1
|
Paolo D, Greco C, Cortellini A, Ramella S, Soda P, Bria A, Sicilia R. Hierarchical embedding attention for overall survival prediction in lung cancer from unstructured EHRs. BMC Med Inform Decis Mak 2025; 25:169. [PMID: 40251623 PMCID: PMC12007135 DOI: 10.1186/s12911-025-02998-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 04/07/2025] [Indexed: 04/20/2025] Open
Abstract
The automated processing of Electronic Health Records (EHRs) poses a significant challenge due to their unstructured nature, rich in valuable, yet disorganized information. Natural Language Processing (NLP), particularly Named Entity Recognition (NER), has been instrumental in extracting structured information from EHR data. However, existing literature primarly focuses on extracting handcrafted clinical features through NLP and NER methods without delving into their learned representations. In this work, we explore the untapped potential of these representations by considering their contextual richness and entity-specific information. Our proposed methodology extracts representations generated by a transformer-based NER model on EHRs data, combines them using a hierarchical attention mechanism, and employs the obtained enriched representation as input for a clinical prediction model. Specifically, this study addresses Overall Survival (OS) in Non-Small Cell Lung Cancer (NSCLC) using unstructured EHRs data collected from an Italian clinical centre encompassing 838 records from 231 lung cancer patients. Whilst our study is applied on EHRs written in Italian, it serves as use case to prove the effectiveness of extracting and employing high level textual representations that capture relevant information as named entities. Our methodology is interpretable because the hierarchical attention mechanism highlights the information in EHRs that the model considers the most crucial during the decision-making process. We validated this interpretability by measuring the agreement of domain experts on the importance assigned by the hierarchical attention mechanism to EHRs information through a questionnaire. Results demonstrate the effectiveness of our method, showcasing statistically significant improvements over traditional manually extracted clinical features.
Collapse
Affiliation(s)
- Domenico Paolo
- Unit of Computer Systems & Bioinformatics, Department of Engineering, University Campus Bio-Medico di Roma, Roma, Italy
| | - Carlo Greco
- Research Unit of Radiation Oncology, Department of Medicine and Surgery, University Campus Bio-Medico di Roma, Roma, Italy
- Operative Research Unit of Radiation Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - Alessio Cortellini
- Operative Research Unit of Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - Sara Ramella
- Research Unit of Radiation Oncology, Department of Medicine and Surgery, University Campus Bio-Medico di Roma, Roma, Italy
- Operative Research Unit of Radiation Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - Paolo Soda
- Unit of Computer Systems & Bioinformatics, Department of Engineering, University Campus Bio-Medico di Roma, Roma, Italy.
- Department of Diagnostics and Intervention, Radiation Physics, Umeå University, Umeå, Sweden.
| | - Alessandro Bria
- Department of Electrical and Information Engineering, University of Cassino and Southern Latium, Cassino, Italy
| | - Rosa Sicilia
- Unit of Computer Systems & Bioinformatics, Department of Engineering, University Campus Bio-Medico di Roma, Roma, Italy
| |
Collapse
|
2
|
Narra LR, Kumar R, Deek MP, Jabbour SK. Updates in Management of Unresectable Stage III Non Small Cell Lung Cancer: A Radiation Oncology Perspective. Cancers (Basel) 2024; 16:4233. [PMID: 39766132 PMCID: PMC11674665 DOI: 10.3390/cancers16244233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/10/2024] [Accepted: 12/14/2024] [Indexed: 01/11/2025] Open
Abstract
Unresectable stage III non-small-cell lung cancer (NSCLC) remains a clinical challenge, due to the need for optimal local and systemic control. The management of unresectable Stage III NSCLC has evolved with advancements in radiation therapy (RT), systemic therapies, and immunotherapy. For patients with locally advanced NSCLC who are not surgical candidates, concurrent chemoradiotherapy (CRT) has modest survival outcomes, due to both local progression and distant metastasis. Efforts to enhance outcomes have led to dose-escalation trials, advances in modern RT techniques such as intensity-modulated RT (IMRT) and proton beam therapy (PBT), and the integration of adaptive RT to optimize target coverage while sparing organs at risk. Concurrent and consolidative immunotherapy, particularly with PD-L1 inhibitors, has shown promise, as evidenced by the PACIFIC trial, which demonstrated improved progression-free survival (PFS) and overall survival (OS) with durvalumab following CRT. Ongoing trials are now investigating novel immunotherapy combinations and targeted therapies in this setting, including dual checkpoint inhibition, DNA repair inhibitors, and molecularly targeted agents like osimertinib for EGFR-mutated NSCLC. Emerging biomarkers, such as circulating tumor DNA and radiomics, offer potential for personalizing treatment and predicting outcomes. Additionally, PBT and MR-guided adaptive RT have shown the potential to reduce toxicities while maintaining efficacy. Integrating these novel approaches may offer opportunities for optimizing treatment responses and minimizing adverse effects in this challenging patient population. Further investigation into patient stratification, biomarker-driven therapy, and refined therapeutic combinations is essential to improve long-term outcomes in unresectable Stage III NSCLC. This narrative review explores the current management strategies for unresectable Stage III NSCLC, from a radiation oncology perspective.
Collapse
Affiliation(s)
| | | | | | - Salma K. Jabbour
- Department of Radiation Oncology, Rutgers Cancer Institute, New Brunswick, NJ 08901, USA; (L.R.N.); (R.K.); (M.P.D.)
| |
Collapse
|
3
|
Demircioğlu A. radMLBench: A dataset collection for benchmarking in radiomics. Comput Biol Med 2024; 182:109140. [PMID: 39270457 DOI: 10.1016/j.compbiomed.2024.109140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 08/20/2024] [Accepted: 09/08/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND New machine learning methods and techniques are frequently introduced in radiomics, but they are often tested on a single dataset, which makes it challenging to assess their true benefit. Currently, there is a lack of a larger, publicly accessible dataset collection on which such assessments could be performed. In this study, a collection of radiomics datasets with binary outcomes in tabular form was curated to allow benchmarking of machine learning methods and techniques. METHODS A variety of journals and online sources were searched to identify tabular radiomics data with binary outcomes, which were then compiled into a homogeneous data collection that is easily accessible via Python. To illustrate the utility of the dataset collection, it was applied to investigate whether feature decorrelation prior to feature selection could improve predictive performance in a radiomics pipeline. RESULTS A total of 50 radiomic datasets were collected, with sample sizes ranging from 51 to 969 and 101 to 11165 features. Using this data, it was observed that decorrelating features did not yield any significant improvement on average. CONCLUSIONS A large collection of datasets, easily accessible via Python, suitable for benchmarking and evaluating new machine learning techniques and methods was curated. Its utility was exemplified by demonstrating that feature decorrelation prior to feature selection does not, on average, lead to significant performance gains and could be omitted, thereby increasing the robustness and reliability of the radiomics pipeline.
Collapse
Affiliation(s)
- Aydin Demircioğlu
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Hufelandstraße 55, D-45147, Essen, Germany.
| |
Collapse
|
4
|
Tran K, Ginzburg D, Hong W, Attenberger U, Ko HS. Post-radiotherapy stage III/IV non-small cell lung cancer radiomics research: a systematic review and comparison of CLEAR and RQS frameworks. Eur Radiol 2024; 34:6527-6543. [PMID: 38625613 PMCID: PMC11399214 DOI: 10.1007/s00330-024-10736-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/07/2024] [Accepted: 03/04/2024] [Indexed: 04/17/2024]
Abstract
BACKGROUND Lung cancer, the second most common cancer, presents persistently dismal prognoses. Radiomics, a promising field, aims to provide novel imaging biomarkers to improve outcomes. However, clinical translation faces reproducibility challenges, despite efforts to address them with quality scoring tools. OBJECTIVE This study had two objectives: 1) identify radiomics biomarkers in post-radiotherapy stage III/IV nonsmall cell lung cancer (NSCLC) patients, 2) evaluate research quality using the CLEAR (CheckList_for_EvaluAtion_of_Radiomics_research), RQS (Radiomics_Quality_Score) frameworks, and formulate an amalgamated CLEAR-RQS tool to enhance scientific rigor. MATERIALS AND METHODS A systematic literature review (Jun-Aug 2023, MEDLINE/PubMed/SCOPUS) was conducted concerning stage III/IV NSCLC, radiotherapy, and radiomic features (RF). Extracted data included study design particulars, such as sample size, radiotherapy/CT technique, selected RFs, and endpoints. CLEAR and RQS were merged into a CLEAR-RQS checklist. Three readers appraised articles utilizing CLEAR, RQS, and CLEAR-RQS metrics. RESULTS Out of 871 articles, 11 met the inclusion/exclusion criteria. The Median cohort size was 91 (range: 10-337) with 9 studies being single-center. No common RF were identified. The merged CLEAR-RQS checklist comprised 61 items. Most unreported items were within CLEAR's "methods" and "open-source," and within RQS's "phantom-calibration," "registry-enrolled prospective-trial-design," and "cost-effective-analysis" sections. No study scored above 50% on RQS. Median CLEAR scores were 55.74% (32.33/58 points), and for RQS, 17.59% (6.3/36 points). CLEAR-RQS article ranking fell between CLEAR and RQS and aligned with CLEAR. CONCLUSION Radiomics research in post-radiotherapy stage III/IV NSCLC exhibits variability and frequently low-quality reporting. The formulated CLEAR-RQS checklist may facilitate education and holds promise for enhancing radiomics research quality. CLINICAL RELEVANCE STATEMENT Current radiomics research in the field of stage III/IV postradiotherapy NSCLC is heterogenous, lacking reproducibility, with no identified imaging biomarker. Radiomics research quality assessment tools may enhance scientific rigor and thereby facilitate radiomics translation into clinical practice. KEY POINTS There is heterogenous and low radiomics research quality in postradiotherapy stage III/IV nonsmall cell lung cancer. Barriers to reproducibility are small cohort size, nonvalidated studies, missing technical parameters, and lack of data, code, and model sharing. CLEAR (CheckList_for_EvaluAtion_of_Radiomics_research), RQS (Radiomics_Quality_Score), and the amalgamated CLEAR-RQS tool are useful frameworks for assessing radiomics research quality and may provide a valuable resource for educational purposes in the field of radiomics.
Collapse
Affiliation(s)
- Kevin Tran
- Department of Cancer Imaging, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
- Faculty of Medicine, Dentistry & Health Sciences, University of Melbourne, Parkville, VIC 3052, Australia
| | - Daniel Ginzburg
- Department of Diagnostic and Interventional Radiology, Venusberg Campus 1, 53127, Bonn, Germany
| | - Wei Hong
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Ulrike Attenberger
- Department of Diagnostic and Interventional Radiology, Venusberg Campus 1, 53127, Bonn, Germany
| | - Hyun Soo Ko
- Department of Cancer Imaging, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia.
- Department of Diagnostic and Interventional Radiology, Venusberg Campus 1, 53127, Bonn, Germany.
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, 305 Grattan St, Melbourne, VIC 3000, Australia.
| |
Collapse
|
5
|
Jeong J, Choi H, Kim M, Kim SS, Goh J, Hwang J, Kim J, Cho HH, Eom K. Computed tomography radiomics models of tumor differentiation in canine small intestinal tumors. Front Vet Sci 2024; 11:1450304. [PMID: 39376912 PMCID: PMC11457012 DOI: 10.3389/fvets.2024.1450304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 09/09/2024] [Indexed: 10/09/2024] Open
Abstract
Radiomics models have been widely exploited in oncology for the investigation of tumor classification, as well as for predicting tumor response to treatment and genomic sequence; however, their performance in veterinary gastrointestinal tumors remains unexplored. Here, we sought to investigate and compare the performance of radiomics models in various settings for differentiating among canine small intestinal adenocarcinoma, lymphoma, and spindle cell sarcoma. Forty-two small intestinal tumors were contoured using four different segmentation methods: pre- or post-contrast, each with or without the inclusion of intraluminal gas. The mesenteric lymph nodes of pre- and post-contrast images were also contoured. The bin settings included bin count and bin width of 16, 32, 64, 128, and 256. Multinomial logistic regression, random forest, and support vector machine models were used to construct radiomics models. Using features from both primary tumors and lymph nodes showed significantly better performance than modeling using only the radiomics features of primary tumors, which indicated that the inclusion of mesenteric lymph nodes aids model performance. The support vector machine model exhibited significantly superior performance compared with the multinomial logistic regression and random forest models. Combining radiologic findings with radiomics features improved performance compared to using only radiomics features, highlighting the importance of radiologic findings in model building. A support vector machine model consisting of radiologic findings, primary tumors, and lymph node radiomics features with bin count 16 in post-contrast images with the exclusion of intraluminal gas showed the best performance among the various models tested. In conclusion, this study suggests that mesenteric lymph node segmentation and radiological findings should be integrated to build a potent radiomics model capable of differentiating among small intestinal tumors.
Collapse
Affiliation(s)
- Jeongyun Jeong
- Department of Veterinary Medical Imaging, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Hyunji Choi
- Department of Veterinary Medical Imaging, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Minjoo Kim
- Shine Animal Medical Center, Seoul, Republic of Korea
| | - Sung-Soo Kim
- VIP Animal Medical Center, Seoul, Republic of Korea
| | - Jinhyong Goh
- Daegu Animal Medical Center, Daegu, Republic of Korea
- Busan Jeil Animal Medical Center, Busan, Republic of Korea
| | | | - Jaehwan Kim
- Department of Veterinary Medical Imaging, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Hwan-Ho Cho
- Department of Electronics Engineering, Incheon National University, Incheon, Republic of Korea
| | - Kidong Eom
- Department of Veterinary Medical Imaging, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
6
|
Horne A, Harada K, Brown KD, Chua KLM, McDonald F, Price G, Putora PM, Rothwell DG, Faivre-Finn C. Treatment Response Biomarkers: Working Toward Personalized Radiotherapy for Lung Cancer. J Thorac Oncol 2024; 19:1164-1185. [PMID: 38615939 DOI: 10.1016/j.jtho.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/05/2024] [Accepted: 04/09/2024] [Indexed: 04/16/2024]
Abstract
Owing to major advances in the field of radiation oncology, patients with lung cancer can now receive technically individualized radiotherapy treatments. Nevertheless, in the era of precision oncology, radiotherapy-based treatment selection needs to be improved as many patients do not benefit or are not offered optimum therapies. Cost-effective robust biomarkers can address this knowledge gap and lead to individuals being offered more bespoke treatments leading to improved outcome. This narrative review discusses some of the current achievements and challenges in the realization of personalized radiotherapy delivery in patients with lung cancer.
Collapse
Affiliation(s)
- Ashley Horne
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom; Department of Radiation Oncology, The Christie NHS Foundation Trust, Manchester, United Kingdom.
| | - Ken Harada
- Department of Radiation Oncology, Showa University Northern Yokohama Hospital, Tsuzuki-ku, Yokohama, Kanagawa, Japan
| | - Katherine D Brown
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom; Department of Research and Innovation, The Christie Hospital NHS Foundation Trust, Manchester, United Kingdom
| | - Kevin Lee Min Chua
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore
| | | | - Gareth Price
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Paul Martin Putora
- Department of Radiation Oncology, Kantonsspital St. Gallen, St. Gallen, Switzerland; Department of Radiation Oncology, Inselspital, University of Bern, Bern, Switzerland
| | - Dominic G Rothwell
- CR-UK National Biomarker Centre, University of Manchester, Manchester, United Kingdom
| | - Corinne Faivre-Finn
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom; Department of Radiation Oncology, The Christie NHS Foundation Trust, Manchester, United Kingdom
| |
Collapse
|
7
|
Fiste O, Gkiozos I, Charpidou A, Syrigos NK. Artificial Intelligence-Based Treatment Decisions: A New Era for NSCLC. Cancers (Basel) 2024; 16:831. [PMID: 38398222 PMCID: PMC10887017 DOI: 10.3390/cancers16040831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/12/2024] [Accepted: 02/17/2024] [Indexed: 02/25/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related mortality among women and men, in developed countries, despite the public health interventions including tobacco-free campaigns, screening and early detection methods, recent therapeutic advances, and ongoing intense research on novel antineoplastic modalities. Targeting oncogenic driver mutations and immune checkpoint inhibition has indeed revolutionized NSCLC treatment, yet there still remains the unmet need for robust and standardized predictive biomarkers to accurately inform clinical decisions. Artificial intelligence (AI) represents the computer-based science concerned with large datasets for complex problem-solving. Its concept has brought a paradigm shift in oncology considering its immense potential for improved diagnosis, treatment guidance, and prognosis. In this review, we present the current state of AI-driven applications on NSCLC management, with a particular focus on radiomics and pathomics, and critically discuss both the existing limitations and future directions in this field. The thoracic oncology community should not be discouraged by the likely long road of AI implementation into daily clinical practice, as its transformative impact on personalized treatment approaches is undeniable.
Collapse
Affiliation(s)
- Oraianthi Fiste
- Oncology Unit, Third Department of Internal Medicine and Laboratory, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.G.); (A.C.); (N.K.S.)
| | | | | | | |
Collapse
|
8
|
Demircioğlu A. The effect of data resampling methods in radiomics. Sci Rep 2024; 14:2858. [PMID: 38310165 PMCID: PMC10838284 DOI: 10.1038/s41598-024-53491-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 02/01/2024] [Indexed: 02/05/2024] Open
Abstract
Radiomic datasets can be class-imbalanced, for instance, when the prevalence of diseases varies notably, meaning that the number of positive samples is much smaller than that of negative samples. In these cases, the majority class may dominate the model's training and thus negatively affect the model's predictive performance, leading to bias. Therefore, resampling methods are often utilized to class-balance the data. However, several resampling methods exist, and neither their relative predictive performance nor their impact on feature selection has been systematically analyzed. In this study, we aimed to measure the impact of nine resampling methods on radiomic models utilizing a set of fifteen publicly available datasets regarding their predictive performance. Furthermore, we evaluated the agreement and similarity of the set of selected features. Our results show that applying resampling methods did not improve the predictive performance on average. On specific datasets, slight improvements in predictive performance (+ 0.015 in AUC) could be seen. A considerable disagreement on the set of selected features was seen (only 28.7% of features agreed), which strongly impedes feature interpretability. However, selected features are similar when considering their correlation (82.9% of features correlated on average).
Collapse
Affiliation(s)
- Aydin Demircioğlu
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany.
| |
Collapse
|
9
|
Zhou Y, Zhang J, Li C, Chen J, Lv F, Deng Y, Chen S, Du Y, Li F. Prediction of non-perfusion volume ratio for uterine fibroids treated with ultrasound-guided high-intensity focused ultrasound based on MRI radiomics combined with clinical parameters. Biomed Eng Online 2023; 22:123. [PMID: 38093245 PMCID: PMC10717163 DOI: 10.1186/s12938-023-01182-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 11/23/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Prediction of non-perfusion volume ratio (NPVR) is critical in selecting patients with uterine fibroids who will potentially benefit from ultrasound-guided high-intensity focused ultrasound (HIFU) treatment, as it reduces the risk of treatment failure. The purpose of this study is to construct an optimal model for predicting NPVR based on T2-weighted magnetic resonance imaging (T2MRI) radiomics features combined with clinical parameters by machine learning. MATERIALS AND METHODS This retrospective study was conducted among 223 patients diagnosed with uterine fibroids from two centers. The patients from one center were allocated to a training cohort (n = 122) and an internal test cohort (n = 46), and the data from the other center (n = 55) was used as an external test cohort. The least absolute shrinkage and selection operator (LASSO) algorithm was employed for feature selection in the training cohort. The support vector machine (SVM) was adopted to construct a radiomics model, a clinical model, and a radiomics-clinical model for NPVR prediction, respectively. The area under the curve (AUC) and the decision curve analysis (DCA) were performed to evaluate the predictive validity and the clinical usefulness of the model, respectively. RESULTS A total of 851 radiomic features were extracted from T2MRI, of which seven radiomics features were screened for NPVR prediction-related radiomics features. The radiomics-clinical model combining radiomics features and clinical parameters showed the best predictive performance in both the internal (AUC = 0.824, 95% CI 0.693-0.954) and external (AUC = 0.773, 95% CI 0.647-0.902) test cohorts, and the DCA also suggested the radiomics-clinical model had the highest net benefit. CONCLUSIONS The radiomics-clinical model could be applied to the NPVR prediction of patients with uterine fibroids treated by HIFU to provide an objective and effective method for selecting potential patients who would benefit from the treatment mostly.
Collapse
Affiliation(s)
- Ye Zhou
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Jinwei Zhang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Chenghai Li
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China.
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China.
| | - Jinyun Chen
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Fajin Lv
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yongbin Deng
- Chongqing Haifu Hospital, Chongqing, 401121, China
| | - Siyao Chen
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Yuling Du
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Faqi Li
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China.
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
10
|
Nibid L, Greco C, Cordelli E, Sabarese G, Fiore M, Liu CZ, Ippolito E, Sicilia R, Miele M, Tortora M, Taffon C, Rakaee M, Soda P, Ramella S, Perrone G. Deep pathomics: A new image-based tool for predicting response to treatment in stage III non-small cell lung cancer. PLoS One 2023; 18:e0294259. [PMID: 38015944 PMCID: PMC10684067 DOI: 10.1371/journal.pone.0294259] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/26/2023] [Indexed: 11/30/2023] Open
Abstract
Despite the advantages offered by personalized treatments, there is presently no way to predict response to chemoradiotherapy in patients with non-small cell lung cancer (NSCLC). In this exploratory study, we investigated the application of deep learning techniques to histological tissue slides (deep pathomics), with the aim of predicting the response to therapy in stage III NSCLC. We evaluated 35 digitalized tissue slides (biopsies or surgical specimens) obtained from patients with stage IIIA or IIIB NSCLC. Patients were classified as responders (12/35, 34.7%) or non-responders (23/35, 65.7%) based on the target volume reduction shown on weekly CT scans performed during chemoradiation treatment. Digital tissue slides were tested by five pre-trained convolutional neural networks (CNNs)-AlexNet, VGG, MobileNet, GoogLeNet, and ResNet-using a leave-two patient-out cross validation approach, and we evaluated the networks' performances. GoogLeNet was globally found to be the best CNN, correctly classifying 8/12 responders and 10/11 non-responders. Moreover, Deep-Pathomics was found to be highly specific (TNr: 90.1) and quite sensitive (TPr: 0.75). Our data showed that AI could surpass the capabilities of all presently available diagnostic systems, supplying additional information beyond that currently obtainable in clinical practice. The ability to predict a patient's response to treatment could guide the development of new and more effective therapeutic AI-based approaches and could therefore be considered an effective and innovative step forward in personalised medicine.
Collapse
Affiliation(s)
- Lorenzo Nibid
- Research Unit of Anatomical Pathology, Department of of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Carlo Greco
- Radiation Oncology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Radiation Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Ermanno Cordelli
- Unit of Computer Systems and Bioinformatics, Department of Engineering, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Giovanna Sabarese
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Michele Fiore
- Radiation Oncology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Radiation Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Charles Z. Liu
- Unit of Computer Systems and Bioinformatics, Department of Engineering, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Edy Ippolito
- Radiation Oncology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Radiation Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Rosa Sicilia
- Unit of Computer Systems and Bioinformatics, Department of Engineering, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Marianna Miele
- Radiation Oncology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Radiation Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Matteo Tortora
- Unit of Computer Systems and Bioinformatics, Department of Engineering, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Chiara Taffon
- Research Unit of Anatomical Pathology, Department of of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Mehrdad Rakaee
- Department of Clinical Pathology, University Hospital of North Norway, Tromsø, Norway
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Paolo Soda
- Unit of Computer Systems and Bioinformatics, Department of Engineering, Università Campus Bio-Medico di Roma, Rome, Italy
- Department of Radiation Sciences, Radiation Physics, Biomedical Engineering, Umeå University, Umeå, Sweden
| | - Sara Ramella
- Radiation Oncology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Radiation Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Giuseppe Perrone
- Research Unit of Anatomical Pathology, Department of of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| |
Collapse
|
11
|
Mumtaz H, Saqib M, Jabeen S, Muneeb M, Mughal W, Sohail H, Safdar M, Mehmood Q, Khan MA, Ismail SM. Exploring alternative approaches to precision medicine through genomics and artificial intelligence - a systematic review. Front Med (Lausanne) 2023; 10:1227168. [PMID: 37849490 PMCID: PMC10577305 DOI: 10.3389/fmed.2023.1227168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/20/2023] [Indexed: 10/19/2023] Open
Abstract
The core idea behind precision medicine is to pinpoint the subpopulations that differ from one another in terms of disease risk, drug responsiveness, and treatment outcomes due to differences in biology and other traits. Biomarkers are found through genomic sequencing. Multi-dimensional clinical and biological data are created using these biomarkers. Better analytic methods are needed for these multidimensional data, which can be accomplished by using artificial intelligence (AI). An updated review of 80 latest original publications is presented on four main fronts-preventive medicine, medication development, treatment outcomes, and diagnostic medicine-All these studies effectively illustrated the significance of AI in precision medicine. Artificial intelligence (AI) has revolutionized precision medicine by swiftly analyzing vast amounts of data to provide tailored treatments and predictive diagnostics. Through machine learning algorithms and high-resolution imaging, AI assists in precise diagnoses and early disease detection. AI's ability to decode complex biological factors aids in identifying novel therapeutic targets, allowing personalized interventions and optimizing treatment outcomes. Furthermore, AI accelerates drug discovery by navigating chemical structures and predicting drug-target interactions, expediting the development of life-saving medications. With its unrivaled capacity to comprehend and interpret data, AI stands as an invaluable tool in the pursuit of enhanced patient care and improved health outcomes. It's evident that AI can open a new horizon for precision medicine by translating complex data into actionable information. To get better results in this regard and to fully exploit the great potential of AI, further research is required on this pressing subject.
Collapse
Affiliation(s)
| | | | | | - Muhammad Muneeb
- Department of Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Wajiha Mughal
- Department of Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Hassan Sohail
- Department of Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Myra Safdar
- Armed Forces Institute of Cardiology and National Institute of Heart Diseases (AFIC-NIHD), Rawalpindi, Pakistan
| | - Qasim Mehmood
- Department of Medicine, King Edward Medical University, Lahore, Pakistan
| | - Muhammad Ahsan Khan
- Department of Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | | |
Collapse
|
12
|
Zhou C, Hou L, Tang X, Liu C, Meng Y, Jia H, Yang H, Zhou S. CT-based radiomics nomogram may predict who can benefit from adaptive radiotherapy in patients with local advanced-NSCLC patients. Radiother Oncol 2023; 183:109637. [PMID: 36963440 DOI: 10.1016/j.radonc.2023.109637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 02/14/2023] [Accepted: 03/17/2023] [Indexed: 03/26/2023]
Abstract
BACKGROUND Although adaptive radiotherapy (ART) has many advantages, ART is not universal in the clinical appliance due to the consumption of a lot of labor, and economic burden. It is necessary to explore a CT stimulation-based radiomics model for screening who can get more benefits from ART in locally advanced non-small cell lung cancer (NSCLC) patients. METHOD 183 cases of NSCLC patients receiving concurrent chemoradiotherapy with an adaptive approach were enrolled as a primary cohort, while 28 cases from another hospital served as an independent external validation cohort. Tumor regression assessment was conducted based on GTV reduction (Criteria A) or according to RECIST Version 1.1(Criteria B). The radiomics features were extracted by the "PyRadiomics" package and further screened by the LASSO method. Then, logistic regression was used to establish the model. Bootstrap and external validation were applied to verify the stability of the model. The receiver operating characteristic (ROC) curve was delineated to assess the predictive efficacy of the radiomics model. Dose-volume histograms were quantitatively compared between the initial and composite ART plans. Clinical endpoints included overall survival (OS) and progression-free survival (PFS). RESULT There were no significant differences in clinical features between tumor regression-resistant (RR) and tumor regression-sensitivity (RS) groups. The AUC values of the Criteria A model and Criteria B model were 0.767 and 0.771, respectively. Bootstrapping validation and external validation confirmed the stability of models. In all patients, there was a significant benefit of ART in the lung, heart, cord, and esophagus compared to non-ART, particularly in RS patients. Furthermore, PFS and OS from ART were significantly longer in RS as defined by Criterion B than in RR patients with the same ART application. CONCLUSION CT-based radiomics can screen out the patients who can gain more benefits from ART, which contribute to guiding and popularizing the application of ART strategy in the clinic within economic benefits and feasibility.
Collapse
Affiliation(s)
- Chao Zhou
- From Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Zhejiang Province 317000, China
| | - Liqiao Hou
- From Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Zhejiang Province 317000, China
| | - Xingni Tang
- From Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Zhejiang Province 317000, China
| | - Changxing Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Yinnan Meng
- From Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Zhejiang Province 317000, China
| | - Haijian Jia
- Department of Radiation Oncology, Enze Hospital Affiliated Hospital of Hangzhou Medical College, Zhejiang Province 317000, China
| | - Haihua Yang
- Department of Radiation Oncology, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi 710018, P.R. China.
| | - Suna Zhou
- From Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Zhejiang Province 317000, China; Department of Radiation Oncology, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Xi'an, Shaanxi 710018, P.R. China.
| |
Collapse
|
13
|
Prata F, Anceschi U, Cordelli E, Faiella E, Civitella A, Tuzzolo P, Iannuzzi A, Ragusa A, Esperto F, Prata SM, Sicilia R, Muto G, Grasso RF, Scarpa RM, Soda P, Simone G, Papalia R. Radiomic Machine-Learning Analysis of Multiparametric Magnetic Resonance Imaging in the Diagnosis of Clinically Significant Prostate Cancer: New Combination of Textural and Clinical Features. Curr Oncol 2023; 30:2021-2031. [PMID: 36826118 PMCID: PMC9955797 DOI: 10.3390/curroncol30020157] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/10/2023] Open
Abstract
BACKGROUND The aim of our study was to develop a radiomic tool for the prediction of clinically significant prostate cancer. METHODS From September 2020 to December 2021, 91 patients who underwent magnetic resonance imaging prostate fusion biopsy at our institution were selected. Prostate cancer aggressiveness was assessed by combining the three orthogonal planes-Llocal binary pattern the 3Dgray level co-occurrence matrix, and other first order statistical features with clinical (semantic) features. The 487 features were used to predict whether the Gleason score was clinically significant (≥7) in the final pathology. A feature selection algorithm was used to determine the most predictive features, and at the end of the process, nine features were chosen through a 10-fold cross validation. RESULTS The feature analysis revealed a detection accuracy of 83.5%, with a clinically significant precision of 84.4% and a clinically significant sensitivity of 91.5%. The resulting area under the curve was 80.4%. CONCLUSIONS Radiomic analysis allowed us to develop a tool that was able to predict a Gleason score of ≥7. This new tool may improve the detection rate of clinically significant prostate cancer and overcome the limitations of the subjective interpretation of magnetic resonance imaging, reducing the number of useless biopsies.
Collapse
Affiliation(s)
- Francesco Prata
- Department of Urology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
- Correspondence: ; Tel.: +39-39-3437-3027; Fax: +39-062-2541-1995
| | - Umberto Anceschi
- Department of Urology, IRCCS “Regina Elena” National Cancer Institute, 00144 Rome, Italy
| | - Ermanno Cordelli
- Unit of Computer Systems and Bioinformatics, Department of Engineering, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Eliodoro Faiella
- Department of Diagnostic and Interventional Radiology, Sant’Anna Hospital, 22042 San Fermo della Battaglia, Italy
| | - Angelo Civitella
- Department of Urology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Piergiorgio Tuzzolo
- Department of Urology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Andrea Iannuzzi
- Department of Urology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Alberto Ragusa
- Department of Urology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Francesco Esperto
- Department of Urology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Salvatore Mario Prata
- Simple Operating Unit of Lower Urinary Tract Surgery, SS. Trinità Hospital, 03039 Sora, Italy
| | - Rosa Sicilia
- Unit of Computer Systems and Bioinformatics, Department of Engineering, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Giovanni Muto
- Department of Urology, Humanitas Gradenigo University, 10153 Turin, Italy
| | - Rosario Francesco Grasso
- Department of Diagnostic and Interventional Radiology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Roberto Mario Scarpa
- Department of Urology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Paolo Soda
- Unit of Computer Systems and Bioinformatics, Department of Engineering, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Giuseppe Simone
- Department of Urology, IRCCS “Regina Elena” National Cancer Institute, 00144 Rome, Italy
| | - Rocco Papalia
- Department of Urology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| |
Collapse
|
14
|
Ge G, Zhang J. Feature selection methods and predictive models in CT lung cancer radiomics. J Appl Clin Med Phys 2023; 24:e13869. [PMID: 36527376 PMCID: PMC9860004 DOI: 10.1002/acm2.13869] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/31/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
Radiomics is a technique that extracts quantitative features from medical images using data-characterization algorithms. Radiomic features can be used to identify tissue characteristics and radiologic phenotyping that is not observable by clinicians. A typical workflow for a radiomics study includes cohort selection, radiomic feature extraction, feature and predictive model selection, and model training and validation. While there has been increasing attention given to radiomic feature extraction, standardization, and reproducibility, currently, there is a lack of rigorous evaluation of feature selection methods and predictive models. Herein, we review the published radiomics investigations in CT lung cancer and provide an overview of the commonly used radiomic feature selection methods and predictive models. We also compare limitations of various methods in clinical applications and present sources of uncertainty associated with those methods. This review is expected to help raise awareness of the impact of radiomic feature and model selection methods on the integrity of radiomics studies.
Collapse
Affiliation(s)
- Gary Ge
- Department of Radiology, University of Kentucky, Lexington, Kentucky, USA
| | - Jie Zhang
- Department of Radiology, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
15
|
Caruso CM, Guarrasi V, Cordelli E, Sicilia R, Gentile S, Messina L, Fiore M, Piccolo C, Beomonte Zobel B, Iannello G, Ramella S, Soda P. A Multimodal Ensemble Driven by Multiobjective Optimisation to Predict Overall Survival in Non-Small-Cell Lung Cancer. J Imaging 2022; 8:298. [PMID: 36354871 PMCID: PMC9697158 DOI: 10.3390/jimaging8110298] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/28/2022] [Accepted: 10/30/2022] [Indexed: 09/10/2024] Open
Abstract
Lung cancer accounts for more deaths worldwide than any other cancer disease. In order to provide patients with the most effective treatment for these aggressive tumours, multimodal learning is emerging as a new and promising field of research that aims to extract complementary information from the data of different modalities for prognostic and predictive purposes. This knowledge could be used to optimise current treatments and maximise their effectiveness. To predict overall survival, in this work, we investigate the use of multimodal learning on the CLARO dataset, which includes CT images and clinical data collected from a cohort of non-small-cell lung cancer patients. Our method allows the identification of the optimal set of classifiers to be included in the ensemble in a late fusion approach. Specifically, after training unimodal models on each modality, it selects the best ensemble by solving a multiobjective optimisation problem that maximises both the recognition performance and the diversity of the predictions. In the ensemble, the labels of each sample are assigned using the majority voting rule. As further validation, we show that the proposed ensemble outperforms the models learning a single modality, obtaining state-of-the-art results on the task at hand.
Collapse
Affiliation(s)
- Camillo Maria Caruso
- Research Unit of Computer Systems and Bioinformatics, Department of Engineering, Università Campus Bio-Medico di Roma, Via Àlvaro del Portillo, 21, 00128 Roma, Italy
| | - Valerio Guarrasi
- Research Unit of Computer Systems and Bioinformatics, Department of Engineering, Università Campus Bio-Medico di Roma, Via Àlvaro del Portillo, 21, 00128 Roma, Italy
- Department of Computer, Control, and Management Engineering, Sapienza University of Rome, 00185 Roma, Italy
| | - Ermanno Cordelli
- Research Unit of Computer Systems and Bioinformatics, Department of Engineering, Università Campus Bio-Medico di Roma, Via Àlvaro del Portillo, 21, 00128 Roma, Italy
| | - Rosa Sicilia
- Research Unit of Computer Systems and Bioinformatics, Department of Engineering, Università Campus Bio-Medico di Roma, Via Àlvaro del Portillo, 21, 00128 Roma, Italy
| | - Silvia Gentile
- Operative Research Unit of Radiation Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
| | - Laura Messina
- Operative Research Unit of Diagnostic Imaging, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
| | - Michele Fiore
- Operative Research Unit of Radiation Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
- Research Unit of Radiation Oncology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Àlvaro del Portillo, 21, 00128 Roma, Italy
| | - Claudia Piccolo
- Operative Research Unit of Diagnostic Imaging, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
| | - Bruno Beomonte Zobel
- Operative Research Unit of Diagnostic Imaging, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
- Research Unit of Diagnostic Imaging, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Àlvaro del Portillo, 21, 00128 Roma, Italy
| | - Giulio Iannello
- Research Unit of Computer Systems and Bioinformatics, Department of Engineering, Università Campus Bio-Medico di Roma, Via Àlvaro del Portillo, 21, 00128 Roma, Italy
| | - Sara Ramella
- Operative Research Unit of Radiation Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
- Research Unit of Radiation Oncology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Àlvaro del Portillo, 21, 00128 Roma, Italy
| | - Paolo Soda
- Research Unit of Computer Systems and Bioinformatics, Department of Engineering, Università Campus Bio-Medico di Roma, Via Àlvaro del Portillo, 21, 00128 Roma, Italy
- Department of Radiation Sciences, Radiation Physics, Biomedical Engineering, Umeå University, 901 87 Umeå, Sweden
| |
Collapse
|
16
|
Iliadou V, Kakkos I, Karaiskos P, Kouloulias V, Platoni K, Zygogianni A, Matsopoulos GK. Early Prediction of Planning Adaptation Requirement Indication Due to Volumetric Alterations in Head and Neck Cancer Radiotherapy: A Machine Learning Approach. Cancers (Basel) 2022; 14:cancers14153573. [PMID: 35892831 PMCID: PMC9331795 DOI: 10.3390/cancers14153573] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/14/2022] [Accepted: 07/20/2022] [Indexed: 11/16/2022] Open
Abstract
Background: During RT cycles, the tumor response pattern could affect tumor coverage and may lead to organs at risk of overdose. As such, early prediction of significant volumetric changes could therefore reduce potential radiation-related adverse effects. Nevertheless, effective machine learning approaches based on the radiomic features of the clinically used CBCT images to determine the tumor volume variations due to RT not having been implemented so far. Methods: CBCT images from 40 HN cancer patients were collected weekly during RT treatment. From the obtained images, the Clinical Target Volume (CTV) and Parotid Glands (PG) regions of interest were utilized to calculate 104 delta-radiomics features. These features were fed on a feature selection and classification procedure for the early prediction of significant volumetric alterations. Results: The proposed framework was able to achieve 0.90 classification performance accuracy while detecting a small subset of discriminative characteristics from the 1st week of RT. The selected features were further analyzed regarding their effects on temporal changes in anatomy and tumor response modeling. Conclusion: The use of machine learning algorithms offers promising perspectives for fast and reliable early prediction of large volumetric deviations as a result of RT treatment, exploiting hidden patterns in the overall anatomical characteristics.
Collapse
Affiliation(s)
- Vasiliki Iliadou
- School of Electrical and Computer Engineering, National Technical University of Athens, 157 73 Athens, Greece; (I.K.); (G.K.M.)
- Correspondence: ; Tel.: +30-21-0772-3577
| | - Ioannis Kakkos
- School of Electrical and Computer Engineering, National Technical University of Athens, 157 73 Athens, Greece; (I.K.); (G.K.M.)
- Department of Biomedical Engineering, University of West Attica, 122 43 Athens, Greece
| | - Pantelis Karaiskos
- Medical Physics Laboratory, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece;
| | - Vassilis Kouloulias
- 2nd Department of Radiology, Radiotherapy Unit, ATTIKON University Hospital, 124 62 Athens, Greece; (V.K.); (K.P.)
| | - Kalliopi Platoni
- 2nd Department of Radiology, Radiotherapy Unit, ATTIKON University Hospital, 124 62 Athens, Greece; (V.K.); (K.P.)
| | - Anna Zygogianni
- 1st Department of Radiology, Radiotherapy Unit, ARETAIEION University Hospital, 115 28 Athens, Greece;
| | - George K. Matsopoulos
- School of Electrical and Computer Engineering, National Technical University of Athens, 157 73 Athens, Greece; (I.K.); (G.K.M.)
| |
Collapse
|
17
|
Cortiula F, Reymen B, Peters S, Van Mol P, Wauters E, Vansteenkiste J, De Ruysscher D, Hendriks LEL. Immunotherapy in unresectable stage III non-small-cell lung cancer: state of the art and novel therapeutic approaches. Ann Oncol 2022; 33:893-908. [PMID: 35777706 DOI: 10.1016/j.annonc.2022.06.013] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/21/2022] [Accepted: 06/21/2022] [Indexed: 12/11/2022] Open
Abstract
The standard of care for patients with stage III non-small-cell lung cancer (NSCLC) is concurrent chemoradiotherapy (CCRT) followed by 1 year of adjuvant durvalumab. Despite the survival benefit granted by immunotherapy in this setting, only 1/3 of patients are alive and disease free at 5 years. Novel treatment strategies are under development to improve patient outcomes in this setting: different anti-programmed cell death protein 1/programmed death-ligand 1 [anti-PD-(L)1] antibodies after CCRT, consolidation immunotherapy after sequential chemoradiotherapy, induction immunotherapy before CCRT and immunotherapy concurrent with CCRT and/or sequential chemoradiotherapy. Cross-trial comparison is particularly challenging in this setting due to the different timing of immunotherapy delivery and different patients' inclusion and exclusion criteria. In this review, we present the results of clinical trials investigating immune therapy in unresectable stage III NSCLC and discuss in-depth their biological rationale, their pitfalls and potential benefits. Particular emphasis is placed on the potential mechanisms of synergism between chemotherapy, radiation therapy and different monoclonal antibodies, and how this affects the tumor immune microenvironment. The designs and questions tackled by ongoing clinical trials are also discussed. Last, we address open questions and unmet clinical needs, such as the necessity for predictive biomarkers (e.g. radiomics and circulating tumor DNA). Identifying distinct subsets of patients to tailor anticancer treatment is a priority, especially in a heterogeneous disease such as stage III NSCLC.
Collapse
Affiliation(s)
- F Cortiula
- Department of Radiation Oncology (Maastro), Maastricht University Medical Centre(+), GROW School for Oncology and Reproduction, Maastricht, the Netherlands; Department of Medical Oncology, Udine University Hospital, Udine, Italy
| | - B Reymen
- Department of Radiation Oncology (Maastro), Maastricht University Medical Centre(+), GROW School for Oncology and Reproduction, Maastricht, the Netherlands
| | - S Peters
- Oncology Department, Lausanne University Hospital, Lausanne, Switzerland
| | - P Van Mol
- Department of Respiratory Diseases KU Leuven, Respiratory Oncology Unit, University Hospitals KU Leuven, Leuven, Belgium
| | - E Wauters
- Department of Respiratory Diseases KU Leuven, Respiratory Oncology Unit, University Hospitals KU Leuven, Leuven, Belgium
| | - J Vansteenkiste
- Department of Respiratory Diseases KU Leuven, Respiratory Oncology Unit, University Hospitals KU Leuven, Leuven, Belgium.
| | - D De Ruysscher
- Department of Radiation Oncology (Maastro), Maastricht University Medical Centre(+), GROW School for Oncology and Reproduction, Maastricht, the Netherlands
| | - L E L Hendriks
- Department of Pulmonary Diseases, Maastricht University Medical Centre(+), GROW School for Oncology and Reproduction, Maastricht, the Netherlands
| |
Collapse
|
18
|
Abdollahi H, Chin E, Clark H, Hyde DE, Thomas S, Wu J, Uribe CF, Rahmim A. Radiomics-guided radiation therapy: opportunities and challenges. Phys Med Biol 2022; 67. [DOI: 10.1088/1361-6560/ac6fab] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 05/13/2022] [Indexed: 11/11/2022]
Abstract
Abstract
Radiomics is an advanced image-processing framework, which extracts image features and considers them as biomarkers towards personalized medicine. Applications include disease detection, diagnosis, prognosis, and therapy response assessment/prediction. As radiation therapy aims for further individualized treatments, radiomics could play a critical role in various steps before, during and after treatment. Elucidation of the concept of radiomics-guided radiation therapy (RGRT) is the aim of this review, attempting to highlight opportunities and challenges underlying the use of radiomics to guide clinicians and physicists towards more effective radiation treatments. This work identifies the value of RGRT in various steps of radiotherapy from patient selection to follow-up, and subsequently provides recommendations to improve future radiotherapy using quantitative imaging features.
Collapse
|
19
|
Ungan G, Lavandier AF, Rouanet J, Hordonneau C, Chauveau B, Pereira B, Boyer L, Garcier JM, Mansard S, Bartoli A, Magnin B. Metastatic melanoma treated by immunotherapy: discovering prognostic markers from radiomics analysis of pretreatment CT with feature selection and classification. Int J Comput Assist Radiol Surg 2022; 17:1867-1877. [PMID: 35650345 DOI: 10.1007/s11548-022-02662-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 04/26/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE Immunotherapy has dramatically improved the prognosis of patients with metastatic melanoma (MM). Yet, there is a lack of biomarkers to predict whether a patient will benefit from immunotherapy. Our aim was to create radiomics models on pretreatment computed tomography (CT) to predict overall survival (OS) and treatment response in patients with MM treated with anti-PD-1 immunotherapy. METHODS We performed a monocentric retrospective analysis of 503 metastatic lesions in 71 patients with 46 radiomics features extracted following lesion segmentation. Predictive accuracies for OS < 1 year versus > 1 year and treatment response versus no response was compared for five feature selection methods (sequential forward selection, recursive, Boruta, relief, random forest) and four classifiers (support vector machine (SVM), random forest, K-nearest neighbor, logistic regression (LR)) used with or without SMOTE data augmentation. A fivefold cross-validation was performed at the patient level, with a tumour-based classification. RESULTS The highest accuracy level for OS predictions was obtained with 3D lesions (0.91) without clinical data integration when combining Boruta feature selection and the LR classifier, The highest accuracy for treatment response prediction was obtained with 3D lesions (0.88) without clinical data integration when combining Boruta feature selection, the LR classifier and SMOTE data augmentation. The accuracy was significantly higher concerning OS prediction with 3D segmentation (0.91 vs 0.86) while clinical data integration led to improved accuracy notably in 2D lesions (0.76 vs 0.87) regarding treatment response prediction. Skewness was the only feature found to be an independent predictor of OS (HR (CI 95%) 1.34, p-value 0.001). CONCLUSION This is the first study to investigate CT texture parameter selection and classification methods for predicting MM prognosis with treatment by immunotherapy. Combining pretreatment CT radiomics features from a single tumor with data selection and classifiers may accurately predict OS and treatment response in MM treated with anti-PD-1.
Collapse
Affiliation(s)
- Gulnur Ungan
- EnCoV, Institut Pascal, UMR 6602 CNRS, Université Clermont Auvergne, 28 place Henri Dunant, 63000, Clermont-Ferrand, France
| | - Anne-Flore Lavandier
- Department of Medical Imaging, CHU Clermont Ferrand, 1 place Lucie Aubrac, 63100, Clermont-Ferrand, France
| | - Jacques Rouanet
- Dermatology Department, CHU Clermont Ferrand, 1 place Lucie Aubrac, 63100, Clermont-Ferrand, France
| | - Constance Hordonneau
- Department of Medical Imaging, CHU Clermont Ferrand, 1 place Lucie Aubrac, 63100, Clermont-Ferrand, France
| | - Benoit Chauveau
- Department of Medical Imaging, CHU Clermont Ferrand, 1 place Lucie Aubrac, 63100, Clermont-Ferrand, France
| | - Bruno Pereira
- Biostatistics Unit, DRCI, CHU Clermont Ferrand, 58 rue Montalembert, 63000, Clermont-Ferrand, France
| | - Louis Boyer
- Department of Medical Imaging, CHU Clermont Ferrand, 1 place Lucie Aubrac, 63100, Clermont-Ferrand, France
| | - Jean-Marc Garcier
- Department of Medical Imaging, CHU Clermont Ferrand, 1 place Lucie Aubrac, 63100, Clermont-Ferrand, France.,Anatomy Department, Université Clermont Auvergne, 28 place Henri Dunant, 63000, Clermont-Ferrand, France
| | - Sandrine Mansard
- Dermatology Department, CHU Clermont Ferrand, 1 place Lucie Aubrac, 63100, Clermont-Ferrand, France
| | - Adrien Bartoli
- EnCoV, Institut Pascal, UMR 6602 CNRS, Université Clermont Auvergne, 28 place Henri Dunant, 63000, Clermont-Ferrand, France
| | - Benoit Magnin
- EnCoV, Institut Pascal, UMR 6602 CNRS, Université Clermont Auvergne, 28 place Henri Dunant, 63000, Clermont-Ferrand, France. .,Department of Medical Imaging, CHU Clermont Ferrand, 1 place Lucie Aubrac, 63100, Clermont-Ferrand, France. .,Anatomy Department, Université Clermont Auvergne, 28 place Henri Dunant, 63000, Clermont-Ferrand, France.
| |
Collapse
|
20
|
Gao H, He ZY, Du XL, Wang ZG, Xiang L. Machine Learning for the Prediction of Synchronous Organ-Specific Metastasis in Patients With Lung Cancer. Front Oncol 2022; 12:817372. [PMID: 35646679 PMCID: PMC9136456 DOI: 10.3389/fonc.2022.817372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 04/11/2022] [Indexed: 12/24/2022] Open
Abstract
Background This study aimed to develop an artificial neural network (ANN) model for predicting synchronous organ-specific metastasis in lung cancer (LC) patients. Methods A total of 62,151 patients who diagnosed as LC without data missing between 2010 and 2015 were identified from Surveillance, Epidemiology, and End Results (SEER) program. The ANN model was trained and tested on an 75/25 split of the dataset. The receiver operating characteristic (ROC) curves, area under the curve (AUC) and sensitivity were used to evaluate and compare the ANN model with the random forest model. Results For distant metastasis in the whole cohort, the ANN model had metrics AUC = 0.759, accuracy = 0.669, sensitivity = 0.906, and specificity = 0.613, which was better than the random forest model. For organ-specific metastasis in the cohort with distant metastasis, the sensitivity in bone metastasis, brain metastasis and liver metastasis were 0.913, 0.906 and 0.925, respectively. The most important variable was separate tumor nodules with 100% importance. The second important variable was visceral pleural invasion for distant metastasis, while histology for organ-specific metastasis. Conclusions Our study developed a “two-step” ANN model for predicting synchronous organ-specific metastasis in LC patients. This ANN model may provide clinicians with more personalized clinical decisions, contribute to rationalize metastasis screening, and reduce the burden on patients and the health care system.
Collapse
Affiliation(s)
- Huan Gao
- School of Medicine and Health Management, Huazhong University of Science and Technology, Wuhan, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhi-yi He
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing-li Du
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zheng-gang Wang
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Zheng-gang Wang, ; Li Xiang,
| | - Li Xiang
- School of Medicine and Health Management, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Zheng-gang Wang, ; Li Xiang,
| |
Collapse
|
21
|
Demircioğlu A. Evaluation of the dependence of radiomic features on the machine learning model. Insights Imaging 2022; 13:28. [PMID: 35201534 PMCID: PMC8873309 DOI: 10.1186/s13244-022-01170-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 02/03/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND In radiomic studies, several models are often trained with different combinations of feature selection methods and classifiers. The features of the best model are usually considered relevant to the problem, and they represent potential biomarkers. Features selected from statistically similarly performing models are generally not studied. To understand the degree to which the selected features of these statistically similar models differ, 14 publicly available datasets, 8 feature selection methods, and 8 classifiers were used in this retrospective study. For each combination of feature selection and classifier, a model was trained, and its performance was measured with AUC-ROC. The best-performing model was compared to other models using a DeLong test. Models that were statistically similar were compared in terms of their selected features. RESULTS Approximately 57% of all models analyzed were statistically similar to the best-performing model. Feature selection methods were, in general, relatively unstable (0.58; range 0.35-0.84). The features selected by different models varied largely (0.19; range 0.02-0.42), although the selected features themselves were highly correlated (0.71; range 0.4-0.92). CONCLUSIONS Feature relevance in radiomics strongly depends on the model used, and statistically similar models will generally identify different features as relevant. Considering features selected by a single model is misleading, and it is often not possible to directly determine whether such features are candidate biomarkers.
Collapse
Affiliation(s)
- Aydin Demircioğlu
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Hufelandstraße 55, 45157, Essen, Germany.
| |
Collapse
|
22
|
Lam SK, Zhang Y, Zhang J, Li B, Sun JC, Liu CYT, Chou PH, Teng X, Ma ZR, Ni RY, Zhou T, Peng T, Xiao HN, Li T, Ren G, Cheung ALY, Lee FKH, Yip CWY, Au KH, Lee VHF, Chang ATY, Chan LWC, Cai J. Multi-Organ Omics-Based Prediction for Adaptive Radiation Therapy Eligibility in Nasopharyngeal Carcinoma Patients Undergoing Concurrent Chemoradiotherapy. Front Oncol 2022; 11:792024. [PMID: 35174068 PMCID: PMC8842229 DOI: 10.3389/fonc.2021.792024] [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: 10/09/2021] [Accepted: 12/01/2021] [Indexed: 12/18/2022] Open
Abstract
Purpose To investigate the role of different multi-organ omics-based prediction models for pre-treatment prediction of Adaptive Radiotherapy (ART) eligibility in patients with nasopharyngeal carcinoma (NPC). Methods and Materials Pre-treatment contrast-enhanced computed tomographic and magnetic resonance images, radiotherapy dose and contour data of 135 NPC patients treated at Hong Kong Queen Elizabeth Hospital were retrospectively analyzed for extraction of multi-omics features, namely Radiomics (R), Morphology (M), Dosiomics (D), and Contouromics (C), from a total of eight organ structures. During model development, patient cohort was divided into a training set and a hold-out test set in a ratio of 7 to 3 via 20 iterations. Four single-omics models (R, M, D, C) and four multi-omics models (RD, RC, RM, RMDC) were developed on the training data using Ridge and Multi-Kernel Learning (MKL) algorithm, respectively, under 10-fold cross validation, and evaluated on hold-out test data using average area under the receiver-operator-characteristics curve (AUC). The best-performing single-omics model was first determined by comparing the AUC distribution across the 20 iterations among the four single-omics models using two-sided student t-test, which was then retrained using MKL algorithm for a fair comparison with the four multi-omics models. Results The R model significantly outperformed all other three single-omics models (all p-value<0.0001), achieving an average AUC of 0.942 (95%CI: 0.938-0.946) and 0.918 (95%CI: 0.903-0.933) in training and hold-out test set, respectively. When trained with MKL, the R model (R_MKL) yielded an increased AUC of 0.984 (95%CI: 0.981-0.988) and 0.927 (95%CI: 0.905-0.948) in training and hold-out test set respectively, while demonstrating no significant difference as compared to all studied multi-omics models in the hold-out test sets. Intriguingly, Radiomic features accounted for the majority of the final selected features, ranging from 64% to 94%, in all the studied multi-omics models. Conclusions Among all the studied models, the Radiomic model was found to play a dominant role for ART eligibility in NPC patients, and Radiomic features accounted for the largest proportion of features in all the multi-omics models.
Collapse
Affiliation(s)
- Sai-Kit Lam
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Yuanpeng Zhang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Jiang Zhang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Bing Li
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Jia-Chen Sun
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Carol Yee-Tung Liu
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Pak-Hei Chou
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Xinzhi Teng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Zong-Rui Ma
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Rui-Yan Ni
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Ta Zhou
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Tao Peng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Hao-Nan Xiao
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Tian Li
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Ge Ren
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Andy Lai-Yin Cheung
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China.,Department of Clinical Oncology, Queen Mary Hospital, Hong Kong, Hong Kong SAR, China
| | - Francis Kar-Ho Lee
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong, Hong Kong SAR, China
| | - Celia Wai-Yi Yip
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong, Hong Kong SAR, China
| | - Kwok-Hung Au
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong, Hong Kong SAR, China
| | - Victor Ho-Fun Lee
- Department of Clinical Oncology, The University of Hong Kong5Comprehensive Oncology Centre, Hong Kong Sanatorium & Hospital, Hong Kong, Hong Kong SAR, China
| | - Amy Tien-Yee Chang
- Comprehensive Oncology Centre, Hong Kong Sanatorium & Hospital, Hong Kong, Hong Kong SAR, China
| | - Lawrence Wing-Chi Chan
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Jing Cai
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
23
|
Lam SK, Zhang J, Zhang YP, Li B, Ni RY, Zhou T, Peng T, Cheung ALY, Chau TC, Lee FKH, Yip CWY, Au KH, Lee VHF, Chang ATY, Chan LWC, Cai J. A Multi-Center Study of CT-Based Neck Nodal Radiomics for Predicting an Adaptive Radiotherapy Trigger of Ill-Fitted Thermoplastic Masks in Patients with Nasopharyngeal Carcinoma. Life (Basel) 2022; 12:life12020241. [PMID: 35207528 PMCID: PMC8876942 DOI: 10.3390/life12020241] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/02/2021] [Accepted: 12/11/2021] [Indexed: 11/30/2022] Open
Abstract
Significant lymph node shrinkage is common in patients with nasopharyngeal carcinoma (NPC) throughout radiotherapy (RT) treatment, causing ill-fitted thermoplastic masks (IfTMs). To deal with this, an ad hoc adaptive radiotherapy (ART) may be required to ensure accurate and safe radiation delivery and to maintain treatment efficacy. Presently, the entire procedure for evaluating an eligible ART candidate is time-consuming, resource-demanding, and highly inefficient. In the artificial intelligence paradigm, the pre-treatment identification of NPC patients at risk for IfTMs has become greatly demanding for achieving efficient ART eligibility screening, while no relevant studies have been reported. Hence, we aimed to investigate the capability of computed tomography (CT)-based neck nodal radiomics for predicting IfTM-triggered ART events in NPC patients via a multi-center setting. Contrast-enhanced CT and the clinical data of 124 and 58 NPC patients from Queen Elizabeth Hospital (QEH) and Queen Mary Hospital (QMH), respectively, were retrospectively analyzed. Radiomic (R), clinical (C), and combined (RC) models were developed using the ridge algorithm in the QEH cohort and evaluated in the QMH cohort using the median area under the receiver operating characteristics curve (AUC). Delong’s test was employed for model comparison. Model performance was further assessed on 1000 replicates in both cohorts separately via bootstrapping. The R model yielded the highest “corrected” AUC of 0.784 (BCa 95%CI: 0.673–0.859) and 0.723 (BCa 95%CI: 0.534–0.859) in the QEH and QMH cohort following bootstrapping, respectively. Delong’s test indicated that the R model performed significantly better than the C model in the QMH cohort (p < 0.0001), while demonstrating no significant difference compared to the RC model (p = 0.5773). To conclude, CT-based neck nodal radiomics was capable of predicting IfTM-triggered ART events in NPC patients in this multi-center study, outperforming the traditional clinical model. The findings of this study provide valuable insights for future study into developing an effective screening strategy for ART eligibility in NPC patients in the long run, ultimately alleviating the workload of clinical practitioners, streamlining ART procedural efficiency in clinics, and achieving personalized RT for NPC patients in the future.
Collapse
Affiliation(s)
- Sai-Kit Lam
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; (S.-K.L.); (J.Z.); (Y.-P.Z.); (B.L.); (R.-Y.N.); (T.Z.); (T.P.); (A.L.-Y.C.); (L.W.-C.C.)
| | - Jiang Zhang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; (S.-K.L.); (J.Z.); (Y.-P.Z.); (B.L.); (R.-Y.N.); (T.Z.); (T.P.); (A.L.-Y.C.); (L.W.-C.C.)
| | - Yuan-Peng Zhang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; (S.-K.L.); (J.Z.); (Y.-P.Z.); (B.L.); (R.-Y.N.); (T.Z.); (T.P.); (A.L.-Y.C.); (L.W.-C.C.)
| | - Bing Li
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; (S.-K.L.); (J.Z.); (Y.-P.Z.); (B.L.); (R.-Y.N.); (T.Z.); (T.P.); (A.L.-Y.C.); (L.W.-C.C.)
| | - Rui-Yan Ni
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; (S.-K.L.); (J.Z.); (Y.-P.Z.); (B.L.); (R.-Y.N.); (T.Z.); (T.P.); (A.L.-Y.C.); (L.W.-C.C.)
| | - Ta Zhou
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; (S.-K.L.); (J.Z.); (Y.-P.Z.); (B.L.); (R.-Y.N.); (T.Z.); (T.P.); (A.L.-Y.C.); (L.W.-C.C.)
| | - Tao Peng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; (S.-K.L.); (J.Z.); (Y.-P.Z.); (B.L.); (R.-Y.N.); (T.Z.); (T.P.); (A.L.-Y.C.); (L.W.-C.C.)
| | - Andy Lai-Yin Cheung
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; (S.-K.L.); (J.Z.); (Y.-P.Z.); (B.L.); (R.-Y.N.); (T.Z.); (T.P.); (A.L.-Y.C.); (L.W.-C.C.)
- Department of Clinical Oncology, Queen Mary Hospital, Hong Kong, China
| | - Tin-Ching Chau
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China; (T.-C.C.); (V.H.-F.L.)
| | - Francis Kar-Ho Lee
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong, China; (F.K.-H.L.); (C.W.-Y.Y.); (K.-H.A.)
| | - Celia Wai-Yi Yip
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong, China; (F.K.-H.L.); (C.W.-Y.Y.); (K.-H.A.)
| | - Kwok-Hung Au
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong, China; (F.K.-H.L.); (C.W.-Y.Y.); (K.-H.A.)
| | - Victor Ho-Fun Lee
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China; (T.-C.C.); (V.H.-F.L.)
| | - Amy Tien-Yee Chang
- Comprehensive Oncology Centre, Hong Kong Sanatorium & Hospital, Hong Kong, China;
| | - Lawrence Wing-Chi Chan
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; (S.-K.L.); (J.Z.); (Y.-P.Z.); (B.L.); (R.-Y.N.); (T.Z.); (T.P.); (A.L.-Y.C.); (L.W.-C.C.)
| | - Jing Cai
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; (S.-K.L.); (J.Z.); (Y.-P.Z.); (B.L.); (R.-Y.N.); (T.Z.); (T.P.); (A.L.-Y.C.); (L.W.-C.C.)
- Correspondence:
| |
Collapse
|
24
|
Abstract
OBJECTIVES A critical problem in radiomic studies is the high dimensionality of the datasets, which stems from small sample sizes and many generic features extracted from the volume of interest. Therefore, feature selection methods are used, which aim to remove redundant as well as irrelevant features. Because there are many feature selection algorithms, it is key to understand their performance in the context of radiomics. MATERIALS AND METHODS A total of 29 feature selection algorithms and 10 classifiers were evaluated on 10 publicly available radiomic datasets. Feature selection methods were compared for training times, for the stability of the selected features, and for ranking, which measures the pairwise similarity of the methods. In addition, the predictive performance of the algorithms was measured by utilizing the area under the receiver operating characteristic curve of the best-performing classifier. RESULTS Feature selections differed largely in training times as well as stability and similarity. No single method was able to outperform another one consistently in predictive performance. CONCLUSION Our results indicated that simpler methods are more stable than complex ones and do not perform worse in terms of area under the receiver operating characteristic curve. Analysis of variance, least absolute shrinkage and selection operator, and minimum redundancy, maximum relevance ensemble appear to be good choices for radiomic studies in terms of predictive performance, as they outperformed most other feature selection methods.
Collapse
|
25
|
Piperdi H, Portal D, Neibart SS, Yue NJ, Jabbour SK, Reyhan M. Adaptive Radiation Therapy in the Treatment of Lung Cancer: An Overview of the Current State of the Field. Front Oncol 2021; 11:770382. [PMID: 34912715 PMCID: PMC8666420 DOI: 10.3389/fonc.2021.770382] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 11/09/2021] [Indexed: 12/25/2022] Open
Abstract
Lung cancer treatment is constantly evolving due to technological advances in the delivery of radiation therapy. Adaptive radiation therapy (ART) allows for modification of a treatment plan with the goal of improving the dose distribution to the patient due to anatomic or physiologic deviations from the initial simulation. The implementation of ART for lung cancer is widely varied with limited consensus on who to adapt, when to adapt, how to adapt, and what the actual benefits of adaptation are. ART for lung cancer presents significant challenges due to the nature of the moving target, tumor shrinkage, and complex dose accumulation because of plan adaptation. This article presents an overview of the current state of the field in ART for lung cancer, specifically, probing topics of: patient selection for the greatest benefit from adaptation, models which predict who and when to adapt plans, best timing for plan adaptation, optimized workflows for implementing ART including alternatives to re-simulation, the best radiation techniques for ART including magnetic resonance guided treatment, algorithms and quality assurance, and challenges and techniques for dose reconstruction. To date, the clinical workflow burden of ART is one of the major reasons limiting its widespread acceptance. However, the growing body of evidence demonstrates overwhelming support for reduced toxicity while improving tumor dose coverage by adapting plans mid-treatment, but this is offset by the limited knowledge about tumor control. Progress made in predictive modeling of on-treatment tumor shrinkage and toxicity, optimizing the timing of adaptation of the plan during the course of treatment, creating optimal workflows to minimize staffing burden, and utilizing deformable image registration represent ways the field is moving toward a more uniform implementation of ART.
Collapse
Affiliation(s)
- Huzaifa Piperdi
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| | - Daniella Portal
- Rutgers Robert Wood Johnson Medical School, Rutgers, The State of New Jersey University, Piscataway, NJ, United States
| | - Shane S. Neibart
- Rutgers Robert Wood Johnson Medical School, Rutgers, The State of New Jersey University, Piscataway, NJ, United States
| | - Ning J. Yue
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| | - Salma K. Jabbour
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
- Rutgers Robert Wood Johnson Medical School, Rutgers, The State of New Jersey University, Piscataway, NJ, United States
| | - Meral Reyhan
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| |
Collapse
|
26
|
Demircioğlu A. Measuring the bias of incorrect application of feature selection when using cross-validation in radiomics. Insights Imaging 2021; 12:172. [PMID: 34817740 PMCID: PMC8613324 DOI: 10.1186/s13244-021-01115-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 10/25/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Many studies in radiomics are using feature selection methods to identify the most predictive features. At the same time, they employ cross-validation to estimate the performance of the developed models. However, if the feature selection is performed before the cross-validation, data leakage can occur, and the results can be biased. To measure the extent of this bias, we collected ten publicly available radiomics datasets and conducted two experiments. First, the models were developed by incorrectly applying the feature selection prior to cross-validation. Then, the same experiment was conducted by applying feature selection correctly within cross-validation to each fold. The resulting models were then evaluated against each other in terms of AUC-ROC, AUC-F1, and Accuracy. RESULTS Applying the feature selection incorrectly prior to the cross-validation showed a bias of up to 0.15 in AUC-ROC, 0.29 in AUC-F1, and 0.17 in Accuracy. CONCLUSIONS Incorrect application of feature selection and cross-validation can lead to highly biased results for radiomic datasets.
Collapse
Affiliation(s)
- Aydin Demircioğlu
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany.
| |
Collapse
|
27
|
Radiomics for Predicting Lung Cancer Outcomes Following Radiotherapy: A Systematic Review. Clin Oncol (R Coll Radiol) 2021; 34:e107-e122. [PMID: 34763965 DOI: 10.1016/j.clon.2021.10.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/24/2021] [Accepted: 10/14/2021] [Indexed: 12/13/2022]
Abstract
Lung cancer's radiomic phenotype may potentially inform clinical decision-making with respect to radical radiotherapy. At present there are no validated biomarkers available for the individualisation of radical radiotherapy in lung cancer and the mortality rate of this disease remains the highest of all other solid tumours. MEDLINE was searched using the terms 'radiomics' and 'lung cancer' according to the Preferred Reporting Items for Systematic Reviews and Met-Analyses (PRISMA) guidance. Radiomics studies were defined as those manuscripts describing the extraction and analysis of at least 10 quantifiable imaging features. Only those studies assessing disease control, survival or toxicity outcomes for patients with lung cancer following radical radiotherapy ± chemotherapy were included. Study titles and abstracts were reviewed by two independent reviewers. The Radiomics Quality Score was applied to the full text of included papers. Of 244 returned results, 44 studies met the eligibility criteria for inclusion. End points frequently reported were local (17%), regional (17%) and distant control (31%), overall survival (79%) and pulmonary toxicity (4%). Imaging features strongly associated with clinical outcomes include texture features belonging to the subclasses Gray level run length matrix, Gray level co-occurrence matrix and kurtosis. The median cohort size for model development was 100 (15-645); in the 11 studies with external validation in a separate independent population, the median cohort size was 84 (21-295). The median number of imaging features extracted was 184 (10-6538). The median Radiomics Quality Score was 11% (0-47). Patient-reported outcomes were not incorporated within any studies identified. No studies externally validated a radiomics signature in a registered prospective study. Imaging-derived indices attained through radiomic analyses could equip thoracic oncologists with biomarkers for treatment response, patterns of failure, normal tissue toxicity and survival in lung cancer. Based on routine scans, their non-invasive nature and cost-effectiveness are major advantages over conventional pathological assessment. Improved tools are required for the appraisal of radiomics studies, as significant barriers to clinical implementation remain, such as standardisation of input scan data, quality of reporting and external validation of signatures in randomised, interventional clinical trials.
Collapse
|
28
|
CT-based radiomics signatures can predict the tumor response of non-small cell lung cancer patients treated with first-line chemotherapy and targeted therapy. Eur Radiol 2021; 32:1538-1547. [PMID: 34564744 DOI: 10.1007/s00330-021-08277-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 07/20/2021] [Accepted: 08/08/2021] [Indexed: 12/29/2022]
Abstract
OBJECTIVES The goal of this study was to evaluate the effectiveness of radiomics signatures on pre-treatment computed tomography (CT) images of lungs to predict the tumor responses of non-small cell lung cancer (NSCLC) patients treated with first-line chemotherapy, targeted therapy, or a combination of both. MATERIALS AND METHODS This retrospective study included 322 NSCLC patients who were treated with first-line chemotherapy, targeted therapy, or a combination of both. Of these patients, 224 were randomly assigned to a cohort to help develop the radiomics signature. A total of 1946 radiomics features were obtained from each patient's CT scan. The top-ranked features were selected by the Minimum Redundancy Maximum Relevance (MRMR) feature-ranking method and used to build a lightweight radiomics signature with the Random Forest (RF) classifier. The independent predictive (IP) features (AUC > 0.6, p value < 0.05) were further identified from the top-ranked features and used to build a refined radiomics signature by the RF classifier. Its prediction performance was tested on the validation cohort, which consisted of the remaining 98 patients. RESULTS The initial lightweight radiomics signature constructed from 15 top-ranked features had an AUC of 0.721 (95% CI, 0.619-0.823). After six IP features were further identified and a refined radiomics signature was built, it had an AUC of 0.746 (95% CI, 0.646-0.846). CONCLUSIONS Radiomics signatures based on pre-treatment CT scans can accurately predict tumor response in NSCLC patients after first-line chemotherapy or targeted therapy treatments. Radiomics features could be used as promising prognostic imaging biomarkers in the future. KEY POINTS The radiomics signature extracted from baseline CT images in patients with NSCLC can predict response to first-line chemotherapy, targeted therapy, or both treatments with an AUC = 0.746 (95% CI, 0.646-0.846). The radiomics signature could be used as a new biomarker for quantitative analysis in radiology, which might provide value in decision-making and to define personalized treatments for cancer patients.
Collapse
|
29
|
Tortora M, Cordelli E, Sicilia R, Miele M, Matteucci P, Iannello G, Ramella S, Soda P. Deep Reinforcement Learning for Fractionated Radiotherapy in Non-Small Cell Lung Carcinoma. Artif Intell Med 2021; 119:102137. [PMID: 34531006 DOI: 10.1016/j.artmed.2021.102137] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 05/28/2021] [Accepted: 08/03/2021] [Indexed: 12/24/2022]
Abstract
Lung cancer is by far the leading cause of cancer death among both men and women. Radiation therapy is one of the main approaches to lung cancer treatment, and its planning is crucial for the therapy outcome. However, the current practice that uniformly delivers the dose does not take into account the patient-specific tumour features that may affect treatment success. Since radiation therapy is by its very nature a sequential procedure, Deep Reinforcement Learning (DRL) is a well-suited methodology to overcome this limitation. In this respect, in this work we present a DRL controller optimizing the daily dose fraction delivered to the patient on the basis of CT scans collected over time during the therapy, offering a personalized treatment not only for volume adaptation, as currently intended, but also for daily fractionation. Furthermore, this contribution introduces a virtual radiotherapy environment based on a set of ordinary differential equations modelling the tissue radiosensitivity by combining both the effect of the radiotherapy treatment and cell growth. Their parameters are estimated from CT scans routinely collected using the Particle Swarm Optimization algorithm. This permits the DRL to learn the optimal behaviour through an iterative trial and error process with the environment. We performed several experiments considering three rewards functions modelling treatment strategies with different tissue aggressiveness and two exploration strategies for the exploration-exploitation dilemma. The results show that our DRL approach can adapt to radiation therapy treatment, optimizing its behaviour according to the different reward functions and outperforming the current clinical practice.
Collapse
Affiliation(s)
- Matteo Tortora
- Unit of Computer Systems & Bioinformatics, Department of Engineering, University Campus Bio-Medico of Rome, Via Alvaro del Portillo 21, 00128, Roma, Italy.
| | - Ermanno Cordelli
- Unit of Computer Systems & Bioinformatics, Department of Engineering, University Campus Bio-Medico of Rome, Via Alvaro del Portillo 21, 00128, Roma, Italy.
| | - Rosa Sicilia
- Unit of Computer Systems & Bioinformatics, Department of Engineering, University Campus Bio-Medico of Rome, Via Alvaro del Portillo 21, 00128, Roma, Italy.
| | - Marianna Miele
- Radiation Oncology, Department of Medicine, University Campus Bio-Medico of Rome, Via Alvaro del Portillo 21, 00128, Roma, Italy.
| | - Paolo Matteucci
- Radiation Oncology, Department of Medicine, University Campus Bio-Medico of Rome, Via Alvaro del Portillo 21, 00128, Roma, Italy.
| | - Giulio Iannello
- Unit of Computer Systems & Bioinformatics, Department of Engineering, University Campus Bio-Medico of Rome, Via Alvaro del Portillo 21, 00128, Roma, Italy.
| | - Sara Ramella
- Radiation Oncology, Department of Medicine, University Campus Bio-Medico of Rome, Via Alvaro del Portillo 21, 00128, Roma, Italy.
| | - Paolo Soda
- Unit of Computer Systems & Bioinformatics, Department of Engineering, University Campus Bio-Medico of Rome, Via Alvaro del Portillo 21, 00128, Roma, Italy.
| |
Collapse
|
30
|
Chetan MR, Gleeson FV. Radiomics in predicting treatment response in non-small-cell lung cancer: current status, challenges and future perspectives. Eur Radiol 2021; 31:1049-1058. [PMID: 32809167 PMCID: PMC7813733 DOI: 10.1007/s00330-020-07141-9] [Citation(s) in RCA: 153] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/03/2020] [Accepted: 08/03/2020] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Radiomics is the extraction of quantitative data from medical imaging, which has the potential to characterise tumour phenotype. The radiomics approach has the capacity to construct predictive models for treatment response, essential for the pursuit of personalised medicine. In this literature review, we summarise the current status and evaluate the scientific and reporting quality of radiomics research in the prediction of treatment response in non-small-cell lung cancer (NSCLC). METHODS A comprehensive literature search was conducted using the PubMed database. A total of 178 articles were screened for eligibility and 14 peer-reviewed articles were included. The radiomics quality score (RQS), a radiomics-specific quality metric emulating the TRIPOD guidelines, was used to assess scientific and reporting quality. RESULTS Included studies reported several predictive markers including first-, second- and high-order features, such as kurtosis, grey-level uniformity and wavelet HLL mean respectively, as well as PET-based metabolic parameters. Quality assessment demonstrated a low median score of + 2.5 (range - 5 to + 9), mainly reflecting a lack of reproducibility and clinical evaluation. There was extensive heterogeneity between studies due to differences in patient population, cancer stage, treatment modality, follow-up timescales and radiomics workflow methodology. CONCLUSIONS Radiomics research has not yet been translated into clinical use. Efforts towards standardisation and collaboration are needed to identify reproducible radiomic predictors of response. Promising radiomic models must be externally validated and their impact evaluated within the clinical pathway before they can be implemented as a clinical decision-making tool to facilitate personalised treatment for patients with NSCLC. KEY POINTS • The included studies reported several promising radiomic markers of treatment response in lung cancer; however, there was a lack of reproducibility between studies. • Quality assessment using the radiomics quality score (RQS) demonstrated a low median total score of + 2.5 (range - 5 to + 9). • Future radiomics research should focus on implementation of standardised radiomics features and software, together with external validation in a prospective setting.
Collapse
Affiliation(s)
- Madhurima R Chetan
- Department of Radiology, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Old Road, Headington, Oxford, OX3 7LE, UK.
- Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Room 6607, Level 6, Oxford, OX3 9DU, UK.
| | - Fergus V Gleeson
- Department of Radiology, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Old Road, Headington, Oxford, OX3 7LE, UK
- Department of Oncology, Old Road Campus Research Building, University of Oxford, Roosevelt Drive, Oxford, OX3 7DQ, UK
| |
Collapse
|
31
|
Hoegen P, Lang C, Akbaba S, Häring P, Splinter M, Miltner A, Bachmann M, Stahl-Arnsberger C, Brechter T, El Shafie RA, Weykamp F, König L, Debus J, Hörner-Rieber J. Cone-Beam-CT Guided Adaptive Radiotherapy for Locally Advanced Non-small Cell Lung Cancer Enables Quality Assurance and Superior Sparing of Healthy Lung. Front Oncol 2020; 10:564857. [PMID: 33363005 PMCID: PMC7756078 DOI: 10.3389/fonc.2020.564857] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 11/04/2020] [Indexed: 12/25/2022] Open
Abstract
Purpose To evaluate the potential of cone-beam-CT (CB-CT) guided adaptive radiotherapy (ART) for locally advanced non-small cell lung cancer (NSCLC) for sparing of surrounding organs-at-risk (OAR). Materials and Methods In 10 patients with locally advanced NSCLC, daily CB-CT imaging was acquired during radio- (n = 4) or radiochemotherapy (n = 6) for simulation of ART. Patients were treated with conventionally fractionated intensity-modulated radiotherapy (IMRT) with total doses of 60–66 Gy (pPlan) (311 fraction CB-CTs). OAR were segmented on every daily CB-CT and the tumor volumes were modified weekly depending on tumor changes. Doses actually delivered were recalculated on daily images (dPlan), and voxel-wise dose accumulation was performed using a deformable registration algorithm. For simulation of ART, treatment plans were adapted using the new contours and re-optimized weekly (aPlan). Results CB-CT showed continuous tumor regression of 1.1 ± 0.4% per day, leading to a residual gross tumor volume (GTV) of 65.3 ± 13.4% after 6 weeks of radiotherapy (p = 0.005). Corresponding PTVs decreased to 83.7 ± 7.8% (p = 0.005). In the actually delivered plans (dPlan), both conformity (p = 0.005) and homogeneity (p = 0.059) indices were impaired compared to the initial plans (pPlan). This resulted in higher actual lung doses than planned: V20Gy was 34.6 ± 6.8% instead of 32.8 ± 4.9% (p = 0.066), mean lung dose was 19.0 ± 3.1 Gy instead of 17.9 ± 2.5 Gy (p = 0.013). The generalized equivalent uniform dose (gEUD) of the lung was 18.9 ± 3.1 Gy instead of 17.8 ± 2.5 Gy (p = 0.013), leading to an increased lung normal tissue complication probability (NTCP) of 15.2 ± 13.9% instead of 9.6 ± 7.3% (p = 0.017). Weekly plan adaptation enabled decreased lung V20Gy of 31.6 ± 6.2% (−3.0%, p = 0.007), decreased mean lung dose of 17.7 ± 2.9 Gy (−1.3 Gy, p = 0.005), and decreased lung gEUD of 17.6 ± 2.9 Gy (−1.3 Gy, p = 0.005). Thus, resulting lung NTCP was reduced to 10.0 ± 9.5% (−5.2%, p = 0.005). Target volume coverage represented by conformity and homogeneity indices could be improved by weekly plan adaptation (CI: p = 0.007, HI: p = 0.114) and reached levels of the initial plan (CI: p = 0.721, HI: p = 0.333). Conclusion IGRT with CB-CT detects continuous GTV and PTV changes. CB-CT-guided ART for locally advanced NSCLC is feasible and enables superior sparing of healthy lung at high levels of plan conformity.
Collapse
Affiliation(s)
- Philipp Hoegen
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.,Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany.,National Center for Tumor Diseases (NCT), Heidelberg, Germany.,Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Clemens Lang
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany.,Medical Physics in Radiotherapy, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sati Akbaba
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.,Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany.,National Center for Tumor Diseases (NCT), Heidelberg, Germany.,Department of Radiation Oncology, Mainz University Hospital, Mainz, Germany
| | - Peter Häring
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany.,Medical Physics in Radiotherapy, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mona Splinter
- Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany.,Medical Physics in Radiotherapy, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Annette Miltner
- Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marion Bachmann
- Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Thomas Brechter
- Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rami A El Shafie
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.,Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany.,National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Fabian Weykamp
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.,Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany.,National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Laila König
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.,Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany.,National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Jürgen Debus
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.,Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany.,National Center for Tumor Diseases (NCT), Heidelberg, Germany.,Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Juliane Hörner-Rieber
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.,Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany.,National Center for Tumor Diseases (NCT), Heidelberg, Germany.,Clinical Cooperation Unit Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
32
|
Bortolotto C, Lancia A, Stelitano C, Montesano M, Merizzoli E, Agustoni F, Stella G, Preda L, Filippi AR. Radiomics features as predictive and prognostic biomarkers in NSCLC. Expert Rev Anticancer Ther 2020; 21:257-266. [PMID: 33216651 DOI: 10.1080/14737140.2021.1852935] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Introduction: Radiomics extracts a large amount of quantitative information from medical images using specific data characterization algorithms. This information, called radiomic features, can be combined with clinical data to build prediction models for prognostic evaluation and treatment selection.Areas covered: We outlined a series of studies investigating the correlation between radiomics features and outcome (prognostic) as well as response to therapy (predictive) in non-small cell lung cancer (NSCLC). We performed our analysis both in the setting of early and advanced stage of disease, with a focus on the different therapies and imaging modalities adopted.Expert opinion: The prognostic and predictive potential of the radiomic approach, combined with clinical models, could help decision-making process and guide toward the creation of an optimal and 'tailored' therapeutic strategy for lung cancer patients. However, due to the low reproducibility of most of the conducted studies and the lack of validated results, such a desirable scenario has not yet been translated to routine clinical practice.
Collapse
Affiliation(s)
| | - Andrea Lancia
- Radiation Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Chiara Stelitano
- Radiology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Marianna Montesano
- Radiation Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Elisa Merizzoli
- Radiation Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | | | - Giulia Stella
- Respiratory Disease Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Lorenzo Preda
- Radiology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | | |
Collapse
|
33
|
Radiomics-Based Prediction of Overall Survival in Lung Cancer Using Different Volumes-Of-Interest. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10186425] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Lung cancer accounts for the largest amount of deaths worldwide with respect to the other oncological pathologies. To guarantee the most effective cure to patients for such aggressive tumours, radiomics is increasing as a novel and promising research field that aims at extracting knowledge from data in terms of quantitative measures that are computed from diagnostic images, with prognostic and predictive ends. This knowledge could be used to optimize current treatments and to maximize their efficacy. To this end, we hereby study the use of such quantitative biomarkers computed from CT images of patients affected by Non-Small Cell Lung Cancer to predict Overall Survival. The main contributions of this work are two: first, we consider different volumes of interest for the same patient to find out whether the volume surrounding the visible lesions can provide useful information; second, we introduce 3D Local Binary Patterns, which are texture measures scarcely explored in radiomics. As further validation, we show that the proposed signature outperforms not only the features automatically computed by a deep learning-based approach, but also another signature at the state-of-the-art using other handcrafted features.
Collapse
|
34
|
de Dios NR, Murcia-Mejía M. Current and future strategies in radiotherapy for small-cell lung cancer. J Clin Transl Res 2020; 6:97-108. [PMID: 33521370 PMCID: PMC7837740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/12/2020] [Accepted: 07/17/2020] [Indexed: 11/24/2022] Open
Abstract
UNLABELLED Small-cell lung cancer (SCLC) accounts for 13% of all lung tumors. The standard treatment in patients with limited-stage (LS) disease is thoracic radiotherapy (TRT) combined with chemotherapy. In extensive-stage (ES) SCLC, the importance of consolidation TRT in patients with a good treatment response has become increasingly recognized. In both LS and ES disease, prophylactic cranial irradiation is recommended in patients who respond to treatment. New therapeutic approaches such as immunotherapy are being increasingly incorporated into the treatment of SCLC, although more slowly than in non-small cell lung cancer. Diverse radiation dose and fractionation schemes, administered in varying combinations with these new drugs, are being investigated. In the present article, we review and update the role of radiotherapy in the treatment of SCLC. We also discuss the main clinical trials currently underway to identify future trends. RELEVANCE FOR PATIENTS Radiotherapy is a critical component of multimodality treatment of SCLC. This article can help physicians to improve medical knowledge and find better ways to treat their SCLC patients.
Collapse
Affiliation(s)
- N. Rodríguez de Dios
- 1Department of Radiation Oncology, Hospital del Mar, Barcelona, Spain,2Hospital del Mar Medical Research Institute, Barcelona, Spain,3Pompeu Fabra University, Barcelona, Spain,
Corresponding author: Núria Rodríguez de Dios Department of Radiation Oncology, Hospital del Mar. Passeig Marítim, 25-29, 08003 Barcelona Tel.: 003493-367-4144
| | - M. Murcia-Mejía
- 4Department of Radiation Oncology, Hospital Sant Joan Reus, Tarragona
| |
Collapse
|
35
|
Yang WC, Hsu FM, Yang PC. Precision radiotherapy for non-small cell lung cancer. J Biomed Sci 2020; 27:82. [PMID: 32693792 PMCID: PMC7374898 DOI: 10.1186/s12929-020-00676-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/17/2020] [Indexed: 02/07/2023] Open
Abstract
Precision medicine is becoming the standard of care in anti-cancer treatment. The personalized precision management of cancer patients highly relies on the improvement of new technology in next generation sequencing and high-throughput big data processing for biological and radiographic information. Systemic precision cancer therapy has been developed for years. However, the role of precision medicine in radiotherapy has not yet been fully implemented. Emerging evidence has shown that precision radiotherapy for cancer patients is possible with recent advances in new radiotherapy technologies, panomics, radiomics and dosiomics. This review focused on the role of precision radiotherapy in non-small cell lung cancer and demonstrated the current landscape.
Collapse
Affiliation(s)
- Wen-Chi Yang
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital, No. 7, Chung-Shan South Rd, Taipei, Taiwan.,Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Feng-Ming Hsu
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital, No. 7, Chung-Shan South Rd, Taipei, Taiwan. .,Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Pan-Chyr Yang
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan. .,Department of Internal Medicine, National Taiwan University Hospital, No.1 Sec 1, Jen-Ai Rd, Taipei, 100, Taiwan.
| |
Collapse
|
36
|
Vaugier L, Ferrer L, Mengue L, Jouglar E. Radiomics for radiation oncologists: are we ready to go? BJR Open 2020; 2:20190046. [PMID: 33178967 PMCID: PMC7594896 DOI: 10.1259/bjro.20190046] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 03/06/2020] [Accepted: 03/09/2020] [Indexed: 12/19/2022] Open
Abstract
Radiomics have emerged as an exciting field of research over the past few years, with very wide potential applications in personalised and precision medicine of the future. Radiomics-based approaches are still however limited in daily clinical practice in oncology. This review focus on how radiomics could be incorporated into the radiation therapy pipeline, and globally help the radiation oncologist, from the tumour diagnosis to follow-up after treatment. Radiomics could impact on all steps of the treatment pipeline, once the limitations in terms of robustness and reproducibility are overcome. Major ongoing efforts should be made to collect and share data in the most standardised manner possible.
Collapse
Affiliation(s)
- Loïg Vaugier
- Department of Radiation Oncology, Institut de Cancérologie de l'Ouest, Nantes - Saint Herblain, France
| | - Ludovic Ferrer
- Department of Medical Physics, Institut de Cancérologie de l'Ouest, Nantes - Saint Herblain, France
| | - Laurence Mengue
- Department of Radiation Oncology, Institut de Cancérologie de l'Ouest, Nantes - Saint Herblain, France
| | - Emmanuel Jouglar
- Department of Radiation Oncology, Institut de Cancérologie de l'Ouest, Nantes - Saint Herblain, France
| |
Collapse
|
37
|
Gatta R, Depeursinge A, Ratib O, Michielin O, Leimgruber A. Integrating radiomics into holomics for personalised oncology: from algorithms to bedside. Eur Radiol Exp 2020; 4:11. [PMID: 32034573 PMCID: PMC7007467 DOI: 10.1186/s41747-019-0143-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 12/06/2019] [Indexed: 12/18/2022] Open
Abstract
Radiomics, artificial intelligence, and deep learning figure amongst recent buzzwords in current medical imaging research and technological development. Analysis of medical big data in assessment and follow-up of personalised treatments has also become a major research topic in the area of precision medicine. In this review, current research trends in radiomics are analysed, from handcrafted radiomics feature extraction and statistical analysis to deep learning. Radiomics algorithms now include genomics and immunomics data to improve patient stratification and prediction of treatment response. Several applications have already shown conclusive results demonstrating the potential of including other “omics” data to existing imaging features. We also discuss further challenges of data harmonisation and management infrastructure to shed a light on the much-needed integration of radiomics and all other “omics” into clinical workflows. In particular, we point to the emerging paradigm shift in the implementation of big data infrastructures to facilitate databanks growth, data extraction and the development of expert software tools. Secured access, sharing, and integration of all health data, called “holomics”, will accelerate the revolution of personalised medicine and oncology as well as expand the role of imaging specialists.
Collapse
Affiliation(s)
- Roberto Gatta
- Personalised Analytic Oncology, Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
| | - Adrien Depeursinge
- Personalised Analytic Oncology, Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland.,University of Applied Sciences and Arts Western Switzerland (HES-SO), Sierre, Switzerland
| | - Osman Ratib
- Service of Medical Imaging, Riviera-Chablais Hospital, Rennaz, Switzerland.,Department of Medical Imaging, Lausanne University Hospital, Lausanne, Switzerland
| | - Olivier Michielin
- Personalised Analytic Oncology, Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland
| | - Antoine Leimgruber
- Personalised Analytic Oncology, Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland. .,Service of Medical Imaging, Riviera-Chablais Hospital, Rennaz, Switzerland.
| |
Collapse
|
38
|
Sajja S, Lee Y, Eriksson M, Nordström H, Sahgal A, Hashemi M, Mainprize JG, Ruschin M. Technical Principles of Dual-Energy Cone Beam Computed Tomography and Clinical Applications for Radiation Therapy. Adv Radiat Oncol 2020; 5:1-16. [PMID: 32051885 PMCID: PMC7004939 DOI: 10.1016/j.adro.2019.07.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 05/21/2019] [Accepted: 07/20/2019] [Indexed: 02/06/2023] Open
Abstract
PURPOSE Medical imaging is an indispensable tool in radiotherapy for dose planning, image guidance and treatment monitoring. Cone beam CT (CBCT) is a low dose imaging technique with high spatial resolution capability as a direct by-product of using flat-panel detectors. However, certain issues such as x-ray scatter, beam hardening and other artifacts limit its utility to the verification of patient positioning using image-guided radiotherapy. METHODS AND MATERIALS Dual-energy (DE)-CBCT has recently demonstrated promise as an improved tool for tumor visualization in benchtop applications. It has the potential to improve soft-tissue contrast and reduce artifacts caused by beam hardening and metal. In this review, the practical aspects of developing a DE-CBCT based clinical and technical workflow are presented based on existing DE-CBCT literature and concepts adapted from the well-established library of work in DE-CT. Furthermore, the potential applications of DE-CBCT on its future role in radiotherapy are discussed. RESULTS AND CONCLUSIONS Based on current literature and an investigation of future applications, there is a clear potential for DE-CBCT technologies to be incorporated into radiotherapy. The applications of DE-CBCT include (but are not limited to): adaptive radiotherapy, brachytherapy, proton therapy, radiomics and theranostics.
Collapse
Affiliation(s)
- Shailaja Sajja
- Sunnybrook Research Institute, Toronto, Ontario, Canada
- QIPCM Imaging Core Lab, Techna Institute, Toronto, Ontario, Canada
| | - Young Lee
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | | | | | - Arjun Sahgal
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | | | | | - Mark Ruschin
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
39
|
A radiomic approach to predicting nodal relapse and disease-specific survival in patients treated with stereotactic body radiation therapy for early-stage non-small cell lung cancer. Strahlenther Onkol 2019; 196:922-931. [PMID: 31722061 DOI: 10.1007/s00066-019-01542-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 10/23/2019] [Indexed: 12/25/2022]
Abstract
PURPOSE To describe the possibility of building a classifier for patients at risk of lymph node relapse and a predictive model for disease-specific survival in patients with early stage non-small cell lung cancer. METHODS A cohort of 102 patients who received stereotactic body radiation treatment was retrospectively investigated. A set of 45 textural features was computed for the tumor volumes on the treatment planning CT images. Patients were split into two independent cohorts (70 patients, 68.9%, for training; and 32 patients, 31.4%, for validation). Three different models were built in the study. A stepwise backward linear discriminant analysis was applied to identify patients at risk of lymph node progression. The performance of the model was assessed by means of standard metrics derived from the confusion matrix. Furthermore, all textural features were correlated to survival data to build two separate predictive models for progression-free survival (PFS) and disease-specific survival (DS-OS). These models were built from the features/predictors found significant in univariate analysis and elastic net regularization by means of a multivarate Cox regression with backward selection. Low- and high-risk groups were identified by maximizing the separation by means of the Youden method. RESULTS In the total cohort (77, 75.5%, males; and 25, 24.5%, females; median age 76.6 years), 15 patients presented nodal progression at the time of analysis; 19 patients (18.6%) died because of disease-specific causes, 25 (24.5%) died from other reasons, 28 (27.5%) were alive without disease, and 30 (29.4%) with either local or distant progression. The specificity, sensitivity, and accuracy of the classifier resulted 83.1 ± 24.5, 87.4 ± 1.2, and 85.4 ± 12.5 in the validation group (coherent with the findings in the training). The area under the curve for the classifier resulted in 0.84 ± 0.04 and 0.73 ± 0.05 for training and validation, respectively. The mean time for DS-OS and PFS for the low- and high-risk subgroups of patients (in the validation groups) were 88.2 month ± 9.0 month vs. 84.1 month ± 7.8 month (low risk) and 52.7 month ± 5.9 month vs. 44.6 month ± 9.2 month (high risk), respectively. CONCLUSION Radiomics analysis based on planning CT images allowed a classifier and predictive models capable of identifying patients at risk of nodal relapse and high-risk of bad prognosis to be built. The radiomics signatures identified were mostly related to tumor heterogeneity.
Collapse
|
40
|
Stieb S, Kiser K, van Dijk L, Livingstone NR, Elhalawani H, Elgohari B, McDonald B, Ventura J, Mohamed ASR, Fuller CD. Imaging for Response Assessment in Radiation Oncology: Current and Emerging Techniques. Hematol Oncol Clin North Am 2019; 34:293-306. [PMID: 31739950 DOI: 10.1016/j.hoc.2019.09.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Imaging in radiation oncology is essential for the evaluation of treatment response in tumors and organs at risk. This influences further treatment decisions and could possibly be used to adapt therapy. This review article focuses on the currently used imaging modalities for response assessment in radiation oncology and gives an overview of new and promising techniques within this field.
Collapse
Affiliation(s)
- Sonja Stieb
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Kendall Kiser
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Lisanne van Dijk
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Nadia Roxanne Livingstone
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Hesham Elhalawani
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Baher Elgohari
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Brigid McDonald
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Juan Ventura
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Abdallah Sherif Radwan Mohamed
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Clifton David Fuller
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
| |
Collapse
|
41
|
Yu TT, Lam SK, To LH, Tse KY, Cheng NY, Fan YN, Lo CL, Or KW, Chan ML, Hui KC, Chan FC, Hui WM, Ngai LK, Lee FKH, Au KH, Yip CWY, Zhang Y, Cai J. Pretreatment Prediction of Adaptive Radiation Therapy Eligibility Using MRI-Based Radiomics for Advanced Nasopharyngeal Carcinoma Patients. Front Oncol 2019; 9:1050. [PMID: 31681588 PMCID: PMC6805774 DOI: 10.3389/fonc.2019.01050] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 09/26/2019] [Indexed: 01/19/2023] Open
Abstract
Background and purpose: Adaptive radiotherapy (ART) can compensate for the dosimetric impacts induced by anatomic and geometric variations in patients with nasopharyngeal carcinoma (NPC); Yet, the need for ART can only be assessed during the radiation treatment and the implementation of ART is resource intensive. Therefore, we aimed to determine tumoral biomarkers using pre-treatment MR images for predicting ART eligibility in NPC patients prior to the start of treatment. Methods: Seventy patients with biopsy-proven NPC (Stage II-IVB) in 2015 were enrolled into this retrospective study. Pre-treatment contrast-enhanced T1-w (CET1-w), T2-w MR images were processed and filtered using Laplacian of Gaussian (LoG) filter before radiomic features extraction. A total of 479 radiomics features, including the first-order (n = 90), shape (n = 14), and texture features (n = 375), were initially extracted from Gross-Tumor-Volume of primary tumor (GTVnp) using CET1-w, T2-w MR images. Patients were randomly divided into a training set (n = 51) and testing set (n = 19). The least absolute shrinkage and selection operator (LASSO) logistic regression model was applied for radiomic model construction in training set to select the most predictive features to predict patients who were replanned and assessed in the testing set. A double cross-validation approach of 100 resampled iterations with 3-fold nested cross-validation was employed in LASSO during model construction. The predictive performance of each model was evaluated using the area under the receiver operator characteristic (ROC) curve (AUC). Results: In the present cohort, 13 of 70 patients (18.6%) underwent ART. Average AUCs in training and testing sets were 0.962 (95%CI: 0.961-0.963) and 0.852 (95%CI: 0.847-0.857) with 8 selected features for CET1-w model; 0.895 (95%CI: 0.893-0.896) and 0.750 (95%CI: 0.745-0.755) with 6 selected features for T2-w model; and 0.984 (95%CI: 0.983-0.984) and 0.930 (95%CI: 0.928-0.933) with 6 selected features for joint T1-T2 model, respectively. In general, the joint T1-T2 model outperformed either CET1-w or T2-w model alone. Conclusions: Our study successfully showed promising capability of MRI-based radiomics features for pre-treatment identification of ART eligibility in NPC patients.
Collapse
Affiliation(s)
- Ting-Ting Yu
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Sai-Kit Lam
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Lok-Hang To
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Ka-Yan Tse
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Nong-Yi Cheng
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Yeuk-Nam Fan
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Cheuk-Lai Lo
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Ka-Wa Or
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Man-Lok Chan
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Ka-Ching Hui
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Fong-Chi Chan
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Wai-Ming Hui
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Lo-Kin Ngai
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Francis Kar-Ho Lee
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong, China
| | - Kwok-Hung Au
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong, China
| | - Celia Wai-Yi Yip
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong, China
| | - Yong Zhang
- Department of Physics, Xiamen University, Xiamen, China
| | - Jing Cai
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hung Hom, Hong Kong
| |
Collapse
|
42
|
D'Amico NC, Sicilia R, Cordelli E, Valbusa G, Grossi E, Zanetti IB, Fazzini D, Scotti G, Beltramo G, Iannello G, Soda P. Early radiomics experiences in predicting CyberKnife response in acoustic neuroma. ACM SIGBIOINFORMATICS RECORD 2019; 8:11-13. [DOI: 10.1145/3307616.3307620] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Vestibular schwannomas, also known as acoustic neuromas, are benign primary intracranial tumor of the myelin-forming cells of the 8th cranial nerve. Stereotactic radiosurgery is one of the available therapies that can effectively control tumor growth, and it can be performed using the CyberKnife robotic device. However, this therapy may have side effects and its efficacy should be assessed up to two years. In this respect, being able to forecast the treatment response using the data collected during the initial and routinely MR images could be a valuable support when planning a personalised therapy. This manuscript therefore introduces a machine learning-based radiomics approach that first computes quantitative biomarkers from MR images and then predicts the treatment response, taking also into consideration the dataset class skewness.
Collapse
Affiliation(s)
- Natascha Claudia D'Amico
- Imaging Department Centro Diagnostico Italiano S.p.A., Milan, Italy and Unit of Computer Systems and Bioinformatics Università Campus Bio-Medico di Roma, Rome, Italy and Università Campus Bio-Medico di Roma - Centro, Diagnostico Italiano S.p.A., Italy
| | - Rosa Sicilia
- Unit of Computer Systems and Bioinformatics Università Campus Bio-Medico di Roma, Rome, Italy and Università Campus Bio-Medico di Roma - Centro, Diagnostico Italiano S.p.A., Italy
| | - Ermanno Cordelli
- Unit of Computer Systems and Bioinformatics Università Campus Bio-Medico di Roma, Rome, Italy and Università Campus Bio-Medico di Roma - Centro, Diagnostico Italiano S.p.A., Italy
| | - Giovanni Valbusa
- Imaging Department Centro Diagnostico Italiano S.p.A., Milan, Italy and Università Campus Bio-Medico di Roma - Centro, Diagnostico Italiano S.p.A., Italy
| | | | - Isa Bossi Zanetti
- CyberKnife Department Centro Diagnostico Italiano S.p.A., Milan, Italy and Università Campus Bio-Medico di Roma - Centro, Diagnostico Italiano S.p.A., Italy
| | - Deborah Fazzini
- Imaging Department Centro Diagnostico Italiano S.p.A., Milan, Italy and Università Campus Bio-Medico di Roma - Centro, Diagnostico Italiano S.p.A., Italy
| | - Giuseppe Scotti
- Imaging Department Centro Diagnostico Italiano S.p.A., Milan, Italy and Università Campus Bio-Medico di Roma - Centro, Diagnostico Italiano S.p.A., Italy
| | | | - Giulio Iannello
- Unit of Computer Systems and Bioinformatics Università Campus Bio-Medico di Roma, Rome, Italy
| | - Paolo Soda
- Unit of Computer Systems and Bioinformatics Università Campus Bio-Medico di Roma, Rome, Italy and Università Campus Bio-Medico di Roma - Centro, Diagnostico Italiano S.p.A., Italy
| |
Collapse
|