2851
|
Zhai TT, Langendijk JA, van Dijk LV, van der Schaaf A, Sommers L, Vemer-van den Hoek JGM, Bijl HP, Halmos GB, Witjes MJH, Oosting SF, Noordzij W, Sijtsema NM, Steenbakkers RJHM. Pre-treatment radiomic features predict individual lymph node failure for head and neck cancer patients. Radiother Oncol 2020; 146:58-65. [PMID: 32114267 DOI: 10.1016/j.radonc.2020.02.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 12/26/2019] [Accepted: 02/09/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND PURPOSE To develop and validate a pre-treatment radiomics-based prediction model to identify pathological lymph nodes (pLNs) at risk of failures after definitive radiotherapy in head and neck squamous cell carcinoma patients. MATERIALS AND METHODS Training and validation cohorts consisted of 165 patients with 558 pLNs and 112 patients with 467 pLNs, respectively. All patients were primarily treated with definitive radiotherapy, with or without systemic treatment. The endpoint was the cumulative incidence of nodal failure. For each pLN, 82 pre-treatment CT radiomic features and 7 clinical features were included in the Cox proportional-hazard analysis. RESULTS There were 68 and 23 nodal failures in the training and validation cohorts, respectively. Multivariable analysis revealed three clinical features (T-stage, gender and WHO Performance-status) and two radiomic features (Least-axis-length representing nodal size and gray level co-occurrence matrix based - Correlation representing nodal heterogeneity) as independent prognostic factors. The model showed good discrimination with a c-index of 0.80 (0.69-0.91) in the validation cohort, significantly better than models based on clinical features (p < 0.001) or radiomics (p = 0.003) alone. High- and low-risk groups were defined by using thresholds of estimated nodal failure risks at 2-year of 60% and 10%, resulting in positive and negative predictive values of 94.4% and 98.7%, respectively. CONCLUSION A pre-treatment prediction model was developed and validated, integrating the quantitative radiomic features of individual lymph nodes with generally used clinical features. Using this prediction model, lymph nodes with a high failure risk can be identified prior to treatment, which might be used to select patients for intensified treatment strategies targeted on individual lymph nodes.
Collapse
Affiliation(s)
- Tian-Tian Zhai
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, The Netherlands; Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China.
| | - Johannes A Langendijk
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Lisanne V van Dijk
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Arjen van der Schaaf
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Linda Sommers
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, The Netherlands
| | | | - Henk P Bijl
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Gyorgy B Halmos
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Max J H Witjes
- Department of Maxillofacial Surgery, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Sjoukje F Oosting
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Walter Noordzij
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Nanna M Sijtsema
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Roel J H M Steenbakkers
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, The Netherlands
| |
Collapse
|
2852
|
Papanikolaou N, Matos C, Koh DM. How to develop a meaningful radiomic signature for clinical use in oncologic patients. Cancer Imaging 2020; 20:33. [PMID: 32357923 PMCID: PMC7195800 DOI: 10.1186/s40644-020-00311-4] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 04/15/2020] [Indexed: 01/08/2023] Open
Abstract
During the last decade, there is an increasing usage of quantitative methods in Radiology in an effort to reduce the diagnostic variability associated with a subjective manner of radiological interpretation. Combined approaches where visual assessment made by the radiologist is augmented by quantitative imaging biomarkers are gaining attention. Advances in machine learning resulted in the rise of radiomics that is a new methodology referring to the extraction of quantitative information from medical images. Radiomics are based on the development of computational models, referred to as “Radiomic Signatures”, trying to address either unmet clinical needs, mostly in the field of oncologic imaging, or to compare radiomics performance with that of radiologists. However, to explore this new technology, initial publications did not consider best practices in the field of machine learning resulting in publications with questionable clinical value. In this paper, our effort was concentrated on how to avoid methodological mistakes and consider critical issues in the workflow of the development of clinically meaningful radiomic signatures.
Collapse
Affiliation(s)
- Nikolaos Papanikolaou
- Computational Clinical Imaging Group, Champalimaud Foundation, Centre for the Unknown, Av. Brasília, Doca de Pedrouços, 1400-038, Lisbon, Portugal.
| | - Celso Matos
- Department of Radiology, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Dow Mu Koh
- Department of Radiology, Royal Marsden Hospital, Sutton, UK
| |
Collapse
|
2853
|
Park JE, Kim HS. [Current Applications and Future Perspectives of Brain Tumor Imaging]. TAEHAN YONGSANG UIHAKHOE CHI 2020; 81:467-487. [PMID: 36238631 PMCID: PMC9431910 DOI: 10.3348/jksr.2020.81.3.467] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/04/2020] [Accepted: 05/07/2020] [Indexed: 11/29/2022]
Abstract
뇌종양의 진단 및 치료 반응 평가의 기본이 되는 영상기법은 해부학적 영상이다. 현재 임상에서 사용 가능한 영상기법들 중 확산 강조 영상 및 관류 영상이 추가적인 정보를 제공하고 있다. 최근에는 종양의 유전체 변이와 이질성 평가가 중요해지면서 라디오믹스와 딥러닝을 이용한 영상분석기법의 임상 응용이 기대되고 있다. 본 종설에서는 뇌종양 영상 임상 적용에서 여전히 중요한 해부학적 영상을 중심으로 한 자기공명영상 촬영 권고안, 최신 영상기법 중 확산 강조 영상 및 관류 영상의 기본 원리, 병태생리학적 배경 및 임상응용, 마지막으로 최근 컴퓨터 기술의 발전으로 많이 연구되고 있는 라디오믹스와 딥러닝의 뇌종양에서의 향후 활용가치에 대해 기술하고자 한다.
Collapse
|
2854
|
Fedorov A, Beichel R, Kalpathy-Cramer J, Clunie D, Onken M, Riesmeier J, Herz C, Bauer C, Beers A, Fillion-Robin JC, Lasso A, Pinter C, Pieper S, Nolden M, Maier-Hein K, Herrmann MD, Saltz J, Prior F, Fennessy F, Buatti J, Kikinis R. Quantitative Imaging Informatics for Cancer Research. JCO Clin Cancer Inform 2020; 4:444-453. [PMID: 32392097 PMCID: PMC7265794 DOI: 10.1200/cci.19.00165] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2020] [Indexed: 01/06/2023] Open
Abstract
PURPOSE We summarize Quantitative Imaging Informatics for Cancer Research (QIICR; U24 CA180918), one of the first projects funded by the National Cancer Institute (NCI) Informatics Technology for Cancer Research program. METHODS QIICR was motivated by the 3 use cases from the NCI Quantitative Imaging Network. 3D Slicer was selected as the platform for implementation of open-source quantitative imaging (QI) tools. Digital Imaging and Communications in Medicine (DICOM) was chosen for standardization of QI analysis outputs. Support of improved integration with community repositories focused on The Cancer Imaging Archive (TCIA). Priorities included improved capabilities of the standard, toolkits and tools, reference datasets, collaborations, and training and outreach. RESULTS Fourteen new tools to support head and neck cancer, glioblastoma, and prostate cancer QI research were introduced and downloaded over 100,000 times. DICOM was amended, with over 40 correction proposals addressing QI needs. Reference implementations of the standard in a popular toolkit and standalone tools were introduced. Eight datasets exemplifying the application of the standard and tools were contributed. An open demonstration/connectathon was organized, attracting the participation of academic groups and commercial vendors. Integration of tools with TCIA was improved by implementing programmatic communication interface and by refining best practices for QI analysis results curation. CONCLUSION Tools, capabilities of the DICOM standard, and datasets we introduced found adoption and utility within the cancer imaging community. A collaborative approach is critical to addressing challenges in imaging informatics at the national and international levels. Numerous challenges remain in establishing and maintaining the infrastructure of analysis tools and standardized datasets for the imaging community. Ideas and technology developed by the QIICR project are contributing to the NCI Imaging Data Commons currently being developed.
Collapse
Affiliation(s)
- Andrey Fedorov
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | | | | | | | | | | | - Christian Herz
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | | | | | | | | | | | | | - Marco Nolden
- German Cancer Research Center, Heidelberg, Germany
| | | | - Markus D. Herrmann
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | | | - Fred Prior
- University of Arkansas for Medical Sciences, Little Rock, AR
| | - Fiona Fennessy
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | | | - Ron Kikinis
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
2855
|
Eresen A, Li Y, Yang J, Shangguan J, Velichko Y, Yaghmai V, Benson AB, Zhang Z. Preoperative assessment of lymph node metastasis in Colon Cancer patients using machine learning: a pilot study. Cancer Imaging 2020; 20:30. [PMID: 32334635 PMCID: PMC7183701 DOI: 10.1186/s40644-020-00308-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 04/15/2020] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Preoperative detection of lymph node (LN) metastasis is critical for planning treatments in colon cancer (CC). The clinical diagnostic criteria based on the size of the LNs are not sensitive to determine metastasis using CT images. In this retrospective study, we investigated the potential value of CT texture features to diagnose LN metastasis using preoperative CT data and patient characteristics by developing quantitative prediction models. METHODS A total of 390 CC patients, undergone surgical resection, were enrolled in this monocentric study. 390 histologically validated LNs were collected from patients and randomly separated into training (312 patients, 155 metastatic and 157 normal LNs) and test cohorts (78 patients, 39 metastatic and 39 normal LNs). Six patient characteristics and 146 quantitative CT imaging features were analyzed and key variables were determined using either exhaustive search or least absolute shrinkage algorithm. Two kernel-based support vector machine classifiers (patient-characteristic model and radiomic-derived model), generated with 10-fold cross-validation, were compared with the clinical model that utilizes long-axis diameter for diagnosis of metastatic LN. The performance of the models was evaluated on the test cohort by computing accuracy, sensitivity, specificity, and area under the receiver operating curve (AUC). RESULTS The clinical model had an overall diagnostic accuracy of 64.87%; specifically, accuracy of 65.38% and 62.82%, sensitivity of 83.87% and 84.62%, and specificity of 47.13% and 41.03% for training and test cohorts, respectively. The patient-demographic model obtained accuracy of 67.31% and 73.08%, the sensitivity of 62.58% and 69.23%, and specificity of 71.97% and 76.23% for training and test cohorts, respectively. Besides, the radiomic-derived model resulted in an accuracy of 81.09% and 79.49%, sensitivity of 83.87% and 74.36%, and specificity of 78.34% and 84.62% for training and test cohorts, respectively. Furthermore, the diagnostic performance of the radiomic-derived model was significantly higher than clinical and patient-demographic models (p < 0.02) according to the DeLong method. CONCLUSIONS The texture of the LNs provided characteristic information about the histological status of the LNs. The radiomic-derived model leveraging LN texture provides better preoperative diagnostic accuracy for the detection of metastatic LNs compared to the clinically accepted diagnostic criteria and patient-demographic model.
Collapse
Affiliation(s)
- Aydin Eresen
- Department of Radiology, Feinberg School of Medicine, Northwestern University, 737 N. Michigan Ave, Suite 1600, Chicago, IL, 60611, USA
| | - Yu Li
- Department of Radiology, Feinberg School of Medicine, Northwestern University, 737 N. Michigan Ave, Suite 1600, Chicago, IL, 60611, USA.,Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jia Yang
- Department of Radiology, Feinberg School of Medicine, Northwestern University, 737 N. Michigan Ave, Suite 1600, Chicago, IL, 60611, USA
| | - Junjie Shangguan
- Department of Radiology, Feinberg School of Medicine, Northwestern University, 737 N. Michigan Ave, Suite 1600, Chicago, IL, 60611, USA
| | - Yury Velichko
- Department of Radiology, Feinberg School of Medicine, Northwestern University, 737 N. Michigan Ave, Suite 1600, Chicago, IL, 60611, USA
| | - Vahid Yaghmai
- Department of Radiology, Feinberg School of Medicine, Northwestern University, 737 N. Michigan Ave, Suite 1600, Chicago, IL, 60611, USA.,Department of Radiological Sciences, School of Medicine, University of California, Irvine, CA, USA.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, 675 N. St. Clair, 21st Floor, Chicago, IL, 60611, USA
| | - Al B Benson
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, 675 N. St. Clair, 21st Floor, Chicago, IL, 60611, USA. .,Division of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| | - Zhuoli Zhang
- Department of Radiology, Feinberg School of Medicine, Northwestern University, 737 N. Michigan Ave, Suite 1600, Chicago, IL, 60611, USA. .,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, 675 N. St. Clair, 21st Floor, Chicago, IL, 60611, USA.
| |
Collapse
|
2856
|
Parr E, Du Q, Zhang C, Lin C, Kamal A, McAlister J, Liang X, Bavitz K, Rux G, Hollingsworth M, Baine M, Zheng D. Radiomics-Based Outcome Prediction for Pancreatic Cancer Following Stereotactic Body Radiotherapy. Cancers (Basel) 2020; 12:cancers12041051. [PMID: 32344538 PMCID: PMC7226523 DOI: 10.3390/cancers12041051] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/09/2020] [Accepted: 04/22/2020] [Indexed: 12/19/2022] Open
Abstract
(1) Background: Radiomics use high-throughput mining of medical imaging data to extract unique information and predict tumor behavior. Currently available clinical prediction models poorly predict treatment outcomes in pancreatic adenocarcinoma. Therefore, we used radiomic features of primary pancreatic tumors to develop outcome prediction models and compared them to traditional clinical models. (2) Methods: We extracted and analyzed radiomic data from pre-radiation contrast-enhanced CTs of 74 pancreatic cancer patients undergoing stereotactic body radiotherapy. A panel of over 800 radiomic features was screened to create overall survival and local-regional recurrence prediction models, which were compared to clinical prediction models and models combining radiomic and clinical information. (3) Results: A 6-feature radiomic signature was identified that achieved better overall survival prediction performance than the clinical model (mean concordance index: 0.66 vs. 0.54 on resampled cross-validation test sets), and the combined model improved the performance slightly further to 0.68. Similarly, a 7-feature radiomic signature better predicted recurrence than the clinical model (mean AUC of 0.78 vs. 0.66). (4) Conclusion: Overall survival and recurrence can be better predicted with models based on radiomic features than with those based on clinical features for pancreatic cancer.
Collapse
Affiliation(s)
- Elsa Parr
- Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (E.P.); (C.L.); (A.K.); (J.M.); (K.B.); (G.R.); (M.H.)
| | - Qian Du
- Biological Sciences, University of Nebraska Lincoln, Lincoln, NE 68521, USA; (Q.D.); (C.Z.)
| | - Chi Zhang
- Biological Sciences, University of Nebraska Lincoln, Lincoln, NE 68521, USA; (Q.D.); (C.Z.)
| | - Chi Lin
- Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (E.P.); (C.L.); (A.K.); (J.M.); (K.B.); (G.R.); (M.H.)
| | - Ahsan Kamal
- Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (E.P.); (C.L.); (A.K.); (J.M.); (K.B.); (G.R.); (M.H.)
| | - Josiah McAlister
- Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (E.P.); (C.L.); (A.K.); (J.M.); (K.B.); (G.R.); (M.H.)
| | - Xiaoying Liang
- Proton Institute, University of Florida, Jacksonville, FL 32206, USA;
| | - Kyle Bavitz
- Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (E.P.); (C.L.); (A.K.); (J.M.); (K.B.); (G.R.); (M.H.)
| | - Gerard Rux
- Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (E.P.); (C.L.); (A.K.); (J.M.); (K.B.); (G.R.); (M.H.)
| | - Michael Hollingsworth
- Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (E.P.); (C.L.); (A.K.); (J.M.); (K.B.); (G.R.); (M.H.)
| | - Michael Baine
- Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (E.P.); (C.L.); (A.K.); (J.M.); (K.B.); (G.R.); (M.H.)
- Correspondence: (M.B.); (D.Z.)
| | - Dandan Zheng
- Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (E.P.); (C.L.); (A.K.); (J.M.); (K.B.); (G.R.); (M.H.)
- Correspondence: (M.B.); (D.Z.)
| |
Collapse
|
2857
|
Elaboration of a multisequence MRI-based radiomics signature for the preoperative prediction of the muscle-invasive status of bladder cancer: a double-center study. Eur Radiol 2020; 30:4816-4827. [PMID: 32318846 DOI: 10.1007/s00330-020-06796-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 12/19/2019] [Accepted: 03/06/2020] [Indexed: 01/03/2023]
Abstract
OBJECTIVES To develop a multisequence MRI-based radiomics signature for the preoperative prediction of the muscle-invasive status of bladder cancer (BCa). METHODS This retrospective study involved 106 eligible patients from two independent clinical centers. All patients underwent a preoperative 3.0 T MRI scan with T2-weighted image (T2WI) and multi-b-value diffusion-weighted image (DWI) sequences. In total, 1404 radiomics features were extracted from the largest region of the reported tumor locations on the T2WI, DWI, and corresponding apparent diffusion coefficient map (ADC) of each patient. A radiomics signature, namely the Radscore, was then generated using the recursive feature elimination approach and a logistic regression algorithm in a training cohort (n = 64). Its performance was then validated in an independent validation cohort (n = 42). The primary imaging and clinical factors in conjunction with the Radscore were used to determine whether the performance could be further improved. RESULTS The Radscore, generated by 36 selected radiomics features, demonstrated a favorable ability to predict muscle-invasive BCa status in both the training (AUC 0.880) and validation (AUC 0.813) cohorts. Subsequently, integrating the two independent predictors (including the Radscore and MRI-determined tumor stalk) into a nomogram exhibited more favorable discriminatory performance, with the AUC improved to 0.924 and 0.877 in both cohorts, respectively. CONCLUSIONS The proposed multisequence MRI-based radiomics signature alone could be an effective tool for quantitative prediction of muscle-invasive status of BCa. Integrating the Radscore with MRI-determined tumor stalk could further improve the discriminatory power, realizing more accurate prediction of nonmuscle-invasive and muscle-invasive BCa. KEY POINTS • DWI is superior to T2WI sequence in reflecting the heterogeneous differences between NMIBC and MIBC, and multisequence MRI helps in the preoperative prediction of muscle-invasive status of BCa. • Co-occurrence (CM), run-length matrix (RLM), and gray-level size zone matrix (GLSZM) features were the favorable feature categories for the prediction of muscle-invasive status of BCa. • The Radscore (proposed multisequence MRI-based radiomics signature) helps predict preoperatively muscle invasion. Combination with the MRI-determined tumor stalk further improves prediction.
Collapse
|
2858
|
Luo Y, Mei D, Gong J, Zuo M, Guo X. Multiparametric MRI-Based Radiomics Nomogram for Predicting Lymphovascular Space Invasion in Endometrial Carcinoma. J Magn Reson Imaging 2020; 52:1257-1262. [PMID: 32315482 DOI: 10.1002/jmri.27142] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 03/03/2020] [Accepted: 03/03/2020] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Lymphovascular space invasion (LVSI) of endometrial carcinoma (EMC) is one of the important prognostic factors, which is not usually visible subjectively. Therefore, clinicians lack imaging-based evidence about LVSI for preoperative treatment decision-making. PURPOSE To develop a multiparametric MRI (mpMRI)-based radiomics nomogram for predicting LVSI in EMC and provide decision-making support to clinicians. STUDY TYPE Retrospective. POPULATION In all, 144 patients with histologically confirmed EMC, 101 patients in a training cohort, and 43 patients in a test cohort. FIELD STRENGTH/SEQUENCE T2 WI, contrast enhanced-T1 WI, and diffusion-weighted imaging (DWI) at 3.0T MRI. ASSESSMENT Tumors were independently segmented images by two radiologists. Two pathologists reviewed the tissue specimens of the tumors to identify the existence of LVSI in consensus. STATISTICAL TESTS The intraclass correlation coefficient was used to test the reliability and least absolute shrinkage and selection operator (LASSO) regression for features selection and then developed a radiomics signature named Rad-score. A nomogram was developed in the training cohort. The diagnostic performance of the nomogram model was assessed by area under the curve (AUC) of the receiver operator characteristic (ROC) in the training and test cohort, respectively. RESULTS LVSI was identified in 32 patients (22.2%). Older age and high grade were correlated with LVSI at univariate analysis. There were five radiomics features that were identified as independent risk factors for LVSI by LASSO regression. Based on age, grade, and Rad-score, the AUC values of the nomogram model to predict LVSI in the training and test cohort were 0.820 (95% confidence interval [CI]: 0.725, 0.916; sensitivity: 82.6%, specificity: 72.9%), 0.807 (95% CI: 0.673, 0.941; sensitivity: 77.8%, specificity: 78.6%), respectively. DATA CONCLUSION The radiomic-based machine-learning model using a nomogram algorithm achieved high diagnostic performance for predicting LVSI of EMC preoperatively, which could enhance risk stratification and provide support for therapeutic decision-making. LEVEL OF EVIDENCE 2. TECHNICAL EFFICACY STAGE 3. J. Magn. Reson. Imaging 2020;52:1257-1262.
Collapse
Affiliation(s)
- Yan Luo
- Department of Radiology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Dongdong Mei
- Department of Radiology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Jingshan Gong
- Department of Radiology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, China.,Department of Radiology, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Min Zuo
- Department of Pathology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Xiaojing Guo
- Department of Pathology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, China
| |
Collapse
|
2859
|
Mazzaschi G, Milanese G, Pagano P, Madeddu D, Gnetti L, Trentini F, Falco A, Frati C, Lorusso B, Lagrasta C, Minari R, Ampollini L, Silva M, Sverzellati N, Quaini F, Roti G, Tiseo M. Integrated CT imaging and tissue immune features disclose a radio-immune signature with high prognostic impact on surgically resected NSCLC. Lung Cancer 2020; 144:30-39. [PMID: 32361033 DOI: 10.1016/j.lungcan.2020.04.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 03/06/2020] [Accepted: 04/07/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Qualitative and quantitative CT imaging features might intercept the multifaceted tumor immune microenvironment (TIME), providing a non-invasive approach to design new prognostic models in NSCLC patients. MATERIALS AND METHODS Our study population consisted of 100 surgically resected NSCLC patients among which 31 served as a validation cohort for quantitative image analysis. TIME was classified according to PD-L1 expression and the magnitude of Tumor Infiltrating Lymphocytes (TILs) and further defined as hot or cold by the tissue analysis of effector (CD8-to-CD3high/PD-1-to-CD8low) or inert (CD8-to-CD3low/PD-1-to-CD8high) phenotypes. CT datasets acted as source for qualitative (semantic, CT-SFs) and quantitative (radiomic, CT-RFs) features which were correlated with clinico-pathological and TIME profiles to determine their impact on survival outcome. RESULTS Specific CT-SFs (texture [TXT], effect [EFC] and margins [MRG]) strongly correlated to PD-L1 and TILs status and showed significant impact on survival outcome (TXT, HR:3.39, 95 % CI 1.12-10-27, P < 0.05; EFC, HR:0.41, 95 % CI 0.18-0.93, P < 0.05; MRG, HR:1.93, 95 % CI 0.88-4.25, P = 0.09). Seven CT derived radiomic features were able to sharply discriminate cases with hot (inflamed) vs cold (desert) TIME, which also exhibited opposite OS (long vs short, HR:0.09, 95 % CI 0.04-0.23, P < 0.001) and DFS (long vs short, HR:0.31, 95 % CI 0.16-0.58, P < 0.001). Moreover, we identified 6 prognostic radiomic features among which ClusterProminence displayed the highest statistical significance (HR:0.13, 95 % CI 0.06-0.31, P < 0.001). These findings were independently validated in an additional cohort of NSCLC (HR:0.11, 95 % CI 0.03-0.40, P = 0.001). Finally, in our training cohort we developed a multiparametric prognostic model, interlacing TIME and clinico-pathological characteristics with CT-SFs (ROC curve AUC:0.83, 95 % CI 0.71-0.92, P < 0.001) or CT-RFs (AUC: 0.91, 95 % CI 0.83-0.99, P < 0.001), which appeared to outperform pTNM staging (AUC: 0.66, 95 % CI 0.51-0.80, P < 0.05) in the risk assessment of NSCLC. CONCLUSION Higher order CT extracted features associated with specific TIME profiles may reveal a radio-immune signature with prognostic impact on resected NSCLC.
Collapse
Affiliation(s)
- Giulia Mazzaschi
- Department of Medicine and Surgery, University of Parma, Medical Oncology Unit, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Gianluca Milanese
- Department of Medicine and Surgery, University of Parma, Institute of Radiologic Science, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Paolo Pagano
- Department of Medicine and Surgery, University of Parma, Institute of Radiologic Science, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Denise Madeddu
- Department of Medicine and Surgery, University of Parma, Pathology Unit, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Letizia Gnetti
- Department of Medicine and Surgery, University of Parma, Pathology Unit, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Francesca Trentini
- Department of Medicine and Surgery, University of Parma, Medical Oncology Unit, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Angela Falco
- Department of Medicine and Surgery, University of Parma, Pathology Unit, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Caterina Frati
- Department of Medicine and Surgery, University of Parma, Pathology Unit, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Bruno Lorusso
- Department of Medicine and Surgery, University of Parma, Pathology Unit, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Costanza Lagrasta
- Department of Medicine and Surgery, University of Parma, Pathology Unit, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Roberta Minari
- Department of Medicine and Surgery, University of Parma, Medical Oncology Unit, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Luca Ampollini
- Department of Medicine and Surgery, University of Parma, Thoracic Surgery, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Mario Silva
- Department of Medicine and Surgery, University of Parma, Institute of Radiologic Science, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Nicola Sverzellati
- Department of Medicine and Surgery, University of Parma, Institute of Radiologic Science, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Federico Quaini
- Department of Medicine and Surgery, Hematology and Bone Marrow Transplantation, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Giovanni Roti
- Department of Medicine and Surgery, Hematology and Bone Marrow Transplantation, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| | - Marcello Tiseo
- Department of Medicine and Surgery, University of Parma, Medical Oncology Unit, University Hospital of Parma, Via Gramsci 14, 43126, Parma, Italy.
| |
Collapse
|
2860
|
[Radiomics models based on non-enhanced MRI can differentiate chondrosarcoma from enchondroma]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2020; 40:483-490. [PMID: 32895139 PMCID: PMC7225098 DOI: 10.12122/j.issn.1673-4254.2020.04.06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
OBJECTIVE To develop and validate radiomics models based on non-enhanced magnetic resonance (MR) imaging for differentiating chondrosarcoma from enchondroma. METHODS We retrospectively evaluated a total of 68 patients (including 27 with chondrosarcoma and 41 with enchondroma), who were randomly divided into training group (n=46) and validation group (n=22). Radiomics features were extracted from T1WI and T2WI-FS sequences of the whole tumor by two radiologists independently and selected by Low Variance, Univariate feature selection, and least absolute shrinkage and selection operator (LASSO). Radiomics models were constructed by multivariate logistic regression analysis based on the features from T1WI and T2WI-FS sequences. The receiver-operating characteristics (ROC) curve and intraclass correlation coefficient (ICC) analyses of the radiomics models and conventional MR imaging were performed to determine their diagnostic accuracy. RESULTS The ICC value for interreader agreement of the radiomics features ranged from 0.779 to 0.923, which indicated good agreement. Ten and 11 features were selected from the T1WI and T2WI-FS sequences to construct radiomics models, respectively. The areas under the curve (AUCs) of T1WI and T2WI-FS models were 0.990 and 0.925 in training group and 0.915 and 0.855 in the validation group, respectively, showing no significant differences between the two sequence-based models (P>0.05). In all the cases, the AUCs of the two radiomics models based on T1WI and T2WI-FS sequences and conventional MR imaging were 0.955, 0.901 and 0.569, respectively, demonstrating a significantly higher diagnostic accuracy of the two sequence-based radiomics models than conventional MR imaging (P<0.01). CONCLUSIONS The radiomics models based on T1WI and T2WI-FS non-enhanced MR imaging can be used for the differentiation of chondrosarcoma from enchondroma.
Collapse
|
2861
|
Feng B, Chen X, Chen Y, Liu K, Li K, Liu X, Yao N, Li Z, Li R, Zhang C, Ji J, Long W. Radiomics nomogram for preoperative differentiation of lung tuberculoma from adenocarcinoma in solitary pulmonary solid nodule. Eur J Radiol 2020; 128:109022. [PMID: 32371184 DOI: 10.1016/j.ejrad.2020.109022] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 03/05/2020] [Accepted: 04/13/2020] [Indexed: 12/19/2022]
Abstract
PURPOSE To investigate the preoperative differential diagnostic performance of a radiomics nomogram in tuberculous granuloma (TBG) and lung adenocarcinoma (LAC) appearing as solitary pulmonary solid nodules (SPSNs). METHOD We retrospectively recruited 426 patients with SPSNs from two centers and assigned them to training (n = 123), internal validation (n = 121), and external validation cohorts (n = 182). A model of deep learning (DL) was built for tumor segmentation from routine computed tomography (CT) images and extraction of 3D radiomics features. We used the least absolute shrinkage and selection operator (LASSO) logistic regression to build a radiomics signature. A clinical model was developed with clinical factors, including age, gender, and CT-based subjective findings (eg, lesion size, lesion location, lesion margin, lobulated sharp, and spiculation sign). We constructed individualized radiomics nomograms incorporating the radiomics signature and clinical factors to validate the diagnostic ability. RESULTS Three factors - radiomics signature, age, and spiculation sign - were found to be independent predictors and were used to build the radiomics nomogram, which showed better diagnostic accuracy than any single model (all net reclassification improvement p < 0.05). The area under curve yielded was 0.9660 (95% confidence interval [CI], 0.9390-0.9931), 0.9342 (95% CI, 0.8944-0.9739), and 0.9064 (95% CI, 0.8639-0.9490) for the training, internal validation, and external validation cohorts, respectively. Decision curve analysis (DCA) and stratification analysis showed the nomogram has potential for generalizability. CONCLUSION The radiomics nomogram we developed can preoperatively distinguish between LAC and TBG in patient with a SPSN.
Collapse
Affiliation(s)
- Bao Feng
- The Department of Radiology, The Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, Guangdong Province, China; School of Electronic Information and Automation, Guilin University of Aerospace Technology, Guilin, Guangxi Province, China
| | - Xiangmeng Chen
- The Department of Radiology, The Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, Guangdong Province, China
| | - Yehang Chen
- School of Electronic Information and Automation, Guilin University of Aerospace Technology, Guilin, Guangxi Province, China
| | - Kunfeng Liu
- The Department of Radiology, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Kunwei Li
- The Department of Radiology, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Xueguo Liu
- The Department of Radiology, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Nan Yao
- The Department of Radiology, The Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, Guangdong Province, China
| | - Zhi Li
- School of Electronic Information and Automation, Guilin University of Aerospace Technology, Guilin, Guangxi Province, China
| | - Ronggang Li
- The Department of Pathology, The Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, Guangdong Province, China
| | - Chaotong Zhang
- The Department of Radiology, The Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, Guangdong Province, China
| | - Jianbo Ji
- School of Electronic Information and Automation, Guilin University of Aerospace Technology, Guilin, Guangxi Province, China
| | - Wansheng Long
- The Department of Radiology, The Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, Guangdong Province, China.
| |
Collapse
|
2862
|
Global updates in the treatment of gastric cancer: a systematic review. Part 2: perioperative management, multimodal therapies, new technologies, standardization of the surgical treatment and educational aspects. Updates Surg 2020; 72:355-378. [PMID: 32306277 DOI: 10.1007/s13304-020-00771-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 04/11/2020] [Indexed: 12/24/2022]
Abstract
Gastric cancer is the fifth malignancy and the third cause of cancer death worldwide, according to the global cancer statistics presented in 2018. Its definition and staging have been revised in the eight edition of the AJCC/TNM classification, which took effect in 2018. Novel molecular classifications for GC have been recently established and the process of translating these classifications into clinical practice is ongoing. The cornerstone of GC treatment is surgical, in a context of multimodal therapy. Surgical treatment is being standardized, and is evolving according to new anatomical concepts and to the recent technological developments. This is leading to a massive improvement in the use of mini-invasive techniques. Mini-invasive techniques aim to be equivalent to open surgery from an oncologic point of view, with better short-term outcomes. The persecution of better short-term outcomes also includes the optimization of the perioperative management, which is being implemented on large scale according to the enhanced recovery after surgery principles. In the era of precision medicine, multimodal treatment is also evolving. The long-time-awaited results of many trials investigating the role for preoperative and postoperative management have been published, changing the clinical practice. Novel investigations focused both on traditional chemotherapeutic regimens and targeted therapies are currently ongoing. Modern platforms increase the possibility for further standardization of the different treatments, promote the use of big data and open new possibilities for surgical learning. This systematic review in two parts assesses all the current updates in GC treatment.
Collapse
|
2863
|
Abdollahi H. Less is More: Intelligent Intensive Care for SARS-CoV-2 Based on the Imaging Data. J Med Imaging Radiat Sci 2020; 51:198-199. [PMID: 32340895 PMCID: PMC7165288 DOI: 10.1016/j.jmir.2020.04.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 04/06/2020] [Indexed: 12/24/2022]
Affiliation(s)
- Hamid Abdollahi
- Faculty of Allied Medicine, Department of Radiologic Sciences and Medical Physics, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
2864
|
Identification of Pancreaticoduodenectomy Resection for Pancreatic Head Adenocarcinoma: A Preliminary Study of Radiomics. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2020; 2020:2761627. [PMID: 32377222 PMCID: PMC7182967 DOI: 10.1155/2020/2761627] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 02/07/2020] [Indexed: 02/07/2023]
Abstract
Background In a pathological examination of pancreaticoduodenectomy for pancreatic head adenocarcinoma, a resection margin without cancer cells in 1 mm is recognized as R0; a resection margin with cancer cells in 1 mm is recognized as R1. The preoperative identification of R0 and R1 is of great significance for surgical decision and prognosis. We conducted a preliminary radiomics study based on preoperative CT (computer tomography) images to evaluate a resection margin which was R0 or R1. Methods We retrospectively analyzed 258 preoperative CT images of 86 patients (34 cases of R0 and 52 cases of R1) who were diagnosed as pancreatic head adenocarcinoma and underwent pancreaticoduodenectomy. The radiomics study consists of five stages: (i) delineate and segment regions of interest (ROIs); (ii) by solving discrete Laplacian equations with Dirichlet boundary conditions, fit the ROIs to rectangular regions; (iii) enhance the textures of the fitted ROIs combining wavelet transform and fractional differential; (iv) extract texture features from the enhanced ROIs combining wavelet transform and statistical analysis methods; and (v) reduce features using principal component analysis (PCA) and classify the resection margins using the support vector machine (SVM), and then investigate the associations between texture features and histopathological characteristics using the Mann-Whitney U-test. To reduce overfitting, the SVM classifier embedded a linear kernel and adopted the leave-one-out cross-validation. Results It achieved an AUC (area under receiver operating characteristic curve) of 0.8614 and an accuracy of 84.88%. Setting p ≤ 0.01 in the Mann-Whitney U-test, two features of the run-length matrix, which are derived from diagonal sub-bands in wavelet decomposition, showed statistically significant differences between R0 and R1. Conclusions It indicates that the radiomics study is rewarding for the aided diagnosis of R0 and R1. Texture features can potentially enhance physicians' diagnostic ability.
Collapse
|
2865
|
Yu H, Meng X, Chen H, Han X, Fan J, Gao W, Du L, Chen Y, Wang Y, Liu X, Zhang L, Ma G, Yang J. Correlation Between Mammographic Radiomics Features and the Level of Tumor-Infiltrating Lymphocytes in Patients With Triple-Negative Breast Cancer. Front Oncol 2020; 10:412. [PMID: 32351879 PMCID: PMC7174560 DOI: 10.3389/fonc.2020.00412] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 03/09/2020] [Indexed: 12/16/2022] Open
Abstract
Objectives: Tumor-infiltrating lymphocytes (TILs) have been identified as a significant prognostic indicator of response to neoadjuvant therapy and immunotherapy for triple-negative breast cancer (TNBC) patients. Herein, we aim to assess the association between TIL levels and mammographic features in TNBC patients. Methods: Forty-three patients with surgically proven TNBC who underwent preoperative mammography from January 2018 to December 2018 were recruited. Pyradiomics software was used to extract 204 quantitative radiomics features, including morphologic, grayscale, and textural features, from the segmented lesion areas. The correlation between radiological characteristics and TIL levels was evaluated by screening the most statistically significant radiological features using Mann-Whitney U-test and Pearson correlation coefficient. The patients were divided into two groups based on tumor TIL levels: patients with TIL levels <50% and those with TIL levels ≥50%. The correlation between TIL levels and clinicopathological characteristics was assessed using the chi-square test or Fisher's exact test. Mann-Whitney U-test and Pearson correlation coefficient were used to analyze the statistical significance and Pearson correlation coefficient of clinical pathological features, age, and radiological features. Results: Of 43 patients, 32 (74.4%) had low TIL levels and 11 (25.6%) had high TIL levels. The histological grade of the low TIL group was higher than that of the high TIL group (p = 0.043). The high TIL group had a more negative threshold Ki-67 level (<14%) than the low TIL group (p = 0.017). The six most important radiomics features [uniformity, variance, grayscale symbiosis matrix (GLCM) correlation, GLCM autocorrelation, gray level difference matrix (GLDM) low gray level emphasis, and neighborhood gray-tone difference matrix (NGTDM) contrast], representing qualitative mammographic image characteristics, were statistically different (p < 0.05) among the low and high TIL groups. Tumors in the high TIL group had a more non-uniform density and a smoother gradient of the tumor pattern than the low TIL group. The changes in Ki-67, age, epidermal growth factor receptor, radiomic characteristics, and Pearson correlation coefficient were statistically significant (p < 0.05). Conclusion: Mammography features not only distinguish high and low TIL levels in TNBC patients but also can act as imaging biomarkers to enhance diagnosis and the response of patients to neoadjuvant therapies and immunotherapies.
Collapse
Affiliation(s)
- Hongwei Yu
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
| | - Xianqi Meng
- Beijing Engineering Research Center of Mixed Reality and Advanced Display, School of Optics and Photonics, Beijing Institute of Technology, Beijing, China
| | - Huang Chen
- Department of Pathology, China-Japan Friendship Hospital, Beijing, China
| | - Xiaowei Han
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingfan Fan
- Beijing Engineering Research Center of Mixed Reality and Advanced Display, School of Optics and Photonics, Beijing Institute of Technology, Beijing, China
| | - Wenwen Gao
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
| | - Lei Du
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yue Chen
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
| | - Yige Wang
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
| | - Xiuxiu Liu
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
| | - Lu Zhang
- Department of Science and Education, Shangluo Central Hospital, Shangluo, China
| | - Guolin Ma
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
| | - Jian Yang
- Beijing Engineering Research Center of Mixed Reality and Advanced Display, School of Optics and Photonics, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
2866
|
Dong D, Fang MJ, Tang L, Shan XH, Gao JB, Giganti F, Wang RP, Chen X, Wang XX, Palumbo D, Fu J, Li WC, Li J, Zhong LZ, De Cobelli F, Ji JF, Liu ZY, Tian J. Deep learning radiomic nomogram can predict the number of lymph node metastasis in locally advanced gastric cancer: an international multicenter study. Ann Oncol 2020; 31:912-920. [PMID: 32304748 DOI: 10.1016/j.annonc.2020.04.003] [Citation(s) in RCA: 228] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 03/11/2020] [Accepted: 04/06/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Preoperative evaluation of the number of lymph node metastasis (LNM) is the basis of individual treatment of locally advanced gastric cancer (LAGC). However, the routinely used preoperative determination method is not accurate enough. PATIENTS AND METHODS We enrolled 730 LAGC patients from five centers in China and one center in Italy, and divided them into one primary cohort, three external validation cohorts, and one international validation cohort. A deep learning radiomic nomogram (DLRN) was built based on the images from multiphase computed tomography (CT) for preoperatively determining the number of LNM in LAGC. We comprehensively tested the DLRN and compared it with three state-of-the-art methods. Moreover, we investigated the value of the DLRN in survival analysis. RESULTS The DLRN showed good discrimination of the number of LNM on all cohorts [overall C-indexes (95% confidence interval): 0.821 (0.785-0.858) in the primary cohort, 0.797 (0.771-0.823) in the external validation cohorts, and 0.822 (0.756-0.887) in the international validation cohort]. The nomogram performed significantly better than the routinely used clinical N stages, tumor size, and clinical model (P < 0.05). Besides, DLRN was significantly associated with the overall survival of LAGC patients (n = 271). CONCLUSION A deep learning-based radiomic nomogram had good predictive value for LNM in LAGC. In staging-oriented treatment of gastric cancer, this preoperative nomogram could provide baseline information for individual treatment of LAGC.
Collapse
Affiliation(s)
- D Dong
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - M-J Fang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - L Tang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Radiology Department, Peking University Cancer Hospital & Institute, Beijing, China
| | - X-H Shan
- Department of Radiology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - J-B Gao
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - F Giganti
- Department of Radiology, University College London Hospital NHS Foundation Trust, London; Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London, London, UK; Department of Radiology, Experimental Imaging Centre, San Raffaele Scientific Institute, Milan, Italy
| | - R-P Wang
- Department of Radiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - X Chen
- Department of Radiology, Guangdong Provincial People's Hospital/Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China; Department of Radiology, Guangzhou First People's Hospital, Guangzhou, Guangdong, China
| | - X-X Wang
- Department of Radiology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - D Palumbo
- Department of Radiology, Experimental Imaging Centre, San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - J Fu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Radiology Department, Peking University Cancer Hospital & Institute, Beijing, China
| | - W-C Li
- Department of Radiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - J Li
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - L-Z Zhong
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - F De Cobelli
- Department of Radiology, Experimental Imaging Centre, San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - J-F Ji
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China.
| | - Z-Y Liu
- Department of Radiology, Guangdong Provincial People's Hospital/Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.
| | - J Tian
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China; Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine, Beihang University, Beijing, China; Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, China.
| |
Collapse
|
2867
|
Lin S, Lin X, Li X, Chen M, Mao R. Making Qualitative Intestinal Stricture Quantitative: Embracing Radiomics in IBD. Inflamm Bowel Dis 2020; 26:743-745. [PMID: 31504502 DOI: 10.1093/ibd/izz197] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Indexed: 12/29/2022]
Abstract
Quantifying intestinal fibrosis is an important but challenging issue in stricturing CD. With the advent of radiomics, MRE and CTE are highly minable data rather than merely pictures, which will greatly facilitate precision medicine in intestinal fibrosis of CD.
Collapse
Affiliation(s)
- Sinan Lin
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, P.R. China.,Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, USA
| | - Xiaoqin Lin
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, P.R. China
| | - Xuehua Li
- Department of Radiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, P.R. China
| | - Minhu Chen
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, P.R. China
| | - Ren Mao
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, P.R. China.,Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, USA
| |
Collapse
|
2868
|
Basler L, Gabryś HS, Hogan SA, Pavic M, Bogowicz M, Vuong D, Tanadini-Lang S, Förster R, Kudura K, Huellner MW, Dummer R, Guckenberger M, Levesque MP. Radiomics, Tumor Volume, and Blood Biomarkers for Early Prediction of Pseudoprogression in Patients with Metastatic Melanoma Treated with Immune Checkpoint Inhibition. Clin Cancer Res 2020; 26:4414-4425. [PMID: 32253232 DOI: 10.1158/1078-0432.ccr-20-0020] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 03/09/2020] [Accepted: 04/01/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE We assessed the predictive potential of positron emission tomography (PET)/CT-based radiomics, lesion volume, and routine blood markers for early differentiation of pseudoprogression from true progression at 3 months. EXPERIMENTAL DESIGN 112 patients with metastatic melanoma treated with immune checkpoint inhibition were included in our study. Median follow-up duration was 22 months. 716 metastases were segmented individually on CT and 2[18F]fluoro-2-deoxy-D-glucose (FDG)-PET imaging at three timepoints: baseline (TP0), 3 months (TP1), and 6 months (TP2). Response was defined on a lesion-individual level (RECIST 1.1) and retrospectively correlated with FDG-PET/CT radiomic features and the blood markers LDH/S100. Seven multivariate prediction model classes were generated. RESULTS Two-year (median) overall survival, progression-free survival, and immune progression-free survival were 69% (not reached), 24% (6 months), and 42% (16 months), respectively. At 3 months, 106 (16%) lesions had progressed, of which 30 (5%) were identified as pseudoprogression at 6 months. Patients with pseudoprogressive lesions and without true progressive lesions had a similar outcome to responding patients and a significantly better 2-year overall survival of 100% (30 months), compared with 15% (10 months) in patients with true progressions/without pseudoprogression (P = 0.002). Patients with mixed progressive/pseudoprogressive lesions were in between at 53% (25 months). The blood prediction model (LDH+S100) achieved an AUC = 0.71. Higher LDH/S100 values indicated a low chance of pseudoprogression. Volume-based models: AUC = 0.72 (TP1) and AUC = 0.80 (delta-volume between TP0/TP1). Radiomics models (including/excluding volume-related features): AUC = 0.79/0.78. Combined blood/volume model: AUC = 0.79. Combined blood/radiomics model (including volume-related features): AUC = 0.78. The combined blood/radiomics model (excluding volume-related features) performed best: AUC = 0.82. CONCLUSIONS Noninvasive PET/CT-based radiomics, especially in combination with blood parameters, are promising biomarkers for early differentiation of pseudoprogression, potentially avoiding added toxicity or delayed treatment switch.
Collapse
Affiliation(s)
- Lucas Basler
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Hubert S Gabryś
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Sabrina A Hogan
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Matea Pavic
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Marta Bogowicz
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Diem Vuong
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Stephanie Tanadini-Lang
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Robert Förster
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Ken Kudura
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Martin W Huellner
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Matthias Guckenberger
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Mitchell P Levesque
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
2869
|
Bibault JE, Xing L, Giraud P, El Ayachy R, Giraud N, Decazes P, Burgun A, Giraud P. Radiomics: A primer for the radiation oncologist. Cancer Radiother 2020; 24:403-410. [PMID: 32265157 DOI: 10.1016/j.canrad.2020.01.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 01/22/2020] [Indexed: 12/14/2022]
Abstract
PURPOSE Radiomics are a set of methods used to leverage medical imaging and extract quantitative features that can characterize a patient's phenotype. All modalities can be used with several different software packages. Specific informatics methods can then be used to create meaningful predictive models. In this review, we will explain the major steps of a radiomics analysis pipeline and then present the studies published in the context of radiation therapy. METHODS A literature review was performed on Medline using the search engine PubMed. The search strategy included the search terms "radiotherapy", "radiation oncology" and "radiomics". The search was conducted in July 2019 and reference lists of selected articles were hand searched for relevance to this review. RESULTS A typical radiomics workflow always includes five steps: imaging and segmenting, data curation and preparation, feature extraction, exploration and selection and finally modeling. In radiation oncology, radiomics studies have been published to explore different clinical outcome in lung (n=5), head and neck (n=5), esophageal (n=3), rectal (n=3), pancreatic (n=2) cancer and brain metastases (n=2). The quality of these retrospective studies is heterogeneous and their results have not been translated to the clinic. CONCLUSION Radiomics has a great potential to predict clinical outcome and better personalize treatment. But the field is still young and constantly evolving. Improvement in bias reduction techniques and multicenter studies will hopefully allow more robust and generalizable models.
Collapse
Affiliation(s)
- J-E Bibault
- Radiation Oncology Department, hôpital européen Georges-Pompidou, Assistance publique-Hôpitaux de Paris, 20, rue Leblanc, 75015 Paris, France; Université de Paris, 85, boulevard Saint-Germain, 75006 Paris, France; Inserm, UMR 1138, Team 22: Information Sciences to support Personalized Medicine, 15, rue de l'École-de-Médecine, 75006 Paris, France.
| | - L Xing
- Laboratory of Artificial Intelligence in Medicine and Biomedical Physics, Stanford University School of Medicine, 875 Blake Wilbur Drive, 94305-5847 Stanford, CA, USA
| | - P Giraud
- Inserm, UMR 1138, Team 22: Information Sciences to support Personalized Medicine, 15, rue de l'École-de-Médecine, 75006 Paris, France
| | - R El Ayachy
- Inserm, UMR 1138, Team 22: Information Sciences to support Personalized Medicine, 15, rue de l'École-de-Médecine, 75006 Paris, France
| | - N Giraud
- Radiation Oncology Department, CHU de Bordeaux, hôpital Haut-Lévêque, avenue Magellan, 33600 Pessac, France
| | - P Decazes
- Nuclear Medicine Department, centre Henri-Becquerel, 1, rue d'Amiens, 76038 Rouen, France; Quantif, EA 4108, université de Rouen, avenue de l'Université, 76801 Saint-Étienne-du-Rouvray, France
| | - A Burgun
- Université de Paris, 85, boulevard Saint-Germain, 75006 Paris, France; Inserm, UMR 1138, Team 22: Information Sciences to support Personalized Medicine, 15, rue de l'École-de-Médecine, 75006 Paris, France; Biomedical Informatics and Public Health Department, hôpital européen Georges-Pompidou, Assistance publique-hôpitaux de Paris, 20, rue Leblanc, 75015 Paris, France
| | - P Giraud
- Radiation Oncology Department, hôpital européen Georges-Pompidou, Assistance publique-Hôpitaux de Paris, 20, rue Leblanc, 75015 Paris, France; Université de Paris, 85, boulevard Saint-Germain, 75006 Paris, France
| |
Collapse
|
2870
|
Fu J, Zhong X, Li N, Van Dams R, Lewis J, Sung K, Raldow AC, Jin J, Qi XS. Deep learning-based radiomic features for improving neoadjuvant chemoradiation response prediction in locally advanced rectal cancer. Phys Med Biol 2020; 65:075001. [PMID: 32092710 DOI: 10.1088/1361-6560/ab7970] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Radiomic features achieve promising results in cancer diagnosis, treatment response prediction, and survival prediction. Our goal is to compare the handcrafted (explicitly designed) and deep learning (DL)-based radiomic features extracted from pre-treatment diffusion-weighted magnetic resonance images (DWIs) for predicting neoadjuvant chemoradiation treatment (nCRT) response in patients with locally advanced rectal cancer (LARC). 43 Patients receiving nCRT were included. All patients underwent DWIs before nCRT and total mesorectal excision surgery 6-12 weeks after completion of nCRT. Gross tumor volume (GTV) contours were drawn by an experienced radiation oncologist on DWIs. The patient-cohort was split into the responder group (n = 22) and the non-responder group (n = 21) based on the post-nCRT response assessed by postoperative pathology, MRI or colonoscopy. Handcrafted and DL-based features were extracted from the apparent diffusion coefficient (ADC) map of the DWI using conventional computer-aided diagnosis methods and a pre-trained convolution neural network, respectively. Least absolute shrinkage and selection operator (LASSO)-logistic regression models were constructed using extracted features for predicting treatment response. The model performance was evaluated with repeated 20 times stratified 4-fold cross-validation using receiver operating characteristic (ROC) curves and compared using the corrected paired t-test. The model built with handcrafted features achieved the mean area under the ROC curve (AUC) of 0.64, while the one built with DL-based features yielded the mean AUC of 0.73. The corrected paired t-test on AUC showed P-value < 0.05. DL-based features extracted from pre-treatment DWIs achieved significantly better classification performance compared with handcrafted features for predicting nCRT response in patients with LARC.
Collapse
Affiliation(s)
- Jie Fu
- Department of Radiation Oncology, University of California, Los Angeles, CA 90095, United States of America
| | | | | | | | | | | | | | | | | |
Collapse
|
2871
|
Hofland J, Kaltsas G, de Herder WW. Advances in the Diagnosis and Management of Well-Differentiated Neuroendocrine Neoplasms. Endocr Rev 2020; 41:bnz004. [PMID: 31555796 PMCID: PMC7080342 DOI: 10.1210/endrev/bnz004] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 02/28/2020] [Indexed: 02/07/2023]
Abstract
Neuroendocrine neoplasms constitute a diverse group of tumors that derive from the sensory and secretory neuroendocrine cells and predominantly arise within the pulmonary and gastrointestinal tracts. The majority of these neoplasms have a well-differentiated grade and are termed neuroendocrine tumors (NETs). This subgroup is characterized by limited proliferation and patients affected by these tumors carry a good to moderate prognosis. A substantial subset of patients presenting with a NET suffer from the consequences of endocrine syndromes as a result of the excessive secretion of amines or peptide hormones, which can impair their quality of life and prognosis. Over the past 15 years, critical developments in tumor grading, diagnostic biomarkers, radionuclide imaging, randomized controlled drug trials, evidence-based guidelines, and superior prognostic outcomes have substantially altered the field of NET care. Here, we review the relevant advances to clinical practice that have significantly upgraded our approach to NET patients, both in diagnostic and in therapeutic options.
Collapse
Affiliation(s)
- Johannes Hofland
- ENETS Center of Excellence, Section of Endocrinology, Department of Internal Medicine, Erasmus MC Cancer Center, Erasmus MC, Rotterdam, The Netherlands
| | - Gregory Kaltsas
- 1st Department of Propaupedic Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Wouter W de Herder
- ENETS Center of Excellence, Section of Endocrinology, Department of Internal Medicine, Erasmus MC Cancer Center, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
2872
|
Hu W, Yang H, Xu H, Mao Y. Radiomics based on artificial intelligence in liver diseases: where we are? Gastroenterol Rep (Oxf) 2020; 8:90-97. [PMID: 32280468 PMCID: PMC7136719 DOI: 10.1093/gastro/goaa011] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/22/2019] [Accepted: 10/27/2019] [Indexed: 12/12/2022] Open
Abstract
Radiomics uses computers to extract a large amount of information from different types of images, form various quantifiable features, and select relevant features using artificial-intelligence algorithms to build models, in order to predict the outcomes of clinical problems (such as diagnosis, treatment, prognosis, etc.). The study of liver diseases by radiomics will contribute to early diagnosis and treatment of liver diseases and improve survival and cure rates of liver diseases. This field is currently in the ascendant and may have great development in the future. Therefore, we summarize the progress of current research in this article and then point out the related deficiencies and the direction of future research.
Collapse
Affiliation(s)
- Wenmo Hu
- Department of Liver Surgery, Peking Union Medical College Hospital, PUMC, Chinese Academy of Medical Sciences, Beijing, P. R. China
| | - Huayu Yang
- Department of Liver Surgery, Peking Union Medical College Hospital, PUMC, Chinese Academy of Medical Sciences, Beijing, P. R. China
| | - Haifeng Xu
- Department of Liver Surgery, Peking Union Medical College Hospital, PUMC, Chinese Academy of Medical Sciences, Beijing, P. R. China
| | - Yilei Mao
- Department of Liver Surgery, Peking Union Medical College Hospital, PUMC, Chinese Academy of Medical Sciences, Beijing, P. R. China
| |
Collapse
|
2873
|
Jiang C, Kong Z, Zhang Y, Liu S, Liu Z, Chen W, Liu P, Liu D, Wang Y, Lyu Y, Zhao D, Wang Y, You H, Feng F, Ma W. Conventional magnetic resonance imaging-based radiomic signature predicts telomerase reverse transcriptase promoter mutation status in grade II and III gliomas. Neuroradiology 2020; 62:803-813. [PMID: 32239241 DOI: 10.1007/s00234-020-02392-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/27/2020] [Indexed: 01/19/2023]
Abstract
PURPOSE Telomerase reverse transcriptase (TERT) promoter mutation status is an important biomarker for the precision diagnosis and prognosis prediction of lower grade glioma (LGG). This study aimed to construct a radiomic signature to noninvasively predict the TERT promoter status in LGGs. METHODS Eighty-three local patients with pathology-confirmed LGG were retrospectively included as a training cohort, and 33 patients from The Cancer Imaging Archive (TCIA) were used as for independent validation. Three types of regions of interest (ROIs), which covered the tumor, peri-tumoral area, and tumor plus peri-tumoral area, were delineated on three-dimensional contrast-enhanced T1 (3D-CE-T1)-weighted and T2-weighted images. One hundred seven shape, first-order, and texture radiomic features from each modality under each ROI were extracted and selected through least absolute shrinkage and selection operator. Radiomic signatures were constructed with multiple classifiers and evaluated using receiver operating characteristic (ROC) analysis. The tumors were also stratified according to IDH status. RESULTS Three radiomic signatures, namely, tumoral radiomic signature, tumoral plus peri-tumoral radiomic signature, and fusion radiomic signature, were built, all of which exhibited good accuracy and balanced sensitivity and specificity. The tumoral signature displayed the best performance, with area under the ROC curves (AUC) of 0.948 (0.903-0.993) in the training cohort and 0.827 (0.667-0.988) in the validation cohort. In the IDH subgroups, the AUCs of the tumoral signature ranged from 0.750 to 0.940. CONCLUSION The MRI-based radiomic signature is reliable for noninvasive evaluation of TERT promoter mutations in LGG regardless of the IDH status. The inclusion of peri-tumoral area did not significantly improve the performance.
Collapse
Affiliation(s)
- Chendan Jiang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ziren Kong
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yiwei Zhang
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, China
| | - Sirui Liu
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, China
| | - Zeyu Liu
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, China
| | - Wenlin Chen
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Penghao Liu
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Delin Liu
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yaning Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuelei Lyu
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, China.,Department of Radiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Dachun Zhao
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hui You
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, China.
| | - Feng Feng
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, China
| | - Wenbin Ma
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
2874
|
Sun Y, Bai H, Xia W, Wang D, Zhou B, Zhao X, Yang G, Xu L, Zhang W, Liu P, Xu J, Meng S, Liu R, Gao X. Predicting the Outcome of Transcatheter Arterial Embolization Therapy for Unresectable Hepatocellular Carcinoma Based on Radiomics of Preoperative Multiparameter MRI. J Magn Reson Imaging 2020; 52:1083-1090. [PMID: 32233054 DOI: 10.1002/jmri.27143] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/04/2020] [Accepted: 03/04/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND In unresectable hepatocellular carcinoma (HCC), methods to predict patients at increased risk of progression are required. PURPOSE To investigate the feasibility of radiomics model in predicting early progression of unresectable HCC after transcatheter arterial chemoembolization (TACE) therapy using preoperative multiparametric magnetic resonance imaging (MP-MRI). STUDY TYPE Retrospective. POPULATION A total of 84 patients with BCLC B stage HCC from one medical center. According to the modified response evaluation criteria in solid tumors, patients who progressed at 6 months after TACE therapy were assigned as the progressive disease (PD) group (n = 32). Patients whose MRI was performed on four devices were divided into a training cohort (n = 67). Patients whose MRI was performed on other than the previous four devices were used as the testing set (n = 17). FIELD STRENGTH/SEQUENCE 3.0T, 1.5T axial T2 -weighted imaging (T2 WI), diffusion-weighted imaging (DWI, b = 0, 500 s/mm2 ), and apparent diffusion coefficient (ADC) ASSESSMENT: PD was confirmed via imaging studies with MRI. Risk factors, including age, alpha fetoprotein (AFP), size, and radiomic-related features of PD were assessed. In addition, the discrimination ability of each radiomics signature was tested on an independent testing set. STATISTICAL TESTS The area under the receiver-operator characteristic (ROC) curve (AUC) was used to evaluate the predictive accuracy of the radiomic signature in both the training and testing sets. The results indicated that the MP-MRI model achieved the greatest benefit. RESULTS In the testing set, the model based on DWI features presented an AUC of (b = 0, 0.786; b = 500, 0.729), followed by T2 WI features (0.729) and ADC (0.714). The AUC of the MP-MRI signature was increased to 0.800 compared to any single MRI signature. The multivariate logistic analysis identified the radiomics signature as independent parameters of PD, while clinical information such as age, AFP, size, etc., had no significance in the PD group. DATA CONCLUSION Preoperative MP-MRI has the potential to predict the outcome of TACE therapy for unresectable HCC. In addition, these image features may be complementary to the current staging systems of HCC patients. LEVEL OF EVIDENCE 2. TECHNICAL EFFICACY STAGE 3. J. Magn. Reson. Imaging 2020;52:1083-1090.
Collapse
Affiliation(s)
- Yuejun Sun
- Department of Interventional Radiology, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Honglin Bai
- University of Science and Technology of China, Hefei, China.,Department of Medical Imaging, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Beijing, China
| | - Wei Xia
- Department of Medical Imaging, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Beijing, China
| | - Dong Wang
- Department of Interventional Radiology, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bo Zhou
- Department of Interventional Radiology, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xingyu Zhao
- University of Science and Technology of China, Hefei, China.,Department of Medical Imaging, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Beijing, China
| | - Guowei Yang
- Department of Interventional Radiology, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ligang Xu
- Department of Interventional Radiology, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Zhang
- Department of Interventional Radiology, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Pingping Liu
- Department of Interventional Radiology, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiacheng Xu
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Siyu Meng
- Department of Electro-Optical Engineering, Changchun University of Science and Technology, Changchun, China
| | - Rong Liu
- Department of Interventional Radiology, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Institution of Medical Imaging, Shanghai, China
| | - Xin Gao
- Department of Medical Imaging, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
2875
|
Jiang Y, Wang H, Wu J, Chen C, Yuan Q, Huang W, Li T, Xi S, Hu Y, Zhou Z, Xu Y, Li G, Li R. Noninvasive imaging evaluation of tumor immune microenvironment to predict outcomes in gastric cancer. Ann Oncol 2020; 31:760-768. [PMID: 32240794 DOI: 10.1016/j.annonc.2020.03.295] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 03/15/2020] [Accepted: 03/18/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The tumor immune microenvironment can provide prognostic and predictive information. A previously validated ImmunoScore of Gastric Cancer (ISGC) evaluates both lymphoid and myeloid cells in the tumor core and invasive margin with immunohistochemical staining of surgical specimens. We aimed to develop a noninvasive radiomics-based predictor of ISGC. PATIENTS AND METHODS In this retrospective study including four independent cohorts of 1778 patients, we extracted 584 quantitative features from the intratumoral and peritumoral regions on contrast-enhanced computed tomography images. A radiomic signature [radiomics ImmunoScore (RIS)] was constructed to predict ISGC using regularized logistic regression. We further evaluated its association with prognosis and chemotherapy response. RESULTS A 13-feature radiomic signature for ISGC was developed and validated in three independent cohorts (area under the curve = 0.786, 0.745, and 0.766). The RIS signature was significantly associated with both disease-free and overall survival in the training and all validation cohorts [hazard ratio (HR) range: 0.296-0.487, all P < 0.001]. In multivariable analysis, the RIS remained an independent prognostic factor adjusting for clinicopathologic variables (adjusted HR range: 0.339-0.605, all P < 0.003). For stage II and stage III disease, patients with a high RIS derived survival benefit from adjuvant chemotherapy {HR = 0.436 [95% confidence interval (CI) 0.253-0.753], P = 0.002; HR = 0.591 (95% CI 0.428-0.818), P < 0.001, respectively}, whereas those with a low RIS did not. CONCLUSION The RIS is a reliable tool for evaluation of immunoscore and retains the prognostic significance in gastric cancer. Future prospective studies are required to confirm its potential to predict treatment response and select patients who will benefit from chemotherapy.
Collapse
Affiliation(s)
- Y Jiang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou; Guangdong Provincial Key Laboratory on Precision and Minimally Invasive Medicine for Gastrointestinal Cancers, Guangzhou, China; Department of Radiation Oncology, Stanford University School of Medicine, Stanford, USA
| | - H Wang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, USA
| | - J Wu
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, USA
| | - C Chen
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Q Yuan
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - W Huang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou; Guangdong Provincial Key Laboratory on Precision and Minimally Invasive Medicine for Gastrointestinal Cancers, Guangzhou, China
| | - T Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou; Guangdong Provincial Key Laboratory on Precision and Minimally Invasive Medicine for Gastrointestinal Cancers, Guangzhou, China
| | - S Xi
- The Seventh Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Y Hu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou; Guangdong Provincial Key Laboratory on Precision and Minimally Invasive Medicine for Gastrointestinal Cancers, Guangzhou, China
| | - Z Zhou
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Y Xu
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - G Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou; Guangdong Provincial Key Laboratory on Precision and Minimally Invasive Medicine for Gastrointestinal Cancers, Guangzhou, China.
| | - R Li
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, USA.
| |
Collapse
|
2876
|
Bourbonne V, Fournier G, Vallières M, Lucia F, Doucet L, Tissot V, Cuvelier G, Hue S, Le Penn Du H, Perdriel L, Bertrand N, Staroz F, Visvikis D, Pradier O, Hatt M, Schick U. External Validation of an MRI-Derived Radiomics Model to Predict Biochemical Recurrence after Surgery for High-Risk Prostate Cancer. Cancers (Basel) 2020; 12:E814. [PMID: 32231077 PMCID: PMC7226108 DOI: 10.3390/cancers12040814] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 03/21/2020] [Accepted: 03/26/2020] [Indexed: 12/29/2022] Open
Abstract
Adjuvant radiotherapy after prostatectomy was recently challenged by early salvage radiotherapy, which highlighted the need for biomarkers to improve risk stratification. Therefore, we developed an MRI ADC map-derived radiomics model to predict biochemical recurrence (BCR) and BCR-free survival (bRFS) after surgery. Our goal in this work was to externally validate this radiomics-based prediction model. EXPERIMENTAL DESIGN A total of 195 patients with a high recurrence risk of prostate cancer (pT3-4 and/or R1 and/or Gleason's score > 7) were retrospectively included in two institutions. Patients with postoperative PSA (Prostate Specific Antigen) > 0.04 ng/mL or lymph node involvement were excluded. Radiomics features were extracted from T2 and ADC delineated tumors. A total of 107 patients from Institution 1 were used to retrain the previously published model. The retrained model was then applied to 88 patients from Institution 2 for external validation. BCR predictions were evaluated using AUC (Area Under the Curve), accuracy, and bRFS using Kaplan-Meier curves. RESULTS With a median follow-up of 46.3 months, 52/195 patients experienced BCR. In the retraining cohort, the clinical prediction model (combining the number of risk factors and postoperative PSA) demonstrated moderate predictive power (accuracy of 63%). The radiomics model (ADC-based SZEGLSZM) predicted BCR with an accuracy of 78% and allowed for significant stratification of patients for bRFS (p < 0.0001). In Institution 2, this radiomics model remained predictive of BCR (accuracy of 0.76%) contrary to the clinical model (accuracy of 0.56%). CONCLUSIONS The recently developed MRI ADC map-based radiomics model was validated in terms of its predictive accuracy of BCR and bRFS after prostatectomy in an external cohort.
Collapse
Affiliation(s)
- Vincent Bourbonne
- Department of Radiation Oncology, CHRU Brest, 29200 Brest, France; (F.L.); (O.P.); (U.S.)
- LaTIM, INSERM, UMR 1101, CHRU Brest, 29200 Brest, France; (M.V.); (D.V.); (M.H.)
| | | | - Martin Vallières
- LaTIM, INSERM, UMR 1101, CHRU Brest, 29200 Brest, France; (M.V.); (D.V.); (M.H.)
- Medical Physics Unit, McGill University, Montreal, QC H3A 0G4, Canada
| | - François Lucia
- Department of Radiation Oncology, CHRU Brest, 29200 Brest, France; (F.L.); (O.P.); (U.S.)
- LaTIM, INSERM, UMR 1101, CHRU Brest, 29200 Brest, France; (M.V.); (D.V.); (M.H.)
| | - Laurent Doucet
- Anatomopathology Department, CHRU Brest, 29200 Brest, France;
| | | | - Gilles Cuvelier
- Urology Department, Cornouaille Hospital, 29000 Quimper, France;
| | - Stephane Hue
- Radiology Department, Cornouaille Hospital, 29000 Quimper, France;
| | | | - Luc Perdriel
- Radiology Department, Clinique St Michel, 29000 Quimper, France;
| | | | - Frederic Staroz
- Anatomopathology Department, Ouest Pathologie, 29000 Quimper, France;
| | - Dimitris Visvikis
- LaTIM, INSERM, UMR 1101, CHRU Brest, 29200 Brest, France; (M.V.); (D.V.); (M.H.)
| | - Olivier Pradier
- Department of Radiation Oncology, CHRU Brest, 29200 Brest, France; (F.L.); (O.P.); (U.S.)
- LaTIM, INSERM, UMR 1101, CHRU Brest, 29200 Brest, France; (M.V.); (D.V.); (M.H.)
| | - Mathieu Hatt
- LaTIM, INSERM, UMR 1101, CHRU Brest, 29200 Brest, France; (M.V.); (D.V.); (M.H.)
| | - Ulrike Schick
- Department of Radiation Oncology, CHRU Brest, 29200 Brest, France; (F.L.); (O.P.); (U.S.)
- LaTIM, INSERM, UMR 1101, CHRU Brest, 29200 Brest, France; (M.V.); (D.V.); (M.H.)
| |
Collapse
|
2877
|
Liver-specific 3D sectioning molds for correlating in vivo CT and MRI with tumor histopathology in woodchucks (Marmota monax). PLoS One 2020; 15:e0230794. [PMID: 32214365 PMCID: PMC7098627 DOI: 10.1371/journal.pone.0230794] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 03/08/2020] [Indexed: 12/19/2022] Open
Abstract
Purpose To evaluate the spatial registration and correlation of liver and tumor histopathology sections with corresponding in vivo CT and MRI using 3D, liver-specific cutting molds in a woodchuck (Marmota monax) hepatic tumor model. Methods Five woodchucks chronically infected with woodchuck hepatitis virus following inoculation at birth and with confirmed hepatic tumors were imaged by contrast enhanced CT or MRI. Virtual 3D liver or tumor models were generated by segmentation of in vivo CT or MR imaging. A specimen-specific cavity was created inside a block containing cutting slots aligned with an imaging plane using computer-aided design software, and the final cutting molds were fabricated using a 3D printer. Livers were resected two days after initial imaging, fixed with formalin or left unfixed, inserted into the 3D molds, and cut into parallel pieces by passing a sharp blade through the parallel slots in the mold. Histopathology sections were acquired and their spatial overlap with in vivo image slices was quantified using the Dice similarity coefficient (DSC). Results Imaging of the woodchucks revealed heterogeneous hepatic tumors of varying size, number, and location. Specimen-specific 3D molds provided accurate co-localization of histopathology of whole livers, liver lobes, and pedunculated tumors with in vivo CT and MR imaging, with or without tissue fixation. Visual inspection of histopathology sections and corresponding in vivo image slices revealed spatial registration of analogous pathologic features. The mean DSC for all specimens was 0.83+/-0.05. Conclusion Use of specimen-specific 3D molds for en bloc liver dissection provided strong spatial overlap and feature correspondence between in vivo image slices and histopathology sections.
Collapse
|
2878
|
Vaugier L, Ferrer L, Mengue L, Jouglar E. Radiomics for radiation oncologists: are we ready to go? BJR Open 2020; 2:20190046. [PMID: 33178967 PMCID: PMC7594896 DOI: 10.1259/bjro.20190046] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 03/06/2020] [Accepted: 03/09/2020] [Indexed: 12/19/2022] Open
Abstract
Radiomics have emerged as an exciting field of research over the past few years, with very wide potential applications in personalised and precision medicine of the future. Radiomics-based approaches are still however limited in daily clinical practice in oncology. This review focus on how radiomics could be incorporated into the radiation therapy pipeline, and globally help the radiation oncologist, from the tumour diagnosis to follow-up after treatment. Radiomics could impact on all steps of the treatment pipeline, once the limitations in terms of robustness and reproducibility are overcome. Major ongoing efforts should be made to collect and share data in the most standardised manner possible.
Collapse
Affiliation(s)
- Loïg Vaugier
- Department of Radiation Oncology, Institut de Cancérologie de l'Ouest, Nantes - Saint Herblain, France
| | - Ludovic Ferrer
- Department of Medical Physics, Institut de Cancérologie de l'Ouest, Nantes - Saint Herblain, France
| | - Laurence Mengue
- Department of Radiation Oncology, Institut de Cancérologie de l'Ouest, Nantes - Saint Herblain, France
| | - Emmanuel Jouglar
- Department of Radiation Oncology, Institut de Cancérologie de l'Ouest, Nantes - Saint Herblain, France
| |
Collapse
|
2879
|
Li Y, Han G, Wu X, Li Z, Zhao K, Zhang Z, Liu Z, Liang C. Normalization of multicenter CT radiomics by a generative adversarial network method. Phys Med Biol 2020; 66. [PMID: 32209747 DOI: 10.1088/1361-6560/ab8319] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 03/25/2020] [Indexed: 12/15/2022]
Abstract
PURPOSE To reduce the variability of radiomics features caused by computed tomography (CT) imaging protocols through using a generative adversarial network (GAN) method. MATERIAL AND METHODS In this study, we defined a set of images acquired with a certain imaging protocol as a domain, and a total of 4 domains (A, B, C, and T [target]) from 3 different scanners were included. In dataset#1, 60 patinets for each domain were collected. Datasets#2 and #3 included 40 slices of spleen for each of the domains. In dataset#4, the slices of 3 colorectal cancer groups (n = 28, 38, and 32) were separately retrieved from 3 different scanners, and each group contained short-term and long-term survivors. 77 features were extracted for evaluation by comparing features distributions. First, we trained the GAN model on dataset#1 to learn how to normalize images from domains A, B, and C to T. Next, by comparing feature distributions between normalized images of the different domains, we identified the appropriate model and assessed it , in dataset #2 and dataset#3, respectively. Finally, to investigate whether our proposed method could facilitate multicenter radiomics analysis, we built the lasso classifier to distinguish short-term from long-term survivors based on a certain group in dataset#4, and validate it in another two groups, which formed a cross-validation between groups in dataset#4. RESULTS After normalization, the percentage of aligned features between domains A vs T, B vs T, and C vs T increased from 10.4 %, 18.2%, and 50.1% to 93.5%, 89.6%, and 77.9%, respectively. In the cross-validation results, average improvement of the area under the receiver operating characteristic curve achieved 11% (3%-32%). CONCLUSION Our proposed GAN-based normalization method could reduce the variability of radiomics features caused by different CT imaging protocols and facilitate multicenter radiomics analysis.
Collapse
Affiliation(s)
- Yajun Li
- South China University of Technology, Guangzhou, Guangdong, CHINA
| | - Guoqiang Han
- College of Electronic and Information Engineering, South China University of Technology, Guangzhou, CHINA
| | - Xiaomei Wu
- South China University of Technology, Guangzhou, Guangdong, CHINA
| | - Zhenhui Li
- Yunnan Cancer Hospital, Kunming, Yunnan, CHINA
| | - Ke Zhao
- South China University of Technology, Guangzhou, Guangdong, CHINA
| | | | - Zaiyi Liu
- Department of Radiology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, CHINA
| | - Changhong Liang
- Radiology, Guangdong General Hospital, Guangzhou, 510080, CHINA
| |
Collapse
|
2880
|
Dong D, Tang L, Li ZY, Fang MJ, Gao JB, Shan XH, Ying XJ, Sun YS, Fu J, Wang XX, Li LM, Li ZH, Zhang DF, Zhang Y, Li ZM, Shan F, Bu ZD, Tian J, Ji JF. Development and validation of an individualized nomogram to identify occult peritoneal metastasis in patients with advanced gastric cancer. Ann Oncol 2020; 30:431-438. [PMID: 30689702 PMCID: PMC6442651 DOI: 10.1093/annonc/mdz001] [Citation(s) in RCA: 283] [Impact Index Per Article: 56.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Occult peritoneal metastasis (PM) in advanced gastric cancer (AGC) patients is highly possible to be missed on computed tomography (CT) images. Patients with occult PMs are subject to late detection or even improper surgical treatment. We therefore aimed to develop a radiomic nomogram to preoperatively identify occult PMs in AGC patients. Patients and methods A total of 554 AGC patients from 4 centers were divided into 1 training, 1 internal validation, and 2 external validation cohorts. All patients’ PM status was firstly diagnosed as negative by CT, but later confirmed by laparoscopy (PM-positive n = 122, PM-negative n = 432). Radiomic signatures reflecting phenotypes of the primary tumor (RS1) and peritoneum region (RS2) were built as predictors of PM from 266 quantitative image features. Individualized nomograms of PM status incorporating RS1, RS2, or clinical factors were developed and evaluated regarding prediction ability. Results RS1, RS2, and Lauren type were significant predictors of occult PM (all P < 0.05). A nomogram of these three factors demonstrated better diagnostic accuracy than the model with RS1, RS2, or clinical factors alone (all net reclassification improvement P < 0.05). The area under curve yielded was 0.958 [95% confidence interval (CI) 0.923–0.993], 0.941 (95% CI 0.904–0.977), 0.928 (95% CI 0.886–0.971), and 0.920 (95% CI 0.862–0.978) for the training, internal, and two external validation cohorts, respectively. Stratification analysis showed that this nomogram had potential generalization ability. Conclusion CT phenotypes of both primary tumor and nearby peritoneum are significantly associated with occult PM status. A nomogram of these CT phenotypes and Lauren type has an excellent prediction ability of occult PM, and may have significant clinical implications on early detection of occult PM for AGC.
Collapse
Affiliation(s)
- D Dong
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Radiology Department, , Peking University Cancer Hospital & Institute, Beijing; University of Chinese Academy of Sciences, Beijing
| | - L Tang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Radiology Department, , Peking University Cancer Hospital & Institute, Beijing
| | - Z-Y Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing
| | - M-J Fang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing; University of Chinese Academy of Sciences, Beijing
| | - J-B Gao
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou
| | - X-H Shan
- Department of Radiology, Affiliated People's Hospital of Jiangsu University, Zhenjiang
| | - X-J Ying
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing
| | - Y-S Sun
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Radiology Department, , Peking University Cancer Hospital & Institute, Beijing
| | - J Fu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Radiology Department, , Peking University Cancer Hospital & Institute, Beijing
| | - X-X Wang
- Department of Radiology, Affiliated People's Hospital of Jiangsu University, Zhenjiang
| | - L-M Li
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou
| | - Z-H Li
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming
| | - D-F Zhang
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming
| | - Y Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing
| | - Z-M Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing
| | - F Shan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing
| | - Z-D Bu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing
| | - J Tian
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing; University of Chinese Academy of Sciences, Beijing; Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine, Beihang University, Beijing, China.
| | - J-F Ji
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing.
| |
Collapse
|
2881
|
Hu HT, Shan QY, Chen SL, Li B, Feng ST, Xu EJ, Li X, Long JY, Xie XY, Lu MD, Kuang M, Shen JX, Wang W. CT-based radiomics for preoperative prediction of early recurrent hepatocellular carcinoma: technical reproducibility of acquisition and scanners. Radiol Med 2020; 125:697-705. [PMID: 32200455 DOI: 10.1007/s11547-020-01174-2] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 03/11/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE To test the technical reproducibility of acquisition and scanners of CT image-based radiomics model for early recurrent hepatocellular carcinoma (HCC). METHODS We included primary HCC patient undergone curative therapies, using early recurrence as endpoint. Four datasets were constructed: 109 images from hospital #1 for training (set 1: 1-mm image slice thickness), 47 images from hospital #1 for internal validation (sets 2 and 3: 1-mm and 10-mm image slice thicknesses, respectively), and 47 images from hospital #2 for external validation (set 4: vastly different from training dataset). A radiomics model was constructed. Radiomics technical reproducibility was measured by overfitting and calibration deviation in external validation dataset. The influence of slice thickness on reproducibility was evaluated in two internal validation datasets. RESULTS Compared with set 1, the model in set 2 indicated favorable prediction efficiency (the area under the curve 0.79 vs. 0.80, P = 0.47) and good calibration (unreliability statistic U: P = 0.33). However, in set 4, significant overfitting (0.63 vs. 0.80, P < 0.01) and calibration deviation (U: P < 0.01) were observed. Similar poor performance was also observed in set 3 (0.56 vs. 0.80, P = 0.02; U: P < 0.01). CONCLUSIONS CT-based radiomics has poor reproducibility between centers. Image heterogeneity, such as slice thickness, can be a significant influencing factor.
Collapse
Affiliation(s)
- Hang-Tong Hu
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, Ultrasomics Artificial Intelligence X-Lab, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhong Shan Road 2, Guangzhou, 510080, China
| | - Quan-Yuan Shan
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, Ultrasomics Artificial Intelligence X-Lab, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhong Shan Road 2, Guangzhou, 510080, China
| | - Shu-Ling Chen
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, Ultrasomics Artificial Intelligence X-Lab, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhong Shan Road 2, Guangzhou, 510080, China
| | - Bin Li
- Clinical trials Unit, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Shi-Ting Feng
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhong Shan Road 2, Guangzhou, 510080, China
| | - Er-Jiao Xu
- Department of Medical Ultrasonics, The Third Affiliated Hospital of Sun Yat-Sen University, Guangdong Key Laboratory of Liver Disease Research, Guangzhou, 510630, Guangdong Province, China
| | - Xin Li
- GE Healthcare, Shanghai, 200030, China
| | - Jian-Yan Long
- Clinical trials Unit, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiao-Yan Xie
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, Ultrasomics Artificial Intelligence X-Lab, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhong Shan Road 2, Guangzhou, 510080, China
| | - Ming-de Lu
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, Ultrasomics Artificial Intelligence X-Lab, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhong Shan Road 2, Guangzhou, 510080, China.,Department of Liver Surgery, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhong Shan Road 2, Guangzhou, 510080, China
| | - Ming Kuang
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, Ultrasomics Artificial Intelligence X-Lab, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhong Shan Road 2, Guangzhou, 510080, China.,Department of Liver Surgery, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhong Shan Road 2, Guangzhou, 510080, China
| | - Jing-Xian Shen
- Department of Radiology, State Key Laboratory of Oncology in South China, The Cancer Center, Sun Yat-sen University, 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, China.
| | - Wei Wang
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, Ultrasomics Artificial Intelligence X-Lab, The First Affiliated Hospital of Sun Yat-Sen University, 58 Zhong Shan Road 2, Guangzhou, 510080, China.
| |
Collapse
|
2882
|
Zhu WS, Shi SY, Yang ZH, Song C, Shen J. Radiomics model based on preoperative gadoxetic acid-enhanced MRI for predicting liver failure. World J Gastroenterol 2020; 26:1208-1220. [PMID: 32231424 PMCID: PMC7093309 DOI: 10.3748/wjg.v26.i11.1208] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 02/18/2020] [Accepted: 02/21/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Postoperative liver failure is the most severe complication in cirrhotic patients with hepatocellular carcinoma (HCC) after major hepatectomy. Current available clinical indexes predicting postoperative residual liver function are not sufficiently accurate.
AIM To determine a radiomics model based on preoperative gadoxetic acid-enhanced magnetic resonance imaging for predicting liver failure in cirrhotic patients with HCC after major hepatectomy.
METHODS For this retrospective study, a radiomics-based model was developed based on preoperative hepatobiliary phase gadoxetic acid-enhanced magnetic resonance images in 101 patients with HCC between June 2012 and June 2018. Sixty-one radiomic features were extracted from hepatobiliary phase images and selected by the least absolute shrinkage and selection operator method to construct a radiomics signature. A clinical prediction model, and radiomics-based model incorporating significant clinical indexes and radiomics signature were built using multivariable logistic regression analysis. The integrated radiomics-based model was presented as a radiomics nomogram. The performances of clinical prediction model, radiomics signature, and radiomics-based model for predicting post-operative liver failure were determined using receiver operating characteristics curve, calibration curve, and decision curve analyses.
RESULTS Five radiomics features from hepatobiliary phase images were selected to construct the radiomics signature. The clinical prediction model, radiomics signature, and radiomics-based model incorporating indocyanine green clearance rate at 15 min and radiomics signature showed favorable performance for predicting postoperative liver failure (area under the curve: 0.809-0.894). The radiomics-based model achieved the highest performance for predicting liver failure (area under the curve: 0.894; 95%CI: 0.823-0.964). The integrated discrimination improvement analysis showed a significant improvement in the accuracy of liver failure prediction when radiomics signature was added to the clinical prediction model (integrated discrimination improvement = 0.117, P = 0.002). The calibration curve and an insignificant Hosmer-Lemeshow test statistic (P = 0.841) demonstrated good calibration of the radiomics-based model. The decision curve analysis showed that patients would benefit more from a radiomics-based prediction model than from a clinical prediction model and radiomics signature alone.
CONCLUSION A radiomics-based model of preoperative gadoxetic acid–enhanced MRI can be used to predict liver failure in cirrhotic patients with HCC after major hepatectomy.
Collapse
Affiliation(s)
- Wang-Shu Zhu
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China
| | - Si-Ya Shi
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China
| | - Ze-Hong Yang
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China
| | - Chao Song
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China
| | - Jun Shen
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, Guangdong Province, China
| |
Collapse
|
2883
|
Dercle L, Fronheiser M, Lu L, Du S, Hayes W, Leung DK, Roy A, Wilkerson J, Guo P, Fojo AT, Schwartz LH, Zhao B. Identification of Non–Small Cell Lung Cancer Sensitive to Systemic Cancer Therapies Using Radiomics. Clin Cancer Res 2020; 26:2151-2162. [DOI: 10.1158/1078-0432.ccr-19-2942] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 11/27/2019] [Accepted: 01/22/2020] [Indexed: 11/16/2022]
|
2884
|
Fiorino C, Guckemberger M, Schwarz M, van der Heide UA, Heijmen B. Technology-driven research for radiotherapy innovation. Mol Oncol 2020; 14:1500-1513. [PMID: 32124546 PMCID: PMC7332218 DOI: 10.1002/1878-0261.12659] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/27/2020] [Accepted: 02/10/2020] [Indexed: 12/16/2022] Open
Abstract
Technology has a pivotal role in the continuous development of radiotherapy. The long road toward modern ‘high‐tech’ radiation oncology has been studded with discoveries and technological innovations that resulted from the interaction of various disciplines. In the last decades, a dramatic technology‐driven revolution has hugely improved the capability of accurately and safely delivering complex‐shaped dose distributions. This has contributed to many clinical improvements, such as the successful management of lung cancer and oligometastatic disease through stereotactic body radiotherapy. Technology‐driven research is an active and lively field with promising potential in several domains, including image guidance, adaptive radiotherapy, integration of artificial intelligence, heavy‐particle therapy, and ‘flash’ ultra‐high dose‐rate radiotherapy. The evolution toward personalized Oncology will deeply influence technology‐driven research, aiming to integrate predictive models and omics analyses into fast and efficient solutions to deliver the best treatment for each single patient. Personalized radiation oncology will need affordable technological solutions for middle‐/low‐income countries, as these are expected to experience the highest increase of cancer incidence and mortality. Moreover, technology solutions for automation of commissioning, quality assurance, safety tests, image segmentation, and plan optimization will be required. Although a large fraction of cancer patients receive radiotherapy, this is certainly not reflected in the worldwide budget for radiotherapy research. Differently from the pharmaceutical companies‐driven research, resources for research in radiotherapy are highly limited to equipment vendors, who can, in turn, initiate a limited number of collaborations with academic research centers. Thus, enhancement of investments in technology‐driven radiotherapy research via public funds, national governments, and the European Union would have a crucial societal impact. It would allow for radiotherapy to further strengthen its role as a highly effective and cost‐efficient cancer treatment modality, and it could facilitate a rapid and equalitarian large‐scale transfer of technology to clinic, with direct impact on patient care.
Collapse
Affiliation(s)
- Claudio Fiorino
- Medical Physics, San Raffaele Scientific Institute, Milano, Italy
| | - Matthias Guckemberger
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Switzerland
| | - Marco Schwarz
- Protontherapy Department, Trento Hospital and TIFPA-INFN, Trento, Italy
| | - Uulke A van der Heide
- Department of Radiation Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands.,Department of Radiation Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ben Heijmen
- Department of Radiation Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| |
Collapse
|
2885
|
Nazari M, Shiri I, Hajianfar G, Oveisi N, Abdollahi H, Deevband MR, Oveisi M, Zaidi H. Noninvasive Fuhrman grading of clear cell renal cell carcinoma using computed tomography radiomic features and machine learning. Radiol Med 2020; 125:754-762. [PMID: 32193870 DOI: 10.1007/s11547-020-01169-z] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 03/05/2020] [Indexed: 01/29/2023]
Abstract
PURPOSE To identify optimal classification methods for computed tomography (CT) radiomics-based preoperative prediction of clear cell renal cell carcinoma (ccRCC) grade. MATERIALS AND METHODS Seventy-one ccRCC patients (31 low grade and 40 high grade) were included in this study. Tumors were manually segmented on CT images followed by the application of three image preprocessing techniques (Laplacian of Gaussian, wavelet filter, and discretization of the intensity values) on delineated tumor volumes. Overall, 2530 radiomics features (tumor shape and size, intensity statistics, and texture) were extracted from each segmented tumor volume. Univariate analysis was performed to assess the association between each feature and the histological condition. Multivariate analysis involved the use of machine learning (ML) algorithms and the following three feature selection algorithms: the least absolute shrinkage and selection operator, Student's t test, and minimum Redundancy Maximum Relevance. These selected features were then used to construct three classification models (SVM, random forest, and logistic regression) to discriminate high from low-grade ccRCC at nephrectomy. Lastly, multivariate model performance was evaluated on the bootstrapped validation cohort using the area under the receiver operating characteristic curve (AUC) metric. RESULTS The univariate analysis demonstrated that among the different image sets, 128 bin-discretized images have statistically significant different texture parameters with a mean AUC of 0.74 ± 3 (q value < 0.05). The three ML-based classifiers showed proficient discrimination between high and low-grade ccRCC. The AUC was 0.78 for logistic regression, 0.62 for random forest, and 0.83 for the SVM model, respectively. CONCLUSION CT radiomic features can be considered as a useful and promising noninvasive methodology for preoperative evaluation of ccRCC Fuhrman grades.
Collapse
Affiliation(s)
- Mostafa Nazari
- Department of Biomedical Engineering and Medical Physics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Isaac Shiri
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211, Geneva 4, Switzerland.
| | - Ghasem Hajianfar
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Science, Tehran, Iran
| | - Niki Oveisi
- School of Population and Public Health, The University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Hamid Abdollahi
- Department of Radiologic Sciences and Medical Physics, Faculty of Allied Medicine, Kerman University, Kerman, Iran
| | - Mohammad Reza Deevband
- Department of Biomedical Engineering and Medical Physics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mehrdad Oveisi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Science, Tehran, Iran.,Department of Computer Science, University of British Columbia, Vancouver, BC, Canada
| | - Habib Zaidi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211, Geneva 4, Switzerland.,Geneva University Neurocenter, Geneva University, Geneva, Switzerland.,Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, Netherlands.,Department of Nuclear Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
2886
|
Dong Y, Zhou L, Xia W, Zhao XY, Zhang Q, Jian JM, Gao X, Wang WP. Preoperative Prediction of Microvascular Invasion in Hepatocellular Carcinoma: Initial Application of a Radiomic Algorithm Based on Grayscale Ultrasound Images. Front Oncol 2020; 10:353. [PMID: 32266138 PMCID: PMC7096379 DOI: 10.3389/fonc.2020.00353] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 02/28/2020] [Indexed: 02/06/2023] Open
Abstract
Objectives: To establish a radiomic algorithm based on grayscale ultrasound images and to make preoperative predictions of microvascular invasion (MVI) in hepatocellular carcinoma (HCC) patients. Methods: In this retrospective study, 322 cases of histopathologically confirmed HCC lesions were included. The classifications based on preoperative grayscale ultrasound images were performed in two stages: (1) classifier #1, MVI-negative and MVI-positive cases; (2) classifier #2, MVI-positive cases were further classified as M1 or M2 cases. The gross-tumoral region (GTR) and peri-tumoral region (PTR) signatures were combined to generate gross- and peri-tumoral region (GPTR) radiomic signatures. The optimal radiomic signatures were further incorporated with vital clinical information. Multivariable logistic regression was used to build radiomic models. Results: Finally, 1,595 radiomic features were extracted from each HCC lesion. At the classifier #1 stage, the radiomic signatures based on features of GTR, PTR, and GPTR showed area under the curve (AUC) values of 0.708 (95% CI, 0.603-0.812), 0.710 (95% CI, 0.609-0.811), and 0.726 (95% CI, 0.625-0.827), respectively. Upon incorporation of vital clinical information, the AUC of the GPTR radiomic algorithm was 0.744 (95% CI, 0.646-0.841). At the classifier #2 stage, the AUC of the GTR radiomic signature was 0.806 (95% CI, 0.667-0.944). Conclusions: Our radiomic algorithm based on grayscale ultrasound images has potential value to facilitate preoperative prediction of MVI in HCC patients. The GTR radiomic signature may be helpful for further discriminating between M1 and M2 levels among MVI-positive patients.
Collapse
Affiliation(s)
- Yi Dong
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Liu Zhou
- Suzhou Institute of Biomedical Engineering and Technology (CAS), Suzhou, China
| | - Wei Xia
- Suzhou Institute of Biomedical Engineering and Technology (CAS), Suzhou, China
| | - Xing-Yu Zhao
- Suzhou Institute of Biomedical Engineering and Technology (CAS), Suzhou, China
| | - Qi Zhang
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jun-Ming Jian
- Suzhou Institute of Biomedical Engineering and Technology (CAS), Suzhou, China
| | - Xin Gao
- Suzhou Institute of Biomedical Engineering and Technology (CAS), Suzhou, China
| | - Wen-Ping Wang
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
2887
|
Pesapane F, Suter MB, Rotili A, Penco S, Nigro O, Cremonesi M, Bellomi M, Jereczek-Fossa BA, Pinotti G, Cassano E. Will traditional biopsy be substituted by radiomics and liquid biopsy for breast cancer diagnosis and characterisation? Med Oncol 2020; 37:29. [PMID: 32180032 DOI: 10.1007/s12032-020-01353-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 02/26/2020] [Indexed: 02/06/2023]
Abstract
The diagnosis of breast cancer currently relies on radiological and clinical evaluation, confirmed by histopathological examination. However, such approach has some limitations as the suboptimal sensitivity, the long turnaround time for recall tests, the invasiveness of the procedure and the risk that some features of target lesions may remain undetected, making re-biopsy a necessity. Recent technological advances in the field of artificial intelligence hold promise in addressing such medical challenges not only in cancer diagnosis, but also in treatment assessment, and monitoring of disease progression. In the perspective of a truly personalised medicine, based on the early diagnosis and individually tailored treatments, two new technologies, namely radiomics and liquid biopsy, are rising as means to obtain information from diagnosis to molecular profiling and response assessment, without the need of a biopsied tissue sample. Radiomics works through the extraction of quantitative peculiar features of cancer from radiological data, while liquid biopsy gets the whole of the malignancy's biology from something as easy as a blood sample. Both techniques hopefully will identify diagnostic and prognostic information of breast cancer potentially reducing the need for invasive (and often difficult to perform) biopsies and favouring an approach that is as personalised as possible for each patient. Nevertheless, such techniques will not substitute tissue biopsy in the near future, and even in further times they will require the aid of other parameters to be correctly interpreted and acted upon.
Collapse
Affiliation(s)
- Filippo Pesapane
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Via Giuseppe Ripamonti, 435, 20141, Milan, MI, Italy.
| | | | - Anna Rotili
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Via Giuseppe Ripamonti, 435, 20141, Milan, MI, Italy
| | - Silvia Penco
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Via Giuseppe Ripamonti, 435, 20141, Milan, MI, Italy
| | - Olga Nigro
- Medical Oncology, ASST Sette Laghi, Viale Borri 57, 21100, Varese, VA, Italy
| | - Marta Cremonesi
- Radiation Research Unit, IEO European Institute of Oncology IRCCS, Via Giuseppe Ripamonti, 435, 20141, Milan, MI, Italy
| | - Massimo Bellomi
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Radiology, IEO European Institute of Oncology IRCCS, Via Giuseppe Ripamonti, 435, 20141, Milan, MI, Italy
| | - Barbara Alicja Jereczek-Fossa
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Radiation Oncology, IEO European Institute of Oncology IRCCS, Via Giuseppe Ripamonti, 435, 20141, Milan, MI, Italy
| | - Graziella Pinotti
- Medical Oncology, ASST Sette Laghi, Viale Borri 57, 21100, Varese, VA, Italy
| | - Enrico Cassano
- Breast Imaging Division, IEO European Institute of Oncology IRCCS, Via Giuseppe Ripamonti, 435, 20141, Milan, MI, Italy
| |
Collapse
|
2888
|
Lin P, Liu WK, Li X, Wan D, Qin H, Li Q, Chen G, He Y, Yang H. MRI-based radiogenomics analysis for predicting genetic alterations in oncogenic signalling pathways in invasive breast carcinoma. Clin Radiol 2020; 75:561.e1-561.e11. [PMID: 32183997 DOI: 10.1016/j.crad.2020.02.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 02/18/2020] [Indexed: 01/23/2023]
Abstract
AIM To investigate the effect of radiomics in the assessment of alterations in canonical cancer pathways in breast cancer. MATERIALS AND METHODS Eighty-eight biopsy-proven breast cancer cases were included in the present study. Radiomics features were extracted from T1-weighted sagittal dynamic contrast-enhanced magnetic resonance imaging (MRI) images. Radiomics signatures were developed to predict genetic alterations in the cell cycle, Myc, PI3K, RTK/RAS, and p53 signalling pathways by using hypothesis testing combined with least absolute shrinkage and selection operator (LASSO) regression analysis. The predictive powers of the models were examined by the area under the curve (AUC) of the receiver operating characteristic curve. RESULTS A total of 5,234 radiomics features were obtained from MRI images based on the tumour region of interest. Hypothesis tests screened 250, 229, 156, 785, and 319 radiomics features that were differentially displayed between cell cycle, Myc, PI3K, RTK/RAS, and p53 alterations and no alteration status. According to the LASSO algorithm, 11, 12, 12, 15, and 13 features were identified for the construction of the radiomics signatures to predict cell cycle, Myc, PI3K, RTK/RAS, and p53 alterations, with AUC values of 0.933, 0.926, 0.956, 0.940, and 0.886, respectively. The cell cycle radiomics score correlated closely with the RTK/RAS and p53 radiomics scores. These signatures were also dysregulated in patients with different oestrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 statuses. CONCLUSION MRI-based radiogenomics analysis exhibits excellent performance in predicting genetic pathways alterations, thus providing a novel approach for non-invasively obtaining genetic-level molecular characteristics of tumours.
Collapse
Affiliation(s)
- P Lin
- Department of Ultrasound, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| | - W K Liu
- Department of Ultrasound, Affiliated Union Hospital of Fujian Medical University, Fuzhou, Fu Jian 350000, China
| | - X Li
- GE Healthcare, Shanghai, China
| | - D Wan
- Department of Ultrasound, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| | - H Qin
- Department of Ultrasound, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| | - Q Li
- Department of Ultrasound, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| | - G Chen
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| | - Y He
- Department of Ultrasound, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| | - H Yang
- Department of Ultrasound, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, China.
| |
Collapse
|
2889
|
Wang Y, Wei W, Liu Z, Liang Y, Liu X, Li Y, Tang Z, Jiang T, Tian J. Predicting the Type of Tumor-Related Epilepsy in Patients With Low-Grade Gliomas: A Radiomics Study. Front Oncol 2020; 10:235. [PMID: 32231995 PMCID: PMC7082349 DOI: 10.3389/fonc.2020.00235] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 02/12/2020] [Indexed: 01/21/2023] Open
Abstract
Purpose: The majority of patients with low-grade gliomas (LGGs) experience tumor-related epilepsy during the disease course. Our study aimed to build a radiomic prediction model for LGG-related epilepsy type based on magnetic resonance imaging (MRI) data. Methods: A total of 205 cases with LGG-related epilepsy were enrolled in the retrospective study and divided into training and validation cohorts (1:1) according to their surgery time. Seven hundred thirty-four radiomic features were extracted from T2-weighted imaging, including six location features. Pearson correlation coefficient, univariate area under curve (AUC) analysis, and least absolute shrinkage and selection operator regression were adopted to select the most relevant features for the epilepsy type to build a radiomic signature. Furthermore, a novel radiomic nomogram was developed for clinical application using the radiomic signature and clinical variables from all patients. Results: Four MRI-based features were selected from the 734 radiomic features, including one location feature. Good discriminative performances were achieved in both training (AUC = 0.859, 95% CI = 0.787–0.932) and validation cohorts (AUC = 0.839, 95% CI = 0.761–0.917) for the type of epilepsy. The accuracies were 80.4 and 80.6%, respectively. The radiomic nomogram also allowed for a high degree of discrimination. All models presented favorable calibration curves and decision curve analyses. Conclusion: Our results suggested that the MRI-based radiomic analysis may predict the type of LGG-related epilepsy to enable individualized therapy for patients with LGG-related epilepsy.
Collapse
Affiliation(s)
- Yinyan Wang
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wei Wei
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China.,School of Electronics and Information, Xi'an Polytechnic University, Xi'an, China.,Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine, Beihang University, Beijing, China.,Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China
| | - Zhenyu Liu
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China.,School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Yuchao Liang
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xing Liu
- Department of Molecular Pathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Yiming Li
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Department of Molecular Pathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Zhenchao Tang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China.,Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine, Beihang University, Beijing, China
| | - Tao Jiang
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Department of Molecular Pathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China.,Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine, Beihang University, Beijing, China.,Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, China.,School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
2890
|
Hettal L, Stefani A, Salleron J, Courrech F, Behm-Ansmant I, Constans JM, Gauchotte G, Vogin G. Radiomics Method for the Differential Diagnosis of Radionecrosis Versus Progression after Fractionated Stereotactic Body Radiotherapy for Brain Oligometastasis. Radiat Res 2020; 193:471-480. [PMID: 32160109 DOI: 10.1667/rr15517.1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Stereotactic radiotherapy (SRT) is recommended for treatment of brain oligometastasis (BoM) in patients with controlled primary disease. Where contrast enhancement enlargement occurs during follow-up, distinguishing between radionecrosis and progression presents a critical challenge. Without pathological confirmation, decision-making may be inappropriate and delayed. Quantitative imaging features extracted from routinely performed examinations are of interest in potentially addressing this problem. We explored the added value of the radiomics method for the differential diagnosis of these two entities. Twenty patients who received SRT for BoM, from any primary location, were included (8 radionecrosis, 12 progressions, pathologically confirmed). We assessed the clinical relevance of 1,766 radiomics features, extracted using IBEX software, from the first T1-weighted postcontrast magnetic resonance imaging (MRI) after SRT showing a lesion modification. We evaluated seven feature-selection methods and 12 classification methods in terms of respective predictive performance. The classification accuracy was measured using Cohen's kappa after leave-one-out cross-validation. In this work, the best predictive power reached was a Cohen's kappa of 0.68 (overall accuracy of 85%), expressing a strong agreement between the algorithm prediction and the histological gold standard. Prediction accuracy was 75% for radionecrosis, and 91% for progression. The area under a curve reached 0.83 using a bagging algorithm trained with the chi-square score features set. These findings indicated that the radiomics method is able to discriminate radionecrosis from progression in an accurate, early and noninvasive way. This promising study is a proof of concept, preceding a larger prospective study for defining a robust model to support decision-making in BoM. In summary, distinguishing between radionecrosis and progression is challenging without pathology. We built a classification model based on imaging data and machine learning. Using this model, we were able predict progression and radionecrosis in, respectively, 91% and 75% of cases.
Collapse
Affiliation(s)
- Liza Hettal
- CNRS UMR 7365 IMoPA, Université de Lorraine, Biopôle, Vandoeuvre-Lès-Nancy, France
| | - Anais Stefani
- Département de Radiothérapie, Institut de Cancérologie de Lorraine, Vandoeuvre-Les-Nancy, France
| | - Julia Salleron
- Département de Cellule Data-biostatistiques, Institut de Cancérologie de Lorraine, Université de Lorraine, Vandoeuvrelès-Nancy, France
| | - Florent Courrech
- Département de Radiothérapie, Institut de Cancérologie de Lorraine, Vandoeuvre-Les-Nancy, France
| | | | | | - Guillaume Gauchotte
- Département d' Anatomie et Cytologie Pathologiques, CHRU Nancy, France.,Département d' INSERM U1256, Université de Lorraine, Nancy, France
| | - Guillaume Vogin
- CNRS UMR 7365 IMoPA, Université de Lorraine, Biopôle, Vandoeuvre-Lès-Nancy, France.,Département de Radiothérapie, Institut de Cancérologie de Lorraine, Vandoeuvre-Les-Nancy, France
| |
Collapse
|
2891
|
Bogowicz M, Jochems A, Deist TM, Tanadini-Lang S, Huang SH, Chan B, Waldron JN, Bratman S, O'Sullivan B, Riesterer O, Studer G, Unkelbach J, Barakat S, Brakenhoff RH, Nauta I, Gazzani SE, Calareso G, Scheckenbach K, Hoebers F, Wesseling FWR, Keek S, Sanduleanu S, Leijenaar RTH, Vergeer MR, Leemans CR, Terhaard CHJ, van den Brekel MWM, Hamming-Vrieze O, van der Heijden MA, Elhalawani HM, Fuller CD, Guckenberger M, Lambin P. Privacy-preserving distributed learning of radiomics to predict overall survival and HPV status in head and neck cancer. Sci Rep 2020; 10:4542. [PMID: 32161279 PMCID: PMC7066122 DOI: 10.1038/s41598-020-61297-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 01/28/2020] [Indexed: 12/23/2022] Open
Abstract
A major challenge in radiomics is assembling data from multiple centers. Sharing data between hospitals is restricted by legal and ethical regulations. Distributed learning is a technique, enabling training models on multicenter data without data leaving the hospitals ("privacy-preserving" distributed learning). This study tested feasibility of distributed learning of radiomics data for prediction of two year overall survival and HPV status in head and neck cancer (HNC) patients. Pretreatment CT images were collected from 1174 HNC patients in 6 different cohorts. 981 radiomic features were extracted using Z-Rad software implementation. Hierarchical clustering was performed to preselect features. Classification was done using logistic regression. In the validation dataset, the receiver operating characteristics (ROC) were compared between the models trained in the centralized and distributed manner. No difference in ROC was observed with respect to feature selection. The logistic regression coefficients were identical between the methods (absolute difference <10-7). In comparison of the full workflow (feature selection and classification), no significant difference in ROC was found between centralized and distributed models for both studied endpoints (DeLong p > 0.05). In conclusion, both feature selection and classification are feasible in a distributed manner using radiomics data, which opens new possibility for training more reliable radiomics models.
Collapse
Grants
- P30 CA016672 NCI NIH HHS
- P50 CA097007 NCI NIH HHS
- R01 DE025248 NIDCR NIH HHS
- R01 CA214825 NCI NIH HHS
- R25 EB025787 NIBIB NIH HHS
- R56 DE025248 NIDCR NIH HHS
- R01 CA218148 NCI NIH HHS
- Swiss National Science Foundation Sinergia grant (310030_173303) and Scientific Exchange grant (IZSEZ0_180524).
- This work was also supported by the Interreg grant EURADIOMICS and the Dutch technology Foundation STW (grant n° 10696 DuCAT and n° P14-19 Radiomics STRaTegy), which is the applied science division of NWO, the Technology Program of the Ministry of Economic Affairs and the Manchester Cancer Research UK major centre grant. The authors also acknowledge financial support from the EU 7th framework program (ARTFORCE - n° 257144, REQUITE - n° 601826), CTMM-TraIT, EUROSTARS (E-DECIDE, DEEPMAM), Kankeronderzoekfonds Limburg from the Health Foundation Limburg, Alpe d’HuZes-KWF (DESIGN), The Dutch Cancer Society, the European Program H2020-2015-17 (ImmunoSABR - n° 733008 and BD2Decide - PHC30-689715), the ERC advanced grant (ERC-ADG-2015, n° 694812 - Hypoximmuno), SME Phase 2 (EU proposal 673780 – RAIL).
- The clinical study used as one of the cohorts was supported by a research grant from Merck (Schweiz) AG.
- Dr. Fuller is a Sabin Family Foundation Fellow. Dr. Fuller receive funding and project-relevant salary support from the National Institutes of Health (NIH), including: National Institute for Dental and Craniofacial Research Award (1R01DE025248-01/R56DE025248-01); National Cancer Institute (NCI) Early Phase Clinical Trials in Imaging and Image-Guided Interventions Program(1R01CA218148-01); National Science Foundation (NSF), Division of Mathematical Sciences; NIH Big Data to Knowledge (BD2K) Program of the National Cancer Institute Early Stage Development of Technologies in Biomedical Computing, Informatics, and Big Data Science Award (1R01CA214825-01); NIH/NCI Cancer Center Support Grant (CCSG) Pilot Research Program Award from the UT MD Anderson CCSG Radiation Oncology and Cancer Imaging Program (P30CA016672) and National Institute of Biomedical Imaging and Bioengineering (NIBIB) Research Education Program (R25EB025787). Dr. Fuller has received direct industry grant support and travel funding from Elekta AB.and Fuller receive funding and project-relevant salary support from NIH/NCI Head and Neck Specialized Programs of Research Excellence (SPORE) Developmental Research Program Award (P50 CA097007-10).
- This project was supported by the Swiss National Science Foundation Sinergia grant (310030_173303)
Collapse
Affiliation(s)
- Marta Bogowicz
- University Hospital Zurich and University of Zurich, Department of Radiation Oncology, Zurich, Switzerland.
- GROW-School for Oncology and Developmental Biology-Maastricht University Medical Centre-, Department of Precision Medicine, The D Lab: Decision Support for Precision Medicine-, Maastricht, The Netherlands.
| | - Arthur Jochems
- GROW-School for Oncology and Developmental Biology-Maastricht University Medical Centre-, Department of Precision Medicine, The D Lab: Decision Support for Precision Medicine-, Maastricht, The Netherlands
| | - Timo M Deist
- GROW-School for Oncology and Developmental Biology-Maastricht University Medical Centre-, Department of Precision Medicine, The D Lab: Decision Support for Precision Medicine-, Maastricht, The Netherlands
| | - Stephanie Tanadini-Lang
- University Hospital Zurich and University of Zurich, Department of Radiation Oncology, Zurich, Switzerland
| | - Shao Hui Huang
- Princess Margaret Cancer Center- University of Toronto, Department of Radiation Oncology, Toronto, Ontario, Canada
| | - Biu Chan
- Princess Margaret Cancer Center- University of Toronto, Department of Radiation Oncology, Toronto, Ontario, Canada
| | - John N Waldron
- Princess Margaret Cancer Center- University of Toronto, Department of Radiation Oncology, Toronto, Ontario, Canada
| | - Scott Bratman
- Princess Margaret Cancer Center- University of Toronto, Department of Radiation Oncology, Toronto, Ontario, Canada
| | - Brian O'Sullivan
- Princess Margaret Cancer Center- University of Toronto, Department of Radiation Oncology, Toronto, Ontario, Canada
| | - Oliver Riesterer
- University Hospital Zurich and University of Zurich, Department of Radiation Oncology, Zurich, Switzerland
- Kantonsspital Aarau, Center for Radiation Oncology- KSA-KSB-, Aarau, Switzerland
| | - Gabriela Studer
- University Hospital Zurich and University of Zurich, Department of Radiation Oncology, Zurich, Switzerland
- Cantonal Hospital Lucerne, Radiation Oncology, Lucerne, Switzerland
| | - Jan Unkelbach
- University Hospital Zurich and University of Zurich, Department of Radiation Oncology, Zurich, Switzerland
| | - Samir Barakat
- GROW-School for Oncology and Developmental Biology-Maastricht University Medical Centre-, Department of Precision Medicine, The D Lab: Decision Support for Precision Medicine-, Maastricht, The Netherlands
| | - Ruud H Brakenhoff
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Otolaryngology/Head and Neck Surgery, Amsterdam, The Netherlands
| | - Irene Nauta
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Otolaryngology/Head and Neck Surgery, Amsterdam, The Netherlands
| | | | - Giuseppina Calareso
- IRCCS Fondazione Istituto Nazionale dei Tumori, Radiology Department, Milan, Italy
| | - Kathrin Scheckenbach
- University Hospital Duesseldorf, Heinrich-Heine-University, Department of Otorhinolaryngology & Head/Neck, Surgery, Duesseldorf, Germany
| | - Frank Hoebers
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology-Maastricht University Medical Centre, Department of Radiation Oncology, Maastricht, The Netherlands
| | - Frederik W R Wesseling
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology-Maastricht University Medical Centre, Department of Radiation Oncology, Maastricht, The Netherlands
| | - Simon Keek
- GROW-School for Oncology and Developmental Biology-Maastricht University Medical Centre-, Department of Precision Medicine, The D Lab: Decision Support for Precision Medicine-, Maastricht, The Netherlands
| | - Sebastian Sanduleanu
- GROW-School for Oncology and Developmental Biology-Maastricht University Medical Centre-, Department of Precision Medicine, The D Lab: Decision Support for Precision Medicine-, Maastricht, The Netherlands
| | - Ralph T H Leijenaar
- GROW-School for Oncology and Developmental Biology-Maastricht University Medical Centre-, Department of Precision Medicine, The D Lab: Decision Support for Precision Medicine-, Maastricht, The Netherlands
| | - Marije R Vergeer
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Radiation Oncology, Amsterdam, The Netherlands
| | - C René Leemans
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Otolaryngology/Head and Neck Surgery, Amsterdam, The Netherlands
| | - Chris H J Terhaard
- University Medical Center Utrecht, Department of Radiotherapy, Utrecht, The Netherlands
| | - Michiel W M van den Brekel
- The Netherlands Cancer Institute, Department of Head and Neck Oncology and Surgery, Amsterdam, The Netherlands
| | - Olga Hamming-Vrieze
- The Netherlands Cancer Institute, Department of Radiation Oncology, Amsterdam, The Netherlands
| | - Martijn A van der Heijden
- The Netherlands Cancer Institute, Department of Head and Neck Oncology and Surgery, Amsterdam, The Netherlands
| | - Hesham M Elhalawani
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Clifton D Fuller
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Matthias Guckenberger
- University Hospital Zurich and University of Zurich, Department of Radiation Oncology, Zurich, Switzerland
| | - Philippe Lambin
- GROW-School for Oncology and Developmental Biology-Maastricht University Medical Centre-, Department of Precision Medicine, The D Lab: Decision Support for Precision Medicine-, Maastricht, The Netherlands
| |
Collapse
|
2892
|
Farchione A, Larici AR, Masciocchi C, Cicchetti G, Congedo MT, Franchi P, Gatta R, Lo Cicero S, Valentini V, Bonomo L, Manfredi R. Exploring technical issues in personalized medicine: NSCLC survival prediction by quantitative image analysis-usefulness of density correction of volumetric CT data. Radiol Med 2020; 125:625-635. [PMID: 32125637 DOI: 10.1007/s11547-020-01157-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 02/10/2020] [Indexed: 02/06/2023]
Abstract
The aim of this study was to apply density correction method to the quantitative image analysis of non-small cell lung cancer (NSCLC) computed tomography (CT) images, determining its influence on overall survival (OS) prediction of surgically treated patients. Clinicopathological (CP) data and preoperative CT scans, pre- and post-contrast medium (CM) administration, of 57 surgically treated NSCLC patients, were retrospectively collected. After CT volumetric density measurement of primary gross tumour volume (GTV), aorta and tracheal air, density correction was conducted on GTV (reference values: aortic blood and tracheal air). For each resulting data set (combining CM administration and normalization), first-order statistical and textural features were extracted. CP and imaging data were correlated with patients 1-, 3- and 5-year OS, alone and combined (uni-/multivariate logistic regression and Akaike information criterion). Predictive performance was evaluated using the ROC curves and AUC values and compared among non-normalized/normalized data sets (DeLong test). The best predictive values were obtained when combining CP and imaging parameters (AUC values: 1 year 0.72; 3 years 0.82; 5 years 0.78). After normalization resulted an improvement in predicting 1-year OS for some of the grey level size zonebased features (large zone low grey level emphasis) and for the combined CP-imaging model, a worse performance for grey level co-occurrence matrix (cluster prominence and shade) and first-order statistical (range) parameters for 1- and 5-year OS, respectively. The negative performance of cluster prominence in predicting 1-year OS was the only statistically significant result (p value 0.05). Density corrections of volumetric CT data showed an opposite influence on the performance of imaging quantitative features in predicting OS of surgically treated NSCLC patients, even if no statistically significant for almost all predictors.
Collapse
Affiliation(s)
- Alessandra Farchione
- Dipartimento Diagnostica per Immagini, Radioterapia oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, Largo Francesco Vito 1, 00168, Rome, Italy.
| | - Anna Rita Larici
- Dipartimento Diagnostica per Immagini, Radioterapia oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, Largo Francesco Vito 1, 00168, Rome, Italy
- Dipartimento Universitario Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy
| | - Carlotta Masciocchi
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, Largo Francesco Vito 1, 00168, Rome, Italy
| | - Giuseppe Cicchetti
- Dipartimento Diagnostica per Immagini, Radioterapia oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, Largo Francesco Vito 1, 00168, Rome, Italy
- Dipartimento Universitario Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy
| | - Maria Teresa Congedo
- Dipartimento Scienze Cardiovascolari e Toraciche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, Largo Francesco Vito 1, 00168, Rome, Italy
| | - Paola Franchi
- UOC Radiologia, Ospedale G. Mazzini, ASL Teramo, Piazza Italia, 64100, Teramo, Italy
| | - Roberto Gatta
- Dipartimento di Scienze Cliniche e Sperimentali, Università degli Studi di Brescia, C/o Piazzale spedali civili 1, 25123, Brescia, Italy
| | - Stefano Lo Cicero
- Dipartimento Diagnostica per Immagini, Radioterapia oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, Largo Francesco Vito 1, 00168, Rome, Italy
- Dipartimento Universitario Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy
| | - Vincenzo Valentini
- Dipartimento Diagnostica per Immagini, Radioterapia oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, Largo Francesco Vito 1, 00168, Rome, Italy
- Dipartimento Universitario Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy
| | - Lorenzo Bonomo
- Dipartimento Diagnostica per Immagini, Radioterapia oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, Largo Francesco Vito 1, 00168, Rome, Italy
- Dipartimento Universitario Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy
| | - Riccardo Manfredi
- Dipartimento Diagnostica per Immagini, Radioterapia oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, Largo Francesco Vito 1, 00168, Rome, Italy
- Dipartimento Universitario Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy
| |
Collapse
|
2893
|
Tseng Y, Ma L, Li S, Luo T, Luo J, Zhang W, Wang J, Chen S. Application of CT-based radiomics in predicting portal pressure and patient outcome in portal hypertension. Eur J Radiol 2020; 126:108927. [PMID: 32146345 DOI: 10.1016/j.ejrad.2020.108927] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 02/25/2020] [Accepted: 02/28/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE Portal venous pressure (PVP) measurement is of clinical significance, especially in patients with portal hypertension. However, the invasive nature and associated complications limits its application. The aim of the study is to propose a noninvasive predictive model of PVP values based on CT-extracted radiomic features. METHODS Radiomics PVP (rPVP) models based on liver, spleen and combined features were established on an experimental cohort of 169 subjects. Radiomics features were extracted from each ROI and reduced via the LASSO regression to achieve an optimal predictive formula. A validation cohort of 62 patients treated for gastroesophageal varices (GOV) was used to confirm the utility of rPVP in predicting variceal recurrence. The association between rPVP and response to treatment was observed. RESULTS Three separate predictive formula for PVP were derived from radiomics features. rPVP was significantly correlated to patient response to endoscopic treatment for GOV. Among which, the model containing both liver and spleen features has the highest predictability of variceal recurrence, with an optimal cut-off value at 29.102 mmHg (AUC 0.866). A Kaplan Meier analysis further confirmed the difference between patients with varying rPVP values. CONCLUSION PVP values can be accurately predicted by a non-invasive, CT derived radiomics model. rPVP serves as a non-invasive and precise reference for predicting treatment outcome for GOV secondary to portal hypertension.
Collapse
Affiliation(s)
- Yujen Tseng
- Department of Gastroenterology,Zhongshan Hosptial, Fudan University, China; Department of Digestive Diseases, Huashan Hospital, Fudan University, China
| | - Lili Ma
- Department of Endoscopy Center, Zhongshan Hospital, Fudan University, China
| | - Shaobo Li
- Shanghai Medical College, Fudan University, China
| | - Tiancheng Luo
- Department of Gastroenterology,Zhongshan Hosptial, Fudan University, China
| | - Jianjun Luo
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, China
| | - Wen Zhang
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, China
| | - Jian Wang
- Department of Gastroenterology,Zhongshan Hosptial, Fudan University, China
| | - Shiyao Chen
- Department of Gastroenterology,Zhongshan Hosptial, Fudan University, China; Department of Endoscopy Center, Zhongshan Hospital, Fudan University, China; Evidence-Based Medicine Center, Fudan University, China.
| |
Collapse
|
2894
|
Mongan J, Moy L, Kahn CE. Checklist for Artificial Intelligence in Medical Imaging (CLAIM): A Guide for Authors and Reviewers. Radiol Artif Intell 2020; 2:e200029. [PMID: 33937821 PMCID: PMC8017414 DOI: 10.1148/ryai.2020200029] [Citation(s) in RCA: 595] [Impact Index Per Article: 119.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 03/05/2020] [Indexed: 12/23/2022]
Affiliation(s)
- John Mongan
- From the Department of Radiology and Biomedical Imaging, University of California–San Francisco, San Francisco, Calif (J.M.); Department of Radiology and Center for Advanced Imaging Innovation and Research, New York University School of Medicine, New York, NY (L.M.); and Department of Radiology, University of Pennsylvania, 3400 Spruce St, 1 Silverstein, Philadelphia, PA 19104 (C.E.K.)
| | - Linda Moy
- From the Department of Radiology and Biomedical Imaging, University of California–San Francisco, San Francisco, Calif (J.M.); Department of Radiology and Center for Advanced Imaging Innovation and Research, New York University School of Medicine, New York, NY (L.M.); and Department of Radiology, University of Pennsylvania, 3400 Spruce St, 1 Silverstein, Philadelphia, PA 19104 (C.E.K.)
| | - Charles E. Kahn
- From the Department of Radiology and Biomedical Imaging, University of California–San Francisco, San Francisco, Calif (J.M.); Department of Radiology and Center for Advanced Imaging Innovation and Research, New York University School of Medicine, New York, NY (L.M.); and Department of Radiology, University of Pennsylvania, 3400 Spruce St, 1 Silverstein, Philadelphia, PA 19104 (C.E.K.)
| |
Collapse
|
2895
|
Brunese L, Mercaldo F, Reginelli A, Santone A. An ensemble learning approach for brain cancer detection exploiting radiomic features. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2020; 185:105134. [PMID: 31675644 DOI: 10.1016/j.cmpb.2019.105134] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 09/27/2019] [Accepted: 10/15/2019] [Indexed: 05/03/2023]
Abstract
BACKGROUND AND OBJECTIVE The brain cancer is one of the most aggressive tumour: the 70% of the patients diagnosed with this malignant cancer will not survive. Early detection of brain tumours can be fundamental to increase survival rates. The brain cancers are classified into four different grades (i.e., I, II, III and IV) according to how normal or abnormal the brain cells look. The following work aims to recognize the different brain cancer grades by analysing brain magnetic resonance images. METHODS A method to identify the components of an ensemble learner is proposed. The ensemble learner is focused on the discrimination between different brain cancer grades using non invasive radiomic features. The considered radiomic features are belonging to five different groups: First Order, Shape, Gray Level Co-occurrence Matrix, Gray Level Run Length Matrix and Gray Level Size Zone Matrix. We evaluate the features effectiveness through hypothesis testing and through decision boundaries, performance analysis and calibration plots thus we select the best candidate classifiers for the ensemble learner. RESULTS We evaluate the proposed method with 111,205 brain magnetic resonances belonging to two freely available data-sets for research purposes. The results are encouraging: we obtain an accuracy of 99% for the benign grade I and the II, III and IV malignant brain cancer detection. CONCLUSION The experimental results confirm that the ensemble learner designed with the proposed method outperforms the current state-of-the-art approaches in brain cancer grade detection starting from magnetic resonance images.
Collapse
Affiliation(s)
- Luca Brunese
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, Campobasso, Italy
| | - Francesco Mercaldo
- Institute for Informatics and Telematics, National Research Council of Italy (CNR), Pisa, Italy; Department of Biosciences and Territory, University of Molise, Pesche (IS), Italy.
| | - Alfonso Reginelli
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Napoli, Italy
| | - Antonella Santone
- Department of Biosciences and Territory, University of Molise, Pesche (IS), Italy
| |
Collapse
|
2896
|
Hoffmann-Vold AM, Fretheim H, Meier C, Maurer B. Circulating biomarkers of systemic sclerosis - interstitial lung disease. JOURNAL OF SCLERODERMA AND RELATED DISORDERS 2020; 5:41-47. [PMID: 35382223 PMCID: PMC8922568 DOI: 10.1177/2397198319894851] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 11/06/2019] [Indexed: 10/24/2023]
Abstract
Interstitial lung disease is a frequent organ manifestation in systemic sclerosis and is associated with high mortality. It is crucial to diagnose interstitial lung disease in systemic sclerosis and to assess severity and identify patients prone to progression at an early stage to ultimately decrease organ damage and improve outcome. Circulating anti-topoisomerase-I autoantibodies have long been associated with the presence and development of systemic sclerosis - interstitial lung disease, evidence on their potential to further predict the clinical course of systemic sclerosis is however conflicting. C-reactive protein is a marker of infection and systemic inflammation with widespread clinical application and is elevated in systemic sclerosis with a tendency towards higher abundancy in patients with early disease. The role of other circulating biomarkers is promising but hampered by the lack of standardized criteria and guidelines for sample/data collection, analyses, reporting and validation and has not reached prime time for clinical application. However, epithelial markers including Krebs von den Lungen-6 and surfactant protein D and several cytokines and chemokines including CCL2 and CCL18 for severity assessment of systemic sclerosis - interstitial lung disease patients at the time of interstitial lung disease diagnosis and to predict interstitial lung disease progression have been reported and seem to be promising candidate biomarkers in the future.
Collapse
Affiliation(s)
- Anna-Maria Hoffmann-Vold
- Department of Rheumatology, Oslo
University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine,
University of Oslo, Oslo, Norway
| | - Håvard Fretheim
- Department of Rheumatology, Oslo
University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine,
University of Oslo, Oslo, Norway
| | - Chantal Meier
- Center of Experimental
Rheumatology, Department of Rheumatology, Zurich University Hospital,
Zurich, Switzerland
| | - Britta Maurer
- Center of Experimental
Rheumatology, Department of Rheumatology, Zurich University Hospital,
Zurich, Switzerland
| |
Collapse
|
2897
|
Huang Z, Zhang W, He D, Cui X, Tian S, Yin H, Song B. Development and validation of a radiomics model based on T2WI images for preoperative prediction of microsatellite instability status in rectal cancer: Study Protocol Clinical Trial (SPIRIT Compliant). Medicine (Baltimore) 2020; 99:e19428. [PMID: 32150094 PMCID: PMC7478495 DOI: 10.1097/md.0000000000019428] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
INTRODUCTION Globally, colorectal cancer (CRC) is the third most commonly diagnosed cancer in males and the second in females. Rectal cancer (RC) accounts for about 28% of all newly diagnosed CRC cases. The treatment of choice for locally advanced RC is a combination of surgical resection and chemotherapy and/or radiotherapy. These patients can potentially be cured, but the clinical outcome depends on the tumor biology. Microsatellite instability (MSI) is an important biomarker in CRC, with crucial diagnostic, prognostic, and predictive implications. It is important to develop a noninvasive, repeatable, and reproducible method to reflect the microsatellite status. Magnetic resonance imaging (MRI) has been recommended as the preferred imaging examination for RC in clinical practice by both the National Comprehensive Cancer Network and the European Society for Medical Oncology guidelines. T2WI is the core sequence of MRI scanning protocol for RC. Radiomics, the high-throughput mining of quantitative image features from standard-of-care medical imaging that enables data to be extracted and applied within clinical-decision support systems to improve diagnostic, prognostic, and predictive accuracy, is gaining importance in cancer research.We proposed a hypothesis: A simple radiomics model based on only T2WI images can accurately evaluate the MSI status of RC preoperatively. OBJECTIVE To develop a radiomics model based on T2WI images for accurate preoperative diagnosis the MSI status of RC. METHOD All patients with RC were retrospectively enrolled. The dataset was randomly split into training cohort (70% of all patients) and testing cohort (30% of all patients). The radiomics features will be extracted from T2WI-MR images of the entire primary tumor region. Least absolute shrinkage and selection operator was used to select the most predictive radiomics features. Logistic regression models were constructed in the training/validation cohort to discriminate the MSI status using clinical factors, radiomics features, or their integration. The diagnostic performance of these 3 models was evaluated in the testing cohort based on their area under the curve, sensitivity, specificity, and accuracy. DISCUSSION This study will help us know whether radiomics model based on T2WI images to preoperative identify MSI status of RC.
Collapse
Affiliation(s)
- Zixing Huang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu
| | - Wei Zhang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu
- Department of Radiology, Sichuan Provincial Corps Hospital, Chinese People’ s Armed Police Forces, Leshan
| | - Du He
- Department of Pathology, West China Hospital, Sichuan University, Chengdu
| | - Xing Cui
- Institute of Advanced Research, Infervision, Beijing, China
| | - Song Tian
- Institute of Advanced Research, Infervision, Beijing, China
| | - Hongkun Yin
- Institute of Advanced Research, Infervision, Beijing, China
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, Chengdu
| |
Collapse
|
2898
|
Complementary role of computed tomography texture analysis for differentiation of pancreatic ductal adenocarcinoma from pancreatic neuroendocrine tumors in the portal-venous enhancement phase. Abdom Radiol (NY) 2020; 45:750-758. [PMID: 31953587 PMCID: PMC8081676 DOI: 10.1007/s00261-020-02406-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Purpose To assess the role of CT-texture analysis (CTTA) for differentiation of pancreatic ductal adenocarcinoma (PDAC) from pancreatic neuroendocrine neoplasm (PNEN) in the portal-venous phase as compared with visual assessment and tumor-to-pancreas attenuation ratios. Methods 53 patients (66.1 ± 8.6y) with PDAC and 42 patients (65.5 ± 12.2y) with PNEN who underwent contrast-enhanced CT for primary staging were evaluated. Volumes of interests (VOIs) were set in the tumor tissue at the portal-venous phase excluding adjacent structures. Based on pyradiomics library, 92 textural features were extracted including 1st, 2nd, and higher order features, and then compared between PNEN and PDAC. The visual assessment classified tumors into hypo-, iso-, or hyperdense to pancreas parenchyma or into homogeneous/heterogeneous. Additionally, attenuation ratios between the tumors and the non-involved pancreas were calculated. Results 8/92 (8.6%) highly significant (p < 0.005) discriminatory textural features between PDAC and PNEN were identified including the 1st order features “median,” “total energy,” “energy,” “10th percentile,” “90th percentile,” “minimum,” “maximum,” and the 2nd order feature “Gray-Level co-occurrence Matrix (GLCM) Informational Measure of Correlation (Imc2).” In PNEN, the higher order feature “GLSZM Small Area High Gray-Level Emphasis” proved significantly higher in G1 compared to G2/3 tumors (p < 0.05). The tumor/parenchyma ratios as well as the visual assessment into hypo-/iso-/hyperdense or homogeneous/heterogeneous did not significantly differ between PDAC and PNEN. Conclusions Our data indicate that CTTA is a feasible tool for differentiation of PNEN from PDAC and also of G1 from G2/3 PNEN in the portal-venous phase. Visual assessment and tumor-to-parenchyma ratios were not useful for discrimination. Electronic supplementary material The online version of this article (10.1007/s00261-020-02406-9) contains supplementary material, which is available to authorized users.
Collapse
|
2899
|
CT-based radiomics and machine learning to predict spread through air space in lung adenocarcinoma. Eur Radiol 2020; 30:4050-4057. [PMID: 32112116 DOI: 10.1007/s00330-020-06694-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 01/02/2020] [Accepted: 01/30/2020] [Indexed: 10/24/2022]
Abstract
PURPOSE Spread through air space (STAS) is a novel invasive pattern of lung adenocarcinoma and is also a risk factor for recurrence and worse prognosis of lung adenocarcinoma. The aims of this study are to develop and validate a computed tomography (CT)‑based radiomics model for preoperative prediction of STAS in lung adenocarcinoma. METHODS AND MATERIALS This retrospective study was approved by an institutional review board and included 462 (mean age, 58.06 years) patients with pathologically confirmed lung adenocarcinoma. STAS was identified in 90 patients (19.5%). Two experienced radiologists segmented and extracted radiomics features on preoperative thin-slice CT images with radiomics extension independently. Intraclass correlation coefficients (ICC) and Pearson's correlation were used to rule out those low reliable (ICC < 0.75) and redundant (r > 0.9) features. Univariate logistic regression was applied to select radiomics features which were associated with STAS. A radiomics-based machine learning predictive model using a random forest (RF) was developed and calibrated with fivefold cross-validation. The diagnostic performance of the model was measured by the area under the curve (AUC) of receiver operating characteristic (ROC). RESULTS With univariate analysis, 12 radiomics features and age were found to be associated with STAS significantly. The RF model achieved an AUC of 0.754 (a sensitivity of 0.880 and a specificity of 0.588) for predicting STAS. CONCLUSION CT-based radiomics model can preoperatively predict STAS in lung adenocarcinoma with good diagnosis performance. KEY POINTS • CT-based radiomics and machine learning model can predict spread through air space (STAS) in lung adenocarcinoma with high accuracy. • The random forest (RF) model achieved an AUC of 0.754 (a sensitivity of 0.880 and a specificity of 0.588) for predicting STAS.
Collapse
|
2900
|
Shen X, Yang F, Yang P, Yang M, Xu L, Zhuo J, Wang J, Lu D, Liu Z, Zheng SS, Niu T, Xu X. A Contrast-Enhanced Computed Tomography Based Radiomics Approach for Preoperative Differentiation of Pancreatic Cystic Neoplasm Subtypes: A Feasibility Study. Front Oncol 2020; 10:248. [PMID: 32185129 PMCID: PMC7058789 DOI: 10.3389/fonc.2020.00248] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 02/13/2020] [Indexed: 12/18/2022] Open
Abstract
Background: Serous cystadenoma (SCA), mucinous cystadenoma (MCN), and intraductal papillary mucinous neoplasm (IPMN) are three subtypes of pancreatic cystic neoplasm (PCN). Due to the potential of malignant-transforming, patients with MCN and IPMN require radical surgery while patients with SCA need periodic surveillance. However, accurate pre-surgery diagnosis between SCA, MCN, and IPMN remains challenging in the clinic. Methods: This study enrolled 164 patients including 76 with SCA, 40 with MCN and 48 with IPMN. Patients were randomly split into a training cohort (n = 115) and validation cohort (n = 41). We performed statistical analysis and Boruta method to screen significantly distinct clinical factors and radiomics features extracted on pre-surgery contrast-enhanced computed tomography (CECT) images among three subtypes. Three reliable machine-learning algorithms, support vector machine (SVM), random forest (RF) and artificial neural network (ANN), were utilized to construct classifiers based on important radiomics features and clinical parameters. Precision, recall, and F1-score were calculated to assess the performance of the constructed classifiers. Results: Nine of 547 radiomics features and eight clinical factors showed a significant difference among SCA, MCN, and IPMN. Five radiomics features (Histogram_Entropy, Histogram_Skeweness, LLL_GLSZM_GLV, Histogram_Uniformity, HHL_Histogram_Kurtosis), and four clinical factors, including serum carbohydrate antigen 19-9, sex, age, and serum carcinoembryonic antigen, were identified important by Boruta method. The SVM classifier achieved an overall accuracy of 73.04% in training cohort and 71.43% in validation cohort, respectively. The RF classifier achieved overall accuracy of 84.35 and 79.59%, respectively. The constructed ANN model showed an overall accuracy of 77.39% in the training dataset and 71.43% in the validation dataset. All the three classifiers showed high F1 score for differentiation among the three subtypes. Conclusion: Our study proved the feasibility and translational value of CECT-based radiomics classifiers for differentiation among SCA, MCN, and IPMN.
Collapse
Affiliation(s)
- Xiaoyong Shen
- Department of Radiology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fan Yang
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Pengfei Yang
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
- College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China
| | - Modan Yang
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lei Xu
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| | - Jianyong Zhuo
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianguo Wang
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Di Lu
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhikun Liu
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shu-sen Zheng
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tianye Niu
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, China
- Nuclear & Radiological Engineering and Medical Physics Programs, Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| | - Xiao Xu
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|