1
|
Öztürk EMA, Ünsal G, Erişir F, Orhan K. Prediction of bone invasion of oral squamous cell carcinoma using a magnetic resonance imaging-based machine learning model. Eur Arch Otorhinolaryngol 2024; 281:6585-6597. [PMID: 39083062 DOI: 10.1007/s00405-024-08862-z] [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: 06/09/2024] [Accepted: 07/20/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVES Radiomics, a recently developed image-processing technology, holds potential in medical diagnostics. This study aimed to propose a machine-learning (ML) model and evaluate its effectiveness in detecting oral squamous cell carcinoma (OSCC) and predicting bone metastasis using magnetic resonance imaging (MRI). MATERIALS-METHODS MRI radiomic features were extracted and analyzed to identify malignant lesions. A total of 86 patients (44 with benign lesions without bone invasion and 42 with malignant lesions with bone invasion) were included. Data and clinical information were managed using the RadCloud Platform (Huiying Medical Technology Co., Ltd., Beijing, China). The study employed a hand-crafted radiomics model, with the dataset randomly split into training and validation sets in an 8:2 ratio using 815 random seeds. RESULTS The results revealed that the ML method support vector machine (SVM) performed best for detecting bone invasion (AUC = 0.999) in the test set. Radiomics tumor features derived from MRI are useful to predicting bone invasion from oral squamous cell carcinoma with high accuracy. CONCLUSIONS This study introduces an ML model utilizing SVM and radiomics to predict bone invasion in OSCC. Despite the promising results, the small sample size necessitates larger multicenter studies to validate and expand these findings.
Collapse
Affiliation(s)
- Elif Meltem Aslan Öztürk
- Department of Dentomaxillofacial Radiology, Faculty of Dentistry, Lokman Hekim University, Ankara, Turkey.
| | - Gürkan Ünsal
- Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Ferhat Erişir
- Department of Otorhinolaryngology and Head and Neck Surgery, Faculty of Medicine, Near East University, Kyrenia, Cyprus
| | - Kaan Orhan
- Department of Dentomaxillofacial Radiology, Faculty of Dentistry, Ankara University, Ankara, Turkey
| |
Collapse
|
2
|
Liu Y, Wang Z, Yang L, Zhang M, Li M, Zhang J, Tang L, Jiang Z, Li X, Deng J, Meng Q, Liu S, Wang K, Qi L. Identification of a rank-based radiomic signature with individualized prognostic value for lung adenocarcinoma in a multi-cohort study. Eur J Radiol 2024; 181:111782. [PMID: 39427495 DOI: 10.1016/j.ejrad.2024.111782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/29/2024] [Accepted: 10/08/2024] [Indexed: 10/22/2024]
Abstract
OBJECTIVES Radiomics provides an opportunity to evaluate cancer prognosis noninvasively. However, the susceptibility of the radiomic quantitative features to multicenter effects, leads to the clinical dilemma of the radiomic signatures. This study aimed to develop a radiomic signature to circumvent multicenter effects, achieving the individualized prognostic assessment of lung adenocarcinoma (LUAD). METHODS Using computed tomography (CT) imaging of 234 stage I-IIIA LUAD patients derived from three public multicenter cohorts, we proposed a rank-based method that utilized the relative rank patterns of quantitative values between radiomic feature pairs within individual patients and established a feature pair signature for LUAD prognosis. We collected a new clinical cohort with 162 LUAD patients for independent validation. RESULTS A rank-based radiomic signature, consisting of 12 feature pairs, was developed, and it could determine the mortality risk for an individual according to the rank patterns of 12 feature pairs within the patient's CT imaging. The prognostic performance of the rank-based signature was effectively validated in the new clinical cohort (log-rank P = 0.0051, C-index = 0.73), whereas other signatures lost their prognostic ability across centers. The novel proposed radiomic nomogram significantly improved the prognostic performance of clinicopathological factors. The further radiogenomic analyses revealed the underlying biological characteristics (e.g., Stemness, Ferroptosis, 'ECM') reflected by the rank-based radiomic signature. CONCLUSIONS This multicenter study illustrates the accuracy and stability of the rank-based radiomic signature for LUAD prognosis, and demonstrates a unique advantage of clinical individualized application. The biological characteristics underlying the rank-based radiomic signature would accelerate its clinical application.
Collapse
Affiliation(s)
- Yixin Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China; Modern Education Technology Center, Harbin Medical University, Harbin, China
| | - Zhihui Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Liping Yang
- PET-CT/MR Department, Harbin Medical University Cancer Hospital, Harbin, China
| | - Meng Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Mengyue Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Juxuan Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Lefan Tang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Zhiyun Jiang
- PET-CT/MR Department, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xin Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Jiaxing Deng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Qingwei Meng
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin 150040, China
| | - Shilong Liu
- Department of Thoracic Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin 150086, China.
| | - Kezheng Wang
- PET-CT/MR Department, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Lishuang Qi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China.
| |
Collapse
|
3
|
Guo T, Cheng B, Li Y, Li Y, Chen S, Lian G, Li J, Gao M, Huang K, Huang Y. A radiomics model for predicting perineural invasion in stage II-III colon cancer based on computer tomography. BMC Cancer 2024; 24:1226. [PMID: 39367321 PMCID: PMC11453003 DOI: 10.1186/s12885-024-12951-x] [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: 06/19/2024] [Accepted: 09/13/2024] [Indexed: 10/06/2024] Open
Abstract
BACKGROUND Colon cancer, a frequently encountered malignancy, exhibits a comparatively poor survival prognosis. Perineural invasion (PNI), highly correlated with tumor progression and metastasis, is a substantial effective predictor of stage II-III colon cancer. Nonetheless, the lack of effective and facile predictive methodologies for detecting PNI prior operation in colon cancer remains a persistent challenge. METHOD Pre-operative computer tomography (CT) images and clinical data of patients diagnosed with stage II-III colon cancer between January 2015 and December 2023 were obtained from two sub-districts of Sun Yat-sen Memorial Hospital (SYSUMH). The LASSO/RF/PCA filters were used to screen radiomics features and LR/SVM models were utilized to construct radiomics model. A comprehensive model, shown as nomogram finally, combining with radiomics score and significant clinical features were developed and validated by area under the curve (AUC) and decision curve analysis (DCA). RESULT The total cohort, comprising 426 individuals, was randomly divided into a development cohort and a validation cohort as a 7:3 ratio. Radiomics scores were extracted from LASSO-SVM models with AUC of 0.898/0.726 in the development and validation cohorts, respectively. Significant clinical features (CA199, CA125, T-stage, and N-stage) were used to establish combining model with radiomics scores. The combined model exhibited superior reliability compared to single radiomics model in AUC value (0.792 vs. 0.726, p = 0.003) in validation cohorts. The radiomics-clinical model demonstrated an AUC of 0.918/0.792, a sensitivity of 0.907/0.813 and a specificity of 0.804/0.716 in the development and validation cohorts, respectively. CONCLUSION The study developed and validated a predictive nomogram model combining radiomics scores and clinical features, and showed good performance in predicting PNI pre-operation in stage II-III colon cancer patients.
Collapse
Affiliation(s)
- Tairan Guo
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
| | - Bing Cheng
- Department of Pathology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
| | - Yunlong Li
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
| | - Yaqing Li
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
| | - Shaojie Chen
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
| | - Guoda Lian
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
| | - Jiajia Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
- Department of Nephrology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Ming Gao
- Department of Radiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China.
| | - Kaihong Huang
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China.
| | - Yuzhou Huang
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China.
| |
Collapse
|
4
|
Bourdillon AT. Computer Vision-Radiomics & Pathognomics. Otolaryngol Clin North Am 2024; 57:719-751. [PMID: 38910065 DOI: 10.1016/j.otc.2024.05.003] [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] [Indexed: 06/25/2024]
Abstract
The role of computer vision in extracting radiographic (radiomics) and histopathologic (pathognomics) features is an extension of molecular biomarkers that have been foundational to our understanding across the spectrum of head and neck disorders. Especially within head and neck cancers, machine learning and deep learning applications have yielded advances in the characterization of tumor features, nodal features, and various outcomes. This review aims to overview the landscape of radiomic and pathognomic applications, informing future work to address gaps. Novel methodologies will be needed to potentially engineer ways of integrating multidimensional data inputs to examine disease features to guide prognosis comprehensively and ultimately clinical management.
Collapse
Affiliation(s)
- Alexandra T Bourdillon
- Department of Otolaryngology-Head & Neck Surgery, University of California-San Francisco, San Francisco, CA 94115, USA.
| |
Collapse
|
5
|
Deasy JO. Data Science Opportunities To Improve Radiotherapy Planning and Clinical Decision Making. Semin Radiat Oncol 2024; 34:379-394. [PMID: 39271273 DOI: 10.1016/j.semradonc.2024.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
Radiotherapy aims to achieve a high tumor control probability while minimizing damage to normal tissues. Personalizing radiotherapy treatments for individual patients, therefore, depends on integrating physical treatment planning with predictive models of tumor control and normal tissue complications. Predictive models could be improved using a wide range of rich data sources, including tumor and normal tissue genomics, radiomics, and dosiomics. Deep learning will drive improvements in classifying normal tissue tolerance, predicting intra-treatment tumor changes, tracking accumulated dose distributions, and quantifying the tumor response to radiotherapy based on imaging. Mechanistic patient-specific computer simulations ('digital twins') could also be used to guide adaptive radiotherapy. Overall, we are entering an era where improved modeling methods will allow the use of newly available data sources to better guide radiotherapy treatments.
Collapse
Affiliation(s)
- Joseph O Deasy
- Department of Medical Physics, Attending Physicist, Chief, Service for Predictive Informatics, Chair, Memorial Sloan Kettering Cancer Center, New York, NY..
| |
Collapse
|
6
|
Choi YH, Kim JE, Lee RW, Kim B, Shin HC, Choe M, Kim Y, Park WY, Jin K, Han S, Paek JH, Kim K. Histopathological correlations of CT-based radiomics imaging biomarkers in native kidney biopsy. BMC Med Imaging 2024; 24:256. [PMID: 39333936 PMCID: PMC11428854 DOI: 10.1186/s12880-024-01434-x] [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: 04/05/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Kidney biopsy is the standard of care for the diagnosis of various kidney diseases. In particular, chronic histopathologic lesions, such as interstitial fibrosis and tubular atrophy, can provide prognostic information regarding chronic kidney disease progression. In this study, we aimed to evaluate historadiological correlations between CT-based radiomic features and chronic histologic changes in native kidney biopsies and to construct and validate a radiomics-based prediction model for chronicity grade. METHODS We included patients aged ≥ 18 years who underwent kidney biopsy and abdominal CT scan within a week before kidney biopsy. Left kidneys were three-dimensionally segmented using a deep learning model based on the 3D Swin UNEt Transformers architecture. We additionally defined isovolumic cortical regions of interest near the lower pole of the left kidneys. Shape, first-order, and high-order texture features were extracted after resampling and kernel normalization. Correlations and diagnostic metrics between extracted features and chronic histologic lesions were examined. A machine learning-based radiomic prediction model for moderate chronicity was developed and compared according to the segmented regions of interest (ROI). RESULTS Overall, moderate correlations with statistical significance (P < 0.05) were found between chronic histopathologic grade and top-ranked radiomic features. Total parenchymal features were more strongly correlated than cortical ROI features, and texture features were more highly ranked. However, conventional imaging markers, including kidney length, were poorly correlated. Top-ranked individual radiomic features had areas under receiver operating characteristic curves (AUCs) of 0.65 to 0.74. Developed radiomics models for moderate-to-severe chronicity achieved AUCs of 0.89 (95% confidence interval [CI] 0.75-0.99) and 0.74 (95% CI 0.52-0.93) for total parenchymal and cortical ROI features, respectively. CONCLUSION Significant historadiological correlations were identified between CT-based radiomic features and chronic histologic changes in native kidney biopsies. Our findings underscore the potential of CT-based radiomic features and their prediction model for the non-invasive assessment of kidney fibrosis.
Collapse
Affiliation(s)
- Yoon Ho Choi
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Jacksonville, FL, USA
| | - Ji-Eun Kim
- Division of Nephrology and Hypertension, Department of Internal Medicine, Inha University Hospital, Inha University College of Medicine, Incheon, Republic of Korea
| | - Ro Woon Lee
- Department of Radiology, Inha University College of Medicine, Incheon, Republic of Korea
| | - Byoungje Kim
- Department of Radiology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Hyeong Chan Shin
- Department of Pathology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Misun Choe
- Department of Pathology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Yaerim Kim
- Division of Nephrology, Department of Internal Medicine, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Woo Yeong Park
- Division of Nephrology, Department of Internal Medicine, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Kyubok Jin
- Division of Nephrology, Department of Internal Medicine, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Seungyeup Han
- Division of Nephrology, Department of Internal Medicine, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Jin Hyuk Paek
- Division of Nephrology, Department of Internal Medicine, Keimyung University School of Medicine, Daegu, Republic of Korea.
| | - Kipyo Kim
- Division of Nephrology and Hypertension, Department of Internal Medicine, Inha University Hospital, Inha University College of Medicine, Incheon, Republic of Korea.
| |
Collapse
|
7
|
Alapati R, Renslo B, Wagoner SF, Karadaghy O, Serpedin A, Kim YE, Feucht M, Wang N, Ramesh U, Bon Nieves A, Lawrence A, Virgen C, Sawaf T, Rameau A, Bur AM. Assessing the Reporting Quality of Machine Learning Algorithms in Head and Neck Oncology. Laryngoscope 2024. [PMID: 39258420 DOI: 10.1002/lary.31756] [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: 02/07/2024] [Revised: 07/25/2024] [Accepted: 08/23/2024] [Indexed: 09/12/2024]
Abstract
OBJECTIVE This study aimed to assess reporting quality of machine learning (ML) algorithms in the head and neck oncology literature using the TRIPOD-AI criteria. DATA SOURCES A comprehensive search was conducted using PubMed, Scopus, Embase, and Cochrane Database of Systematic Reviews, incorporating search terms related to "artificial intelligence," "machine learning," "deep learning," "neural network," and various head and neck neoplasms. REVIEW METHODS Two independent reviewers analyzed each published study for adherence to the 65-point TRIPOD-AI criteria. Items were classified as "Yes," "No," or "NA" for each publication. The proportion of studies satisfying each TRIPOD-AI criterion was calculated. Additionally, the evidence level for each study was evaluated independently by two reviewers using the Oxford Centre for Evidence-Based Medicine (OCEBM) Levels of Evidence. Discrepancies were reconciled through discussion until consensus was reached. RESULTS The study highlights the need for improvements in ML algorithm reporting in head and neck oncology. This includes more comprehensive descriptions of datasets, standardization of model performance reporting, and increased sharing of ML models, data, and code with the research community. Adoption of TRIPOD-AI is necessary for achieving standardized ML research reporting in head and neck oncology. CONCLUSION Current reporting of ML algorithms hinders clinical application, reproducibility, and understanding of the data used for model training. To overcome these limitations and improve patient and clinician trust, ML developers should provide open access to models, code, and source data, fostering iterative progress through community critique, thus enhancing model accuracy and mitigating biases. LEVEL OF EVIDENCE NA Laryngoscope, 2024.
Collapse
Affiliation(s)
- Rahul Alapati
- Department of Otolaryngology-Head & Neck Surgery, University of Kansas Medical Center, Kansas City, Kansas, U.S.A
| | - Bryan Renslo
- Department of Otolaryngology-Head & Neck Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, U.S.A
| | - Sarah F Wagoner
- Department of Otolaryngology-Head & Neck Surgery, University of Kansas Medical Center, Kansas City, Kansas, U.S.A
| | - Omar Karadaghy
- Department of Otolaryngology-Head & Neck Surgery, University of Kansas Medical Center, Kansas City, Kansas, U.S.A
| | - Aisha Serpedin
- Department of Otolaryngology-Head & Neck Surgery, Weill Cornell, New York City, New York, U.S.A
| | - Yeo Eun Kim
- Department of Otolaryngology-Head & Neck Surgery, Weill Cornell, New York City, New York, U.S.A
| | - Maria Feucht
- Department of Otolaryngology-Head & Neck Surgery, University of Kansas Medical Center, Kansas City, Kansas, U.S.A
| | - Naomi Wang
- Department of Otolaryngology-Head & Neck Surgery, University of Kansas Medical Center, Kansas City, Kansas, U.S.A
| | - Uma Ramesh
- Department of Otolaryngology-Head & Neck Surgery, University of Kansas Medical Center, Kansas City, Kansas, U.S.A
| | - Antonio Bon Nieves
- Department of Otolaryngology-Head & Neck Surgery, University of Kansas Medical Center, Kansas City, Kansas, U.S.A
| | - Amelia Lawrence
- Department of Otolaryngology-Head & Neck Surgery, University of Kansas Medical Center, Kansas City, Kansas, U.S.A
| | - Celina Virgen
- Department of Otolaryngology-Head & Neck Surgery, University of Kansas Medical Center, Kansas City, Kansas, U.S.A
| | - Tuleen Sawaf
- Department of Otolaryngology-Head & Neck Surgery, University of Maryland, Baltimore, Maryland, U.S.A
| | - Anaïs Rameau
- Department of Otolaryngology-Head & Neck Surgery, Weill Cornell, New York City, New York, U.S.A
| | - Andrés M Bur
- Department of Otolaryngology-Head & Neck Surgery, University of Kansas Medical Center, Kansas City, Kansas, U.S.A
| |
Collapse
|
8
|
Alabi RO, Elmusrati M, Leivo I, Almangush A, Mäkitie AA. Artificial Intelligence-Driven Radiomics in Head and Neck Cancer: Current Status and Future Prospects. Int J Med Inform 2024; 188:105464. [PMID: 38728812 DOI: 10.1016/j.ijmedinf.2024.105464] [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: 10/15/2023] [Revised: 04/20/2024] [Accepted: 04/22/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND Radiomics is a rapidly growing field used to leverage medical radiological images by extracting quantitative features. These are supposed to characterize a patient's phenotype, and when combined with artificial intelligence techniques, to improve the accuracy of diagnostic models and clinical outcome prediction. OBJECTIVES This review aims at examining the application areas of artificial intelligence-based radiomics (AI-based radiomics) for the management of head and neck cancer (HNC). It further explores the workflow of AI-based radiomics for personalized and precision oncology in HNC. Finally, it examines the current challenges of AI-based radiomics in daily clinical oncology and offers possible solutions to these challenges. METHODS Comprehensive electronic databases (PubMed, Medline via Ovid, Scopus, Web of Science, CINAHL, and Cochrane Library) were searched following the Preferred Reporting Items for Systematic Review and Meta-Analysis (PRISMA) guidelines. The quality of included studies and their risk of biases were evaluated using the Transparent Reporting of a Multivariable Prediction Model for Individual Prognosis or Diagnosis (TRIPOD)and Prediction Model Risk of Bias Assessment Tool (PROBAST). RESULTS Out of the 659 search hits retrieved, 45 fulfilled the inclusion criteria. Our review revealed that the application of AI-based radiomics model as an ancillary tool for improved decision-making in HNC management includes radiomics-based cancer diagnosis and radiomics-based cancer prognosis. The radiomics-based cancer diagnosis includes tumor staging, tumor grading, and classification of malignant and benign tumors. Similarly, radiomics-based cancer prognosis includes prediction for treatment response, recurrence, metastasis, and survival. In addition, the challenges in the implementation of these models for clinical evaluations include data imbalance, feature engineering (extraction and selection), model generalizability, multi-modal fusion, and model interpretability. CONCLUSION Considering the highly subjective and interobserver variability that is peculiar to the interpretation of medical images by expert clinicians, AI-based radiomics seeks to offer potentially useful quantitative information, which is not visible to the human eye or unintentionally often remain ignored during clinical imaging practice. By enabling the extraction of this type of information, AI-based radiomics has the potential to revolutionize HNC oncology, providing a platform for more personalized, higher quality, and cost-effective care for HNC patients.
Collapse
Affiliation(s)
- Rasheed Omobolaji Alabi
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Industrial Digitalization, School of Technology and Innovations, University of Vaasa, Vaasa, Finland.
| | - Mohammed Elmusrati
- Department of Industrial Digitalization, School of Technology and Innovations, University of Vaasa, Vaasa, Finland
| | - Ilmo Leivo
- University of Turku, Institute of Biomedicine, Pathology, Turku, Finland
| | - Alhadi Almangush
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland; University of Turku, Institute of Biomedicine, Pathology, Turku, Finland; Department of Pathology, University of Helsinki, Helsinki, Finland; Faculty of Dentistry, Misurata University, Misurata, Libya
| | - Antti A Mäkitie
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Otorhinolaryngology - Head and Neck Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Division of Ear, Nose and Throat Diseases, Department of Clinical Sciences, Intervention and Technology, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
9
|
Lam V, O'Brien O, Amin O, Nigar E, Kumar M, Lingam RK. Oral cavity cancer and its pre-treatment radiological evaluation: A pictorial overview. Eur J Radiol 2024; 176:111494. [PMID: 38776803 DOI: 10.1016/j.ejrad.2024.111494] [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: 04/12/2024] [Accepted: 05/02/2024] [Indexed: 05/25/2024]
Abstract
PURPOSE Oral cavity cancer, primarily squamous cell carcinoma (SCC), is a prevalent malignancy globally, necessitating accurate clinical assessment and staging to enable effective treatment planning. Diagnosis requires biopsy and is followed by surgical resection and reconstruction as the primary therapeutic modality. Imaging plays a pivotal role during this process, aiding in the evaluation of tumour extent, nodal involvement and distant metastases. However, despite its value, both radiologists and clinicians must recognise its inherent limitations. METHODS This pictorial review article aims to illustrate the application of various imaging modalities in the pre-treatment evaluation of oral cavity SCC and highlights potential pitfalls. It underscores the importance of understanding the anatomical subsites of the oral cavity, the diverse patterns of spread tumours exhibit at each site, alongside the role of imaging in facilitating informed management strategies, while also acknowledging its limitations. RESULTS The review delves into fundamentals of current staging including nodal involvement, while, emphasising imaging strategies and potential limitations. Finally, it touches on the potential of novel radiomic techniques in characterising tumours and predicting treatment response. CONCLUSIONS Pre-treatment oral cavity cancer staging reflects an ongoing quest for enhanced diagnostic accuracy and prognostic prediction. Recognising the value of imaging alongside its limitations fosters a multidisciplinary approach to treatment planning, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Vincent Lam
- Department of Radiology, Leicester Royal Infirmary, University Hospitals of Leicester NHS Trust, Infirmary Square, Leicester LE1 5WW, United Kingdom
| | - Owen O'Brien
- Department of Radiology, Northwick Park Hospital, London North West University Healthcare NHS Trust, Watford Road, London HA1 3UJ, United Kingdom
| | - Omed Amin
- Department of Radiology, Northwick Park Hospital, London North West University Healthcare NHS Trust, Watford Road, London HA1 3UJ, United Kingdom; Department of Radiology, Chelsea and Westminster NHS Foundation Trust, 369 Fulham Rd, London SW10 9NH, United Kingdom
| | - Ezra Nigar
- Department of Pathology, Northwick Park Hospital, London North West University Healthcare NHS Trust, Watford Road, London HA1 3UJ, United Kingdom
| | - Mahesh Kumar
- Department of Oral and Maxillofacial Surgery, Northwick Park Hospital, London North West University Healthcare NHS Trust, Watford Road, London HA1 3UJ, United Kingdom; Department of Oral and Maxillofacial Surgery, Hillingdon Hospital, The Hillingdon Hospitals NHS Foundation Trust, Pield Heath Rd, Uxbridge UB8 3NN, United Kingdom
| | - Ravi Kumar Lingam
- Department of Radiology, Northwick Park Hospital, London North West University Healthcare NHS Trust, Watford Road, London HA1 3UJ, United Kingdom.
| |
Collapse
|
10
|
Al Hasan MM, Ghazimoghadam S, Tunlayadechanont P, Mostafiz MT, Gupta M, Roy A, Peters K, Hochhegger B, Mancuso A, Asadizanjani N, Forghani R. Automated Segmentation of Lymph Nodes on Neck CT Scans Using Deep Learning. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024:10.1007/s10278-024-01114-w. [PMID: 38937342 DOI: 10.1007/s10278-024-01114-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 04/01/2024] [Accepted: 04/03/2024] [Indexed: 06/29/2024]
Abstract
Early and accurate detection of cervical lymph nodes is essential for the optimal management and staging of patients with head and neck malignancies. Pilot studies have demonstrated the potential for radiomic and artificial intelligence (AI) approaches in increasing diagnostic accuracy for the detection and classification of lymph nodes, but implementation of many of these approaches in real-world clinical settings would necessitate an automated lymph node segmentation pipeline as a first step. In this study, we aim to develop a non-invasive deep learning (DL) algorithm for detecting and automatically segmenting cervical lymph nodes in 25,119 CT slices from 221 normal neck contrast-enhanced CT scans from patients without head and neck cancer. We focused on the most challenging task of segmentation of small lymph nodes, evaluated multiple architectures, and employed U-Net and our adapted spatial context network to detect and segment small lymph nodes measuring 5-10 mm. The developed algorithm achieved a Dice score of 0.8084, indicating its effectiveness in detecting and segmenting cervical lymph nodes despite their small size. A segmentation framework successful in this task could represent an essential initial block for future algorithms aiming to evaluate small objects such as lymph nodes in different body parts, including small lymph nodes looking normal to the naked human eye but harboring early nodal metastases.
Collapse
Affiliation(s)
- Md Mahfuz Al Hasan
- Radiomics and Augmented Intelligence Laboratory (RAIL), Department of Radiology and the Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, 1600 SW Archer Road, Gainesville, FL, 32610-0374, USA
- Department of Electrical and Computer Engineering, University of Florida College of Medicine, Gainesville, FL, USA
| | - Saba Ghazimoghadam
- Augmented Intelligence and Precision Health Laboratory, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Padcha Tunlayadechanont
- Augmented Intelligence and Precision Health Laboratory, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Diagnostic and Therapeutic Radiology and Research, Faculty of Medicine Ramathibodi Hospital, Ratchathewi, Bangkok, Thailand
| | - Mohammed Tahsin Mostafiz
- Radiomics and Augmented Intelligence Laboratory (RAIL), Department of Radiology and the Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, 1600 SW Archer Road, Gainesville, FL, 32610-0374, USA
- Department of Electrical and Computer Engineering, University of Florida College of Medicine, Gainesville, FL, USA
| | - Manas Gupta
- Radiomics and Augmented Intelligence Laboratory (RAIL), Department of Radiology and the Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, 1600 SW Archer Road, Gainesville, FL, 32610-0374, USA
| | - Antika Roy
- Radiomics and Augmented Intelligence Laboratory (RAIL), Department of Radiology and the Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, 1600 SW Archer Road, Gainesville, FL, 32610-0374, USA
- Department of Electrical and Computer Engineering, University of Florida College of Medicine, Gainesville, FL, USA
| | - Keith Peters
- Radiomics and Augmented Intelligence Laboratory (RAIL), Department of Radiology and the Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, 1600 SW Archer Road, Gainesville, FL, 32610-0374, USA
- Department of Radiology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Bruno Hochhegger
- Radiomics and Augmented Intelligence Laboratory (RAIL), Department of Radiology and the Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, 1600 SW Archer Road, Gainesville, FL, 32610-0374, USA
- Department of Radiology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Anthony Mancuso
- Radiomics and Augmented Intelligence Laboratory (RAIL), Department of Radiology and the Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, 1600 SW Archer Road, Gainesville, FL, 32610-0374, USA
- Department of Radiology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Navid Asadizanjani
- Radiomics and Augmented Intelligence Laboratory (RAIL), Department of Radiology and the Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, 1600 SW Archer Road, Gainesville, FL, 32610-0374, USA
- Department of Electrical and Computer Engineering, University of Florida College of Medicine, Gainesville, FL, USA
| | - Reza Forghani
- Radiomics and Augmented Intelligence Laboratory (RAIL), Department of Radiology and the Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, 1600 SW Archer Road, Gainesville, FL, 32610-0374, USA.
- Department of Radiology, University of Florida College of Medicine, Gainesville, FL, USA.
- Division of Medical Physics, University of Florida College of Medicine, Gainesville, FL, USA.
- Department of Neurology, Division of Movement Disorders, University of Florida College of Medicine, Gainesville, FL, USA.
- Augmented Intelligence and Precision Health Laboratory, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
| |
Collapse
|
11
|
Jayawickrama SM, Ranaweera PM, Pradeep RGGR, Jayasinghe YA, Senevirathna K, Hilmi AJ, Rajapakse RMG, Kanmodi KK, Jayasinghe RD. Developments and future prospects of personalized medicine in head and neck squamous cell carcinoma diagnoses and treatments. Cancer Rep (Hoboken) 2024; 7:e2045. [PMID: 38522008 PMCID: PMC10961052 DOI: 10.1002/cnr2.2045] [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: 08/28/2023] [Revised: 02/07/2024] [Accepted: 03/05/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND Precision healthcare has entered a new era because of the developments in personalized medicine, especially in the diagnosis and treatment of head and neck squamous cell carcinoma (HNSCC). This paper explores the dynamic landscape of personalized medicine as applied to HNSCC, encompassing both current developments and future prospects. RECENT FINDINGS The integration of personalized medicine strategies into HNSCC diagnosis is driven by the utilization of genetic data and biomarkers. Epigenetic biomarkers, which reflect modifications to DNA that can influence gene expression, have emerged as valuable indicators for early detection and risk assessment. Treatment approaches within the personalized medicine framework are equally promising. Immunotherapy, gene silencing, and editing techniques, including RNA interference and CRISPR/Cas9, offer innovative means to modulate gene expression and correct genetic aberrations driving HNSCC. The integration of stem cell research with personalized medicine presents opportunities for tailored regenerative approaches. The synergy between personalized medicine and technological advancements is exemplified by artificial intelligence (AI) and machine learning (ML) applications. These tools empower clinicians to analyze vast datasets, predict patient responses, and optimize treatment strategies with unprecedented accuracy. CONCLUSION The developments and prospects of personalized medicine in HNSCC diagnosis and treatment offer a transformative approach to managing this complex malignancy. By harnessing genetic insights, biomarkers, immunotherapy, gene editing, stem cell therapies, and advanced technologies like AI and ML, personalized medicine holds the key to enhancing patient outcomes and ushering in a new era of precision oncology.
Collapse
Affiliation(s)
| | | | | | | | - Kalpani Senevirathna
- Centre for Research in Oral Cancer, Faculty of Dental SciencesUniversity of PeradeniyaKandySri Lanka
| | | | | | - Kehinde Kazeem Kanmodi
- School of DentistryUniversity of RwandaKigaliRwanda
- Faculty of DentistryUniversity of PuthisastraPhnom PenhCambodia
- Cephas Health Research Initiative IncIbadanNigeria
- School of Health and Life SciencesTeesside UniversityMiddlesbroughUK
| | - Ruwan Duminda Jayasinghe
- Centre for Research in Oral Cancer, Faculty of Dental SciencesUniversity of PeradeniyaKandySri Lanka
- Faculty of DentistryUniversity of PuthisastraPhnom PenhCambodia
- School of Health and Life SciencesTeesside UniversityMiddlesbroughUK
- Department of Oral Medicine and Periodontology, Faculty of Dental SciencesUniversity of PeradeniyaKandySri Lanka
| |
Collapse
|
12
|
Peiliang Wang MD, Yikun Li MM, Mengyu Zhao MM, Jinming Yu MD, Feifei Teng MD. Distinguishing immune checkpoint inhibitor-related pneumonitis from radiation pneumonitis by CT radiomics features in non-small cell lung cancer. Int Immunopharmacol 2024; 128:111489. [PMID: 38266450 DOI: 10.1016/j.intimp.2024.111489] [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: 10/22/2023] [Revised: 12/26/2023] [Accepted: 01/02/2024] [Indexed: 01/26/2024]
Abstract
PURPOSE To develop a CT-based model to classify pneumonitis etiology in patients with non-small cell lung cancer(NSCLC) after radiotherapy(RT) and Immune checkpoint inhibitors(ICIs). METHODS We retrospectively identified 130 NSCLC patients who developed pneumonitis after receipt of ICIs only (n = 50), thoracic RT only (n = 50) (ICIs only + thoracic RT only, the training cohort, n = 100), and RT + ICIs (the test cohort, n = 30). Clinical and CT radiomics features were described and compared between different groups. We constructed a random forest (RF) classifier and a linear discriminant analysis (LDA) classifier by CT radiomics to discern pneumonitis etiology. RESULTS The patients in RT + ICIs group have more high grade (grade 3-4) pneumonitis compared to patients in ICIs only or RT only group (p < 0.05). Pneumonitis after the combined therapy was not a simple superposition mode of RT-related pneumonitis(RP) and ICI-related pneumonitis(CIP), resulting in the distinct characteristics of both RT and ICIs-related pneumonitis. The RF classifier showed favorable discrimination between RP and CIP with an area under the receiver operating curve (AUC) of 0.859 (95 %CI: 0.788-0.929) in the training cohort and 0.851 (95 % CI: 0.700-1) in the test cohort. The LDA classifier achieved an AUC of 0.881 (95 %CI: 0.815-0.947) in the training cohort and 0.842 (95 %CI: 0.686-0.997) in the test cohort. Our analysis revealed four principal CT-based features shared across both models:original_glrlm_LongRunLowGrayLevelEmphasis, wavelet-HLL_firstorder_Median, wavelet-LLL_ngtdm_Busyness, and wavelet-LLL_glcm_JointAverage. CONCLUSION CT radiomics-based classifiers could provide a noninvasive method to identify the predominant etiology in NSCLC patients who developed pneumonitis after RT alone, ICIs alone or RT + ICIs.
Collapse
Affiliation(s)
- M D Peiliang Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Cheeloo College of Medicine, Shandong University, Jinan 250117, China
| | - M M Yikun Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - M M Mengyu Zhao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - M D Jinming Yu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Cheeloo College of Medicine, Shandong University, Jinan 250117, China
| | - M D Feifei Teng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Cheeloo College of Medicine, Shandong University, Jinan 250117, China.
| |
Collapse
|
13
|
Lin CH, Yan JL, Yap WK, Kang CJ, Chang YC, Tsai TY, Chang KP, Liao CT, Hsu CL, Chou WC, Wang HM, Huang PW, Fan KH, Huang BS, Tung-Chieh Chang J, Tu SJ, Lin CY. Prognostic value of interim CT-based peritumoral and intratumoral radiomics in laryngeal and hypopharyngeal cancer patients undergoing definitive radiotherapy. Radiother Oncol 2023; 189:109938. [PMID: 37806562 DOI: 10.1016/j.radonc.2023.109938] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 09/20/2023] [Accepted: 10/02/2023] [Indexed: 10/10/2023]
Abstract
BACKGROUND AND PURPOSE We aimed to investigate the prognostic value of peritumoral and intratumoral computed tomography (CT)-based radiomics during the course of radiotherapy (RT) in patients with laryngeal and hypopharyngeal cancer (LHC). MATERIALS AND METHODS A total of 92 eligible patients were 1:1 randomly assigned into training and validation cohorts. Pre-RT and mid-RT radiomic features were extracted from pre-treatment and interim CT. LASSO-Cox regression was used for feature selection and model construction. Time-dependent area under the receiver operating curve (AUC) analysis was applied to evaluate the models' prognostic performances. Risk stratification ability on overall survival (OS) and progression-free survival (PFS) were assessed using the Kaplan-Meier method and Cox regression. The associations between radiomics and clinical parameters as well as circulating lymphocyte counts were also evaluated. RESULTS The mid-RT peritumoral (AUC: 0.77) and intratumoral (AUC: 0.79) radiomic models yielded better performance for predicting OS than the pre-RT intratumoral model (AUC: 0.62) in validation cohort. This was confirmed by Kaplan-Meier analysis, in which risk stratification depended on the mid-RT peritumoral (p = 0.009) and intratumoral (p = 0.003) radiomics could be improved for OS, in comparison to the pre-RT intratumoral radiomics (p = 0.199). Multivariate analysis identified mid-RT peritumoral and intratumoral radiomic models as independent prognostic factors for both OS and PFS. Mid-RT peritumoral and intratumoral radiomics were correlated with treatment-related lymphopenia. CONCLUSION Mid-RT peritumoral and intratumoral radiomic models are promising image biomarkers that could have clinical utility for predicting OS and PFS in patients with LHC treated with RT.
Collapse
Affiliation(s)
- Chia-Hsin Lin
- Proton and Radiation Therapy Center, Chang Gung Memorial Hospital-Linkou Medical Center, Department of Radiation Oncology, Chang Gung University, Taoyuan, Taiwan.
| | - Jiun-Lin Yan
- Department of Neurosurgery, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan; School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Wing-Keen Yap
- Proton and Radiation Therapy Center, Chang Gung Memorial Hospital-Linkou Medical Center, Department of Radiation Oncology, Chang Gung University, Taoyuan, Taiwan.
| | - Chung-Jan Kang
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital, Medical College of Chang Gung University, Taoyuan, Taiwan.
| | - Yun-Chen Chang
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital, Medical College of Chang Gung University, Taoyuan, Taiwan.
| | - Tsung-You Tsai
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital, Medical College of Chang Gung University, Taoyuan, Taiwan.
| | - Kai-Ping Chang
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital, Medical College of Chang Gung University, Taoyuan, Taiwan.
| | - Chun-Ta Liao
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital, Medical College of Chang Gung University, Taoyuan, Taiwan.
| | - Cheng-Lung Hsu
- Department of Hematology-Oncology, Chang Gung Memorial Hospital, Medical College of Chang Gung University, Taoyuan, Taiwan.
| | - Wen-Chi Chou
- Department of Hematology-Oncology, Chang Gung Memorial Hospital, Medical College of Chang Gung University, Taoyuan, Taiwan.
| | - Hung-Ming Wang
- Department of Hematology-Oncology, Chang Gung Memorial Hospital, Medical College of Chang Gung University, Taoyuan, Taiwan.
| | - Pei-Wei Huang
- Department of Hematology-Oncology, Chang Gung Memorial Hospital, Medical College of Chang Gung University, Taoyuan, Taiwan.
| | - Kang-Hsing Fan
- Department of Radiation Oncology, New Taipei Municipal Tucheng Hospital, New Taipei City, Taiwan.
| | - Bing-Shen Huang
- Proton and Radiation Therapy Center, Chang Gung Memorial Hospital-Linkou Medical Center, Department of Radiation Oncology, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Clinical Medical Science, Chang Gung University, Taoyuan, Taiwan.
| | - Joseph Tung-Chieh Chang
- Proton and Radiation Therapy Center, Chang Gung Memorial Hospital-Linkou Medical Center, Department of Radiation Oncology, Chang Gung University, Taoyuan, Taiwan; Department of Radiation Oncology, Xiamen Chang Gung Memorial Hospital, Xiamen, Fujian, China.
| | - Shu-Ju Tu
- Department of Medical Imaging and Radiological Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Medical Imaging and Intervention, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| | - Chien-Yu Lin
- Proton and Radiation Therapy Center, Chang Gung Memorial Hospital-Linkou Medical Center, Department of Radiation Oncology, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
14
|
Nguyen TM, Bertolus C, Giraud P, Burgun A, Saintigny P, Bibault JE, Foy JP. A Radiomics Approach to Identify Immunologically Active Tumor in Patients with Head and Neck Squamous Cell Carcinomas. Cancers (Basel) 2023; 15:5369. [PMID: 38001629 PMCID: PMC10670096 DOI: 10.3390/cancers15225369] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/05/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND We recently developed a gene-expression-based HOT score to identify the hot/cold phenotype of head and neck squamous cell carcinomas (HNSCCs), which is associated with the response to immunotherapy. Our goal was to determine whether radiomic profiling from computed tomography (CT) scans can distinguish hot and cold HNSCC. METHOD We included 113 patients from The Cancer Genome Atlas (TCGA) and 20 patients from the Groupe Hospitalier Pitié-Salpêtrière (GHPS) with HNSCC, all with available pre-treatment CT scans. The hot/cold phenotype was computed for all patients using the HOT score. The IBEX software (version 4.11.9, accessed on 30 march 2020) was used to extract radiomic features from the delineated tumor region in both datasets, and the intraclass correlation coefficient (ICC) was computed to select robust features. Machine learning classifier models were trained and tested in the TCGA dataset and validated using the area under the receiver operator characteristic curve (AUC) in the GHPS cohort. RESULTS A total of 144 radiomic features with an ICC >0.9 was selected. An XGBoost model including these selected features showed the best performance prediction of the hot/cold phenotype with AUC = 0.86 in the GHPS validation dataset. CONCLUSIONS AND RELEVANCE We identified a relevant radiomic model to capture the overall hot/cold phenotype of HNSCC. This non-invasive approach could help with the identification of patients with HNSCC who may benefit from immunotherapy.
Collapse
Affiliation(s)
- Tan Mai Nguyen
- Sorbonne Université, Department of Maxillo-Facial Surgery, Hôpital Pitié-Salpêtrière, Assistance Publique des Hôpitaux de Paris, 75013 Paris, France; (T.M.N.); (C.B.)
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, 69008 Lyon, France;
- INSERM, UMR S1138, Cordeliers Research Center, Université Paris Cité, 75005 Paris, France; (P.G.); (A.B.); (J.-E.B.)
| | - Chloé Bertolus
- Sorbonne Université, Department of Maxillo-Facial Surgery, Hôpital Pitié-Salpêtrière, Assistance Publique des Hôpitaux de Paris, 75013 Paris, France; (T.M.N.); (C.B.)
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, 69008 Lyon, France;
| | - Paul Giraud
- INSERM, UMR S1138, Cordeliers Research Center, Université Paris Cité, 75005 Paris, France; (P.G.); (A.B.); (J.-E.B.)
- Sorbonne Université, Department of Radiation Oncology, Hôpital Pitié-Salpêtrière, Assistance Publique des Hôpitaux de Paris, 75013 Paris, France
| | - Anita Burgun
- INSERM, UMR S1138, Cordeliers Research Center, Université Paris Cité, 75005 Paris, France; (P.G.); (A.B.); (J.-E.B.)
| | - Pierre Saintigny
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, 69008 Lyon, France;
- Department of Medical Oncology, Centre Léon Bérard, 69008 Lyon, France
| | - Jean-Emmanuel Bibault
- INSERM, UMR S1138, Cordeliers Research Center, Université Paris Cité, 75005 Paris, France; (P.G.); (A.B.); (J.-E.B.)
- Department of Radiation Oncology, Hôpital Européen Georges-Pompidou, Université Paris Cité, 75015 Paris, France
| | - Jean-Philippe Foy
- Sorbonne Université, Department of Maxillo-Facial Surgery, Hôpital Pitié-Salpêtrière, Assistance Publique des Hôpitaux de Paris, 75013 Paris, France; (T.M.N.); (C.B.)
- Sorbonne Université, INSERM UMRS 938, Centre de Recherche de Saint Antoine, Team Cancer Biology and Therapeutics, 75011 Paris, France
| |
Collapse
|
15
|
Altinok O, Guvenis A. Interpretable radiomics method for predicting human papillomavirus status in oropharyngeal cancer using Bayesian networks. Phys Med 2023; 114:102671. [PMID: 37708571 DOI: 10.1016/j.ejmp.2023.102671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 06/14/2023] [Accepted: 09/06/2023] [Indexed: 09/16/2023] Open
Abstract
OBJECTIVES To develop a simple interpretable Bayesian Network (BN) to classify HPV status in patients with oropharyngeal cancer. METHODS Two hundred forty-six patients, 216 of whom were HPV positive, were used in this study. We extracted 851 radiomics markers from patients' contrast-enhanced Computed Tomography (CT) images. Mens eX Machina (MXM) approach selected two most relevant predictors: sphericity and max2DDiameterRow. The area under the curve (AUC) demonstrated BN model performance in 30% of the data reserved for testing. A Support Vector Machine (SVM) based method was also implemented for comparison purposes. RESULTS The Mens eX Machina (MXM) approach selected two most relevant predictors: sphericity and max2DDiameterRow. Areas under the Curves (AUC) were found 0.78 and 0.72 on the training and test data, respectively. When using support vector machine (SVM) and 25 features, the AUC was found 0.83 on the test data. CONCLUSIONS The straightforward structure and power of interpretability of our BN model will help clinicians make treatment decisions and enable the non-invasive detection of HPV status from contrast-enhanced CT images. Higher accuracy can be obtained using more complex structures at the expense of lower interpretability. ADVANCES IN KNOWLEDGE Radiomics is being studied lately as a simple imaging data based HPV status detection technique which can be an alternative to laboratory approaches. However, it generally lacks interpretability. This work demonstrated the feasibility of using Bayesian networks based radiomics for predicting HPV positivity in an interpretable way.
Collapse
Affiliation(s)
- Oya Altinok
- Institute of Biomedical Engineering, Bogazici University, Istanbul, Turkey; Biomedical Engineering, Namik Kemal University, Tekirdağ, Turkey.
| | - Albert Guvenis
- Institute of Biomedical Engineering, Bogazici University, Istanbul, Turkey
| |
Collapse
|
16
|
Zhong NN, Wang HQ, Huang XY, Li ZZ, Cao LM, Huo FY, Liu B, Bu LL. Enhancing head and neck tumor management with artificial intelligence: Integration and perspectives. Semin Cancer Biol 2023; 95:52-74. [PMID: 37473825 DOI: 10.1016/j.semcancer.2023.07.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 07/11/2023] [Accepted: 07/15/2023] [Indexed: 07/22/2023]
Abstract
Head and neck tumors (HNTs) constitute a multifaceted ensemble of pathologies that primarily involve regions such as the oral cavity, pharynx, and nasal cavity. The intricate anatomical structure of these regions poses considerable challenges to efficacious treatment strategies. Despite the availability of myriad treatment modalities, the overall therapeutic efficacy for HNTs continues to remain subdued. In recent years, the deployment of artificial intelligence (AI) in healthcare practices has garnered noteworthy attention. AI modalities, inclusive of machine learning (ML), neural networks (NNs), and deep learning (DL), when amalgamated into the holistic management of HNTs, promise to augment the precision, safety, and efficacy of treatment regimens. The integration of AI within HNT management is intricately intertwined with domains such as medical imaging, bioinformatics, and medical robotics. This article intends to scrutinize the cutting-edge advancements and prospective applications of AI in the realm of HNTs, elucidating AI's indispensable role in prevention, diagnosis, treatment, prognostication, research, and inter-sectoral integration. The overarching objective is to stimulate scholarly discourse and invigorate insights among medical practitioners and researchers to propel further exploration, thereby facilitating superior therapeutic alternatives for patients.
Collapse
Affiliation(s)
- Nian-Nian Zhong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Han-Qi Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Xin-Yue Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Zi-Zhan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Lei-Ming Cao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Fang-Yi Huo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Bing Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| | - Lin-Lin Bu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| |
Collapse
|
17
|
Song C, Chen X, Tang C, Xue P, Jiang Y, Qiao Y. Artificial intelligence for HPV status prediction based on disease-specific images in head and neck cancer: A systematic review and meta-analysis. J Med Virol 2023; 95:e29080. [PMID: 37691329 DOI: 10.1002/jmv.29080] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 07/14/2023] [Accepted: 08/03/2023] [Indexed: 09/12/2023]
Abstract
Accurate early detection of the human papillomavirus (HPV) status in head and neck cancer (HNC) is crucial to identify at-risk populations, stratify patients, personalized treatment options, and predict prognosis. Artificial intelligence (AI) is an emerging tool to dissect imaging features. This systematic review and meta-analysis aimed to evaluate the performance of AI to predict the HPV positivity through the HPV-associated diseased images in HNC patients. A systematic literature search was conducted in databases including Ovid-MEDLINE, Embase, and Web of Science Core Collection for studies continuously published from inception up to October 30, 2022. Search strategies included keywords such as "artificial intelligence," "head and neck cancer," "HPV," and "sensitivity & specificity." Duplicates, articles without HPV predictions, letters, scientific reports, conference abstracts, or reviews were excluded. Binary diagnostic data were then extracted to generate contingency tables and then used to calculate the pooled sensitivity (SE), specificity (SP), area under the curve (AUC), and their 95% confidence interval (CI). A random-effects model was used for meta-analysis, four subgroup analyses were further explored. Totally, 22 original studies were included in the systematic review, 15 of which were eligible to generate 33 contingency tables for meta-analysis. The pooled SE and SP for all studies were 79% (95% CI: 75-82%) and 74% (95% CI: 69-78%) respectively, with an AUC of 0.83 (95% CI: 0.79-0.86). When only selecting one contingency table with the highest accuracy from each study, our analysis revealed a pooled SE of 79% (95% CI: 75-83%), SP of 75% (95% CI: 69-79%), and an AUC of 0.84 (95% CI: 0.81-0.87). The respective heterogeneities were moderate (I2 for SE and SP were 51.70% and 51.01%) and only low (35.99% and 21.44%). This evidence-based study showed an acceptable and promising performance for AI algorithms to predict HPV status in HNC but was not comparable to the routine p16 immunohistochemistry. The exploitation and optimization of AI algorithms warrant further research. Compared with previous studies, future studies anticipate to make progress in the selection of databases, improvement of international reporting guidelines, and application of high-quality deep learning algorithms.
Collapse
Affiliation(s)
- Cheng Song
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xu Chen
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chao Tang
- Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Peng Xue
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Jiang
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Youlin Qiao
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
18
|
Chen W, Sá RC, Bai Y, Napel S, Gevaert O, Lauderdale DS, Giger ML. Machine learning with multimodal data for COVID-19. Heliyon 2023; 9:e17934. [PMID: 37483733 PMCID: PMC10362086 DOI: 10.1016/j.heliyon.2023.e17934] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/03/2023] [Indexed: 07/25/2023] Open
Abstract
In response to the unprecedented global healthcare crisis of the COVID-19 pandemic, the scientific community has joined forces to tackle the challenges and prepare for future pandemics. Multiple modalities of data have been investigated to understand the nature of COVID-19. In this paper, MIDRC investigators present an overview of the state-of-the-art development of multimodal machine learning for COVID-19 and model assessment considerations for future studies. We begin with a discussion of the lessons learned from radiogenomic studies for cancer diagnosis. We then summarize the multi-modality COVID-19 data investigated in the literature including symptoms and other clinical data, laboratory tests, imaging, pathology, physiology, and other omics data. Publicly available multimodal COVID-19 data provided by MIDRC and other sources are summarized. After an overview of machine learning developments using multimodal data for COVID-19, we present our perspectives on the future development of multimodal machine learning models for COVID-19.
Collapse
Affiliation(s)
- Weijie Chen
- Medical Imaging and Data Resource Center (MIDRC), USA
- Division of Imaging, Diagnostics, and Software Reliability, Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, USA
| | - Rui C. Sá
- Medical Imaging and Data Resource Center (MIDRC), USA
- Department of Medicine, University of California, San Diego, USA
| | - Yuntong Bai
- Medical Imaging and Data Resource Center (MIDRC), USA
- Division of Imaging, Diagnostics, and Software Reliability, Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, USA
| | - Sandy Napel
- Medical Imaging and Data Resource Center (MIDRC), USA
- Department of Radiology, Stanford University, USA
| | - Olivier Gevaert
- Medical Imaging and Data Resource Center (MIDRC), USA
- Department of Medicine and Department of Biomedical Data Science, Stanford University, USA
| | - Diane S. Lauderdale
- Medical Imaging and Data Resource Center (MIDRC), USA
- Department of Public Health Sciences, University of Chicago, USA
| | - Maryellen L. Giger
- Medical Imaging and Data Resource Center (MIDRC), USA
- Department of Radiology, University of Chicago, USA
| |
Collapse
|
19
|
Zhang YP, Zhang XY, Cheng YT, Li B, Teng XZ, Zhang J, Lam S, Zhou T, Ma ZR, Sheng JB, Tam VCW, Lee SWY, Ge H, Cai J. Artificial intelligence-driven radiomics study in cancer: the role of feature engineering and modeling. Mil Med Res 2023; 10:22. [PMID: 37189155 DOI: 10.1186/s40779-023-00458-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 05/04/2023] [Indexed: 05/17/2023] Open
Abstract
Modern medicine is reliant on various medical imaging technologies for non-invasively observing patients' anatomy. However, the interpretation of medical images can be highly subjective and dependent on the expertise of clinicians. Moreover, some potentially useful quantitative information in medical images, especially that which is not visible to the naked eye, is often ignored during clinical practice. In contrast, radiomics performs high-throughput feature extraction from medical images, which enables quantitative analysis of medical images and prediction of various clinical endpoints. Studies have reported that radiomics exhibits promising performance in diagnosis and predicting treatment responses and prognosis, demonstrating its potential to be a non-invasive auxiliary tool for personalized medicine. However, radiomics remains in a developmental phase as numerous technical challenges have yet to be solved, especially in feature engineering and statistical modeling. In this review, we introduce the current utility of radiomics by summarizing research on its application in the diagnosis, prognosis, and prediction of treatment responses in patients with cancer. We focus on machine learning approaches, for feature extraction and selection during feature engineering and for imbalanced datasets and multi-modality fusion during statistical modeling. Furthermore, we introduce the stability, reproducibility, and interpretability of features, and the generalizability and interpretability of models. Finally, we offer possible solutions to current challenges in radiomics research.
Collapse
Affiliation(s)
- Yuan-Peng Zhang
- Department of Medical Informatics, Nantong University, Nantong, 226001, Jiangsu, China
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, 999077, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, Guangdong, China
| | - Xin-Yun Zhang
- Department of Medical Informatics, Nantong University, Nantong, 226001, Jiangsu, China
| | - Yu-Ting Cheng
- Department of Medical Informatics, Nantong University, Nantong, 226001, Jiangsu, China
| | - Bing Li
- Department of Radiation Oncology, the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008, Henan, China
| | - Xin-Zhi Teng
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Jiang Zhang
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Saikit Lam
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Ta Zhou
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Zong-Rui Ma
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Jia-Bao Sheng
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Victor C W Tam
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Shara W Y Lee
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Hong Ge
- Department of Radiation Oncology, the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008, Henan, China
| | - Jing Cai
- Department of Health Technology and Informatics, the Hong Kong Polytechnic University, Hong Kong, 999077, China.
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, Guangdong, China.
| |
Collapse
|
20
|
Chen Q, Wei R, Li S. A preoperative nomogram model for the prediction of lymph node metastasis in buccal mucosa cancer. Cancer Med 2023. [PMID: 37184116 DOI: 10.1002/cam4.6076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 03/27/2023] [Accepted: 05/03/2023] [Indexed: 05/16/2023] Open
Abstract
OBJECTIVES We sought to construct a nomogram model predicting lymph node metastasis (LNM) in patients with squamous cell carcinoma of the buccal mucosa based on preoperative clinical characteristics. METHODS Patients who underwent radical resection of a primary tumor in the buccal mucosa with neck dissection were enrolled. Clinical characteristics independently associated with LNM in multivariate analyses were adopted to build the model. Patients at low risk of LNM were defined by a predicted probability of LNM of less than 5%. RESULTS Patients who underwent surgery in an earlier period (January 2015-November 2019) were defined as the model development cohort (n = 325), and those who underwent surgery later (November 2019-March 2021) were defined as the validation cohort (n = 140). Age, tumor differentiation, tumor thickness, and clinical N stage assessed by computed tomography/magnetic resonance imaging (cN) were independent predictors of LNM. The nomogram model based on these four predictors showed good discrimination accuracy in both the model development and validation cohorts, with areas under the receiver-operating characteristic curve (AUC) of 0.814 and 0.828, respectively. LNM prediction by the nomogram model was superior to cN in AUC comparisons (0.815 vs. 0.753) and decision curve analysis of the whole cohort. Seventy-one patients were defined as having a low risk of LNM, among whom the actual metastasis rate was only 1.4%. CONCLUSIONS A robust nomogram model for preoperative LNM prediction is built.
Collapse
Affiliation(s)
- Qian Chen
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Rui Wei
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Shan Li
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
21
|
Salahuddin Z, Chen Y, Zhong X, Woodruff HC, Rad NM, Mali SA, Lambin P. From Head and Neck Tumour and Lymph Node Segmentation to Survival Prediction on PET/CT: An End-to-End Framework Featuring Uncertainty, Fairness, and Multi-Region Multi-Modal Radiomics. Cancers (Basel) 2023; 15:1932. [PMID: 37046593 PMCID: PMC10093277 DOI: 10.3390/cancers15071932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/17/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
Automatic delineation and detection of the primary tumour (GTVp) and lymph nodes (GTVn) using PET and CT in head and neck cancer and recurrence-free survival prediction can be useful for diagnosis and patient risk stratification. We used data from nine different centres, with 524 and 359 cases used for training and testing, respectively. We utilised posterior sampling of the weight space in the proposed segmentation model to estimate the uncertainty for false positive reduction. We explored the prognostic potential of radiomics features extracted from the predicted GTVp and GTVn in PET and CT for recurrence-free survival prediction and used SHAP analysis for explainability. We evaluated the bias of models with respect to age, gender, chemotherapy, HPV status, and lesion size. We achieved an aggregate Dice score of 0.774 and 0.760 on the test set for GTVp and GTVn, respectively. We observed a per image false positive reduction of 19.5% and 7.14% using the uncertainty threshold for GTVp and GTVn, respectively. Radiomics features extracted from GTVn in PET and from both GTVp and GTVn in CT are the most prognostic, and our model achieves a C-index of 0.672 on the test set. Our framework incorporates uncertainty estimation, fairness, and explainability, demonstrating the potential for accurate detection and risk stratification.
Collapse
Affiliation(s)
- Zohaib Salahuddin
- The D-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Yi Chen
- The D-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6200 MD Maastricht, The Netherlands
- Key Laboratory of Intelligent Medical Image Analysis and Precise Diagnosis, College of Computer Science and Technology, Guizhou University, Guiyang 550025, China
| | - Xian Zhong
- The D-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6200 MD Maastricht, The Netherlands
- Department of Medical Ultrasonics, Institute of Diagnostic and Interventional Ultrasound, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Henry C. Woodruff
- The D-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6200 MD Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, GROW—School for Oncology and Reproduction, Maastricht University Medical Center+, 6229 HX Maastricht, The Netherlands
| | - Nastaran Mohammadian Rad
- The D-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Shruti Atul Mali
- The D-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Philippe Lambin
- The D-Lab, Department of Precision Medicine, GROW—School for Oncology and Reproduction, Maastricht University, 6200 MD Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, GROW—School for Oncology and Reproduction, Maastricht University Medical Center+, 6229 HX Maastricht, The Netherlands
| |
Collapse
|
22
|
Histogram analysis of synthetic magnetic resonance imaging: Correlations with histopathological factors in head and neck squamous cell carcinoma. Eur J Radiol 2023; 160:110715. [PMID: 36753947 DOI: 10.1016/j.ejrad.2023.110715] [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: 12/31/2022] [Accepted: 01/24/2023] [Indexed: 01/30/2023]
Abstract
PURPOSE To analyse the association between histogram parameters derived from synthetic MRI (SyMRI) and different histopathological factors in head and neck squamous cell carcinoma (HNSCC). METHOD Sixty-one patients with histologically proven primary HNSCC were prospectively enrolled. The correlations between histogram parameters of SyMRI (T1, T2 and proton density (PD) maps) and histopathological factors were analysed using Spearman analysis. The Mann-Whitney U test or Student's t test was utilized to differentiate histological grades and human papillomavirus (HPV) status. The ROC curves and leave-one-out cross-validation (LOOCV) were used to evaluate the differentiation performance. Bootstrapping was applied to avoid overfitting. RESULTS Several histogram parameters were associated with histological grade: T1 map (r = 0.291) and PD map (r = 0.294 - 0.382/-0.343), and PD_75th Percentile showed the highest differentiation performance (AUC: 0.721 (ROC) and 0.719 (LOOCV)). Moderately negative correlations were found between p16 status and the histogram parameters: T1 map (r = -0.587 - -0.390), T2 map (r = -0.649 - -0.357) and PD map (r = -0.537 - -0.338). In differentiating HPV infection, Entropy was the most discriminative parameter in each map and T2_Entropy showed the highest diagnostic performance (AUC: 0.851 [ROC] and 0.851 [LOOCV]). Additionally, several histogram parameters were correlated with Ki-67 (r = -0.379/-0.397), epidermal growth factor receptor (EGFR) (r = 0.318/0.322) status and p53 (r = 0.452 - 0.665/-0.607) status. CONCLUSIONS Histogram parameters derived from SyMRI may serve as a potential biomarker for discriminating relevant histopathological features, including histological differentiation grade, HPV infection, Ki-67, EGFR and p53 statuses.
Collapse
|
23
|
Shah D, Gehani A, Mahajan A, Chakrabarty N. Advanced Techniques in Head and Neck Cancer Imaging: Guide to Precision Cancer Management. Crit Rev Oncog 2023; 28:45-62. [PMID: 37830215 DOI: 10.1615/critrevoncog.2023047799] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
Precision treatment requires precision imaging. With the advent of various advanced techniques in head and neck cancer treatment, imaging has become an integral part of the multidisciplinary approach to head and neck cancer care from diagnosis to staging and also plays a vital role in response evaluation in various tumors. Conventional anatomic imaging (CT scan, MRI, ultrasound) remains basic and focuses on defining the anatomical extent of the disease and its spread. Accurate assessment of the biological behavior of tumors, including tumor cellularity, growth, and response evaluation, is evolving with recent advances in molecular, functional, and hybrid/multiplex imaging. Integration of these various advanced diagnostic imaging and nonimaging methods aids understanding of cancer pathophysiology and provides a more comprehensive evaluation in this era of precision treatment. Here we discuss the current status of various advanced imaging techniques and their applications in head and neck cancer imaging.
Collapse
Affiliation(s)
- Diva Shah
- Senior Consultant Radiologist, Department of Radiodiagnosis, HCG Cancer Centre, Ahmedabad, 380060, Gujarat, India
| | - Anisha Gehani
- Department of Radiology and Imaging Sciences, Tata Medical Centre, New Town, WB 700160, India
| | - Abhishek Mahajan
- Department of Radiology, The Clatterbridge Cancer Centre NHS Foundation Trust, Liverpool, L7 8YA, United Kingdom
| | - Nivedita Chakrabarty
- Department of Radiodiagnosis, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), 400012, Mumbai, India
| |
Collapse
|
24
|
Siow TY, Yeh CH, Lin G, Lin CY, Wang HM, Liao CT, Toh CH, Chan SC, Lin CP, Ng SH. MRI Radiomics for Predicting Survival in Patients with Locally Advanced Hypopharyngeal Cancer Treated with Concurrent Chemoradiotherapy. Cancers (Basel) 2022; 14:cancers14246119. [PMID: 36551604 PMCID: PMC9775984 DOI: 10.3390/cancers14246119] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/06/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
A reliable prognostic stratification of patients with locally advanced hypopharyngeal cancer who had been treated with concurrent chemoradiotherapy (CCRT) is crucial for informing tailored management strategies. The purpose of this retrospective study was to develop robust and objective magnetic resonance imaging (MRI) radiomics-based models for predicting overall survival (OS) and progression-free survival (PFS) in this patient population. The study participants included 198 patients (median age: 52.25 years (interquartile range = 46.88-59.53 years); 95.96% men) who were randomly divided into a training cohort (n = 132) and a testing cohort (n = 66). Radiomic parameters were extracted from post-contrast T1-weighted MR images. Radiomic features for model construction were selected from the training cohort using least absolute shrinkage and selection operator-Cox regression models. Prognostic performances were assessed by calculating the integrated area under the receiver operating characteristic curve (iAUC). The ability of radiomic models to predict OS (iAUC = 0.580, 95% confidence interval (CI): 0.558-0.591) and PFS (iAUC = 0.625, 95% CI = 0.600-0.633) was validated in the testing cohort. The combination of radiomic signatures with traditional clinical parameters outperformed clinical variables alone in the prediction of survival outcomes (observed iAUC increments = 0.279 [95% CI = 0.225-0.334] and 0.293 [95% CI = 0.232-0.351] for OS and PFS, respectively). In summary, MRI radiomics has value for predicting survival outcomes in patients with hypopharyngeal cancer treated with CCRT, especially when combined with clinical prognostic variables.
Collapse
Affiliation(s)
- Tiing Yee Siow
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 333423, Taiwan
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Chih-Hua Yeh
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 333423, Taiwan
| | - Gigin Lin
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 333423, Taiwan
| | - Chien-Yu Lin
- Department of Radiation Oncology and Proton Therapy Center, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 333423, Taiwan
| | - Hung-Ming Wang
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 333423, Taiwan
| | - Chun-Ta Liao
- Department of Otorhinolaryngology, Head and Neck Surgery, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 333423, Taiwan
| | - Cheng-Hong Toh
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 333423, Taiwan
| | - Sheng-Chieh Chan
- Department of Nuclear Medicine, Hualien Tzu Chi Hospital, Tzu Chi University School of Medicine, Buddhist Tzu Chi Medical Foundation, Hualien 970473, Taiwan
| | - Ching-Po Lin
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Correspondence: (C.-P.L.); (S.-H.N.)
| | - Shu-Hang Ng
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan 333423, Taiwan
- Correspondence: (C.-P.L.); (S.-H.N.)
| |
Collapse
|
25
|
Zhan PC, Lyu PJ, Li Z, Liu X, Wang HX, Liu NN, Zhang Y, Huang W, Chen Y, Gao JB. CT-Based Radiomics Analysis for Noninvasive Prediction of Perineural Invasion of Perihilar Cholangiocarcinoma. Front Oncol 2022; 12:900478. [PMID: 35795043 PMCID: PMC9252420 DOI: 10.3389/fonc.2022.900478] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 05/20/2022] [Indexed: 12/02/2022] Open
Abstract
Purpose The study aimed to construct and evaluate a CT-Based radiomics model for noninvasive detecting perineural invasion (PNI) of perihilar cholangiocarcinoma (pCCA) preoperatively. Materials and Methods From February 2012 to October 2021, a total of 161 patients with pCCA who underwent resection were retrospectively enrolled in this study. Patients were allocated into the training cohort and the validation cohort according to the diagnostic time. Venous phase images of contrast-enhanced CT were used for radiomics analysis. The intraclass correlation efficient (ICC), the correlation analysis, and the least absolute shrinkage and selection operator (LASSO) regression were applied to select radiomics features and built radiomics signature. Logistic regression analyses were performed to establish a clinical model, a radiomics model, and a combined model. The performance of the predictive models was measured by area under the receiver operating characteristic curve (AUC), and pairwise ROC comparisons between models were tested using the Delong method. Finally, the model with the best performance was presented as a nomogram, and its calibration and clinical usefulness were assessed. Results Finally, 15 radiomics features were selected to build a radiomics signature, and three models were developed through logistic regression. In the training cohort, the combined model showed a higher predictive capability (AUC = 0.950) than the radiomics model and the clinical model (AUC: radiomics = 0.914, clinical = 0.756). However, in the validation cohort, the AUC of the radiomics model (AUC = 0.885) was significantly higher than the other two models (AUC: combined = 0.791, clinical = 0.567). After comprehensive consideration, the radiomics model was chosen to develop the nomogram. The calibration curve and decision curve analysis (DCA) suggested that the nomogram had a good consistency and clinical utility. Conclusion We developed a CT-based radiomics model with good performance to noninvasively predict PNI of pCCA preoperatively.
Collapse
Affiliation(s)
- Peng-Chao Zhan
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Imaging Diagnosis and Treatment for Digestive System Tumor, Zhengzhou, China
| | - Pei-jie Lyu
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhen Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xing Liu
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hui-Xia Wang
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Na-Na Liu
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Imaging Diagnosis and Treatment for Digestive System Tumor, Zhengzhou, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenpeng Huang
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Imaging Diagnosis and Treatment for Digestive System Tumor, Zhengzhou, China
| | - Yan Chen
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jian-bo Gao
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Imaging Diagnosis and Treatment for Digestive System Tumor, Zhengzhou, China
| |
Collapse
|
26
|
Pham N, Ju C, Kong T, Mukherji SK. Artificial Intelligence in Head and Neck Imaging. Semin Ultrasound CT MR 2022; 43:170-175. [PMID: 35339257 DOI: 10.1053/j.sult.2022.02.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Artificial intelligence (AI) can be applied to head and neck imaging to augment image quality and various clinical tasks including segmentation of tumor volumes, tumor characterization, tumor prognostication and treatment response, and prediction of metastatic lymph node disease. Head and neck oncology care is well positioned for the application of AI since treatment is guided by a wealth of information derived from CT, MRI, and PET imaging data. AI-based methods can integrate complex imaging, histologic, molecular, and clinical data to model tumor biology and behavior, and potentially identify associations, far beyond what conventional qualitative imaging can provide alone.
Collapse
Affiliation(s)
- Nancy Pham
- Neuroradiology, Radiology Department, University of California Los Angeles David Geffen School of Medicine, Los Angeles, CA; Neuroradiology, Radiology Department, University of Illinois.
| | - Connie Ju
- Neuroradiology, Radiology Department, University of California Los Angeles David Geffen School of Medicine, Los Angeles, CA
| | - Tracie Kong
- Neuroradiology, Radiology Department, University of California Los Angeles David Geffen School of Medicine, Los Angeles, CA
| | - Suresh K Mukherji
- Neuroradiology, Radiology Department, University of California Los Angeles David Geffen School of Medicine, Los Angeles, CA
| |
Collapse
|
27
|
Shen H, Huang Y, Yuan X, Liu D, Tu C, Wang Y, Li X, Wang X, Chen Q, Zhang J. Using quantitative parameters derived from pretreatment dual-energy computed tomography to predict histopathologic features in head and neck squamous cell carcinoma. Quant Imaging Med Surg 2022; 12:1243-1256. [PMID: 35111620 DOI: 10.21037/qims-21-650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 09/16/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) patients with a high tumor grade, lymphovascular invasion (LVI), or perineural invasion (PNI) tend to demonstrate a poor prognosis in clinical series. Thus, the identification of histopathological features, including tumor grade, LVI, and PNI, before treatment could be used to stratify the prognosis of patients with HNSCC. This study aimed to assess whether quantitative parameters derived from pretreatment dual-energy computed tomography (DECT) can predict the histopathological features of patients with HNSCC. METHODS In this study, 72 consecutive patients with pathologically confirmed HNSCC were enrolled and underwent dual-phase (noncontrast-enhanced phase and contrast-enhanced phase) DECT examinations. Normalized iodine concentration (NIC), the slope of the spectral Hounsfield unit curve (λHU), and normalized effective atomic number (NZeff) were calculated. The attenuation values on 40-140 keV noise-optimized virtual monoenergetic images [VMIs (+)] in the contrast-enhanced phase were recorded. The diagnostic performance of the quantitative parameters for predicting histopathological features, including tumor grade, LVI, and PNI, was assessed by receiver operating characteristic curves. RESULTS The NIC, λHU, NZeff, and attenuation value on the VMIs (+) at 40 keV (A40) in the grade III group, LVI-positive group, and PNI-positive group were significantly higher than those in the grade I and II groups, the LVI-negative group, and the PNI-negative group (all P values <0.05). A multivariate logistic regression model combining these 4 quantitative parameters improved the diagnostic performance of the model in predicting tumor grade, LVI, and PNI (areas under the curve: 0.969, 0.944, and 0.931, respectively). CONCLUSIONS Quantitative parameters derived from pretreatment DECT, including NIC, λHU, NZeff, and A4,0 were found to be imaging markers for predicting the histopathological characteristics of HNSCC. Combining all these characteristics improved the predictive performance of the model.
Collapse
Affiliation(s)
- Hesong Shen
- Department of Radiology, Chongqing University Cancer Hospital and Chongqing Cancer Institute and Chongqing Cancer Hospital, Chongqing, China
| | - Yuanying Huang
- Department of Oncology and Hematology, Chongqing General Hospital, University of the Chinese Academy of Sciences, Chongqing, China
| | - Xiaoqian Yuan
- Department of Radiology, Chongqing University Cancer Hospital and Chongqing Cancer Institute and Chongqing Cancer Hospital, Chongqing, China
| | - Daihong Liu
- Department of Radiology, Chongqing University Cancer Hospital and Chongqing Cancer Institute and Chongqing Cancer Hospital, Chongqing, China
| | - Chunrong Tu
- Department of Radiology, Chongqing University Cancer Hospital and Chongqing Cancer Institute and Chongqing Cancer Hospital, Chongqing, China
| | - Yu Wang
- Department of Radiology, Chongqing University Cancer Hospital and Chongqing Cancer Institute and Chongqing Cancer Hospital, Chongqing, China
| | - Xiaoqin Li
- Department of Radiology, Chongqing University Cancer Hospital and Chongqing Cancer Institute and Chongqing Cancer Hospital, Chongqing, China
| | - Xiaoxia Wang
- Department of Radiology, Chongqing University Cancer Hospital and Chongqing Cancer Institute and Chongqing Cancer Hospital, Chongqing, China
| | - Qiuzhi Chen
- Department of Radiology, Chongqing University Cancer Hospital and Chongqing Cancer Institute and Chongqing Cancer Hospital, Chongqing, China
| | - Jiuquan Zhang
- Department of Radiology, Chongqing University Cancer Hospital and Chongqing Cancer Institute and Chongqing Cancer Hospital, Chongqing, China
| |
Collapse
|
28
|
George MM, Tolley NS. AIM in Otolaryngology and Head and Neck Surgery. Artif Intell Med 2022. [DOI: 10.1007/978-3-030-64573-1_198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
|
29
|
Sukhadia SS, Tyagi A, Venkataraman V, Mukherjee P, Prasad P, Gevaert O, Nagaraj SH. ImaGene: a web-based software platform for tumor radiogenomic evaluation and reporting. BIOINFORMATICS ADVANCES 2022; 2:vbac079. [PMID: 36699376 PMCID: PMC9714320 DOI: 10.1093/bioadv/vbac079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 09/26/2022] [Accepted: 11/09/2022] [Indexed: 11/12/2022]
Abstract
Summary Radiographic imaging techniques provide insight into the imaging features of tumor regions of interest, while immunohistochemistry and sequencing techniques performed on biopsy samples yield omics data. Relationships between tumor genotype and phenotype can be identified from these data through traditional correlation analyses and artificial intelligence (AI) models. However, the radiogenomics community lacks a unified software platform with which to conduct such analyses in a reproducible manner. To address this gap, we developed ImaGene, a web-based platform that takes tumor omics and imaging datasets as inputs, performs correlation analysis between them, and constructs AI models. ImaGene has several modifiable configuration parameters and produces a report displaying model diagnostics. To demonstrate the utility of ImaGene, we utilized data for invasive breast carcinoma (IBC) and head and neck squamous cell carcinoma (HNSCC) and identified potential associations between imaging features and nine genes (WT1, LGI3, SP7, DSG1, ORM1, CLDN10, CST1, SMTNL2, and SLC22A31) for IBC and eight genes (NR0B1, PLA2G2A, MAL, CLDN16, PRDM14, VRTN, LRRN1, and MECOM) for HNSCC. ImaGene has the potential to become a standard platform for radiogenomic tumor analyses due to its ease of use, flexibility, and reproducibility, playing a central role in the establishment of an emerging radiogenomic knowledge base. Availability and implementation www.ImaGene.pgxguide.org, https://github.com/skr1/Imagene.git. Supplementary information Supplementary data are available at https://github.com/skr1/Imagene.git.
Collapse
Affiliation(s)
- Shrey S Sukhadia
- Centre for Genomics and Personalised Health, Queensland University of Technology, Brisbane, QLD 4000, Australia.,Translational Research Institute, Brisbane, QLD 4000, Australia
| | - Aayush Tyagi
- Yardi School of Artificial Intelligence, Indian Institute of Technology, New Delhi 110016, India
| | - Vivek Venkataraman
- Centre for Genomics and Personalised Health, Queensland University of Technology, Brisbane, QLD 4000, Australia.,Translational Research Institute, Brisbane, QLD 4000, Australia
| | - Pritam Mukherjee
- Stanford Center for Biomedical Informatics Research, Department of Medicine and Biomedical Data Science, Stanford University, Stanford, CA 94305-5101, USA
| | - Pratosh Prasad
- Department of Electrical Communication Engineering, Indian Institute of Science, Bangalore 560012, India
| | - Olivier Gevaert
- Stanford Center for Biomedical Informatics Research, Department of Medicine and Biomedical Data Science, Stanford University, Stanford, CA 94305-5101, USA
| | - Shivashankar H Nagaraj
- Centre for Genomics and Personalised Health, Queensland University of Technology, Brisbane, QLD 4000, Australia.,Translational Research Institute, Brisbane, QLD 4000, Australia
| |
Collapse
|
30
|
Zhang C, Gu J, Zhu Y, Meng Z, Tong T, Li D, Liu Z, Du Y, Wang K, Tian J. AI in spotting high-risk characteristics of medical imaging and molecular pathology. PRECISION CLINICAL MEDICINE 2021; 4:271-286. [PMID: 35692858 PMCID: PMC8982528 DOI: 10.1093/pcmedi/pbab026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 02/07/2023] Open
Abstract
Medical imaging provides a comprehensive perspective and rich information for disease diagnosis. Combined with artificial intelligence technology, medical imaging can be further mined for detailed pathological information. Many studies have shown that the macroscopic imaging characteristics of tumors are closely related to microscopic gene, protein and molecular changes. In order to explore the function of artificial intelligence algorithms in in-depth analysis of medical imaging information, this paper reviews the articles published in recent years from three perspectives: medical imaging analysis method, clinical applications and the development of medical imaging in the direction of pathological molecular prediction. We believe that AI-aided medical imaging analysis will be extensively contributing to precise and efficient clinical decision.
Collapse
Affiliation(s)
- Chong Zhang
- Department of Big Data Management and Application, School of International Economics and Management, Beijing Technology and Business University, Beijing 100048, China
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Jionghui Gu
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yangyang Zhu
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zheling Meng
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tong Tong
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dongyang Li
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhenyu Liu
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Du
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kun Wang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine and Engineering, Beihang University, Beijing 100191, China
| |
Collapse
|
31
|
Abstract
Artificial intelligence (AI) algorithms, particularly deep learning, have developed to the point that they can be applied in image recognition tasks. The use of AI in medical imaging can guide radiologists to more accurate image interpretation and diagnosis in radiology. The software will provide data that we cannot extract from the images. The rapid development in computational capabilities supports the wide applications of AI in a range of cancers. Among those are its widespread applications in head and neck cancer.
Collapse
|
32
|
Lo Muzio FP, Rozzi G, Rossi S, Luciani GB, Foresti R, Cabassi A, Fassina L, Miragoli M. Artificial Intelligence Supports Decision Making during Open-Chest Surgery of Rare Congenital Heart Defects. J Clin Med 2021; 10:5330. [PMID: 34830612 PMCID: PMC8623430 DOI: 10.3390/jcm10225330] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 12/21/2022] Open
Abstract
The human right ventricle is barely monitored during open-chest surgery due to the absence of intraoperative imaging techniques capable of elaborating its complex function. Accordingly, artificial intelligence could not be adopted for this specific task. We recently proposed a video-based approach for the real-time evaluation of the epicardial kinematics to support medical decisions. Here, we employed two supervised machine learning algorithms based on our technique to predict the patients' outcomes before chest closure. Videos of the beating hearts were acquired before and after pulmonary valve replacement in twelve Tetralogy of Fallot patients and recordings were properly labeled as the "unhealthy" and "healthy" classes. We extracted frequency-domain-related features to train different supervised machine learning models and selected their best characteristics via 10-fold cross-validation and optimization processes. Decision surfaces were built to classify two additional patients having good and unfavorable clinical outcomes. The k-nearest neighbors and support vector machine showed the highest prediction accuracy; the patients' class was identified with a true positive rate ≥95% and the decision surfaces correctly classified the additional patients in the "healthy" (good outcome) or "unhealthy" (unfavorable outcome) classes. We demonstrated that classifiers employed with our video-based technique may aid cardiac surgeons in decision making before chest closure.
Collapse
Affiliation(s)
- Francesco Paolo Lo Muzio
- Department of Surgery, Dentistry, Pediatrics and Gynecology, University of Verona, 37134 Verona, Italy; (F.P.L.M.); (G.R.); (G.B.L.)
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (S.R.); (R.F.); (A.C.)
| | - Giacomo Rozzi
- Department of Surgery, Dentistry, Pediatrics and Gynecology, University of Verona, 37134 Verona, Italy; (F.P.L.M.); (G.R.); (G.B.L.)
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (S.R.); (R.F.); (A.C.)
- Humanitas Research Hospital—IRCCS, Via Manzoni 56, 20089 Rozzano, MI, Italy
| | - Stefano Rossi
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (S.R.); (R.F.); (A.C.)
| | - Giovanni Battista Luciani
- Department of Surgery, Dentistry, Pediatrics and Gynecology, University of Verona, 37134 Verona, Italy; (F.P.L.M.); (G.R.); (G.B.L.)
| | - Ruben Foresti
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (S.R.); (R.F.); (A.C.)
| | - Aderville Cabassi
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (S.R.); (R.F.); (A.C.)
| | - Lorenzo Fassina
- Department of Electrical, Computer and Biomedical Engineering (DIII), University of Pavia, 27100 Pavia, Italy
| | - Michele Miragoli
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (S.R.); (R.F.); (A.C.)
- Humanitas Research Hospital—IRCCS, Via Manzoni 56, 20089 Rozzano, MI, Italy
| |
Collapse
|
33
|
Prediction of treatment outcome using MRI radiomics and machine learning in oropharyngeal cancer patients after surgical treatment. Oral Oncol 2021; 122:105559. [PMID: 34649039 DOI: 10.1016/j.oraloncology.2021.105559] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/07/2021] [Accepted: 09/28/2021] [Indexed: 11/22/2022]
Abstract
OBJECTIVES In this study, we aimed to analyze preoperative MRI images of oropharyngeal cancer patients who underwent surgical treatment, extracted radiomics features, and constructed a disease recurrence and death prediction model using radiomics features and machine-learning techniques. MATERIALS AND METHODS A total of 157 patients participated in this study, and 107 stable radiomics features were selected and used for constructing a predictive model. RESULTS The performance of the combined model (clinical and radiomics) yielded the following results: AUC of 0.786, accuracy of 0.854, precision of 0.429, recall of 0.500, and f1 score of 0.462. The combined model showed better performance than either the clinical and radiomics only models for predicting disease recurrence. For predicting death, the combined model performance has an AUC of 0.841, accuracy of 0.771, precision of 0.308, recall of 0.667, and f1 score of 0.421. The combined model showed superior performance over the predictive model using only clinical variables. A Cox proportional hazard model using the combined variables for predicting patient death yielded a c-index value that was significantly better than that of the model including only clinical variables. CONCLUSIONS A predictive model using clinical variables and MRI radiomics features showed excellent performance in predicting disease recurrence and death in oropharyngeal cancer patients. In the future, a multicenter study is necessary to verify the model's performance and confirm its clinical usefulness.
Collapse
|
34
|
Gul M, Bonjoc KJC, Gorlin D, Wong CW, Salem A, La V, Filippov A, Chaudhry A, Imam MH, Chaudhry AA. Diagnostic Utility of Radiomics in Thyroid and Head and Neck Cancers. Front Oncol 2021; 11:639326. [PMID: 34307123 PMCID: PMC8293690 DOI: 10.3389/fonc.2021.639326] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 06/08/2021] [Indexed: 11/21/2022] Open
Abstract
Radiomics is an emerging field in radiology that utilizes advanced statistical data characterizing algorithms to evaluate medical imaging and objectively quantify characteristics of a given disease. Due to morphologic heterogeneity and genetic variation intrinsic to neoplasms, radiomics have the potential to provide a unique insight into the underlying tumor and tumor microenvironment. Radiomics has been gaining popularity due to potential applications in disease quantification, predictive modeling, treatment planning, and response assessment - paving way for the advancement of personalized medicine. However, producing a reliable radiomic model requires careful evaluation and construction to be translated into clinical practices that have varying software and/or medical equipment. We aim to review the diagnostic utility of radiomics in otorhinolaryngology, including both cancers of the head and neck as well as the thyroid.
Collapse
Affiliation(s)
- Maryam Gul
- Amaze Research Foundation, Department of Biomarker Discovery, Anaheim, CA, United States
| | - Kimberley-Jane C. Bonjoc
- Department of Diagnostic and Interventional Radiology, City of Hope National Medical Center, Duarte, CA, United States
| | - David Gorlin
- Department of Diagnostic and Interventional Radiology, City of Hope National Medical Center, Duarte, CA, United States
| | - Chi Wah Wong
- Department of Diagnostic and Interventional Radiology, City of Hope National Medical Center, Duarte, CA, United States
| | - Amirah Salem
- Department of Diagnostic and Interventional Radiology, City of Hope National Medical Center, Duarte, CA, United States
| | - Vincent La
- Department of Diagnostic and Interventional Radiology, City of Hope National Medical Center, Duarte, CA, United States
| | - Aleksandr Filippov
- Department of Diagnostic and Interventional Radiology, City of Hope National Medical Center, Duarte, CA, United States
| | - Abbas Chaudhry
- Amaze Research Foundation, Department of Biomarker Discovery, Anaheim, CA, United States
| | - Muhammad H. Imam
- Florida Cancer Specialists, Department of Oncology, Orlando, FL, United States
| | - Ammar A. Chaudhry
- Department of Diagnostic and Interventional Radiology, City of Hope National Medical Center, Duarte, CA, United States
| |
Collapse
|
35
|
Keek SA, Wesseling FWR, Woodruff HC, van Timmeren JE, Nauta IH, Hoffmann TK, Cavalieri S, Calareso G, Primakov S, Leijenaar RTH, Licitra L, Ravanelli M, Scheckenbach K, Poli T, Lanfranco D, Vergeer MR, Leemans CR, Brakenhoff RH, Hoebers FJP, Lambin P. A Prospectively Validated Prognostic Model for Patients with Locally Advanced Squamous Cell Carcinoma of the Head and Neck Based on Radiomics of Computed Tomography Images. Cancers (Basel) 2021; 13:3271. [PMID: 34210048 PMCID: PMC8269129 DOI: 10.3390/cancers13133271] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/15/2021] [Accepted: 06/23/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Locoregionally advanced head and neck squamous cell carcinoma (HNSCC) patients have high relapse and mortality rates. Imaging-based decision support may improve outcomes by optimising personalised treatment, and support patient risk stratification. We propose a multifactorial prognostic model including radiomics features to improve risk stratification for advanced HNSCC, compared to TNM eighth edition, the gold standard. PATIENT AND METHODS Data of 666 retrospective- and 143 prospective-stage III-IVA/B HNSCC patients were collected. A multivariable Cox proportional-hazards model was trained to predict overall survival (OS) using diagnostic CT-based radiomics features extracted from the primary tumour. Separate analyses were performed using TNM8, tumour volume, clinical and biological variables, and combinations thereof with radiomics features. Patient risk stratification in three groups was assessed through Kaplan-Meier (KM) curves. A log-rank test was performed for significance (p-value < 0.05). The prognostic accuracy was reported through the concordance index (CI). RESULTS A model combining an 11-feature radiomics signature, clinical and biological variables, TNM8, and volume could significantly stratify the validation cohort into three risk groups (p < 0∙01, CI of 0.79 as validation). CONCLUSION A combination of radiomics features with other predictors can predict OS very accurately for advanced HNSCC patients and improves on the current gold standard of TNM8.
Collapse
Affiliation(s)
- Simon A. Keek
- The D-Lab, Department of Precision Medicine, GROW-School for Oncology, Maastricht University, Maastricht, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands; (S.A.K.); (H.C.W.); (S.P.)
| | - Frederik W. R. Wesseling
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Postbus 3035, 6202 NA Maastricht, The Netherlands; (F.W.R.W.); (F.J.P.H.)
| | - Henry C. Woodruff
- The D-Lab, Department of Precision Medicine, GROW-School for Oncology, Maastricht University, Maastricht, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands; (S.A.K.); (H.C.W.); (S.P.)
- Department of Radiology and Nuclear Medicine, GROW-School for Oncology, Maastricht University Medical Centre+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Janita E. van Timmeren
- Department of Radiation Oncology, University Hospital Zürich, University of Zürich, Rämistrasse 100, 8091 Zürich, Switzerland;
| | - Irene H. Nauta
- Amsterdam UMC, Otolaryngology/Head and Neck Surgery, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Postbus 7057, 1007 MB Amsterdam, The Netherlands; (I.H.N.); (C.R.L.); (R.H.B.)
| | - Thomas K. Hoffmann
- Department of Otorhinolaryngology, Head Neck Surgery, i2SOUL Consortium, University of Ulm, Frauensteige 14a (Haus 18), 89075 Ulm, Germany;
| | - Stefano Cavalieri
- Head and Neck Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, via Giacomo Venezian, University of Milan, 1 20133 Milano, Italy; (S.C.); (L.L.)
| | - Giuseppina Calareso
- Radiology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori via Giacomo Venezian, 1 20133 Milano, Italy;
| | - Sergey Primakov
- The D-Lab, Department of Precision Medicine, GROW-School for Oncology, Maastricht University, Maastricht, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands; (S.A.K.); (H.C.W.); (S.P.)
| | | | - Lisa Licitra
- Head and Neck Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, via Giacomo Venezian, University of Milan, 1 20133 Milano, Italy; (S.C.); (L.L.)
- Department of Oncology and Hemato-Oncology, University of Milan, via S. Sofia 9/1, 20122 Milano, Italy
| | - Marco Ravanelli
- Department of Medicine and Surgery, University of Brescia, Viale Europa, 11-25123 Brescia, Italy;
| | - Kathrin Scheckenbach
- Department. of Otorhinolaryngology-Head and Neck Surgery, University Hospital Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany;
| | - Tito Poli
- Maxillofacial Surgery Unit, Department of Medicine and Surgery, University of Parma-University Hospital of Parma, via Università, 12-I, 43121 Parma, Italy; (T.P.); (D.L.)
| | - Davide Lanfranco
- Maxillofacial Surgery Unit, Department of Medicine and Surgery, University of Parma-University Hospital of Parma, via Università, 12-I, 43121 Parma, Italy; (T.P.); (D.L.)
| | - Marije R. Vergeer
- Amsterdam UMC, Cancer Center Amsterdam, Department of Radiation Oncology, Vrije Universiteit Amsterdam, Postbus 7057, 1007 MB Amsterdam, The Netherlands;
| | - C. René Leemans
- Amsterdam UMC, Otolaryngology/Head and Neck Surgery, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Postbus 7057, 1007 MB Amsterdam, The Netherlands; (I.H.N.); (C.R.L.); (R.H.B.)
| | - Ruud H. Brakenhoff
- Amsterdam UMC, Otolaryngology/Head and Neck Surgery, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Postbus 7057, 1007 MB Amsterdam, The Netherlands; (I.H.N.); (C.R.L.); (R.H.B.)
| | - Frank J. P. Hoebers
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Postbus 3035, 6202 NA Maastricht, The Netherlands; (F.W.R.W.); (F.J.P.H.)
| | - Philippe Lambin
- The D-Lab, Department of Precision Medicine, GROW-School for Oncology, Maastricht University, Maastricht, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands; (S.A.K.); (H.C.W.); (S.P.)
- Department of Radiology and Nuclear Medicine, GROW-School for Oncology, Maastricht University Medical Centre+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| |
Collapse
|
36
|
Zhang MH, Hasse A, Carroll T, Pearson AT, Cipriani NA, Ginat DT. Differentiating low and high grade mucoepidermoid carcinoma of the salivary glands using CT radiomics. Gland Surg 2021; 10:1646-1654. [PMID: 34164309 DOI: 10.21037/gs-20-830] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background The purpose of this study is to determine if Haralick texture analysis on CT imaging of mucoepidermoid carcinomas (MEC) can differentiate low-grade and high-grade tumors. Methods A retrospective review of 18 patients with MEC of the salivary glands, corresponding CT imaging and pathology report was performed. Tumors were manually segmented and image analysis was performed to calculate radiomic features. Radiomic features were compared between low-grade and high-grade MEC. A multivariable logistic regression model and receiver operating characteristic analysis was performed. Results A total of 18 patients (mean age, 51, range 9-83 years, 8 men and 10 women) were included. Nine patients had low-grade pathology and nine patients had high-grade pathology. Of the 18 cases, 7 (39%) occurred in the parotid gland and 11 (61%) occurred in minor salivary glands. No individual feature was significantly different between low-grade and high-grade MEC. A logistic regression model including surface regularity, energy and information measure II of correlation was performed and was able to predict high-grade MEC accurately (sensitivity 89%, specificity 68%). The area under the receiver operating characteristic curve was 0.802. Conclusions High-grade MEC tend to have a low energy, high correlation texture as well as surface irregularity. Together, these three features may comprise a tumor phenotype that is able to predict high-grade pathology in MECs.
Collapse
Affiliation(s)
- Michael H Zhang
- Pritzker School of Medicine, The University of Chicago, Chicago IL, USA
| | - Adam Hasse
- Graduate Program in Medical Physics, The University of Chicago, Chicago, IL, USA
| | - Timothy Carroll
- Graduate Program in Medical Physics, The University of Chicago, Chicago, IL, USA
| | | | | | - Daniel T Ginat
- Department of Radiology, The University of Chicago, Chicago IL, USA
| |
Collapse
|
37
|
Yoon H, Ha S, Kwon SJ, Park SY, Kim J, O JH, Yoo IR. Prognostic value of tumor metabolic imaging phenotype by FDG PET radiomics in HNSCC. Ann Nucl Med 2021; 35:370-377. [PMID: 33554314 DOI: 10.1007/s12149-021-01586-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 12/28/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Tumor metabolic phenotype can be assessed with integrated image pattern analysis of 18F-fluoro-deoxy-glucose (FDG) Positron Emission Tomography/Computed Tomography (PET/CT), called radiomics. This study was performed to assess the prognostic value of radiomics PET parameters in head and neck squamous cell carcinoma (HNSCC) patients. METHODS 18F-fluoro-deoxy-glucose (FDG) PET/CT data of 215 patients from HNSCC collection free database in The Cancer Imaging Archive (TCIA), and 122 patients in Seoul St. Mary's Hospital with baseline FDG PET/CT for locally advanced HNSCC were reviewed. Data from TCIA database were used as a training cohort, and data from Seoul St. Mary's Hospital as a validation cohort. With the training cohort, primary tumors were segmented by Nestles' adaptive thresholding method. Segmental tumors in PET images were preprocessed using relative resampling of 64 bins. Forty-two PET parameters, including conventional parameters and texture parameters, were measured. Binary groups of homogeneous imaging phenotypes, clustered by K-means method, were compared for overall survival (OS) and disease-free survival (DFS) by log-rank test. Selected individual radiomics parameters were tested along with clinical factors, including age and sex, by Cox-regression test for OS and DFS, and the significant parameters were tested with multivariate analysis. Significant parameters on multivariate analysis were again tested with multivariate analysis in the validation cohort. RESULTS A total of 119 patients, 70 from training, and 49 from validation cohort, were included in the study. The median follow-up period was 62 and 52 months for the training and the validation cohort, respectively. In the training cohort. binary groups with different metabolic radiomics phenotypes showed significant difference in OS (p = 0.036), and borderline difference in DFS (p = 0.086). Gray-Level Non-Uniformity for zone (GLNUGLZLM) was the most significant prognostic factor for both OS (hazard ratio [HR] 3.1, 95% confidence interval [CI] 1.4-7.3, p = 0.008) and DFS (HR 4.5, CI 1.3-16, p = 0.020). Multivariate analysis revealed GLNUGLZLM as an independent prognostic factor for OS (HR 3.7, 95% CI 1.1-7.5, p = 0.032). GLNUGLZLM remained as an independent prognostic factor in the validation cohort (HR 14.8. 95% CI 3.3-66, p < 0.001). CONCLUSIONS Baseline FDG PET radiomics contain risk information for survival prognosis in HNSCC patients. The metabolic heterogeneity parameter, GLNUGLZLM, may assist clinicians in patient risk assessment as a feasible prognostic factor.
Collapse
Affiliation(s)
- Hyukjin Yoon
- Division of Nuclear Medicine, Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Seunggyun Ha
- Division of Nuclear Medicine, Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea.
| | - Soo Jin Kwon
- Division of Nuclear Medicine, Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Sonya Youngju Park
- Division of Nuclear Medicine, Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jihyun Kim
- Division of Nuclear Medicine, Department of Radiology, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Daejeon, South Korea
| | - Joo Hyun O
- Division of Nuclear Medicine, Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Ie Ryung Yoo
- Division of Nuclear Medicine, Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
38
|
Wang X, Li BB. Deep Learning in Head and Neck Tumor Multiomics Diagnosis and Analysis: Review of the Literature. Front Genet 2021; 12:624820. [PMID: 33643386 PMCID: PMC7902873 DOI: 10.3389/fgene.2021.624820] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 01/07/2021] [Indexed: 12/24/2022] Open
Abstract
Head and neck tumors are the sixth most common neoplasms. Multiomics integrates multiple dimensions of clinical, pathologic, radiological, and biological data and has the potential for tumor diagnosis and analysis. Deep learning (DL), a type of artificial intelligence (AI), is applied in medical image analysis. Among the DL techniques, the convolution neural network (CNN) is used for image segmentation, detection, and classification and in computer-aided diagnosis. Here, we reviewed multiomics image analysis of head and neck tumors using CNN and other DL neural networks. We also evaluated its application in early tumor detection, classification, prognosis/metastasis prediction, and the signing out of the reports. Finally, we highlighted the challenges and potential of these techniques.
Collapse
Affiliation(s)
- Xi Wang
- Department of Oral Pathology, Peking University School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing, China
- Research Unit of Precision Pathologic Diagnosis in Tumors of the Oral and Maxillofacial Regions, Chinese Academy of Medical Sciences, Beijing, China
| | - Bin-bin Li
- Department of Oral Pathology, Peking University School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing, China
- Research Unit of Precision Pathologic Diagnosis in Tumors of the Oral and Maxillofacial Regions, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
39
|
Peng Z, Wang Y, Wang Y, Jiang S, Fan R, Zhang H, Jiang W. Application of radiomics and machine learning in head and neck cancers. Int J Biol Sci 2021; 17:475-486. [PMID: 33613106 PMCID: PMC7893590 DOI: 10.7150/ijbs.55716] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 12/10/2020] [Indexed: 02/07/2023] Open
Abstract
With the continuous development of medical image informatics technology, more and more high-throughput quantitative data could be extracted from digital medical images, which has resulted in a new kind of omics-Radiomics. In recent years, in addition to genomics, proteomics and metabolomics, radiomic has attracted the interest of more and more researchers. Compared to other omics, radiomics can be perfectly integrated with clinical data, even with the pathology and molecular biomarker, so that the study can be closer to the clinical reality and more revealing of the tumor development. Mass data will also be generated in this process. Machine learning, due to its own characteristics, has a unique advantage in processing massive radiomic data. By analyzing mass amounts of data with strong clinical relevance, people can construct models that more accurately reflect tumor development and progression, thereby providing the possibility of personalized and sequential treatment of patients. As one of the cancer types whose treatment and diagnosis rely on imaging examination, radiomics has a very broad application prospect in head and neck cancers (HNC). Until now, there have been some notable results in HNC. In this review, we will introduce the concepts and workflow of radiomics and machine learning and their current applications in head and neck cancers, as well as the directions and applications of artificial intelligence in the treatment and diagnosis of HNC.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Weihong Jiang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha 410078, Hunan, China
| |
Collapse
|
40
|
George MM, Tolley NS. AIM in Otolaryngology and Head & Neck Surgery. Artif Intell Med 2021. [DOI: 10.1007/978-3-030-58080-3_198-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|