51
|
Wang L, Kelly B, Lee EH, Wang H, Zheng J, Zhang W, Halabi S, Liu J, Tian Y, Han B, Huang C, Yeom KW, Deng K, Song J. Multi-classifier-based identification of COVID-19 from chest computed tomography using generalizable and interpretable radiomics features. Eur J Radiol 2021; 136:109552. [PMID: 33497881 PMCID: PMC7810032 DOI: 10.1016/j.ejrad.2021.109552] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/09/2020] [Accepted: 01/12/2021] [Indexed: 12/11/2022]
Abstract
PURPOSE To investigate the efficacy of radiomics in diagnosing patients with coronavirus disease (COVID-19) and other types of viral pneumonia with clinical symptoms and CT signs similar to those of COVID-19. METHODS Between 18 January 2020 and 20 May 2020, 110 SARS-CoV-2 positive and 108 SARS-CoV-2 negative patients were retrospectively recruited from three hospitals based on the inclusion criteria. Manual segmentation of pneumonia lesions on CT scans was performed by four radiologists. The latest version of Pyradiomics was used for feature extraction. Four classifiers (linear classifier, k-nearest neighbour, least absolute shrinkage and selection operator [LASSO], and random forest) were used to differentiate SARS-CoV-2 positive and SARS-CoV-2 negative patients. Comparison of the performance of the classifiers and radiologists was evaluated by ROC curve and Kappa score. RESULTS We manually segmented 16,053 CT slices, comprising 32,625 pneumonia lesions, from the CT scans of all patients. Using Pyradiomics, 120 radiomic features were extracted from each image. The key radiomic features screened by different classifiers varied and lead to significant differences in classification accuracy. The LASSO achieved the best performance (sensitivity: 72.2%, specificity: 75.1%, and AUC: 0.81) on the external validation dataset and attained excellent agreement (Kappa score: 0.89) with radiologists (average sensitivity: 75.6%, specificity: 78.2%, and AUC: 0.81). All classifiers indicated that "Original_Firstorder_RootMeanSquared" and "Original_Firstorder_Uniformity" were significant features for this task. CONCLUSIONS We identified radiomic features that were significantly associated with the classification of COVID-19 pneumonia using multiple classifiers. The quantifiable interpretation of the differences in features between the two groups extends our understanding of CT imaging characteristics of COVID-19 pneumonia.
Collapse
Affiliation(s)
- Lu Wang
- School of Medical Informatics, China Medical University Puhe Rd, Shenbei New District, Shenyang, Liaoning, 110122, China
| | - Brendan Kelly
- Department of Radiology, School of Medicine, Stanford University 725 Welch Rd MC 5654, Palo Alto, CA, 94305, United States
| | - Edward H. Lee
- Department of Radiology, School of Medicine, Stanford University 725 Welch Rd MC 5654, Palo Alto, CA, 94305, United States
| | - Hongmei Wang
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, No. 1 Swan Lake Road Hefei, Anhui, 230036, China
| | - Jimmy Zheng
- Department of Radiology, School of Medicine, Stanford University 725 Welch Rd MC 5654, Palo Alto, CA, 94305, United States
| | - Wei Zhang
- Department of Radiology, the Lu’an Affiliated Hospital, Anhui Medical University, No. 21 Wanxi Rd, Lu’an, Anhui, 237005, China
| | - Safwan Halabi
- Department of Radiology, School of Medicine, Stanford University 725 Welch Rd MC 5654, Palo Alto, CA, 94305, United States
| | - Jining Liu
- Bengbu Medical College, Department of Imaging Medicine, 2600 Donghai Avenue, Bengbu, Anhui, 233030, China
| | - Yulong Tian
- Wannan Medical College, Department of Imaging Medicine and Nuclear Medicine, 22 Wenchang West Rd, Higher Education Park, Wuhu, Anhui, 241002, China
| | - Baoqin Han
- Wannan Medical College, Department of Imaging Medicine and Nuclear Medicine, 22 Wenchang West Rd, Higher Education Park, Wuhu, Anhui, 241002, China
| | - Chuanbin Huang
- Wannan Medical College, Department of Imaging Medicine and Nuclear Medicine, 22 Wenchang West Rd, Higher Education Park, Wuhu, Anhui, 241002, China
| | - Kristen W. Yeom
- Department of Radiology, School of Medicine, Stanford University 725 Welch Rd MC 5654, Palo Alto, CA, 94305, United States
| | - Kexue Deng
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, No. 1 Swan Lake Road Hefei, Anhui, 230036, China,Corresponding author
| | - Jiangdian Song
- School of Medical Informatics, China Medical University Puhe Rd, Shenbei New District, Shenyang, Liaoning, 110122, China; Department of Radiology, School of Medicine, Stanford University 1201 Welch Rd, Lucas Center, Palo Alto, CA, 94305, United States.
| |
Collapse
|
52
|
Deep Learning Based HPV Status Prediction for Oropharyngeal Cancer Patients. Cancers (Basel) 2021; 13:cancers13040786. [PMID: 33668646 PMCID: PMC7917758 DOI: 10.3390/cancers13040786] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/07/2021] [Accepted: 02/09/2021] [Indexed: 12/30/2022] Open
Abstract
Simple Summary Determination of human papillomavirus (HPV) status for oropharyngeal cancer patients depicts a essential diagnostic factor and is important for treatment decisions. Current histological methods are invasive, time consuming and costly. We tested the ability of deep learning models for HPV status testing based on routinely acquired diagnostic CT images. A network trained for sports video clip classification was modified and then fine tuned for HPV status prediction. In this way, very basic information about image structures is induced into the model before training is started, while still allowing for exploitation of full 3D information in the CT images. Usage of this approach helps the network to cope with a small number of training examples and makes it more robust. For comparison, two other models were trained, one not relying on a pre-training task and another one pre-trained on 2D Data. The pre-trained video model preformed best. Abstract Infection with the human papillomavirus (HPV) has been identified as a major risk factor for oropharyngeal cancer (OPC). HPV-related OPCs have been shown to be more radiosensitive and to have a reduced risk for cancer related death. Hence, the histological determination of HPV status of cancer patients depicts an essential diagnostic factor. We investigated the ability of deep learning models for imaging based HPV status detection. To overcome the problem of small medical datasets, we used a transfer learning approach. A 3D convolutional network pre-trained on sports video clips was fine-tuned, such that full 3D information in the CT images could be exploited. The video pre-trained model was able to differentiate HPV-positive from HPV-negative cases, with an area under the receiver operating characteristic curve (AUC) of 0.81 for an external test set. In comparison to a 3D convolutional neural network (CNN) trained from scratch and a 2D architecture pre-trained on ImageNet, the video pre-trained model performed best. Deep learning models are capable of CT image-based HPV status determination. Video based pre-training has the ability to improve training for 3D medical data, but further studies are needed for verification.
Collapse
|
53
|
Wang X, Li BB. Deep Learning in Head and Neck Tumor Multiomics Diagnosis and Analysis: Review of the Literature. Front Genet 2021; 12:624820. [PMID: 33643386 PMCID: PMC7902873 DOI: 10.3389/fgene.2021.624820] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 01/07/2021] [Indexed: 12/24/2022] Open
Abstract
Head and neck tumors are the sixth most common neoplasms. Multiomics integrates multiple dimensions of clinical, pathologic, radiological, and biological data and has the potential for tumor diagnosis and analysis. Deep learning (DL), a type of artificial intelligence (AI), is applied in medical image analysis. Among the DL techniques, the convolution neural network (CNN) is used for image segmentation, detection, and classification and in computer-aided diagnosis. Here, we reviewed multiomics image analysis of head and neck tumors using CNN and other DL neural networks. We also evaluated its application in early tumor detection, classification, prognosis/metastasis prediction, and the signing out of the reports. Finally, we highlighted the challenges and potential of these techniques.
Collapse
Affiliation(s)
- Xi Wang
- Department of Oral Pathology, Peking University School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing, China
- Research Unit of Precision Pathologic Diagnosis in Tumors of the Oral and Maxillofacial Regions, Chinese Academy of Medical Sciences, Beijing, China
| | - Bin-bin Li
- Department of Oral Pathology, Peking University School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing, China
- Research Unit of Precision Pathologic Diagnosis in Tumors of the Oral and Maxillofacial Regions, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
54
|
Corradini S, Niyazi M, Verellen D, Valentini V, Walsh S, Grosu AL, Lauber K, Giaccia A, Unger K, Debus J, Pieters BR, Guckenberger M, Senan S, Budach W, Rad R, Mayerle J, Belka C. X-change symposium: status and future of modern radiation oncology-from technology to biology. Radiat Oncol 2021; 16:27. [PMID: 33541387 PMCID: PMC7863262 DOI: 10.1186/s13014-021-01758-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/28/2021] [Indexed: 02/06/2023] Open
Abstract
Future radiation oncology encompasses a broad spectrum of topics ranging from modern clinical trial design to treatment and imaging technology and biology. In more detail, the application of hybrid MRI devices in modern image-guided radiotherapy; the emerging field of radiomics; the role of molecular imaging using positron emission tomography and its integration into clinical routine; radiation biology with its future perspectives, the role of molecular signatures in prognostic modelling; as well as special treatment modalities such as brachytherapy or proton beam therapy are areas of rapid development. More clinically, radiation oncology will certainly find an important role in the management of oligometastasis. The treatment spectrum will also be widened by the rational integration of modern systemic targeted or immune therapies into multimodal treatment strategies. All these developments will require a concise rethinking of clinical trial design. This article reviews the current status and the potential developments in the field of radiation oncology as discussed by a panel of European and international experts sharing their vision during the "X-Change" symposium, held in July 2019 in Munich (Germany).
Collapse
Affiliation(s)
- Stefanie Corradini
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.
| | - Maximilian Niyazi
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Dirk Verellen
- Department of Radiotherapy, Iridium Network, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Vincenzo Valentini
- Department of Radiation Oncology and Hematology, Fondazione Policlinico Universitario A.Gemelli IRCCS, Università Cattolica S. Cuore, Rome, Italy
| | | | - Anca-L Grosu
- Department of Radiation Oncology, Medical Center, Medical Faculty, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | - Kirsten Lauber
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Amato Giaccia
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University, Stanford, USA
| | - Kristian Unger
- Integrative Biology Group, Helmholtz Zentrum Munich, Munich, Germany
| | - Jürgen Debus
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Bradley R Pieters
- Department of Radiation Oncology, Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Matthias Guckenberger
- Department of Radiation Oncology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Suresh Senan
- Department of Radiation Oncology, Amsterdam University Medical Centers, Location VUmc, Amsterdam, The Netherlands
| | - Wilfried Budach
- Department of Radiation Oncology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Roland Rad
- Center for Translational Cancer Research (TranslaTUM), TU Munich, Munich, Germany
| | - Julia Mayerle
- Department of Internal Medicine II, University Hospital, LMU, Munich, Germany
| | - Claus Belka
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| |
Collapse
|
55
|
Bos P, van den Brekel MWM, Gouw ZAR, Al‐Mamgani A, Waktola S, Aerts HJWL, Beets‐Tan RGH, Castelijns JA, Jasperse B. Clinical variables and magnetic resonance imaging-based radiomics predict human papillomavirus status of oropharyngeal cancer. Head Neck 2021; 43:485-495. [PMID: 33029923 PMCID: PMC7821378 DOI: 10.1002/hed.26505] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 09/15/2020] [Accepted: 09/24/2020] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Human papillomavirus (HPV)-positive oropharyngeal squamous cell carcinoma (OPSCC) have better prognosis and treatment response compared to HPV-negative OPSCC. This study aims to noninvasively predict HPV status of OPSCC using clinical and/or radiological variables. METHODS Seventy-seven magnetic resonance radiomic features were extracted from T1-weighted postcontrast images of the primary tumor of 153 patients. Logistic regression models were created to predict HPV status, determined with immunohistochemistry, based on clinical variables, radiomic features, and its combination. Model performance was evaluated using area under the curve (AUC). RESULTS Model performance showed AUCs of 0.794, 0.764, and 0.871 for the clinical, radiomic, and combined models, respectively. Smoking, higher T-classification (T3 and T4), larger, less round, and heterogeneous tumors were associated with HPV-negative tumors. CONCLUSION Models based on clinical variables and/or radiomic tumor features can predict HPV status in OPSCC patients with good performance and can be considered when HPV testing is not available.
Collapse
Affiliation(s)
- Paula Bos
- Department of RadiologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Department of Head and Neck Oncology and SurgeryThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Michiel W. M. van den Brekel
- Department of Head and Neck Oncology and SurgeryThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Department of Oral and Maxillofacial SurgeryAmsterdam University Medical Center (AUMC)Amsterdamthe Netherlands
| | - Zeno A. R. Gouw
- Department of Radiation OncologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Abrahim Al‐Mamgani
- Department of Radiation OncologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Selam Waktola
- Department of RadiologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Hugo J. W. L. Aerts
- Department of Radiation OncologyDana‐Farber Cancer Institute, Harvard Medical SchoolBostonMassachusettsUSA
| | - Regina G. H. Beets‐Tan
- Department of RadiologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- GROW School for Oncology and Developmental BiologyUniversity of MaastrichtMaastrichtThe Netherlands
- Department of Regional Health ResearchUniversity of Southern DenmarkDenmark
| | - Jonas A. Castelijns
- Department of RadiologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Bas Jasperse
- Department of RadiologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Department of RadiologyAmsterdam University Medical CenterAmsterdamThe Netherlands
| |
Collapse
|
56
|
Technical feasibility of radiomics signature analyses for improving detection of occult tonsillar cancer. Sci Rep 2021; 11:192. [PMID: 33420249 PMCID: PMC7794329 DOI: 10.1038/s41598-020-80597-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/21/2020] [Indexed: 02/02/2023] Open
Abstract
Diagnosis of occult palatine tonsil squamous cell carcinoma (SCC) using conventional magnetic resonance imaging (MRI) is difficult in patients with cervical nodal metastasis from an unknown primary site at presentation. We aimed to establish a radiomics approach based on MRI features extracted from the volume of interest in these patients. An Elastic Net model was developed to differentiate between normal palatine tonsils and occult palatine tonsil SCC. The diagnostic performances of the model with radiomics features extracted from T1-weighted image (WI), T2WI, contrast-enhanced T1WI, and an apparent diffusion coefficient (ADC) map had area under the receiver operating characteristic (AUROC) curve values of 0.831, 0.840, 0.781, and 0.807, respectively, for differential diagnosis. The model with features from the ADC alone showed the highest sensitivity of 90.0%, while the model with features from T1WI + T2WI + contrast-enhanced T1WI showed the highest AUROC of 0.853. The added sensitivity of the radiomics feature analysis were 34.6% over that of conventional MRI to detect occult palatine tonsil SCC. Therefore, we concluded that adding radiomics feature analysis to MRI may improve the detection sensitivity for occult palatine tonsil SCC in patients with a cervical nodal metastasis from cancer of an unknown primary site.
Collapse
|
57
|
Guiot J, Vaidyanathan A, Deprez L, Zerka F, Danthine D, Frix AN, Thys M, Henket M, Canivet G, Mathieu S, Eftaxia E, Lambin P, Tsoutzidis N, Miraglio B, Walsh S, Moutschen M, Louis R, Meunier P, Vos W, Leijenaar RTH, Lovinfosse P. Development and Validation of an Automated Radiomic CT Signature for Detecting COVID-19. Diagnostics (Basel) 2020; 11:E41. [PMID: 33396587 PMCID: PMC7823620 DOI: 10.3390/diagnostics11010041] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 12/28/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) outbreak has reached pandemic status. Drastic measures of social distancing are enforced in society and healthcare systems are being pushed to and beyond their limits. To help in the fight against this threat on human health, a fully automated AI framework was developed to extract radiomics features from volumetric chest computed tomography (CT) exams. The detection model was developed on a dataset of 1381 patients (181 COVID-19 patients plus 1200 non COVID control patients). A second, independent dataset of 197 RT-PCR confirmed COVID-19 patients and 500 control patients was used to assess the performance of the model. Diagnostic performance was assessed by the area under the receiver operating characteristic curve (AUC). The model had an AUC of 0.882 (95% CI: 0.851-0.913) in the independent test dataset (641 patients). The optimal decision threshold, considering the cost of false negatives twice as high as the cost of false positives, resulted in an accuracy of 85.18%, a sensitivity of 69.52%, a specificity of 91.63%, a negative predictive value (NPV) of 94.46% and a positive predictive value (PPV) of 59.44%. Benchmarked against RT-PCR confirmed cases of COVID-19, our AI framework can accurately differentiate COVID-19 from routine clinical conditions in a fully automated fashion. Thus, providing rapid accurate diagnosis in patients suspected of COVID-19 infection, facilitating the timely implementation of isolation procedures and early intervention.
Collapse
Affiliation(s)
- Julien Guiot
- Department of Pneumology, University Hospital of Liège, 4020 Liège, Belgium; (A.-N.F.); (M.H.); (R.L.)
| | - Akshayaa Vaidyanathan
- Research and Development, Oncoradiomics SA, 4000 Liège, Belgium; (A.V.); (F.Z.); (N.T.); (B.M.); (S.W.); (W.V.); (R.T.H.L.)
- The D-Lab, Department of Precision Medicine, Maastricht University, 6229 Maastricht, The Netherlands;
| | - Louis Deprez
- Department of Radiology, University Hospital of Liège, 4020 Liège, Belgium; (L.D.); (D.D.); (E.E.); (P.M.)
| | - Fadila Zerka
- Research and Development, Oncoradiomics SA, 4000 Liège, Belgium; (A.V.); (F.Z.); (N.T.); (B.M.); (S.W.); (W.V.); (R.T.H.L.)
- The D-Lab, Department of Precision Medicine, Maastricht University, 6229 Maastricht, The Netherlands;
| | - Denis Danthine
- Department of Radiology, University Hospital of Liège, 4020 Liège, Belgium; (L.D.); (D.D.); (E.E.); (P.M.)
| | - Anne-Noëlle Frix
- Department of Pneumology, University Hospital of Liège, 4020 Liège, Belgium; (A.-N.F.); (M.H.); (R.L.)
| | - Marie Thys
- Department of Medico-Economic Information, University Hospital of Liège, 4020 Liège, Belgium;
| | - Monique Henket
- Department of Pneumology, University Hospital of Liège, 4020 Liège, Belgium; (A.-N.F.); (M.H.); (R.L.)
| | - Gregory Canivet
- Department of Computer Applications, University Hospital of Liège, 4020 Liège, Belgium; (G.C.); (S.M.)
| | - Stephane Mathieu
- Department of Computer Applications, University Hospital of Liège, 4020 Liège, Belgium; (G.C.); (S.M.)
| | - Evanthia Eftaxia
- Department of Radiology, University Hospital of Liège, 4020 Liège, Belgium; (L.D.); (D.D.); (E.E.); (P.M.)
| | - Philippe Lambin
- The D-Lab, Department of Precision Medicine, Maastricht University, 6229 Maastricht, The Netherlands;
| | - Nathan Tsoutzidis
- Research and Development, Oncoradiomics SA, 4000 Liège, Belgium; (A.V.); (F.Z.); (N.T.); (B.M.); (S.W.); (W.V.); (R.T.H.L.)
| | - Benjamin Miraglio
- Research and Development, Oncoradiomics SA, 4000 Liège, Belgium; (A.V.); (F.Z.); (N.T.); (B.M.); (S.W.); (W.V.); (R.T.H.L.)
| | - Sean Walsh
- Research and Development, Oncoradiomics SA, 4000 Liège, Belgium; (A.V.); (F.Z.); (N.T.); (B.M.); (S.W.); (W.V.); (R.T.H.L.)
| | - Michel Moutschen
- Department of Infectious Diseases, University Hospital of Liège, 4020 Liège, Belgium;
| | - Renaud Louis
- Department of Pneumology, University Hospital of Liège, 4020 Liège, Belgium; (A.-N.F.); (M.H.); (R.L.)
| | - Paul Meunier
- Department of Radiology, University Hospital of Liège, 4020 Liège, Belgium; (L.D.); (D.D.); (E.E.); (P.M.)
| | - Wim Vos
- Research and Development, Oncoradiomics SA, 4000 Liège, Belgium; (A.V.); (F.Z.); (N.T.); (B.M.); (S.W.); (W.V.); (R.T.H.L.)
| | - Ralph T. H. Leijenaar
- Research and Development, Oncoradiomics SA, 4000 Liège, Belgium; (A.V.); (F.Z.); (N.T.); (B.M.); (S.W.); (W.V.); (R.T.H.L.)
| | - Pierre Lovinfosse
- Department of Nuclear Medicine and Oncological Imaging, University Hospital of Liège, 4020 Liège, Belgium;
| |
Collapse
|
58
|
Interpretable Machine Learning Model for Locoregional Relapse Prediction in Oropharyngeal Cancers. Cancers (Basel) 2020; 13:cancers13010057. [PMID: 33379188 PMCID: PMC7795920 DOI: 10.3390/cancers13010057] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/21/2020] [Accepted: 12/21/2020] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND There is no evidence to support surgery or radiotherapy as the best treatment for resectable oropharyngeal cancers with a negative HPV status. Predictive algorithms may help to decide which strategy to choose, but they will only be accepted by caregivers and European authorities if they are interpretable. As a proof of concept, we developed a predictive and interpretable algorithm to predict locoregional relapse at 18 months for oropharyngeal cancers as a first step towards that goal. METHODS The model was based on clinical and Pyradiomics features extracted from the dosimetric CT scan. Intraclass correlation was used to filter out features dependant on delineation. Correlated redundant features were also removed. An XGBoost model was cross-validated and optimised on the HN1 cohort (79 patients), and performances were assessed on the ART ORL cohort (45 patients). The Shapley Values were used to provide an overall and local explanation of the model. RESULTS On the ART ORL cohort, the model trained on HN1 yielded a precision-or predictive positive value-of 0.92, a recall of 0.42, an area under the curve of the receiver operating characteristic of 0.68 and an accuracy of 0.64. The most contributory features were shape Voxel Volume, grey level size zone matrix Small Area Emphasis (glszmSAE), gldm Dependence Non Uniformity Normalized (gldmDNUN), Sex and Age. CONCLUSIONS We developed an interpretable and generalizable model that could yield a good precision-positive predictive value-for relapse at 18 months on a different test cohort.
Collapse
|
59
|
Vuong D, Tanadini-Lang S, Wu Z, Marks R, Unkelbach J, Hillinger S, Eboulet EI, Thierstein S, Peters S, Pless M, Guckenberger M, Bogowicz M. Radiomics Feature Activation Maps as a New Tool for Signature Interpretability. Front Oncol 2020; 10:578895. [PMID: 33364192 PMCID: PMC7753181 DOI: 10.3389/fonc.2020.578895] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/22/2020] [Indexed: 12/25/2022] Open
Abstract
Introduction In the field of personalized medicine, radiomics has shown its potential to support treatment decisions. However, the limited feature interpretability hampers its introduction into the clinics. Here, we propose a new methodology to create radiomics feature activation maps, which allows to identify the spatial-anatomical locations responsible for signature activation based on local radiomics. The feasibility of this technique will be studied for histological subtype differentiation (adenocarcinoma versus squamous cell carcinoma) in non-small cell lung cancer (NSCLC) using computed tomography (CT) radiomics. Materials and Methods Pre-treatment CT scans were collected from a multi-centric Swiss trial (training, n=73, IIIA/N2 NSCLC, SAKK 16/00) and an independent cohort (validation, n=32, IIIA/N2/IIIB NSCLC). Based on the gross tumor volume (GTV), four peritumoral region of interests (ROI) were defined: lung_exterior (expansion into the lung), iso_exterior (expansion into lung and soft tissue), gradient (GTV border region), GTV+Rim (GTV and iso_exterior). For each ROI, 154 radiomic features were extracted using an in-house developed software implementation (Z-Rad, Python v2.7.14). Features robust against delineation variability served as an input for a multivariate logistic regression analysis. Model performance was quantified using the area under the receiver operating characteristic curve (AUC) and verified using five-fold cross validation and internal validation. Local radiomic features were extracted from the GTV+Rim ROI using non-overlapping 3x3x3 voxel patches previously marked as GTV or rim. A binary activation map was created for each patient using the median global feature value from the training. The ratios of activated/non-activated patches of GTV and rim regions were compared between histological subtypes (Wilcoxon test). Results Iso_exterior, gradient, GTV+Rim showed good performances for histological subtype prediction (AUCtraining=0.68-0.72 and AUCvalidation=0.73-0.74) whereas GTV and lung_exterior models failed validation. GTV+Rim model feature activation maps showed that local texture feature distribution differed significantly between histological subtypes in the rim (p=0.0481) but not in the GTV (p=0.461). Conclusion In this exploratory study, radiomics-based prediction of NSCLC histological subtypes was predominantly based on the peritumoral region indicating that radiomics activation maps can be useful for tracing back the spatial location of regions responsible for signature activation.
Collapse
Affiliation(s)
- Diem Vuong
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Stephanie Tanadini-Lang
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Ze Wu
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Robert Marks
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Jan Unkelbach
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Sven Hillinger
- Department of Thoracic Surgery, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Eric Innocents Eboulet
- Department of Clinical Trial Management, Swiss Group for Clinical Cancer Research (SAKK) Coordinating Center, Bern, Switzerland
| | - Sandra Thierstein
- Department of Clinical Trial Management, Swiss Group for Clinical Cancer Research (SAKK) Coordinating Center, Bern, Switzerland
| | - Solange Peters
- Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Miklos Pless
- Department of Medical Oncology, Kantonsspital Winterthur, Winterthur, Switzerland
| | - Matthias Guckenberger
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Marta Bogowicz
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| |
Collapse
|
60
|
Bagher-Ebadian H, Chetty IJ. Technical Note: ROdiomiX: A validated software for radiomics analysis of medical images in radiation oncology. Med Phys 2020; 48:354-365. [PMID: 33169367 DOI: 10.1002/mp.14590] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 10/27/2020] [Accepted: 11/03/2020] [Indexed: 12/19/2022] Open
Abstract
PURPOSE This study introduces an in-house-designed software platform (ROdiomiX) for the radiomics analysis of medical images in radiation oncology. ROdiomiX is a MATLAB-based framework for the computation of radiomic features and feature aggregation techniques in compliance with the Image-Biomarker-Standardization-Initiative (IBSI) guidelines, which includes preprocessing protocols and quantitative benchmark results for analysis of computational phantom images. METHODS AND MATERIALS The ROdiomiX software system consists of a series of computation cores implemented on the basis of the guidelines proposed by the IBSI. It is capable of quantitative computation of the following 11 different feature categories: Local-Intensity, Intensity-Histogram, Intensity-Based-Statistical, Intensity-Volume-Histogram, Gray-Level-Co-occurrence, Gray-Level-Run-Length, Gray-Level-Size-Zone, Gray-Level-Distance-Zone, Neighborhood-Grey-Tone-Difference, Neighboring-Grey-Level-Dependence, and Morphological feature. ROdiomiX was validated against benchmark values for the IBSI 3D digital phantom, as well as one designed in-house (HFH). The intraclass correlation coefficient (ICC) for estimating the degree of absolute agreement between ROdiomiX computation and benchmark values for different features at the 95% confidence level (CL) was used for comparison. RESULTS Among the 11 feature categories with 149 total features including 10 different feature aggregation methods (following the IBSI guidelines), the percent difference between absolute feature values computed by the ROdiomiX software and benchmark values reported for IBSI and HFH digital phantoms were 0.14% + 0.43% and 0.11% + 0.27%, respectively. The ICC values were >0.997 for all ten feature categories for both the IBSI and HFH digital phantoms. CONCLUSION The authors successfully developed a platform for computation of quantitative radiomic features. The image preprocessing and computational software cores were designed following the procedures specified by the IBSI. Benchmarking testing was in excellent agreement against the IBSI- and HFH-designed computational phantoms.
Collapse
Affiliation(s)
- Hassan Bagher-Ebadian
- Department of Radiation Oncology, Henry Ford Health System, 2799 West Grand Blvd, Detroit, MI, 48202, USA
| | - Indrin J Chetty
- Department of Radiation Oncology, Henry Ford Health System, 2799 West Grand Blvd, Detroit, MI, 48202, USA
| |
Collapse
|
61
|
Klein S, Quaas A, Quantius J, Löser H, Meinel J, Peifer M, Wagner S, Gattenlöhner S, Wittekindt C, von Knebel Doeberitz M, Prigge ES, Langer C, Noh KW, Maltseva M, Reinhardt HC, Büttner R, Klussmann JP, Wuerdemann N. Deep Learning Predicts HPV Association in Oropharyngeal Squamous Cell Carcinomas and Identifies Patients with a Favorable Prognosis Using Regular H&E Stains. Clin Cancer Res 2020; 27:1131-1138. [PMID: 33262137 DOI: 10.1158/1078-0432.ccr-20-3596] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 10/31/2020] [Accepted: 11/25/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Human papillomavirus (HPV) in oropharyngeal squamous cell carcinoma (OPSCC) is tumorigenic and has been associated with a favorable prognosis compared with OPSCC caused by tobacco, alcohol, and other carcinogens. Meanwhile, machine learning has evolved as a powerful tool to predict molecular and cellular alterations of medical images of various sources. EXPERIMENTAL DESIGN We generated a deep learning-based HPV prediction score (HPV-ps) on regular hematoxylin and eosin (H&E) stains and assessed its performance to predict HPV association using 273 patients from two different sites (OPSCC; Giessen, n = 163; Cologne, n = 110). Then, the prognostic relevance in a total of 594 patients (Giessen, Cologne, HNSCC TCGA) was evaluated. In addition, we investigated whether four board-certified pathologists could identify HPV association (n = 152) and compared the results to the classifier. RESULTS Although pathologists were able to diagnose HPV association from H&E-stained slides (AUC = 0.74, median of four observers), the interrater reliability was minimal (Light Kappa = 0.37; P = 0.129), as compared with AUC = 0.8 using the HPV-ps within two independent cohorts (n = 273). The HPV-ps identified individuals with a favorable prognosis in a total of 594 patients from three cohorts (Giessen, OPSCC, HR = 0.55, P < 0.0001; Cologne, OPSCC, HR = 0.44, P = 0.0027; TCGA, non-OPSCC head and neck, HR = 0.69, P = 0.0073). Interestingly, the HPV-ps further stratified patients when combined with p16 status (Giessen, HR = 0.06, P < 0.0001; Cologne, HR = 0.3, P = 0.046). CONCLUSIONS Detection of HPV association in OPSCC using deep learning with help of regular H&E stains may either be used as a single biomarker, or in combination with p16 status, to identify patients with OPSCC with a favorable prognosis, potentially outperforming combined HPV-DNA/p16 status as a biomarker for patient stratification.
Collapse
Affiliation(s)
- Sebastian Klein
- Institute of Pathology, Medical Faculty, University Hospital Cologne, Cologne, Germany.
| | - Alexander Quaas
- Institute of Pathology, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Jennifer Quantius
- Institute of Pathology, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Heike Löser
- Institute of Pathology, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Jörn Meinel
- Institute of Pathology, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Martin Peifer
- Department of Translational Genomics, Center of Integrated Oncology Cologne-Bonn, Medical Faculty, University of Cologne, Cologne, Germany.,Center for Molecular Medicine, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Steffen Wagner
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Giessen, Giessen, Germany
| | | | - Claus Wittekindt
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Giessen, Giessen, Germany
| | - Magnus von Knebel Doeberitz
- Department of Applied Tumor Biology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Elena-Sophie Prigge
- Department of Applied Tumor Biology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christine Langer
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Giessen, Giessen, Germany
| | - Ka-Won Noh
- Institute of Pathology, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Margaret Maltseva
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Hans Christian Reinhardt
- Clinic for Hematology and Stem Cell Transplantation, University Hospital Essen, Essen, Germany.,West German Cancer Center, University Hospital Essen, Essen, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Reinhard Büttner
- Institute of Pathology, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Jens Peter Klussmann
- Center for Molecular Medicine, Medical Faculty, University Hospital Cologne, Cologne, Germany.,Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Nora Wuerdemann
- Center for Molecular Medicine, Medical Faculty, University Hospital Cologne, Cologne, Germany.,Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty, University Hospital Cologne, Cologne, Germany
| |
Collapse
|
62
|
Suh CH, Lee KH, Choi YJ, Chung SR, Baek JH, Lee JH, Yun J, Ham S, Kim N. Oropharyngeal squamous cell carcinoma: radiomic machine-learning classifiers from multiparametric MR images for determination of HPV infection status. Sci Rep 2020; 10:17525. [PMID: 33067484 PMCID: PMC7568530 DOI: 10.1038/s41598-020-74479-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 08/23/2020] [Indexed: 02/06/2023] Open
Abstract
We investigated the ability of machine-learning classifiers on radiomics from pre-treatment multiparametric magnetic resonance imaging (MRI) to accurately predict human papillomavirus (HPV) status in patients with oropharyngeal squamous cell carcinoma (OPSCC). This retrospective study collected data of 60 patients (48 HPV-positive and 12 HPV-negative) with newly diagnosed histopathologically proved OPSCC, who underwent head and neck MRIs consisting of axial T1WI, T2WI, CE-T1WI, and apparent diffusion coefficient (ADC) maps from diffusion-weighted imaging (DWI). The median age was 59 years (the range being 35 to 85 years), and 83.3% of patients were male. The imaging data were randomised into a training set (32 HPV-positive and 8 HPV-negative OPSCC) and a test set (16 HPV-positive and 4 HPV-negative OPSCC) in each fold. 1618 quantitative features were extracted from manually delineated regions-of-interest of primary tumour and one definite lymph node in each sequence. After feature selection by using the least absolute shrinkage and selection operator (LASSO), three different machine-learning classifiers (logistic regression, random forest, and XG boost) were trained and compared in the setting of various combinations between four sequences. The highest diagnostic accuracies were achieved when using all sequences, and the difference was significant only when the combination did not include the ADC map. Using all sequences, logistic regression and the random forest classifier yielded higher accuracy compared with the that of the XG boost classifier, with mean area under curve (AUC) values of 0.77, 0.76, and 0.71, respectively. The machine-learning classifier of non-invasive and quantitative radiomics signature could guide the classification of the HPV status.
Collapse
Affiliation(s)
- Chong Hyun Suh
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, 86 Asanbyeongwon-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Kyung Hwa Lee
- Department of Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea.,Department of Biomedical Engineering, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Young Jun Choi
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, 86 Asanbyeongwon-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea.
| | - Sae Rom Chung
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, 86 Asanbyeongwon-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Jung Hwan Baek
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, 86 Asanbyeongwon-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Jeong Hyun Lee
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, 86 Asanbyeongwon-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Jihye Yun
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, 86 Asanbyeongwon-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Sungwon Ham
- Department of Biomedical Engineering, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Namkug Kim
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, 86 Asanbyeongwon-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea. .,Department of Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, 86 Asanbyeongwon-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea.
| |
Collapse
|
63
|
Bologna M, Corino V, Calareso G, Tenconi C, Alfieri S, Iacovelli NA, Cavallo A, Cavalieri S, Locati L, Bossi P, Romanello DA, Ingargiola R, Rancati T, Pignoli E, Sdao S, Pecorilla M, Facchinetti N, Trama A, Licitra L, Mainardi L, Orlandi E. Baseline MRI-Radiomics Can Predict Overall Survival in Non-Endemic EBV-Related Nasopharyngeal Carcinoma Patients. Cancers (Basel) 2020; 12:E2958. [PMID: 33066161 PMCID: PMC7601980 DOI: 10.3390/cancers12102958] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/07/2020] [Accepted: 10/08/2020] [Indexed: 02/06/2023] Open
Abstract
Advanced stage nasopharyngeal cancer (NPC) shows highly variable treatment outcomes, suggesting the need for independent prognostic factors. This study aims at developing a magnetic resonance imaging (MRI)-based radiomic signature as a prognostic marker for different clinical endpoints in NPC patients from non-endemic areas. A total 136 patients with advanced NPC and available MRI imaging (T1-weighted and T2-weighted) were selected. For each patient, 2144 radiomic features were extracted from the main tumor and largest lymph node. A multivariate Cox regression model was trained on a subset of features to obtain a radiomic signature for overall survival (OS), which was also applied for the prognosis of other clinical endpoints. Validation was performed using 10-fold cross-validation. The added prognostic value of the radiomic features to clinical features and volume was also evaluated. The radiomics-based signature had good prognostic power for OS and loco-regional recurrence-free survival (LRFS), with C-index of 0.68 and 0.72, respectively. In all the cases, the addition of radiomics to clinical features improved the prognostic performance. Radiomic features can provide independent prognostic information in NPC patients from non-endemic areas.
Collapse
Affiliation(s)
- Marco Bologna
- Department of Electronics, Information and Bioengineering (DEIB) Politecnico di Milano, 20133 Milan, Italy; (V.C.); (L.M.)
| | - Valentina Corino
- Department of Electronics, Information and Bioengineering (DEIB) Politecnico di Milano, 20133 Milan, Italy; (V.C.); (L.M.)
| | - Giuseppina Calareso
- Department of Radiology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (G.C.); (S.S.)
| | - Chiara Tenconi
- Department of Oncology and Hemato-Oncology, Università degli studi di Milano, 20133 Milan, Italy; (C.T.); (L.L.)
- Medical Physics Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (A.C.); (D.A.R.); (R.I.); (E.P.)
| | - Salvatore Alfieri
- Head and Neck Cancer Medical Oncology 3 Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy; (S.A.); (S.C.); (L.L.)
| | - Nicola Alessandro Iacovelli
- Radiotherapy 2 Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy; (N.A.I.); (N.F.); (E.O.)
| | - Anna Cavallo
- Medical Physics Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (A.C.); (D.A.R.); (R.I.); (E.P.)
| | - Stefano Cavalieri
- Head and Neck Cancer Medical Oncology 3 Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy; (S.A.); (S.C.); (L.L.)
| | - Laura Locati
- Head and Neck Cancer Medical Oncology 3 Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy; (S.A.); (S.C.); (L.L.)
| | - Paolo Bossi
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, ASST Spedali Civili, 25123 Brescia, Italy;
| | - Domenico Attilio Romanello
- Medical Physics Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (A.C.); (D.A.R.); (R.I.); (E.P.)
| | - Rossana Ingargiola
- Medical Physics Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (A.C.); (D.A.R.); (R.I.); (E.P.)
| | - Tiziana Rancati
- Prostate Cancer Program, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| | - Emanuele Pignoli
- Medical Physics Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (A.C.); (D.A.R.); (R.I.); (E.P.)
| | - Silvana Sdao
- Department of Radiology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (G.C.); (S.S.)
| | - Mattia Pecorilla
- Post-Graduate School in Radiodiagnostics, Università degli Studi di Milano, 20133 Milan, Italy;
| | - Nadia Facchinetti
- Radiotherapy 2 Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy; (N.A.I.); (N.F.); (E.O.)
| | - Annalisa Trama
- Research Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy;
| | - Lisa Licitra
- Department of Oncology and Hemato-Oncology, Università degli studi di Milano, 20133 Milan, Italy; (C.T.); (L.L.)
- Head and Neck Cancer Medical Oncology 3 Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy; (S.A.); (S.C.); (L.L.)
| | - Luca Mainardi
- Department of Electronics, Information and Bioengineering (DEIB) Politecnico di Milano, 20133 Milan, Italy; (V.C.); (L.M.)
| | - Ester Orlandi
- Radiotherapy 2 Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy; (N.A.I.); (N.F.); (E.O.)
| |
Collapse
|
64
|
Bologna M, Corino V, Tenconi C, Facchinetti N, Calareso G, Iacovelli N, Cavallo A, Alfieri S, Cavalieri S, Fallai C, Valdagni R, Rancati T, Trama A, Licitra L, Orlandi E, Mainardi L. Methodology and technology for the development of a prognostic MRI-based radiomic model for the outcome of head and neck cancer patients. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2020; 2020:1152-1155. [PMID: 33018191 DOI: 10.1109/embc44109.2020.9176565] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The purpose of this study was to establish a methodology and technology for the development of an MRI-based radiomic signature for prognosis of overall survival (OS) in nasopharyngeal cancer from non-endemic areas. The signature was trained using 1072 features extracted from the main tumor in T1-weighted and T2-weighted images of 142 patients. A model with 2 radiomic features was obtained (RAD model). Tumor volume and a signature obtained by training the model on permuted survival data (RADperm model) were used as a reference. A 10-fold cross-validation was used to validate the signature. Harrel's C-index was used as performance metric. A statistical comparison of the RAD, RADperm and volume was performed using Wilcoxon signed rank tests. The C-index for the RAD model was higher compared to the one of the RADperm model (0.69±0.08 vs 0.47±0.05), which ensures absence of overfitting. Also, the signature obtained with the RAD model had an improved C-index compared to tumor volume alone (0.69±0.08 vs 0.65±0.06), suggesting that the radiomic signature provides additional prognostic information.
Collapse
|
65
|
Katsoulakis E, Yu Y, Apte AP, Leeman JE, Katabi N, Morris L, Deasy JO, Chan TA, Lee NY, Riaz N, Hatzoglou V, Oh JH. Radiomic analysis identifies tumor subtypes associated with distinct molecular and microenvironmental factors in head and neck squamous cell carcinoma. Oral Oncol 2020; 110:104877. [PMID: 32619927 DOI: 10.1016/j.oraloncology.2020.104877] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 06/17/2020] [Accepted: 06/22/2020] [Indexed: 01/03/2023]
Abstract
PURPOSE To identify whether radiomic features from pre-treatment computed tomography (CT) scans can predict molecular differences between head and neck squamous cell carcinoma (HNSCC) using The Cancer Imaging Archive (TCIA) and The Cancer Genome Atlas (TCGA). METHODS 77 patients from the TCIA with HNSCC had imaging suitable for analysis. Radiomic features were extracted and unsupervised consensus clustering was performed to identify subtypes. Genomic data was extracted from the matched patients in the TCGA database. We explored relationships between radiomic features and molecular profiles of tumors, including the tumor immune microenvironment. A machine learning method was used to build a model predictive of CD8 + T-cells. An independent cohort of 83 HNSCC patients was used to validate the radiomic clusters. RESULTS We initially extracted 104 two-dimensional radiomic features, and after feature stability tests and removal of volume dependent features, reduced this to 67 features for subsequent analysis. Consensus clustering based on these features resulted in two distinct clusters. The radiomic clusters differed by primary tumor subsite (p = 0.0096), HPV status (p = 0.0127), methylation-based clustering results (p = 0.0025), and tumor immune microenvironment. A random forest model using radiomic features predicted CD8 + T-cells independent of HPV status with R2 = 0.30 (p < 0.0001) on cross validation. Consensus clustering on the validation cohort resulted in two distinct clusters that differ in tumor subsite (p = 1.3 × 10-7) and HPV status (p = 4.0 × 10-7). CONCLUSION Radiomic analysis can identify biologic features of tumors such as HPV status and T-cell infiltration and may be able to provide other information in the near future to help with patient stratification.
Collapse
Affiliation(s)
- Evangelia Katsoulakis
- Department of Radiation Oncology, Veterans Affairs, James A Haley, Tampa, FL 33612, USA
| | - Yao Yu
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Aditya P Apte
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jonathan E Leeman
- Department of Radiation Oncology, Dana Farber Cancer Institute/Brigham and Women's Hospital, Boston, MA 02189, USA
| | - Nora Katabi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Luc Morris
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Joseph O Deasy
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Timothy A Chan
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Nancy Y Lee
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Nadeem Riaz
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Vaios Hatzoglou
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - Jung Hun Oh
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
66
|
Konings H, Stappers S, Geens M, De Winter BY, Lamote K, van Meerbeeck JP, Specenier P, Vanderveken OM, Ledeganck KJ. A Literature Review of the Potential Diagnostic Biomarkers of Head and Neck Neoplasms. Front Oncol 2020; 10:1020. [PMID: 32670885 PMCID: PMC7332560 DOI: 10.3389/fonc.2020.01020] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 05/22/2020] [Indexed: 12/19/2022] Open
Abstract
Head and neck neoplasms have a poor prognosis because of their late diagnosis. Finding a biomarker to detect these tumors in an early phase could improve the prognosis and survival rate. This literature review provides an overview of biomarkers, covering the different -omics fields to diagnose head and neck neoplasms in the early phase. To date, not a single biomarker, nor a panel of biomarkers for the detection of head and neck tumors has been detected with clinical applicability. Limitations for the clinical implementation of the investigated biomarkers are mainly the heterogeneity of the study groups (e.g., small population in which the biomarker was tested, and/or only including high-risk populations) and a low sensitivity and/or specificity of the biomarkers under study. Further research on biomarkers to diagnose head and neck neoplasms in an early stage, is therefore needed.
Collapse
Affiliation(s)
- Heleen Konings
- Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Sofie Stappers
- Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Margot Geens
- Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Benedicte Y De Winter
- Laboratorium of Experimental Medicine and Pediatrics and Member of the Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Kevin Lamote
- Laboratorium of Experimental Medicine and Pediatrics and Member of the Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium.,Department of Pneumology, Antwerp University Hospital, Edegem, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Jan P van Meerbeeck
- Laboratorium of Experimental Medicine and Pediatrics and Member of the Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium.,Department of Pneumology, Antwerp University Hospital, Edegem, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Pol Specenier
- Department of Oncology, Antwerp University Hospital, Edegem, Belgium.,Center for Oncological Research (CORE), University of Antwerp, Antwerp, Belgium
| | - Olivier M Vanderveken
- Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Department of Otorhinolaryngology-Head and Neck Surgery, Antwerp University Hospital, Edegem, Belgium.,Department of Translational Neurosciences, Antwerp University, Antwerp, Belgium
| | - Kristien J Ledeganck
- Laboratorium of Experimental Medicine and Pediatrics and Member of the Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
67
|
Outcome prediction of head and neck squamous cell carcinoma by MRI radiomic signatures. Eur Radiol 2020; 30:6311-6321. [PMID: 32500196 PMCID: PMC7554007 DOI: 10.1007/s00330-020-06962-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/07/2020] [Accepted: 05/15/2020] [Indexed: 12/31/2022]
Abstract
OBJECTIVES Head and neck squamous cell carcinoma (HNSCC) shows a remarkable heterogeneity between tumors, which may be captured by a variety of quantitative features extracted from diagnostic images, termed radiomics. The aim of this study was to develop and validate MRI-based radiomic prognostic models in oral and oropharyngeal cancer. MATERIALS AND METHODS Native T1-weighted images of four independent, retrospective (2005-2013), patient cohorts (n = 102, n = 76, n = 89, and n = 56) were used to delineate primary tumors, and to extract 545 quantitative features from. Subsequently, redundancy filtering and factor analysis were performed to handle collinearity in the data. Next, radiomic prognostic models were trained and validated to predict overall survival (OS) and relapse-free survival (RFS). Radiomic features were compared to and combined with prognostic models based on standard clinical parameters. Performance was assessed by integrated area under the curve (iAUC). RESULTS In oral cancer, the radiomic model showed an iAUC of 0.69 (OS) and 0.70 (RFS) in the validation cohort, whereas the iAUC in the oropharyngeal cancer validation cohort was 0.71 (OS) and 0.74 (RFS). By integration of radiomic and clinical variables, the most accurate models were defined (iAUC oral cavity, 0.72 (OS) and 0.74 (RFS); iAUC oropharynx, 0.81 (OS) and 0.78 (RFS)), and these combined models outperformed prognostic models based on standard clinical variables only (p < 0.001). CONCLUSIONS MRI radiomics is feasible in HNSCC despite the known variability in MRI vendors and acquisition protocols, and radiomic features added information to prognostic models based on clinical parameters. KEY POINTS • MRI radiomics can predict overall survival and relapse-free survival in oral and HPV-negative oropharyngeal cancer. • MRI radiomics provides additional prognostic information to known clinical variables, with the best performance of the combined models. • Variation in MRI vendors and acquisition protocols did not influence performance of radiomic prognostic models.
Collapse
|
68
|
Radiomic biomarkers for head and neck squamous cell carcinoma. Strahlenther Onkol 2020; 196:868-878. [PMID: 32495038 DOI: 10.1007/s00066-020-01638-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 05/13/2020] [Indexed: 12/22/2022]
Abstract
Tumor heterogeneity is a well-known prognostic factor in head and neck squamous cell carcinoma (HNSCC). A major limitation of tissue- and blood-derived tumor markers is the lack of spatial resolution to image tumor heterogeneity. Tissue markers derived from tumor biopsies usually represent only a small tumor subregion at a single timepoint and are therefore often not representative of the tumors' biology or the biological alterations during and after treatment. Similarly, liquid biopsies give an overall picture of the tumors' secreted factors but completely lack any spatial resolution. Radiomics has the potential to give complete three-dimensional information about the tumor. We conducted a comprehensive literature search to assess the correlation of radiomics to tumor biology and treatment outcome in HNSCC and to assess current limitations of the radiomic biomarkers. In total, 25 studies that explored the ability of radiomics to predict tumor biology and phenotype in HNSCC and 28 studies that explored radiomics to predict post-treatment events were identified. Out of these 53 studies, only three failed to show a significant correlation. The major technical challenges are currently artifacts due to metal implants, non-standardized contrast injection, and delineation uncertainties. All studies to date were retrospective and none of the above-mentioned radiomics signatures have been validated in an independent cohort using an independent software implementation, which shows that transferability due to the numerous technical challenges is currently a major limitation. However, radiomics is a very young field and these studies hopefully pave the way for clinical implementation of radiomics for HNSCC in the future.
Collapse
|
69
|
Mukherjee P, Cintra M, Huang C, Zhou M, Zhu S, Colevas AD, Fischbein N, Gevaert O. CT-based Radiomic Signatures for Predicting Histopathologic Features in Head and Neck Squamous Cell Carcinoma. Radiol Imaging Cancer 2020; 2:e190039. [PMID: 32550599 DOI: 10.1148/rycan.2020190039] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 01/08/2020] [Accepted: 01/22/2020] [Indexed: 12/15/2022]
Abstract
Purpose To determine the performance of CT-based radiomic features for noninvasive prediction of histopathologic features of tumor grade, extracapsular spread, perineural invasion, lymphovascular invasion, and human papillomavirus status in head and neck squamous cell carcinoma (HNSCC). Materials and Methods In this retrospective study, which was approved by the local institutional ethics committee, CT images and clinical data from patients with pathologically proven HNSCC from The Cancer Genome Atlas (n = 113) and an institutional test cohort (n = 71) were analyzed. A machine learning model was trained with 2131 extracted radiomic features to predict tumor histopathologic characteristics. In the model, principal component analysis was used for dimensionality reduction, and regularized regression was used for classification. Results The trained radiomic model demonstrated moderate capability of predicting HNSCC features. In the training cohort and the test cohort, the model achieved a mean area under the receiver operating characteristic curve (AUC) of 0.75 (95% confidence interval [CI]: 0.68, 0.81) and 0.66 (95% CI: 0.45, 0.84), respectively, for tumor grade; a mean AUC of 0.64 (95% CI: 0.55, 0.62) and 0.70 (95% CI: 0.47, 0.89), respectively, for perineural invasion; a mean AUC of 0.69 (95% CI: 0.56, 0.81) and 0.65 (95% CI: 0.38, 0.87), respectively, for lymphovascular invasion; a mean AUC of 0.77 (95% CI: 0.65, 0.88) and 0.67 (95% CI: 0.15, 0.80), respectively, for extracapsular spread; and a mean AUC of 0.71 (95% CI: 0.29, 1.0) and 0.80 (95% CI: 0.65, 0.92), respectively, for human papillomavirus status. Conclusion Radiomic CT models have the potential to predict characteristics typically identified on pathologic assessment of HNSCC.Supplemental material is available for this article.© RSNA, 2020.
Collapse
Affiliation(s)
- Pritam Mukherjee
- Department of Medicine, Stanford Center for Biomedical Informatics Research (BMIR), Stanford, Calif (P.M., M.C., C.H., M.Z., O.G.); Department of Radiology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil (M.C.); Department of Nutrition and Food Hygiene, Chronic Disease Research Institute, School of Public Health, School of Medicine, Zhejiang University, Zhejiang, China (C.H., S.Z.); Division of Oncology, Department of Medicine (A.D.C.), Department of Radiology (N.F.), and Department of Biomedical Data Science (O.G.), Stanford University, 1265 Welch Rd, Stanford, CA 94305-5479
| | - Murilo Cintra
- Department of Medicine, Stanford Center for Biomedical Informatics Research (BMIR), Stanford, Calif (P.M., M.C., C.H., M.Z., O.G.); Department of Radiology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil (M.C.); Department of Nutrition and Food Hygiene, Chronic Disease Research Institute, School of Public Health, School of Medicine, Zhejiang University, Zhejiang, China (C.H., S.Z.); Division of Oncology, Department of Medicine (A.D.C.), Department of Radiology (N.F.), and Department of Biomedical Data Science (O.G.), Stanford University, 1265 Welch Rd, Stanford, CA 94305-5479
| | - Chao Huang
- Department of Medicine, Stanford Center for Biomedical Informatics Research (BMIR), Stanford, Calif (P.M., M.C., C.H., M.Z., O.G.); Department of Radiology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil (M.C.); Department of Nutrition and Food Hygiene, Chronic Disease Research Institute, School of Public Health, School of Medicine, Zhejiang University, Zhejiang, China (C.H., S.Z.); Division of Oncology, Department of Medicine (A.D.C.), Department of Radiology (N.F.), and Department of Biomedical Data Science (O.G.), Stanford University, 1265 Welch Rd, Stanford, CA 94305-5479
| | - Mu Zhou
- Department of Medicine, Stanford Center for Biomedical Informatics Research (BMIR), Stanford, Calif (P.M., M.C., C.H., M.Z., O.G.); Department of Radiology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil (M.C.); Department of Nutrition and Food Hygiene, Chronic Disease Research Institute, School of Public Health, School of Medicine, Zhejiang University, Zhejiang, China (C.H., S.Z.); Division of Oncology, Department of Medicine (A.D.C.), Department of Radiology (N.F.), and Department of Biomedical Data Science (O.G.), Stanford University, 1265 Welch Rd, Stanford, CA 94305-5479
| | - Shankuan Zhu
- Department of Medicine, Stanford Center for Biomedical Informatics Research (BMIR), Stanford, Calif (P.M., M.C., C.H., M.Z., O.G.); Department of Radiology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil (M.C.); Department of Nutrition and Food Hygiene, Chronic Disease Research Institute, School of Public Health, School of Medicine, Zhejiang University, Zhejiang, China (C.H., S.Z.); Division of Oncology, Department of Medicine (A.D.C.), Department of Radiology (N.F.), and Department of Biomedical Data Science (O.G.), Stanford University, 1265 Welch Rd, Stanford, CA 94305-5479
| | - A Dimitrios Colevas
- Department of Medicine, Stanford Center for Biomedical Informatics Research (BMIR), Stanford, Calif (P.M., M.C., C.H., M.Z., O.G.); Department of Radiology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil (M.C.); Department of Nutrition and Food Hygiene, Chronic Disease Research Institute, School of Public Health, School of Medicine, Zhejiang University, Zhejiang, China (C.H., S.Z.); Division of Oncology, Department of Medicine (A.D.C.), Department of Radiology (N.F.), and Department of Biomedical Data Science (O.G.), Stanford University, 1265 Welch Rd, Stanford, CA 94305-5479
| | - Nancy Fischbein
- Department of Medicine, Stanford Center for Biomedical Informatics Research (BMIR), Stanford, Calif (P.M., M.C., C.H., M.Z., O.G.); Department of Radiology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil (M.C.); Department of Nutrition and Food Hygiene, Chronic Disease Research Institute, School of Public Health, School of Medicine, Zhejiang University, Zhejiang, China (C.H., S.Z.); Division of Oncology, Department of Medicine (A.D.C.), Department of Radiology (N.F.), and Department of Biomedical Data Science (O.G.), Stanford University, 1265 Welch Rd, Stanford, CA 94305-5479
| | - Olivier Gevaert
- Department of Medicine, Stanford Center for Biomedical Informatics Research (BMIR), Stanford, Calif (P.M., M.C., C.H., M.Z., O.G.); Department of Radiology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil (M.C.); Department of Nutrition and Food Hygiene, Chronic Disease Research Institute, School of Public Health, School of Medicine, Zhejiang University, Zhejiang, China (C.H., S.Z.); Division of Oncology, Department of Medicine (A.D.C.), Department of Radiology (N.F.), and Department of Biomedical Data Science (O.G.), Stanford University, 1265 Welch Rd, Stanford, CA 94305-5479
| |
Collapse
|
70
|
Haider SP, Burtness B, Yarbrough WG, Payabvash S. Applications of radiomics in precision diagnosis, prognostication and treatment planning of head and neck squamous cell carcinomas. CANCERS OF THE HEAD & NECK 2020; 5:6. [PMID: 32391171 PMCID: PMC7197186 DOI: 10.1186/s41199-020-00053-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 03/09/2020] [Indexed: 12/15/2022]
Abstract
Recent advancements in computational power, machine learning, and artificial intelligence technology have enabled automated evaluation of medical images to generate quantitative diagnostic and prognostic biomarkers. Such objective biomarkers are readily available and have the potential to improve personalized treatment, precision medicine, and patient selection for clinical trials. In this article, we explore the merits of the most recent addition to the “-omics” concept for the broader field of head and neck cancer – “Radiomics”. This review discusses radiomics studies focused on (molecular) characterization, classification, prognostication and treatment guidance for head and neck squamous cell carcinomas (HNSCC). We review the underlying hypothesis, general concept and typical workflow of radiomic analysis, and elaborate on current and future challenges to be addressed before routine clinical application.
Collapse
Affiliation(s)
- Stefan P Haider
- 1Department of Radiology and Biomedical Imaging, Division of Neuroradiology, Yale School of Medicine, New Haven, CT USA.,2Department of Otorhinolaryngology, University Hospital of Ludwig Maximilians University of Munich, Munich, Germany
| | - Barbara Burtness
- 3Department of Internal Medicine, Division of Medical Oncology, Yale School of Medicine, New Haven, CT USA
| | - Wendell G Yarbrough
- 4Department of Otolaryngology/Head and Neck Surgery, University of North Carolina School of Medicine, Chapel Hill, NC USA
| | - Seyedmehdi Payabvash
- 1Department of Radiology and Biomedical Imaging, Division of Neuroradiology, Yale School of Medicine, New Haven, CT USA
| |
Collapse
|
71
|
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: 5.3] [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
|
72
|
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: 6.8] [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
|
73
|
Human Papillomavirus Testing in Head and Neck Squamous Cell Carcinoma in 2020: Where Are We Now and Where Are We Going? Head Neck Pathol 2020; 14:321-329. [PMID: 32124415 PMCID: PMC7235114 DOI: 10.1007/s12105-019-01117-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 12/17/2019] [Indexed: 12/24/2022]
Abstract
High risk human papillomavirus (HPV) has transformed head and neck oncology in the past several decades. Now that we have recognized that HPV-positive oropharyngeal squamous cell carcinoma (OPSCC) is a unique cancer type with distinct clinicopathologic features and favorable prognosis, it has become essential to test patients in routine practice. We have progressed greatly in our knowledge of this disease and gone, over the past two to three decades, from doing testing in highly variable amounts and methods to, now, with the help of national and international guidelines and patient staging requirements, to a situation where almost all patients with OPSCC are getting accurate classification through at least p16 immunohistochemistry. However, we are still struggling with how to accurately test specimens from cervical lymph nodes, and, in particular, on fine needle aspiration. In addition, many patients with non-oropharyngeal SCC are getting clinically unnecessary p16 and/or HPV-specific testing. The trends suggest progressive improvement in practices, but many practical questions still remain. On the horizon are myriad non-tissue-based tests, such as HPV serology and plasma DNA, DNA-based testing of fine needle aspirate fluid, computerized analysis of digitized pathology and radiology images, and machine learning from clinical and pathologic features, that may render pathologists largely obsolete for establishing HPV status for our patients' tumors. This review takes a brief look back in time to where we have been, then characterizes current practices in 2020 and lingering questions, and, finally, looks ahead into the possible future of HPV testing in patients with head and neck SCC.
Collapse
|
74
|
Rogers W, Thulasi Seetha S, Refaee TAG, Lieverse RIY, Granzier RWY, Ibrahim A, Keek SA, Sanduleanu S, Primakov SP, Beuque MPL, Marcus D, van der Wiel AMA, Zerka F, Oberije CJG, van Timmeren JE, Woodruff HC, Lambin P. Radiomics: from qualitative to quantitative imaging. Br J Radiol 2020; 93:20190948. [PMID: 32101448 DOI: 10.1259/bjr.20190948] [Citation(s) in RCA: 161] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Historically, medical imaging has been a qualitative or semi-quantitative modality. It is difficult to quantify what can be seen in an image, and to turn it into valuable predictive outcomes. As a result of advances in both computational hardware and machine learning algorithms, computers are making great strides in obtaining quantitative information from imaging and correlating it with outcomes. Radiomics, in its two forms "handcrafted and deep," is an emerging field that translates medical images into quantitative data to yield biological information and enable radiologic phenotypic profiling for diagnosis, theragnosis, decision support, and monitoring. Handcrafted radiomics is a multistage process in which features based on shape, pixel intensities, and texture are extracted from radiographs. Within this review, we describe the steps: starting with quantitative imaging data, how it can be extracted, how to correlate it with clinical and biological outcomes, resulting in models that can be used to make predictions, such as survival, or for detection and classification used in diagnostics. The application of deep learning, the second arm of radiomics, and its place in the radiomics workflow is discussed, along with its advantages and disadvantages. To better illustrate the technologies being used, we provide real-world clinical applications of radiomics in oncology, showcasing research on the applications of radiomics, as well as covering its limitations and its future direction.
Collapse
Affiliation(s)
- William Rogers
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands.,Department of Thoracic Oncology, IRCCS Foundation National Cancer Institute, Milan, Italy
| | - Sithin Thulasi Seetha
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands.,Department of Thoracic Oncology, IRCCS Foundation National Cancer Institute, Milan, Italy
| | - Turkey A G Refaee
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands.,Department of Diagnostic Radiology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Relinde I Y Lieverse
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Renée W Y Granzier
- Department of Radiology and Nuclear Imaging, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands.,Department of Surgery, Maastricht University Medical Centre, Grow-School for Oncology and Developmental Biology, Maastricht, The Netherlands
| | - Abdalla Ibrahim
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands.,Department of Radiology and Nuclear Imaging, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands.,Department of Nuclear Medicine and Comprehensive diagnostic center Aachen (CDCA), University Hospital RWTH Aachen University, Aachen, Germany.,Division of Nuclear Medicine and Oncological Imaging, Department of Medical Physics, Hospital Center Universitaire De Liege, Liege, Belgium
| | - Simon A Keek
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Sebastian Sanduleanu
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Sergey P Primakov
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Manon P L Beuque
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Damiënne Marcus
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Alexander M A van der Wiel
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Fadila Zerka
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Cary J G Oberije
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Janita E van Timmeren
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands.,Department of Radiation Oncology, University Hospital Zürich, Zürich, Switzerland.,University of Zürich, Zürich, Switzerland
| | - Henry C Woodruff
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands.,Department of Radiology and Nuclear Imaging, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Philippe Lambin
- The D-Lab & The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands.,Department of Radiology and Nuclear Imaging, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
75
|
Bologna M, Corino VDA, Mainardi LT. Assessment of the effect of intensity standardization on the reliability of T1-weighted MRI radiomic features: experiment on a virtual phantom. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2020; 2019:413-416. [PMID: 31945926 DOI: 10.1109/embc.2019.8857825] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The effect of time of repetition (TR) and time of echo (TE) on radiomic features was evaluated using a virtual phantom. Forty-two T1-weighted MRI images of the same virtual phantom were simulated with TR and TE in a range used in clinical practice. Fifty-eight radiomic features were considered for this analysis. Features were extracted from 3 different regions of interest (ROIs) from the original images and from images that underwent intensity standardization (linear intensity standardization, Z-score standardization and histogram matching). Intraclass correlation coefficient (ICC) was used to assess the reliability of the radiomic features and a threshold of 0.75 was used to discriminate features with good or bad reliability. The coefficient of determination R2 was used to quantify correlation between features and image acquisition parameters. The majority of radiomic features (76%) had good reliability (ICC>0.75) and 66% of the features were uncorrelated with TR and TE (R2<; 0.5). Intensity standardization (in particular histogram matching) significantly reduced the correlation. Intensity standardization also increased the reliability of FOS features, but histogram matching significantly reduced the reliability of GLCM features.
Collapse
|
76
|
Bagher-Ebadian H, Lu M, Siddiqui F, Ghanem AI, Wen N, Wu Q, Liu C, Movsas B, Chetty IJ. Application of radiomics for the prediction of HPV status for patients with head and neck cancers. Med Phys 2020; 47:563-575. [PMID: 31853980 DOI: 10.1002/mp.13977] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 10/28/2019] [Accepted: 11/22/2019] [Indexed: 12/19/2022] Open
Abstract
PURPOSE To perform radiomic analysis of primary tumors extracted from pretreatment contrast-enhanced computed tomography (CE-CT) images for patients with oropharyngeal cancers to identify discriminant features and construct an optimal classifier for the characterization and prediction of human papilloma virus (HPV) status. MATERIALS AND METHODS One hundred and eighty seven patients with oropharyngeal cancers with known HPV status (confirmed by immunohistochemistry-p16 protein testing) were retrospectively studied as follows: Group A: 95 patients (19HPV- and 76HPV+) from the MICAII grand challenge. Group B: 92 patients (52HPV- and 40HPV+) from our institution. Radiomic features (172) were extracted from pretreatment diagnostic CE-CT images of the gross tumor volume (GTV). Levene and Kolmogorov-Smirnov's tests with absolute biserial correlation (>0.48) were used to identify the discriminant features between the HPV+ and HPV- groups. The discriminant features were used to train and test eight different classifiers. Area under receiver operating characteristic (AUC), positive predictive and negative predictive values (PPV and NPV, respectively) were used to evaluate the performance of the classifiers. Principal component analysis (PCA) was applied on the discriminant feature set and seven PCs were used to train and test a generalized linear model (GLM) classifier. RESULTS Among 172 radiomic features only 12 radiomic features (from 3 categories) were significantly different (P < 0.05, |BSC| > 0.48) between the HPV+ and HPV- groups. Among the eight classifiers trained and applied for prediction of HPV status, the GLM showed the highest performance for each discriminant feature and the combined 12 features: AUC/PPV/NPV = 0.878/0.834/0.811. The GLM high prediction power was AUC/PPV/NPV = 0.849/0.731/0.788 and AUC/PPV/NPV = 0.869/0.807/0.870 for unseen test datasets for groups A and B, respectively. After eliminating the correlation among discriminant features by applying PCA analysis, the performance of the GLM was improved by 3.3%, 2.2%, and 1.8% for AUC, PPV, and NPV, respectively. CONCLUSIONS Results imply that GTV's for HPV+ patients exhibit higher intensities, smaller lesion size, greater sphericity/roundness, and higher spatial intensity-variation/heterogeneity. Results are suggestive that radiomic features primarily associated with the spatial arrangement and morphological appearance of the tumor on contrast-enhanced diagnostic CT datasets may be potentially used for classification of HPV status.
Collapse
Affiliation(s)
| | - Mei Lu
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI, USA
| | - Farzan Siddiqui
- Department of Radiation Oncology, Henry Ford Health System, Detroit, MI, USA
| | - Ahmed I Ghanem
- Department of Radiation Oncology, Henry Ford Health System, Detroit, MI, USA.,Department of Clinical Oncology, Alexandria University, Alexandria, Egypt
| | - Ning Wen
- Department of Radiation Oncology, Henry Ford Health System, Detroit, MI, USA
| | - Qixue Wu
- Department of Radiation Oncology, Henry Ford Health System, Detroit, MI, USA
| | - Chang Liu
- Department of Radiation Oncology, Henry Ford Health System, Detroit, MI, USA
| | - Benjamin Movsas
- Department of Radiation Oncology, Henry Ford Health System, Detroit, MI, USA
| | - Indrin J Chetty
- Department of Radiation Oncology, Henry Ford Health System, Detroit, MI, USA
| |
Collapse
|
77
|
Sollini M, Antunovic L, Chiti A, Kirienko M. Towards clinical application of image mining: a systematic review on artificial intelligence and radiomics. Eur J Nucl Med Mol Imaging 2019; 46:2656-2672. [PMID: 31214791 PMCID: PMC6879445 DOI: 10.1007/s00259-019-04372-x] [Citation(s) in RCA: 161] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 05/23/2019] [Indexed: 02/06/2023]
Abstract
PURPOSE The aim of this systematic review was to analyse literature on artificial intelligence (AI) and radiomics, including all medical imaging modalities, for oncological and non-oncological applications, in order to assess how far the image mining research stands from routine medical application. To do this, we applied a trial phases classification inspired from the drug development process. METHODS Among the articles we considered for inclusion from PubMed were multimodality AI and radiomics investigations, with a validation analysis aimed at relevant clinical objectives. Quality assessment of selected papers was performed according to the QUADAS-2 criteria. We developed the phases classification criteria for image mining studies. RESULTS Overall 34,626 articles were retrieved, 300 were selected applying the inclusion/exclusion criteria, and 171 high-quality papers (QUADAS-2 ≥ 7) were identified and analysed. In 27/171 (16%), 141/171 (82%), and 3/171 (2%) studies the development of an AI-based algorithm, radiomics model, and a combined radiomics/AI approach, respectively, was described. A total of 26/27(96%) and 1/27 (4%) AI studies were classified as phase II and III, respectively. Consequently, 13/141 (9%), 10/141 (7%), 111/141 (79%), and 7/141 (5%) radiomics studies were classified as phase 0, I, II, and III, respectively. All three radiomics/AI studies were categorised as phase II trials. CONCLUSIONS The results of the studies are promising but still not mature enough for image mining tools to be implemented in the clinical setting and be widely used. The transfer learning from the well-known drug development process, with some specific adaptations to the image mining discipline could represent the most effective way for radiomics and AI algorithms to become the standard of care tools.
Collapse
Affiliation(s)
- Martina Sollini
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090, Pieve Emanuele, Milan, Italy
| | - Lidija Antunovic
- Nuclear Medicine, Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy
| | - Arturo Chiti
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090, Pieve Emanuele, Milan, Italy
- Nuclear Medicine, Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy
| | - Margarita Kirienko
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090, Pieve Emanuele, Milan, Italy.
| |
Collapse
|
78
|
Moan JM, Amdal CD, Malinen E, Svestad JG, Bogsrud TV, Dale E. The prognostic role of 18F-fluorodeoxyglucose PET in head and neck cancer depends on HPV status. Radiother Oncol 2019; 140:54-61. [DOI: 10.1016/j.radonc.2019.05.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 05/13/2019] [Accepted: 05/15/2019] [Indexed: 11/25/2022]
|
79
|
Combined CT radiomics of primary tumor and metastatic lymph nodes improves prediction of loco-regional control in head and neck cancer. Sci Rep 2019; 9:15198. [PMID: 31645603 PMCID: PMC6811564 DOI: 10.1038/s41598-019-51599-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 10/02/2019] [Indexed: 12/20/2022] Open
Abstract
Loco-regional control (LRC) is a major clinical endpoint after definitive radiochemotherapy (RCT) of head and neck cancer (HNC). Radiomics has been shown a promising biomarker in cancer research, however closer related to primary tumor control than composite endpoints. Radiomics studies often focus on the analysis of primary tumor (PT). We hypothesize that the combination of PT and lymph nodes (LN) radiomics better predicts LRC in HNC treated with RCT. Radiomics analysis was performed in CT images of 128 patients using Z-Rad implementation (training n = 77, validation n = 51). 285 features were extracted from PT and involved LN. Features were preselected with the maximum relevance minimum redundancy method and the multivariate Cox model was trained using least absolute shrinkage and selection operator. The mixed model was based on the combination of PT and LN radiomics, whereas the PT model included only the PT features. The mixed model showed significantly higher performance than the PT model (p < 0.01), c-index of 0.67 and 0.63, respectively; and better risk group stratification. The clinical nodal status was not a significant predictor in the combination with PT radiomics. This study shows that the LRC can be better predicted by expansion of radiomics analysis with LN features.
Collapse
|
80
|
Abstract
OBJECTIVE. Imaging plays an important role in the diagnosis and staging of malignancies. Many common lymphoproliferative and other solid tumor malignancies can be viral-related. CONCLUSION. This review discusses the imaging findings that can be associated with common viral-induced malignancies. Knowledge of these imaging presentations can help narrow the differential diagnosis to reach a specific diagnosis through a precise workup and proper management.
Collapse
|
81
|
Stepp WH, Farquhar D, Sheth S, Mazul A, Mamdani M, Hackman TG, Hayes DN, Zevallos JP. RNA Oncoimmune Phenotyping of HPV-Positive p16-Positive Oropharyngeal Squamous Cell Carcinomas by Nodal Status. JAMA Otolaryngol Head Neck Surg 2019; 144:967-975. [PMID: 29710215 DOI: 10.1001/jamaoto.2018.0602] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Importance Clinical trials that deintensify treatment for patients with suspected human papillomavirus (HPV)-positive oropharyngeal squamous cell carcinoma (OPSCC) use p16 expression to identify HPV-mediated tumors and guide treatment. While p16 staining has a strong correlation with good outcomes, approximately 12% of p16-positive patients have recurrent disease. Biomarkers that reveal tumor-specific characteristics, such as nodal involvement, may change therapy decisions. Objective To assess whether if a tumor-specific genetic signature exists for node-negative vs node-positive HPV 16-positive/p16-positive OPSCCs. Design, Setting, and Participants This was a retrospective cohort study with randomized case selection for p16 OPSCCs undertaken at a university-based, tertiary care cancer center. Samples were collected from patients with p16-positive OPSCC. A total of 21 HPV 16/p16-positive tumors were used in this study. Main Outcomes and Measures Gene expression profiles of node-negative vs node-positive tumor samples were evaluated using a differential expression analysis approach and the sensitivity and specificity of a molecular signature was determined. Results Among the 21 patients in the study (3 women, 18 men; mean [SD] age, 54.6 [9.6] years), 6 had node-negative disease and 15 had node-positive disease. Using differential expression analysis, we found 146 genes that were significantly different in patients with node-negative disease vs those with node-positive disease, of which 15 genes were used to create a genetic signature that could distinguish node-negative-like from node-positive-like disease. The resultant molecular signature has a sensitivity of 88.2% (95% CI, 63.6%-98.5%) and specificity of 85.7% (95% CI, 42.1%-99.6%). The positive likelihood ratio of this signature was 6.1 (95% CI, 1.0-38.2) and the negative likelihood ratio was 0.1 (95% CI, 0.04-0.5). Given this population's prevalence of node-positive disease of 70.8%, the positive- and negative-predicative values for this gene signature were 93.7% (95% CI, 70.8%-98.9%) and 75.0% (95% CI, 44.1%-92.0%), respectively. In addition, we developed a gene signature using agnostic, machine learning software that identified a 40-gene profile that predicts node-negative disease from node-positive disease (area under the curve, 0.93; 95% CI, 0.63-1.00). Conclusions and Relevance Many HPV-16 and p16-positive tumors are treated as "lower-risk," but they do not have similar genetic compositions at the biological level. The identification of subgroups with unique expression patterns, such as those with nodal metastases, may guide physicians toward alternative or more aggressive therapies. In our study, unguided clustering suggested that that the larger biological characteristics of a tumor could be a better prognostic biomarker.
Collapse
Affiliation(s)
- Wesley H Stepp
- Department of Otolaryngology, University of North Carolina, School of Medicine, Chapel Hill
| | - Douglas Farquhar
- Department of Otolaryngology, University of North Carolina, School of Medicine, Chapel Hill
| | - Siddharth Sheth
- Division of Medical Oncology, Department of Medicine, University of North Carolina, School of Medicine, Chapel Hill
| | - Angela Mazul
- Department of Epidemiology, University of North Carolina, Gillings School of Public Health, Chapel Hill.,now at Division of Hematology-Oncology, Department of Medicine, University of Tennessee Health Science Center, Memphis
| | - Mohammed Mamdani
- Department of Otolaryngology, University of North Carolina, School of Medicine, Chapel Hill
| | - Trevor G Hackman
- Department of Otolaryngology, University of North Carolina, School of Medicine, Chapel Hill
| | - D Neil Hayes
- Division of Medical Oncology, Department of Medicine, University of North Carolina, School of Medicine, Chapel Hill.,now at Division of Hematology-Oncology, Department of Medicine, University of Tennessee Health Science Center, Memphis
| | - Jose P Zevallos
- Department of Epidemiology, University of North Carolina, Gillings School of Public Health, Chapel Hill.,now at Department of Otolaryngology, Washington University in St Louis, School of Medicine, St Louis, Missouri
| |
Collapse
|
82
|
Bogowicz M, Tanadini-Lang S, Veit-Haibach P, Pruschy M, Bender S, Sharma A, Hüllner M, Studer G, Stieb S, Hemmatazad H, Glatz S, Guckenberger M, Riesterer O. Perfusion CT radiomics as potential prognostic biomarker in head and neck squamous cell carcinoma. Acta Oncol 2019; 58:1514-1518. [PMID: 31304860 DOI: 10.1080/0284186x.2019.1629013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- M. Bogowicz
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - S. Tanadini-Lang
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - P. Veit-Haibach
- Department of Nuclear Medicine, University Hospital Zurich and University of Zurich, Zurich, Switzerland
- Department of Diagnostic and Interventional Radiology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - M. Pruschy
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - S. Bender
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - A. Sharma
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - M. Hüllner
- Department of Nuclear Medicine, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - G. Studer
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
- Institute for Radiation Oncology, Cantonal Hospital Lucerne, Lucerne, Switzerland
| | - S. Stieb
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - H. Hemmatazad
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - S. Glatz
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - M. Guckenberger
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - O. Riesterer
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
- Center for Radiation Oncology, KSA-KSB, Cantonal Hospital Aarau, Aarau, Switzerland
| |
Collapse
|
83
|
Chen M, Cao J, Bai Y, Tong C, Lin J, Jindal V, Barchi LC, Nadalin S, Yang SX, Pesce A, Panaro F, Ariche A, Kai K, Memeo R, Bekaii-Saab T, Cai X. Development and Validation of a Nomogram for Early Detection of Malignant Gallbladder Lesions. Clin Transl Gastroenterol 2019; 10:e00098. [PMID: 31663905 PMCID: PMC6884352 DOI: 10.14309/ctg.0000000000000098] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES Preoperative decision-making for differentiating malignant from benign lesions in the gallbladder remains challenging. We aimed to create a diagnostic nomogram to identify gallbladder cancer (GBC), especially for incidental GBC (IGBC), before surgical resection. METHODS A total of 587 consecutive patients with pathologically confirmed gallbladder lesions from a hospital were randomly assigned to a training cohort (70%) and an internal validation cohort (30%), with 287 patients from other centers as an external validation cohort. Radiological features were developed by the least absolute shrinkage and selection operator logistic regression model. Significant radiological features and independent clinical factors, identified by multivariate analyses, were used to construct a nomogram. RESULTS A diagnostic nomogram was established by age, CA19.9, and 6 radiological features. The values of area under the curve in the internal and external validation cohorts were up to 0.91 and 0.89, respectively. The calibration curves for probability of GBC showed optimal agreement between nomogram prediction and actual observation. Compared with previous methods, it demonstrated superior sensitivity (91.5%) and accuracy (85.1%) in the diagnosis of GBC. The accuracy using the nomogram was significantly higher in GBC groups compared with that by radiologists in the training cohort (P < 0.001) and similarly in each cohort. Notably, most of the IGBC, which were misdiagnosed as benign lesions, were successfully identified using this nomogram. DISCUSSION A novel nomogram provides a powerful tool for detecting the presence of cancer in gallbladder masses, with an increase in accuracy and sensitivity. It demonstrates an unprecedented potential for IGBC identification.
Collapse
Affiliation(s)
- Mingyu Chen
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Jiasheng Cao
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Yang Bai
- Department of General Surgery, Jinhua Municipal Central Hospital, Jinhua, China
| | - Chenhao Tong
- Department of General Surgery, Shaoxing People's Hospital, Zhejiang University, Shaoxing, China
| | - Jian Lin
- Department of General Surgery, Longyou People's Hospital, Quzhou, China
| | - Vishal Jindal
- Department of Internal Medicine, St. Vincent Hospital, Worcester, Massachusetts, USA
| | - Leandro Cardoso Barchi
- Digestive Surgery Division, Department of Gastroenterology, University of Sao Paulo School of Medicine, São Paulo, Brazil
| | - Silvio Nadalin
- Department of General, Visceral and Transplant Surgery, University Hospital Tuebingen, Tuebingen, Germany
| | - Sherry X. Yang
- National Clinical Target Validation Laboratory, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Antonio Pesce
- Department of Medical and Surgical Sciences and Advanced Technologies “G.F. Ingrassia” Policlinico-Vittorio Emanuele Hospital, Unit of General Surgery, University of Catania, Catania, Italy
| | - Fabrizio Panaro
- Division of Transplantation, Department of General Surgery, University of Montpellier-College of Medicine, Saint Eloi Hospital, Montpellier, France
| | - Arie Ariche
- Department of Surgery, Hadassah Medical Center, Mount Scopus, Jerusalem, Israel
| | - Keita Kai
- Department of Pathology, Saga University Hospital, Saga, Japan
| | - Riccardo Memeo
- Department of Emergency and Organ Transplantation, General Surgery and Transplantation, University Aldo Moro of Bari, Bari, Italy
| | | | - Xiujun Cai
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | | |
Collapse
|
84
|
Bogowicz M, Vuong D, Huellner MW, Pavic M, Andratschke N, Gabrys HS, Guckenberger M, Tanadini-Lang S. CT radiomics and PET radiomics: ready for clinical implementation? THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2019; 63:355-370. [PMID: 31527578 DOI: 10.23736/s1824-4785.19.03192-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Today, rapid technical and clinical developments result in an increasing number of treatment options for oncological diseases. Thus, decision support systems are needed to offer the right treatment to the right patient. Imaging biomarkers hold great promise in patient-individual treatment guidance. Routinely performed for diagnosis and staging, imaging datasets are expected to hold more information than used in the clinical practice. Radiomics describes the extraction of a large number of meaningful quantitative features from medical images, such as computed tomography (CT) and positron emission tomography (PET). Due to the non-invasive nature and ability to capture 3D image-based heterogeneity, radiomic features are potential surrogate markers of the cancer phenotype. Several radiomic studies are published per day, owing to encouraging results of many radiomics-based patient outcome models. Despite this comparably large number of studies, radiomics is mainly studied in proof of principle concept. Hence, a translation of radiomics from a hot topic research field into an essential clinical decision-making tool is lacking, but of high clinical interest. EVIDENCE ACQUISITION Herein, we present a literature review addressing the clinical evidence of CT and PET radiomics. An extensive literature review was conducted in PubMed, including papers on robustness and clinical applications. EVIDENCE SYNTHESIS We summarize image-modality related influences on the robustness of radiomic features and provide an overview of clinical evidence reported in the literature. Today, more evidence has been provided for CT imaging, however, PET imaging offers the promise of direct imaging of biological processes and functions. We provide a summary of future research directions, which needs to be addressed in order to successfully introduce radiomics into clinical medicine. In comparison to CT, more focus should be directed towards harmonization of PET acquisition and reconstruction protocols, which is important for transferable modelling. CONCLUSIONS Both CT and PET radiomics are promising pre-treatment and intra-treatment biomarkers for outcome prediction. Most studies are performed in retrospective setting, however their validation in prospective data collections is ongoing.
Collapse
Affiliation(s)
- Marta Bogowicz
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland -
| | - Diem Vuong
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Martin W Huellner
- Department of Nuclear Medicine, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Matea Pavic
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Nicolaus Andratschke
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Hubert S Gabrys
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Matthias Guckenberger
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Stephanie Tanadini-Lang
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| |
Collapse
|
85
|
Sheikh K, Lee SH, Cheng Z, Lakshminarayanan P, Peng L, Han P, McNutt TR, Quon H, Lee J. Predicting acute radiation induced xerostomia in head and neck Cancer using MR and CT Radiomics of parotid and submandibular glands. Radiat Oncol 2019; 14:131. [PMID: 31358029 PMCID: PMC6664784 DOI: 10.1186/s13014-019-1339-4] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 07/17/2019] [Indexed: 12/24/2022] Open
Abstract
PURPOSE To analyze baseline CT/MR-based image features of salivary glands to predict radiation-induced xerostomia 3-months after head-and-neck cancer (HNC) radiotherapy. METHODS A retrospective analysis was performed on 266 HNC patients who were treated using radiotherapy at our institution between 2009 and 2018. CT and T1 post-contrast MR images along with NCI-CTCAE xerostomia grade (3-month follow-up) were prospectively collected at our institution. CT and MR images were registered on which parotid/submandibular glands were contoured. Image features were extracted for ipsilateral/contralateral parotid and submandibular glands relative to the location of the primary tumor. Dose-volume-histogram (DVH) parameters were also acquired. Features were pre-selected based on Spearman correlation before modelling by examining the correlation with xerostomia (p < 0.05). A shrinkage regression analysis of the pre-selected features was performed using LASSO. The internal validity of the variable selection was estimated by repeating the entire variable selection procedure using a leave-one-out-cross-validation. The most frequently selected variables were considered in the final model. A generalized linear regression with repeated ten-fold cross-validation was developed to predict radiation-induced xerostomia at 3-months after radiotherapy. This model was tested in an independent dataset (n = 50) of patients who were treated at the same institution in 2017-2018. We compared the prediction performances under eight conditions (DVH-only, CT-only, MR-only, CT + MR, DVH + CT, DVH + CT + MR, Clinical+CT + MR, and Clinical+DVH + CT + MR) using the area under the receiver operating characteristic curve (ROC-AUC). RESULTS Among extracted features, 7 CT, 5 MR, and 2 DVH features were selected. The internal cohort (n = 216) ROC-AUC values for DVH, CT, MR, and Clinical+DVH + CT + MR features were 0.73 ± 0.01, 0.69 ± 0.01, 0.70 ± 0.01, and 0.79 ± 0.01, respectively. The validation cohort (n = 50) ROC-AUC values for DVH, CT, MR, and Clinical+DVH + CT + MR features were 0.63, 0.57, 0.66, and 0.68, respectively. The DVH-ROC was not significantly different than the CT-ROC (p = 0.8) or MR-ROC (p = 0.4). However, the CT + MR-ROC was significantly different than the CT-ROC (p = 0.03), but not the Clinical+DVH + CT + MR model (p = 0.5). CONCLUSION Our results suggest that baseline CT and MR image features may reflect baseline salivary gland function and potential risk for radiation injury. The integration of baseline image features into prediction models has the potential to improve xerostomia risk stratification with the ultimate goal of truly personalized HNC radiotherapy.
Collapse
Affiliation(s)
- Khadija Sheikh
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, 401 North Broadway, Suite 1440, Baltimore, MD 21287-5678 USA
| | - Sang Ho Lee
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, 401 North Broadway, Suite 1440, Baltimore, MD 21287-5678 USA
| | - Zhi Cheng
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, 401 North Broadway, Suite 1440, Baltimore, MD 21287-5678 USA
| | - Pranav Lakshminarayanan
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, 401 North Broadway, Suite 1440, Baltimore, MD 21287-5678 USA
| | - Luke Peng
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, 401 North Broadway, Suite 1440, Baltimore, MD 21287-5678 USA
| | - Peijin Han
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, 401 North Broadway, Suite 1440, Baltimore, MD 21287-5678 USA
| | - Todd R. McNutt
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, 401 North Broadway, Suite 1440, Baltimore, MD 21287-5678 USA
| | - Harry Quon
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, 401 North Broadway, Suite 1440, Baltimore, MD 21287-5678 USA
| | - Junghoon Lee
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, 401 North Broadway, Suite 1440, Baltimore, MD 21287-5678 USA
| |
Collapse
|
86
|
Alterio D, Marvaso G, Ferrari A, Volpe S, Orecchia R, Jereczek-Fossa BA. Modern radiotherapy for head and neck cancer. Semin Oncol 2019; 46:233-245. [PMID: 31378376 DOI: 10.1053/j.seminoncol.2019.07.002] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 07/15/2019] [Indexed: 02/07/2023]
Abstract
Radiation therapy (RT) plays a key role in curative-intent treatments for head and neck cancers. Its use is indicated as a sole therapy in early stage tumors or in combination with surgery or concurrent chemotherapy in advanced stages. Recent technologic advances have resulted in both improved oncologic results and expansion of the indications for RT in clinical practice. Despite this, RT administered to the head and neck region is still burdened by a high rate of acute and late side effects. Moreover, about 50% of patients with high-risk disease experience loco-regional recurrence within 3 years of follow-up. Therefore, in recent decades, efforts have been dedicated to optimize the cost/benefit ratio of RT in this subset of patients. The aim of the present review was to highlight modern concepts of RT for head and neck cancers considering both the technological advances that have been achieved and recent knowledge that has informed the biological interaction between radiation and both tumor and healthy tissues.
Collapse
Affiliation(s)
- Daniela Alterio
- Division of Radiotherapy, IEO European Institute of Oncology, IRCCS, Milan, Italy
| | - Giulia Marvaso
- Division of Radiotherapy, IEO European Institute of Oncology, IRCCS, Milan, Italy.
| | - Annamaria Ferrari
- Division of Radiotherapy, IEO European Institute of Oncology, IRCCS, Milan, Italy
| | - Stefania Volpe
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | | | - Barbara Alicja Jereczek-Fossa
- Division of Radiotherapy, IEO European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| |
Collapse
|
87
|
Huang C, Cintra M, Brennan K, Zhou M, Colevas AD, Fischbein N, Zhu S, Gevaert O. Development and validation of radiomic signatures of head and neck squamous cell carcinoma molecular features and subtypes. EBioMedicine 2019; 45:70-80. [PMID: 31255659 PMCID: PMC6642281 DOI: 10.1016/j.ebiom.2019.06.034] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 06/18/2019] [Accepted: 06/18/2019] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Radiomics-based non-invasive biomarkers are promising to facilitate the translation of therapeutically related molecular subtypes for treatment allocation of patients with head and neck squamous cell carcinoma (HNSCC). METHODS We included 113 HNSCC patients from The Cancer Genome Atlas (TCGA-HNSCC) project. Molecular phenotypes analyzed were RNA-defined HPV status, five DNA methylation subtypes, four gene expression subtypes and five somatic gene mutations. A total of 540 quantitative image features were extracted from pre-treatment CT scans. Features were selected and used in a regularized logistic regression model to build binary classifiers for each molecular subtype. Models were evaluated using the average area under the Receiver Operator Characteristic curve (AUC) of a stratified 10-fold cross-validation procedure repeated 10 times. Next, an HPV model was trained with the TCGA-HNSCC, and tested on a Stanford cohort (N = 53). FINDINGS Our results show that quantitative image features are capable of distinguishing several molecular phenotypes. We obtained significant predictive performance for RNA-defined HPV+ (AUC = 0.73), DNA methylation subtypes MethylMix HPV+ (AUC = 0.79), non-CIMP-atypical (AUC = 0.77) and Stem-like-Smoking (AUC = 0.71), and mutation of NSD1 (AUC = 0.73). We externally validated the HPV prediction model (AUC = 0.76) on the Stanford cohort. When compared to clinical models, radiomic models were superior to subtypes such as NOTCH1 mutation and DNA methylation subtype non-CIMP-atypical while were inferior for DNA methylation subtype CIMP-atypical and NSD1 mutation. INTERPRETATION Our study demonstrates that radiomics can potentially serve as a non-invasive tool to identify treatment-relevant subtypes of HNSCC, opening up the possibility for patient stratification, treatment allocation and inclusion in clinical trials. FUND: Dr. Gevaert reports grants from National Institute of Dental & Craniofacial Research (NIDCR) U01 DE025188, grants from National Institute of Biomedical Imaging and Bioengineering of the National Institutes of Health (NIBIB), R01 EB020527, grants from National Cancer Institute (NCI), U01 CA217851, during the conduct of the study; Dr. Huang and Dr. Zhu report grants from China Scholarship Council (Grant NO:201606320087), grants from China Medical Board Collaborating Program (Grant NO:15-216), the Cyrus Tang Foundation, and the Zhejiang University Education Foundation during the conduct of the study; Dr. Cintra reports grants from São Paulo State Foundation for Teaching and Research (FAPESP), during the conduct of the study.
Collapse
Affiliation(s)
- Chao Huang
- Chronic Disease Research Institute, School of Public Health, and Women's Hospital, School of Medicine, Zhejiang University, Zhejiang, Hangzhou, China; Department of Nutrition and Food Hygiene, School of Public Health, Zhejiang University, Zhejiang, Hangzhou, China; Department of Medicine, Stanford Center for Biomedical Informatics Research (BMIR), USA
| | - Murilo Cintra
- Department of Medicine, Stanford Center for Biomedical Informatics Research (BMIR), USA; Department of Radiology, Stanford University, USA; Ribeirão Preto Medical School, University of São Paulo, Brazil
| | - Kevin Brennan
- Department of Medicine, Stanford Center for Biomedical Informatics Research (BMIR), USA
| | - Mu Zhou
- Department of Medicine, Stanford Center for Biomedical Informatics Research (BMIR), USA
| | | | | | - Shankuan Zhu
- Chronic Disease Research Institute, School of Public Health, and Women's Hospital, School of Medicine, Zhejiang University, Zhejiang, Hangzhou, China; Department of Nutrition and Food Hygiene, School of Public Health, Zhejiang University, Zhejiang, Hangzhou, China.
| | - Olivier Gevaert
- Department of Medicine, Stanford Center for Biomedical Informatics Research (BMIR), USA; Department of Biomedical Data Science, Stanford University, USA.
| |
Collapse
|
88
|
Gabelloni M, Faggioni L, Neri E. Imaging biomarkers in upper gastrointestinal cancers. BJR Open 2019; 1:20190001. [PMID: 33178936 PMCID: PMC7592483 DOI: 10.1259/bjro.20190001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 02/23/2019] [Accepted: 03/29/2019] [Indexed: 12/02/2022] Open
Abstract
In parallel with the increasingly widespread availability of high performance imaging platforms and recent progresses in pathobiological characterisation and treatment of gastrointestinal malignancies, imaging biomarkers have become a major research topic due to their potential to provide additional quantitative information to conventional imaging modalities that can improve accuracy at staging and follow-up, predict outcome, and guide treatment planning in an individualised manner. The aim of this review is to briefly examine the status of current knowledge about imaging biomarkers in the field of upper gastrointestinal cancers, highlighting their potential applications and future perspectives in patient management from diagnosis onwards.
Collapse
Affiliation(s)
- Michela Gabelloni
- Department of Translational Research, Diagnostic and Interventional Radiology, University of Pisa, Pisa, Italy
| | - Lorenzo Faggioni
- Department of Translational Research, Diagnostic and Interventional Radiology, University of Pisa, Pisa, Italy
| | - Emanuele Neri
- Department of Translational Research, Diagnostic and Interventional Radiology, University of Pisa, Pisa, Italy
| |
Collapse
|
89
|
Cozzi L, Franzese C, Fogliata A, Franceschini D, Navarria P, Tomatis S, Scorsetti M. Predicting survival and local control after radiochemotherapy in locally advanced head and neck cancer by means of computed tomography based radiomics. Strahlenther Onkol 2019; 195:805-818. [PMID: 31222468 DOI: 10.1007/s00066-019-01483-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 06/06/2019] [Indexed: 12/31/2022]
Abstract
PURPOSE To appraise the ability of a radiomics signature to predict clinical outcome after definitive radiochemotherapy (RCT) of stage III-IV head and neck cancer. METHODS A cohort of 110 patients was included in a retrospective analysis. Radiomics texture features were extracted from the gross tumor volumes contoured on planning computed tomography (CT) images. The cohort of patients was randomly divided into a training (70 patients) and a validation (40 patients) cohorts. Textural features were correlated to survival and control data to build predictive models. All the significant predictors of the univariate analysis were included in a multivariate model. The quality of the models was appraised by means of the concordance index (CI). RESULTS A signature with 3 features was identified as predictive of overall survival (OS) with CI = 0.88 and 0.90 for the training and validation cohorts, respectively. A signature with 2 features was identified for progression-free survival (PFS; CI = 0.72 and 0.80); 2 features also characterized the signature for local control (LC; CI = 0.72 and 0.82). In all cases, the stratification in high- and low-risk groups for the training and validation cohorts led to significant differences in the actuarial curves. In the validation cohort the mean OS times (in months) were 78.9 ± 2.1 vs 67.4 ± 6.0 in the low- and high-risk groups, respectively, the PFS was 73.1 ± 3.7 and 50.7 ± 7.2, while the LC was 78.7 ± 2.1 and 63.9 ± 6.5. CONCLUSION CT-based radiomic signatures that correlate with survival and control after RCT were identified and allow low- and high-risk groups of patients to be identified.
Collapse
Affiliation(s)
- Luca Cozzi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (Milan), Italy. .,Radiotherapy and Radiosurgery, Humanitas Clinical and Research Center, Rozzano (Milan), Italy.
| | - Ciro Franzese
- Radiotherapy and Radiosurgery, Humanitas Clinical and Research Center, Rozzano (Milan), Italy
| | - Antonella Fogliata
- Radiotherapy and Radiosurgery, Humanitas Clinical and Research Center, Rozzano (Milan), Italy
| | - Davide Franceschini
- Radiotherapy and Radiosurgery, Humanitas Clinical and Research Center, Rozzano (Milan), Italy
| | - Pierina Navarria
- Radiotherapy and Radiosurgery, Humanitas Clinical and Research Center, Rozzano (Milan), Italy
| | - Stefano Tomatis
- Radiotherapy and Radiosurgery, Humanitas Clinical and Research Center, Rozzano (Milan), Italy
| | - Marta Scorsetti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (Milan), Italy.,Radiotherapy and Radiosurgery, Humanitas Clinical and Research Center, Rozzano (Milan), Italy
| |
Collapse
|
90
|
Budach V, Tinhofer I. Novel prognostic clinical factors and biomarkers for outcome prediction in head and neck cancer: a systematic review. Lancet Oncol 2019; 20:e313-e326. [DOI: 10.1016/s1470-2045(19)30177-9] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 02/21/2019] [Accepted: 02/25/2019] [Indexed: 01/16/2023]
|
91
|
Mirestean CC, Pagute O, Buzea C, Iancu RI, Iancu DT. Radiomic Machine Learning and Texture Analysis - New Horizons for Head and Neck Oncology. MAEDICA 2019; 14:126-130. [PMID: 31523292 PMCID: PMC6709390 DOI: 10.26574/maedica.2019.14.2.126] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Radiomics is a relatively new concept that consists of extracting data from images and applies advanced characterization algorithms to generate imaging features. These features are biomarkers with prognostic and predictive value, which provide a characterization of tumor phenotypes in a non-invasive manner. The clinical application of radiomics is hampered by challenges such as lack of image acquisition and analysis standardization. Textural features extracted from computed tomography (CT), magnetic resonance imaging (MRI) and positron emission tomography-computed tomography (PET-CT) images of patients diagnosed with head and neck cancers can be used in the pre-therapeutic evaluation of the response to multimodal chemo-radiotherapy. For patients with positive HPV-oropharyngeal cancers, the correlation of the radiomic textural features from the tumor with p16 values from the pathological sample can identify tumor specific signatures in CT imaging, an entity with favorable prognosis and a better response to chemo-radiotherapy. Pretreatment contrast CT-scans were extracted and radiomics analysis of gross tumor volume were performed using MaZda package apart from MaZda software containing B11 program for texture analysis and visualization. Data set was randomly divided into a training dataset and a test dataset and machine learning algorithms were applied to identify a textural radiomic signature. Radiomic texture analysis and machine learning algorithms demonstrate a predictive potential related to the capability of stratification for subclasses of platinum-chemotherapy resistance and radioresistant head and neck cancers requiring an intensification of multimodal treatment.
Collapse
Affiliation(s)
| | | | - Calin Buzea
- Regional Institute of Oncology, Iasi, Romania
| | | | | |
Collapse
|
92
|
Payabvash S, Chan A, Jabehdar Maralani P, Malhotra A. Quantitative diffusion magnetic resonance imaging for prediction of human papillomavirus status in head and neck squamous-cell carcinoma: A systematic review and meta-analysis. Neuroradiol J 2019; 32:232-240. [PMID: 31084347 DOI: 10.1177/1971400919849808] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
PURPOSE Head and neck squamous-cell carcinoma (HNSCC) related to human papillomavirus (HPV) infection represents a distinct biological and prognostic subtype compared to the HPV-negative form. Prior studies suggest a correlation between the apparent diffusion coefficient (ADC) values on diffusion-weighted imaging (DWI) of primary tumor lesion and HPV status in HNSCC. In this meta-analysis, we compared the average ADC of primary lesion between HPV-positive and HPV-negative HNSCC. METHODS A comprehensive literature search of PubMed and Embase was performed. Studies comparing the average ADC on echo-planar DWI of primary tumor lesions between HPV-positive and HPV-negative HNSCC were included. The standardized mean difference was calculated using fixed- and random-effects models. Tau-squared estimates of total heterogeneity and Higgins inconsistency index (I2 test) were determined. RESULTS A total of five studies, pooling data of 264 patients, were included for meta-analysis. Among these five studies, three had included oral cavity, hypopharyngeal, and/or laryngeal HNSCC in addition to oropharyngeal subsite. Primary lesions were comprised of 185 HPV-negative and 79 HPV-positive HNSCC. The meta-analysis showed lower average ADC values in HPV-positive HNSCC compared to the HPV-negative form, with a standardized mean difference of 0.961 (95% confidence interval 0.644-1.279; p < 0.0001). Since there was no significant heterogeneity in analysis (p = 0.3852), both random- and fixed-effects models resulted in the same estimates of overall effect. CONCLUSIONS HPV-positive HNSCC primary lesions have a lower average ADC compared to the HPV-negative form, highlighting the potential application of quantitative diffusion magnetic resonance imaging as a noninvasive imaging biomarker for prediction of HPV status.
Collapse
Affiliation(s)
| | - Aimee Chan
- 2 Department of Medical Imaging, University of Toronto, Canada
| | | | - Ajay Malhotra
- 1 Department of Radiology and Biomedical Imaging, Yale School of Medicine, USA
| |
Collapse
|
93
|
Abstract
Oropharyngeal carcinoma associated with the human papillomavirus is increasing in incidence and represents a unique head and neck disease with favorable treatment outcomes. This review evaluates the evolving role of radiotherapy in regional management with an overall goal of treatment de-escalation in the appropriate patient. Determining the optimal approach and selection factors for treatment de-escalation is under active investigation. Response to induction chemotherapy, refining adverse pathologic factors after a primary surgical approach, decreasing radiation dose with or without chemotherapy in the definitive or adjuvant settings as well as more selective nodal level irradiation all are current strategies for treatment de-escalation. This review details the likely changes in regional radiotherapy management for oropharyngeal carcinoma in the modern human papillomavirus era and discusses future approaches to patient selection with the goal of reducing toxicities while maintaining function preservation and quality of life in group of patients who are younger and healthier than traditional head and neck cancer patients.
Collapse
Affiliation(s)
- Moses Tam
- Department of Radiation Oncology, New York University Langone Health, New York, NY.
| | - Kenneth Hu
- Department of Radiation Oncology, New York University Langone Health, New York, NY
| |
Collapse
|
94
|
Liu Z, Wang S, Dong D, Wei J, Fang C, Zhou X, Sun K, Li L, Li B, Wang M, Tian J. The Applications of Radiomics in Precision Diagnosis and Treatment of Oncology: Opportunities and Challenges. Theranostics 2019; 9:1303-1322. [PMID: 30867832 PMCID: PMC6401507 DOI: 10.7150/thno.30309] [Citation(s) in RCA: 504] [Impact Index Per Article: 100.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 01/10/2019] [Indexed: 12/14/2022] Open
Abstract
Medical imaging can assess the tumor and its environment in their entirety, which makes it suitable for monitoring the temporal and spatial characteristics of the tumor. Progress in computational methods, especially in artificial intelligence for medical image process and analysis, has converted these images into quantitative and minable data associated with clinical events in oncology management. This concept was first described as radiomics in 2012. Since then, computer scientists, radiologists, and oncologists have gravitated towards this new tool and exploited advanced methodologies to mine the information behind medical images. On the basis of a great quantity of radiographic images and novel computational technologies, researchers developed and validated radiomic models that may improve the accuracy of diagnoses and therapy response assessments. Here, we review the recent methodological developments in radiomics, including data acquisition, tumor segmentation, feature extraction, and modelling, as well as the rapidly developing deep learning technology. Moreover, we outline the main applications of radiomics in diagnosis, treatment planning and evaluations in the field of oncology with the aim of developing quantitative and personalized medicine. Finally, we discuss the challenges in the field of radiomics and the scope and clinical applicability of these methods.
Collapse
Affiliation(s)
- Zhenyu Liu
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100080, China
| | - Shuo Wang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100080, China
| | - Di Dong
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100080, China
| | - Jingwei Wei
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100080, China
| | - Cheng Fang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xuezhi Zhou
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, 100190, China
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Kai Sun
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, 100190, China
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Longfei Li
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, 100190, China
- Collaborative Innovation Center for Internet Healthcare, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Bo Li
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Meiyun Wang
- Department of Radiology, Henan Provincial People's Hospital & the People's Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, 100190, China
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, 100191, China
| |
Collapse
|
95
|
Neri E, Del Re M, Paiar F, Erba P, Cocuzza P, Regge D, Danesi R. Radiomics and liquid biopsy in oncology: the holons of systems medicine. Insights Imaging 2018; 9:915-924. [PMID: 30430428 PMCID: PMC6269342 DOI: 10.1007/s13244-018-0657-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 08/10/2018] [Accepted: 08/28/2018] [Indexed: 12/15/2022] Open
Abstract
Abstract Radiomics is a process of extraction and analysis of quantitative features from diagnostic images. Liquid biopsy is a test done on a sample of blood to look for cancer cells or for pieces of tumourigenic DNA circulating in the blood. Radiomics and liquid biopsy have great potential in oncology, since both are minimally invasive, easy to perform, and can be repeated in patient follow-up visits, enabling the extraction of valuable information regarding tumour type, aggressiveness, progression, and response to treatment. Both methods are in their infancy, with major evidence of application in lung and gastrointestinal cancer, while still undergoing evaluation in other cancer types. In this paper, the main oncologic applications of radiomics and liquid biopsy are reviewed, and a synergistic approach incorporating both tests for cancer diagnosis and follow-up is discussed within the context of systems medicine. Teaching Points • Radiomics is a process of extraction and analysis of quantitative features from diagnostic images. • Most clinical applications of radiomics are in the field of oncologic imaging. • Radiomics applies to all imaging modalities. • A cluster of radiomic features is a “radiomic signature”. • Machine learning may improve the efficacy of radiomics analysis.
Collapse
Affiliation(s)
- Emanuele Neri
- Diagnostic and Interventional Radiology, Department of Translational Research, University of Pisa, Pisa, Italy.
| | - Marzia Del Re
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Fabiola Paiar
- Radiation Oncology Unit, Department of Translational Research, University of Pisa, Pisa, Italy
| | - Paola Erba
- Nuclear Medicine Unit, Department of Translational Research, University of Pisa, Pisa, Italy
| | - Paola Cocuzza
- Radiation Oncology Unit, Department of Translational Research, University of Pisa, Pisa, Italy
| | - Daniele Regge
- Radiology Unit, Candiolo Cancer Institute - FPO, IRCCS, Candiolo, Turin, Italy
| | - Romano Danesi
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
96
|
Morin O, Vallières M, Jochems A, Woodruff HC, Valdes G, Braunstein SE, Wildberger JE, Villanueva-Meyer JE, Kearney V, Yom SS, Solberg TD, Lambin P. A Deep Look Into the Future of Quantitative Imaging in Oncology: A Statement of Working Principles and Proposal for Change. Int J Radiat Oncol Biol Phys 2018; 102:1074-1082. [PMID: 30170101 DOI: 10.1016/j.ijrobp.2018.08.032] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 08/21/2018] [Accepted: 08/21/2018] [Indexed: 12/13/2022]
Abstract
The adoption of enterprise digital imaging, along with the development of quantitative imaging methods and the re-emergence of statistical learning, has opened the opportunity for more personalized cancer treatments through transformative data science research. In the last 5 years, accumulating evidence has indicated that noninvasive advanced imaging analytics (i.e., radiomics) can reveal key components of tumor phenotype for multiple lesions at multiple time points over the course of treatment. Many groups using homegrown software have extracted engineered and deep quantitative features on 3-dimensional medical images for better spatial and longitudinal understanding of tumor biology and for the prediction of diverse outcomes. These developments could augment patient stratification and prognostication, buttressing emerging targeted therapeutic approaches. Unfortunately, the rapid growth in popularity of this immature scientific discipline has resulted in many early publications that miss key information or use underpowered patient data sets, without production of generalizable results. Quantitative imaging research is complex, and key principles should be followed to realize its full potential. The fields of quantitative imaging and radiomics in particular require a renewed focus on optimal study design and reporting practices, standardization, interpretability, data sharing, and clinical trials. Standardization of image acquisition, feature calculation, and statistical analysis (i.e., machine learning) are required for the field to move forward. A new data-sharing paradigm enacted among open and diverse participants (medical institutions, vendors and associations) should be embraced for faster development and comprehensive clinical validation of imaging biomarkers. In this review and critique of the field, we propose working principles and fundamental changes to the current scientific approach, with the goal of high-impact research and development of actionable prediction models that will yield more meaningful applications of precision cancer medicine.
Collapse
Affiliation(s)
- Olivier Morin
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California.
| | | | - Arthur Jochems
- The D-Lab, Grow Research Institute for Oncology, Maastricht University, Maastricht, The Netherlands
| | - Henry C Woodruff
- The D-Lab, Grow Research Institute for Oncology, Maastricht University, Maastricht, The Netherlands
| | - Gilmer Valdes
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California
| | - Steve E Braunstein
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California
| | - Joachim E Wildberger
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | | | - Vasant Kearney
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California
| | - Sue S Yom
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California
| | - Timothy D Solberg
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California
| | - Philippe Lambin
- The D-Lab, Grow Research Institute for Oncology, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|