abstract
Radiomics is the method of choice for investigating the association between cancer imaging phenotype, cancer genotype and clinical outcome prediction in the era of precision medicine. The fast dispersal of this new methodology has benefited from the existing advances of the core technologies involved in radiomics workflow: image acquisition, tumor segmentation, feature extraction and machine learning. However, despite the rapidly increasing body of publications, there is no real clinical use of a developed radiomics signature so far. Reasons are multifaceted. One of the major challenges is the lack of reproducibility and generalizability of the reported radiomics signatures (features and models). Sources of variation exist in each step of the workflow; some are controllable or can be controlled to certain degrees, while others are uncontrollable or even unknown. Insufficient transparency in reporting radiomics studies further prevents translation of the developed radiomics signatures from the bench to the bedside. This review article first addresses sources of variation, which is illustrated using demonstrative examples. Then, it reviews a number of published studies and progresses made to date in the investigation and improvement of feature reproducibility and model performance. Lastly, it discusses potential strategies and practical considerations to reduce feature variability and improve the quality of radiomics study. This review focuses on CT image acquisition, tumor segmentation, quantitative feature extraction, and the disease of lung cancer.
Collapse
Affiliation(s)
- Binsheng Zhao
- Department of Radiology, Columbia University Irving Medical Center, New York, NY, United States
| |
Collapse
|
89
|
Nakanishi R, Oki E, Hasuda H, Sano E, Miyashita Y, Sakai A, Koga N, Kuriyama N, Nonaka K, Fujimoto Y, Jogo T, Hokonohara K, Hu Q, Hisamatsu Y, Ando K, Kimura Y, Yoshizumi T, Mori M. ASO Author Reflection: Radiomics-Based Prediction for the Responder to First-Line Oxaliplatin-Based Chemotherapy in Patients with Colorectal Liver Metastasis. Ann Surg Oncol 2021; 28:2986-2987. [PMID: 33725205 DOI: 10.1245/s10434-020-09584-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 12/18/2020] [Indexed: 11/18/2022]
Affiliation(s)
- Ryota Nakanishi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hirofumi Hasuda
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Eiki Sano
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yu Miyashita
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Akihiro Sakai
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Naomichi Koga
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Naotaka Kuriyama
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kentaro Nonaka
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yoshiaki Fujimoto
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Tomoko Jogo
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kentaro Hokonohara
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Qingjiang Hu
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yuichi Hisamatsu
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Koji Ando
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yasue Kimura
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Tomoharu Yoshizumi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Masaki Mori
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
90
|
Lu L, Sun SH, Afran A, Yang H, Lu ZF, So J, Schwartz LH, Zhao B. Identifying Robust Radiomics Features for Lung Cancer by Using In-Vivo and Phantom Lung Lesions. Tomography 2021; 7:55-64. [PMID: 33681463 PMCID: PMC7934702 DOI: 10.3390/tomography7010005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 12/17/2020] [Indexed: 02/06/2023] Open
Abstract
We propose a novel framework for determining radiomics feature robustness by considering the effects of both biological and noise signals. This framework is preliminarily tested in a study predicting the epidermal growth factor receptor (EGFR) mutation status in non-small cell lung cancer (NSCLC) patients. Pairs of CT images (baseline, 3-week post therapy) of 46 NSCLC patients with known EGFR mutation status were collected and a FDA-customized anthropomorphic thoracic phantom was scanned on two vendors' scanners at four different tube currents. Delta radiomics features were extracted from the NSCLC patient CTs and reproducible, non-redundant, and informative features were identified. The feature value differences between EGFR mutant and EGFR wildtype patients were quantitatively measured as the biological signal. Similarly, radiomics features were extracted from the phantom CTs. A pairwise comparison between settings resulted in a feature value difference that was quantitatively measured as the noise signal. Biological signals were compared to noise signals at each setting to determine if the distributions were significantly different by two-sample t-test, and thus robust. Four optimal features were selected to predict EGFR mutation status, Tumor-Mass, Sigmoid-Offset-Mean, Gabor-Energy and DWT-Energy, which quantified tumor mass, tumor-parenchyma density transition at boundary, line-like pattern inside tumor and intratumoral heterogeneity, respectively. The first three variables showed robustness across the majority of studied CT acquisition parameters. The textual feature DWT-Energy was less robust. The proposed framework was able to determine robustness of radiomics features at specific settings by comparing biological signal to noise signal. Identification of robust radiomics features may improve the generalizability of radiomics models in future studies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Binsheng Zhao
- Department of Radiology, New York Presbyterian Hospital, Columbia University Medical Center, New York, NY 10032, USA; (L.L.); (S.H.S.); (A.A.); (H.Y.); (Z.F.L.); (J.S.); (L.H.S.)
| |
Collapse
|
91
|
Badic B, Tixier F, Cheze Le Rest C, Hatt M, Visvikis D. Radiogenomics in Colorectal Cancer. Cancers (Basel) 2021; 13:cancers13050973. [PMID: 33652647 PMCID: PMC7956421 DOI: 10.3390/cancers13050973] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 02/07/2021] [Accepted: 02/20/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Colorectal carcinoma is characterized by intratumoral heterogeneity that can be assessed by radiogenomics. Radiomics, high-throughput quantitative data extracted from medical imaging, combined with molecular analysis, through genomic and transcriptomic data, is expected to lead to significant advances in personalized medicine. However, a radiogenomics approach in colorectal cancer is still in its early stages and many problems remain to be solved. Here we review the progress and challenges in this field at its current stage, as well as future developments. Abstract The steady improvement of high-throughput technologies greatly facilitates the implementation of personalized precision medicine. Characterization of tumor heterogeneity through image-derived features—radiomics and genetic profile modifications—genomics, is a rapidly evolving field known as radiogenomics. Various radiogenomics studies have been dedicated to colorectal cancer so far, highlighting the potential of these approaches to enhance clinical decision-making. In this review, a general outline of colorectal radiogenomics literature is provided, discussing the current limitations and suggested further developments.
Collapse
Affiliation(s)
- Bogdan Badic
- National Institute of Health and Medical Research, LaTIM—Laboratory of Medical Information Processing (INSERM LaTIM), UMR 1101, Université Bretagne Occidentale, 29238 Brest, France; (F.T.); (C.C.L.R.); (M.H.); (D.V.)
- Correspondence: ; Tel.: +33-298-347-215
| | - Florent Tixier
- National Institute of Health and Medical Research, LaTIM—Laboratory of Medical Information Processing (INSERM LaTIM), UMR 1101, Université Bretagne Occidentale, 29238 Brest, France; (F.T.); (C.C.L.R.); (M.H.); (D.V.)
| | - Catherine Cheze Le Rest
- National Institute of Health and Medical Research, LaTIM—Laboratory of Medical Information Processing (INSERM LaTIM), UMR 1101, Université Bretagne Occidentale, 29238 Brest, France; (F.T.); (C.C.L.R.); (M.H.); (D.V.)
- Department of Nuclear Medicine, University Hospital of Poitiers, 86021 Poitiers, France
| | - Mathieu Hatt
- National Institute of Health and Medical Research, LaTIM—Laboratory of Medical Information Processing (INSERM LaTIM), UMR 1101, Université Bretagne Occidentale, 29238 Brest, France; (F.T.); (C.C.L.R.); (M.H.); (D.V.)
| | - Dimitris Visvikis
- National Institute of Health and Medical Research, LaTIM—Laboratory of Medical Information Processing (INSERM LaTIM), UMR 1101, Université Bretagne Occidentale, 29238 Brest, France; (F.T.); (C.C.L.R.); (M.H.); (D.V.)
| |
Collapse
|
92
|
Kuo CY, Chiu HM. Application of artificial intelligence in gastroenterology: Potential role in clinical practice. J Gastroenterol Hepatol 2021; 36:267-272. [PMID: 33624890 DOI: 10.1111/jgh.15403] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 01/11/2021] [Indexed: 02/06/2023]
Abstract
Artificial intelligence (AI) based on deep learning boosted medical research in the past years and is expected to enormously change the style of health care in many aspects in the foreseeable future. Nowadays, there are exploding volumes of healthcare-related data being generated daily. Because of its time-sensitive characteristics, being able to process large amounts of data in real-time fashion is crucial in healthcare settings. In gastroenterology practice, AI can manage and interpret the sheer amount of data with different formats coming from a myriad of sources, including currently used endoscopic or imaging devices, digital record systems, and electronic health records, or from other sources such as governmental databases, social media, or wearable devices over a long period. Traditional gastroenterology is nowadays beginning to transform to a new personalized, predictive, and preventive paradigm. Evidence-based practices and recent studies are coming out every day, and big data-based approaches and the progress in basic sciences and its emerging applications are now becoming the indispensable part of precision medicine. In gastroenterology, AI can be applied in disease diagnosis, treatment guidance, outcome prediction, and reducing workload of the healthcare staff. As the healthcare community begins to embrace AI technology, how to seamlessly construct an interoperable platform to accommodate data with high variety and veracity with high velocity and implement AI in the clinical workflow would be the future challenges.
Collapse
Affiliation(s)
- Chen-Ya Kuo
- Department of Internal Medicine, Fu Jen Catholic University Hospital, New Taipei City, Taiwan
| | - Han-Mo Chiu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
93
|
Hewitt DB, Pawlik TM, Cloyd JM. Who Will Benefit? Using Radiomics to Predict Response to Oxaliplatin-Based Chemotherapy in Patients with Colorectal Liver Metastases. Ann Surg Oncol 2021; 28:2931-2933. [PMID: 33475881 DOI: 10.1245/s10434-020-09586-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 12/30/2020] [Indexed: 11/18/2022]
Affiliation(s)
- Daniel Brock Hewitt
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Timothy M Pawlik
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Jordan M Cloyd
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
94
|
Nakanishi R, Oki E, Hasuda H, Sano E, Miyashita Y, Sakai A, Koga N, Kuriyama N, Nonaka K, Fujimoto Y, Jogo T, Hokonohara K, Hu Q, Hisamatsu Y, Ando K, Kimura Y, Yoshizumi T, Mori M. Radiomics Texture Analysis for the Identification of Colorectal Liver Metastases Sensitive to First-Line Oxaliplatin-Based Chemotherapy. Ann Surg Oncol 2021; 28:2975-2985. [PMID: 33454878 DOI: 10.1245/s10434-020-09581-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 12/01/2020] [Indexed: 01/06/2023]
Abstract
OBJECTIVE The aim of this study was to develop a radiomics-based prediction model for the response of colorectal liver metastases to oxaliplatin-based chemotherapy. METHODS Forty-two consecutive patients treated with oxaliplatin-based first-line chemotherapy for colorectal liver metastasis at our institution from August 2013 to October 2019 were enrolled in this retrospective study. Overall, 126 liver metastases were chronologically divided into the training (n = 94) and validation (n = 32) cohorts. Regions of interest were manually segmented, and the best response to chemotherapy was decided based on Response Evaluation Criteria in Solid Tumors (RECIST). Patients who achieved clinical complete and partial response according to RECIST were defined as good responders. Radiomics features were extracted from the pretreatment enhanced computed tomography scans, and a radiomics score was calculated using the least absolute shrinkage and selection operator regression model in a trial cohort. RESULTS The radiomics score significantly discriminated good responders in both the trial (area under the curve [AUC] 0.8512, 95% confidence interval [CI] 0.7719-0.9305; p < 0.0001) and validation (AUC 0.7792, 95% CI 0.6176-0.9407; p < 0.0001) cohorts. Multivariate analysis revealed that high radiomics scores greater than - 0.06 (odds ratio [OR] 23.803, 95% CI 8.432-80.432; p < 0.0001), clinical non-T4 (OR 6.054, 95% CI 2.164-18.394; p = 0.0005), and metachronous disease (OR 11.787, 95% CI 2.333-70.833; p = 0.0025) were independently associated with good response. CONCLUSIONS Radiomics signatures may be a potential biomarker for the early prediction of chemosensitivity in colorectal liver metastases. This approach may support the treatment strategy for colorectal liver metastasis.
Collapse
Affiliation(s)
- Ryota Nakanishi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hirofumi Hasuda
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eiki Sano
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yu Miyashita
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Akihiro Sakai
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Naomichi Koga
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Naotaka Kuriyama
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kentaro Nonaka
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshiaki Fujimoto
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomoko Jogo
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kentaro Hokonohara
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Qingjiang Hu
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuichi Hisamatsu
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koji Ando
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasue Kimura
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomoharu Yoshizumi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaki Mori
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
95
|
Xue P, Tang C, Li Q, Li Y, Shen Y, Zhao Y, Chen J, Wu J, Li L, Wang W, Li Y, Cui X, Zhang S, Zhang W, Zhang X, Ma K, Zheng Y, Qian T, Ng MTA, Liu Z, Qiao Y, Jiang Y, Zhao F. Development and validation of an artificial intelligence system for grading colposcopic impressions and guiding biopsies. BMC Med 2020; 18:406. [PMID: 33349257 PMCID: PMC7754595 DOI: 10.1186/s12916-020-01860-y] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/19/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Colposcopy diagnosis and directed biopsy are the key components in cervical cancer screening programs. However, their performance is limited by the requirement for experienced colposcopists. This study aimed to develop and validate a Colposcopic Artificial Intelligence Auxiliary Diagnostic System (CAIADS) for grading colposcopic impressions and guiding biopsies. METHODS Anonymized digital records of 19,435 patients were obtained from six hospitals across China. These records included colposcopic images, clinical information, and pathological results (gold standard). The data were randomly assigned (7:1:2) to a training and a tuning set for developing CAIADS and to a validation set for evaluating performance. RESULTS The agreement between CAIADS-graded colposcopic impressions and pathology findings was higher than that of colposcopies interpreted by colposcopists (82.2% versus 65.9%, kappa 0.750 versus 0.516, p < 0.001). For detecting pathological high-grade squamous intraepithelial lesion or worse (HSIL+), CAIADS showed higher sensitivity than the use of colposcopies interpreted by colposcopists at either biopsy threshold (low-grade or worse 90.5%, 95% CI 88.9-91.4% versus 83.5%, 81.5-85.3%; high-grade or worse 71.9%, 69.5-74.2% versus 60.4%, 57.9-62.9%; all p < 0.001), whereas the specificities were similar (low-grade or worse 51.8%, 49.8-53.8% versus 52.0%, 50.0-54.1%; high-grade or worse 93.9%, 92.9-94.9% versus 94.9%, 93.9-95.7%; all p > 0.05). The CAIADS also demonstrated a superior ability in predicting biopsy sites, with a median mean-intersection-over-union (mIoU) of 0.758. CONCLUSIONS The CAIADS has potential in assisting beginners and for improving the diagnostic quality of colposcopy and biopsy in the detection of cervical precancer/cancer.
Collapse
Affiliation(s)
- Peng Xue
- Department of Epidemiology and Biostatistics, School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Department of Cancer Epidemiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chao Tang
- School of Public Health, Dalian Medical University, Dalian, China
| | - Qing Li
- Diagnosis and Treatment for Cervical Lesions Center, Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, China
| | | | - Yu Shen
- Zonsun Healthcare, Shenzhen, China
| | - Yuqian Zhao
- Center for Cancer Prevention Research, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | | | | | - Longyu Li
- Jiangxi Maternal and Child Health Hospital, Nanchang, China
| | - Wei Wang
- Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yucong Li
- Chongqing University Cancer Hospital, Chongqing, China
| | - Xiaoli Cui
- Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Shaokai Zhang
- Affiliated Cancer Hospital of Zhengzhou University/Henan Cancer Hospital, Zhengzhou, China
| | - Wenhua Zhang
- Department of Cancer Epidemiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xun Zhang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kai Ma
- Tencent Jarvis Lab, Shenzhen, China
| | | | | | | | - Zhihua Liu
- Department of Gynecology, Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Youlin Qiao
- Department of Epidemiology and Biostatistics, School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Department of Cancer Epidemiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yu Jiang
- Department of Epidemiology and Biostatistics, School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Fanghui Zhao
- Department of Cancer Epidemiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
96
|
Kudou M, Kosuga T, Otsuji E. Artificial intelligence in gastrointestinal cancer: Recent advances and future perspectives. Artif Intell Gastroenterol 2020; 1:71-85. [DOI: 10.35712/aig.v1.i4.71] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 10/28/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023] Open
Abstract
Artificial intelligence (AI) using machine or deep learning algorithms is attracting increasing attention because of its more accurate image recognition ability and prediction performance than human-aid analyses. The application of AI models to gastrointestinal (GI) clinical oncology has been investigated for the past decade. AI has the capacity to automatically detect and diagnose GI tumors with similar diagnostic accuracy to expert clinicians. AI may also predict malignant potential, such as tumor histology, metastasis, patient survival, resistance to cancer treatments and the molecular biology of tumors, through image analyses of radiological or pathological imaging data using complex deep learning models beyond human cognition. The introduction of AI-assisted diagnostic systems into clinical settings is expected in the near future. However, limitations associated with the evaluation of GI tumors by AI models have yet to be resolved. Recent studies on AI-assisted diagnostic models of gastric and colorectal cancers in the endoscopic, pathological, and radiological fields were herein reviewed. The limitations and future perspectives for the application of AI systems in clinical settings have also been discussed. With the establishment of a multidisciplinary team containing AI experts in each medical institution and prospective studies, AI-assisted medical systems will become a promising tool for GI cancer.
Collapse
Affiliation(s)
- Michihiro Kudou
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
- Department of Surgery, Kyoto Okamoto Memorial Hospital, Kyoto 613-0034, Japan
| | - Toshiyuki Kosuga
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
- Department of Surgery, Saiseikai Shiga Hospital, Ritto 520-3046, Japan
| | - Eigo Otsuji
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| |
Collapse
|
97
|
Staal FCR, van der Reijd DJ, Taghavi M, Lambregts DMJ, Beets-Tan RGH, Maas M. Radiomics for the Prediction of Treatment Outcome and Survival in Patients With Colorectal Cancer: A Systematic Review. Clin Colorectal Cancer 2020; 20:52-71. [PMID: 33349519 DOI: 10.1016/j.clcc.2020.11.001] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 09/03/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023]
Abstract
Prediction of outcome in patients with colorectal cancer (CRC) is challenging as a result of lack of a robust biomarker and heterogeneity between and within tumors. The aim of this review was to assess the current possibilities and limitations of radiomics (on computed tomography [CT], magnetic resonance imaging [MRI], and positron emission tomography [PET]) for the prediction of treatment outcome and long-term outcome in CRC. Medline/PubMed was searched up to August 2020 for studies that used radiomics for the prediction of response to treatment and survival in patients with CRC (based on pretreatment imaging). The Quality Assessment of Diagnostic Accuracy Studies (QUADAS) tool and Radiomics Quality Score (RQS) were used for quality assessment. A total of 76 studies met the inclusion criteria and were included for further analysis. Radiomics analyses were performed on MRI in 41 studies, on CT in 30 studies, and on 18F-FDG-PET/CT in 10 studies. Heterogeneous results were reported regarding radiomics methods and included features. High-quality studies (n = 13), consisting mainly of MRI-based radiomics to predict response in rectal cancer, were able to predict response with good performance. Radiomics literature in CRC is highly heterogeneous, but it nonetheless holds promise for the prediction of outcome. The most evidence is available for MRI-based radiomics in rectal cancer. Future radiomics research in CRC should focus on independent validation of existing models rather than on developing new models.
Collapse
Affiliation(s)
- Femke C R Staal
- Department of Radiology, Antoni van Leeuwenhoek, The Netherlands Cancer Institute, Amsterdam, The Netherlands; GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Denise J van der Reijd
- Department of Radiology, Antoni van Leeuwenhoek, The Netherlands Cancer Institute, Amsterdam, The Netherlands; GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Marjaneh Taghavi
- Department of Radiology, Antoni van Leeuwenhoek, The Netherlands Cancer Institute, Amsterdam, The Netherlands; GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Doenja M J Lambregts
- Department of Radiology, Antoni van Leeuwenhoek, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Regina G H Beets-Tan
- Department of Radiology, Antoni van Leeuwenhoek, The Netherlands Cancer Institute, Amsterdam, The Netherlands; GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands; Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Monique Maas
- Department of Radiology, Antoni van Leeuwenhoek, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
98
|
Chang E, Joel M, Chang HY, Du J, Khanna O, Omuro A, Chiang V, Aneja S. Comparison of Radiomic Feature Aggregation Methods for Patients with Multiple Tumors. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020:2020.11.04.20226159. [PMID: 33173902 PMCID: PMC7654896 DOI: 10.1101/2020.11.04.20226159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Background Radiomic feature analysis has been shown to be effective at modeling cancer outcomes. It has not yet been established how to best combine these radiomic features in patients with multifocal disease. As the number of patients with multifocal metastatic cancer continues to rise, there is a need for improving personalized patient-level prognostication to better inform treatment. Methods We compared six mathematical methods of combining radiomic features of 3596 tumors in 831 patients with multiple brain metastases and evaluated the performance of these aggregation methods using three survival models: a standard Cox proportional hazards model, a Cox proportional hazards model with LASSO regression, and a random survival forest. Results Across all three survival models, the weighted average of the largest three metastases had the highest concordance index (95% confidence interval) of 0.627 (0.595-0.661) for the Cox proportional hazards model, 0.628 (0.591-0.666) for the Cox proportional hazards model with LASSO regression, and 0.652 (0.565-0.727) for the random survival forest model. Conclusions Radiomic features can be effectively combined to establish patient-level outcomes in patients with multifocal brain metastases. Future studies are needed to confirm that the volume-weighted average of the largest three tumors is an effective method for combining radiomic features across other imaging modalities and disease sites.
Collapse
Affiliation(s)
- Enoch Chang
- Department of Therapeutic Radiology, Yale School of Medicine
| | | | | | | | | | | | | | - Sanjay Aneja
- Department of Therapeutic Radiology, Yale School of Medicine
- Center for Outcomes Research and Evaluation, Yale School of Medicine
| |
Collapse
|
99
|
Chu Y, Li J, Zeng Z, Huang B, Zhao J, Liu Q, Wu H, Fu J, Zhang Y, Zhang Y, Cai J, Zeng F. A Novel Model Based on CXCL8-Derived Radiomics for Prognosis Prediction in Colorectal Cancer. Front Oncol 2020; 10:575422. [PMID: 33178604 PMCID: PMC7592598 DOI: 10.3389/fonc.2020.575422] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 08/14/2020] [Indexed: 12/24/2022] Open
Abstract
Introduction: Prognosis prediction is essential to improve therapeutic strategies and to achieve better clinical outcomes in colorectal cancer (CRC) patients. Radiomics based on high-throughput mining of quantitative medical imaging is an emerging field in recent years. However, the relationship among prognosis, radiomics features, and gene expression remains unknown. Methods: We retrospectively analyzed 141 patients (from study 1) diagnosed with CRC from February 2018 to October 2019 and randomly divided them into training (N = 99) and testing (N = 42) cohorts. Radiomics features in venous phase image were extracted from preoperative computed tomography (CT) images. Gene expression was detected by RNA-sequencing on tumor tissues. The least absolute shrinkage and selection operator (LASSO) regression model was used for selecting imaging features and building the radiomics model. A total of 45 CRC patients (study 2) with immunohistochemical (IHC) staining of CXCL8 diagnosed with CRC from January 2014 to October 2018 were included in the independent testing cohort. A clinical model was validated for prognosis prediction in prognostic testing cohort (163 CRC patients from 2014 to 2018, study 3). We performed a combined radiomics model that was composed of radiomics score, tumor stage, and CXCL8-derived radiomics model to make comparison with the clinical model. Results: In our study, we identified the CXCL8 as a hub gene in affecting prognosis, which is mainly through regulating cytokine-cytokine receptor interaction and neutrophil migration pathway. The radiomics model incorporated 12 radiomics features screened by LASSO according to CXCL8 expression in the training cohort and showed good performance in testing and IHC testing cohorts. Finally, the CXCL8-derived radiomics model combined with tumor stage performed high ability in predicting the prognosis of CRC patients in the prognostic testing cohort, with an area under the curve (AUC) of 0.774 [95% confidence interval (CI): 0.674-0.874]. Kaplan-Meier analysis of the overall survival probability in CRC patients stratified by combined model revealed that high-risk patients have a poor prognosis compared with low-risk patients (Log-rank P < 0.0001). Conclusion: We demonstrated that the radiomics model reflected by CXCL8 combined with tumor stage information is a reliable approach to predict the prognosis in CRC patients and has a potential ability in assisting clinical decision-making.
Collapse
Affiliation(s)
- Yanpeng Chu
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, China.,Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Jie Li
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, China
| | - Zhaoping Zeng
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, China
| | - Bin Huang
- Department of Gastrointestinal Surgery, Nanchong Central Hospital, Nanchong, China
| | - Jiaojiao Zhao
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, China
| | - Qin Liu
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, China
| | - Huaping Wu
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, China
| | - Jiangping Fu
- Department of Oncology, Dazhou Central Hospital, Dazhou, China
| | - Yin Zhang
- Department of Oncology, Dazhou Central Hospital, Dazhou, China
| | - Yefan Zhang
- Department of Hepatobiliary Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianqiang Cai
- Department of Hepatobiliary Surgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fanxin Zeng
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, China.,School of Medicine, Sichuan University of Arts and Science, Dazhou, China
| |
Collapse
|
100
|
Radiomics of Liver Metastases: A Systematic Review. Cancers (Basel) 2020; 12:cancers12102881. [PMID: 33036490 PMCID: PMC7600822 DOI: 10.3390/cancers12102881] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/03/2020] [Accepted: 10/05/2020] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Patients with liver metastases can be scheduled for different therapies (e.g., chemotherapy, surgery, radiotherapy, and ablation). The choice of the most appropriate treatment should rely on adequate understanding of tumor biology and prediction of survival, but reliable biomarkers are lacking. Radiomics is an innovative approach to medical imaging: it identifies invisible-to-the-human-eye radiological patterns that can predict tumor aggressiveness and patients outcome. We reviewed the available literature to elucidate the role of radiomics in patients with liver metastases. Thirty-two papers were analyzed, mostly (56%) concerning metastases from colorectal cancer. Even if available studies are still preliminary, radiomics provided effective prediction of response to chemotherapy and of survival, allowing more accurate and earlier prediction than standard predictors. Entropy and homogeneity were the radiomic features with the strongest clinical impact. In the next few years, radiomics is expected to give a consistent contribution to the precision medicine approach to patients with liver metastases. Abstract Multidisciplinary management of patients with liver metastases (LM) requires a precision medicine approach, based on adequate profiling of tumor biology and robust biomarkers. Radiomics, defined as the high-throughput identification, analysis, and translational applications of radiological textural features, could fulfill this need. The present review aims to elucidate the contribution of radiomic analyses to the management of patients with LM. We performed a systematic review of the literature through the most relevant databases and web sources. English language original articles published before June 2020 and concerning radiomics of LM extracted from CT, MRI, or PET-CT were considered. Thirty-two papers were identified. Baseline higher entropy and lower homogeneity of LM were associated with better survival and higher chemotherapy response rates. A decrease in entropy and an increase in homogeneity after chemotherapy correlated with radiological tumor response. Entropy and homogeneity were also highly predictive of tumor regression grade. In comparison with RECIST criteria, radiomic features provided an earlier prediction of response to chemotherapy. Lastly, texture analyses could differentiate LM from other liver tumors. The commonest limitations of studies were small sample size, retrospective design, lack of validation datasets, and unavailability of univocal cut-off values of radiomic features. In conclusion, radiomics can potentially contribute to the precision medicine approach to patients with LM, but interdisciplinarity, standardization, and adequate software tools are needed to translate the anticipated potentialities into clinical practice.
Collapse
|