1
|
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
|
2
|
Qin K, Gong C, Cheng Y, Li L, Liu C, Yang F, Rao J, Li Q. Radiomics-based model for prediction of TGF-β1 expression in head and neck squamous cell carcinoma. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2024; 14:239-252. [PMID: 39309414 PMCID: PMC11411193 DOI: 10.62347/jmkv7596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 08/08/2024] [Indexed: 09/25/2024]
Abstract
OBJECTIVE To explore the connection between TGF-β1 expression and the survival of patients with head and neck squamous cell carcinoma (HNSCC), as well as whether non-invasive CT-based Radiomics can predict TGF-β1 expression in HNSCC patients. METHODS Data on transcriptional profiling and clinical information were acquired from the TCGA database and subsequently categorized based on the TGF-β1 expression cutoff value. Based on the completeness of enhanced arterial phase CT scans, 139 HNSCC patients were selected. The PyRadiomics package was used to extract radiomic features, and the 3D Slicer software was used for image segmentation. Using the mRMR_RFE and Repeat LASSO algorithms, the optimal features for establishing the corresponding gradient enhancement prediction models were identified. RESULTS A survival analysis was performed on 483 patients, who were divided into two groups based on the TGF-β1 expression cut-off. The Kaplan-Meier curve indicated that TGF-β1 was a significant independent risk factor that reduced patient survival. To construct gradient enhancement prediction models, we used the mRMR_RFE algorithm and the Repeat_LASSO algorithm to obtain two features (glrlm and ngtdm) and three radiation features (glrlm, first order_10percentile, and gldm). In both the training and validation cohorts, the two established models demonstrated strong predictive potential. Furthermore, there was no statistically significant difference in the calibration curve, DCA diagram, or AUC values between the mRMR_RFE_GBM model and the LASSO_GBM model, suggesting that both models fit well. CONCLUSION Based on these findings, TGF-β1 was shown to be significantly associated with a poor prognosis and to be a potential risk factor for HNSCC. Furthermore, by employing the mRMR_RFE_GBM and Repeat_LASSO_GBM models, we were able to effectively predict TGF-β1 expression levels in HNSCC through non-invasive CT-based Radiomics.
Collapse
Affiliation(s)
- Kai Qin
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, China
| | - Chen Gong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, China
| | - Yi Cheng
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, China
| | - Li Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, China
| | - Chengxia Liu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, China
| | - Feng Yang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, China
| | - Jie Rao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, China
| | - Qianxia Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, Hubei, China
| |
Collapse
|
3
|
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
|
4
|
Rao Y, Ma Y, Wang J, Xiao W, Wu J, Shi L, Guo L, Fan L. Performance of radiomics in the differential diagnosis of parotid tumors: a systematic review. Front Oncol 2024; 14:1383323. [PMID: 39119093 PMCID: PMC11306159 DOI: 10.3389/fonc.2024.1383323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 07/01/2024] [Indexed: 08/10/2024] Open
Abstract
Purpose A systematic review and meta-analysis were conducted to evaluate the diagnostic precision of radiomics in the differential diagnosis of parotid tumors, considering the increasing utilization of radiomics in tumor diagnosis. Although some researchers have attempted to apply radiomics in this context, there is ongoing debate regarding its accuracy. Methods Databases of PubMed, Cochrane, EMBASE, and Web of Science up to May 29, 2024 were systematically searched. The quality of included primary studies was assessed using the Radiomics Quality Score (RQS) checklist. The meta-analysis was performed utilizing a bivariate mixed-effects model. Results A total of 39 primary studies were incorporated. The machine learning model relying on MRI radiomics for diagnosis malignant tumors of the parotid gland, demonstrated a sensitivity of 0.80 [95% CI: 0.74, 0.86], SROC of 0.89 [95% CI: 0.27-0.99] in the validation set. The machine learning model based on MRI radiomics for diagnosis malignant tumors of the parotid gland, exhibited a sensitivity of 0.83[95% CI: 0.76, 0.88], SROC of 0.89 [95% CI: 0.17-1.00] in the validation set. The models also demonstrated high predictive accuracy for benign lesions. Conclusion There is great potential for radiomics-based models to improve the accuracy of diagnosing benign and malignant tumors of the parotid gland. To further enhance this potential, future studies should consider implementing standardized radiomics-based features, adopting more robust feature selection methods, and utilizing advanced model development tools. These measures can significantly improve the diagnostic accuracy of artificial intelligence algorithms in distinguishing between benign and malignant tumors of the parotid gland. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42023434931.
Collapse
Affiliation(s)
- Yilin Rao
- Department of Prosthodontics, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Yuxi Ma
- Department of Prosthodontics, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Jinghan Wang
- Department of Prosthodontics, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Weiwei Xiao
- Department of Prosthodontics, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Jiaqi Wu
- Department of Prosthodontics, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Liang Shi
- Department of Prosthodontics, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Ling Guo
- Department of Prosthodontics, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Liyuan Fan
- Department of Prosthodontics, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
5
|
de Oliveira LAP, Lopes DLG, Gomes JPP, da Silveira RV, Nozaki DVA, Santos LF, Castellano G, de Castro Lopes SLP, Costa ALF. Enhanced Diagnostic Precision: Assessing Tumor Differentiation in Head and Neck Squamous Cell Carcinoma Using Multi-Slice Spiral CT Texture Analysis. J Clin Med 2024; 13:4038. [PMID: 39064078 PMCID: PMC11277332 DOI: 10.3390/jcm13144038] [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: 05/13/2024] [Revised: 06/28/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
This study explores the efficacy of texture analysis by using preoperative multi-slice spiral computed tomography (MSCT) to non-invasively determine the grade of cellular differentiation in head and neck squamous cell carcinoma (HNSCC). In a retrospective study, MSCT scans of patients with HNSCC were analyzed and classified based on its histological grade as moderately differentiated, well-differentiated, or poorly differentiated. The location of the tumor was categorized as either in the bone or in soft tissues. Segmentation of the lesion areas was conducted, followed by texture analysis. Eleven GLCM parameters across five different distances were calculated. Median values and correlations of texture parameters were examined in relation to tumor differentiation grade by using Spearman's correlation coefficient and Kruskal-Wallis and Dunn tests. Forty-six patients were included, predominantly female (87%), with a mean age of 66.7 years. Texture analysis revealed significant parameter correlations with histopathological grades of tumor differentiation. The study identified no significant age correlation with tumor differentiation, which underscores the potential of texture analysis as an age-independent biomarker. The strong correlations between texture parameters and histopathological grades support the integration of this technique into the clinical decision-making process.
Collapse
Affiliation(s)
- Lays Assolini Pinheiro de Oliveira
- Department of Anesthesiology, Oncology and Radiology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas 13084-971, SP, Brazil;
- Postgraduate Program in Dentistry, Dentomaxillofacial Radiology and Imaging Laboratory, Department of Dentistry, Cruzeiro do Sul University (UNICSUL), São Paulo 01506-000, SP, Brazil;
| | - Diana Lorena Garcia Lopes
- Postgraduate Program in Dentistry, Dentomaxillofacial Radiology and Imaging Laboratory, Department of Dentistry, Cruzeiro do Sul University (UNICSUL), São Paulo 01506-000, SP, Brazil;
| | - João Pedro Perez Gomes
- Department of Stomatology, Division of Oral Radiology, School of Dentistry, University of São Paulo (USP), São Paulo 05508-000, SP, Brazil;
| | - Rafael Vinicius da Silveira
- Institute of Physics Gleb Wataghin, Universidade Estadual de Campinas (UNICAMP), Campinas 13084-971, SP, Brazil; (R.V.d.S.); (G.C.)
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas 13083-970, SP, Brazil
| | | | - Lana Ferreira Santos
- Department of Diagnosis and Surgery, São José dos Campos School of Dentistry, São Paulo State University (UNESP), São José dos Campos, São Paulo 12245-000, SP, Brazil; (L.F.S.); (S.L.P.d.C.L.)
| | - Gabriela Castellano
- Institute of Physics Gleb Wataghin, Universidade Estadual de Campinas (UNICAMP), Campinas 13084-971, SP, Brazil; (R.V.d.S.); (G.C.)
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas 13083-970, SP, Brazil
| | - Sérgio Lúcio Pereira de Castro Lopes
- Department of Diagnosis and Surgery, São José dos Campos School of Dentistry, São Paulo State University (UNESP), São José dos Campos, São Paulo 12245-000, SP, Brazil; (L.F.S.); (S.L.P.d.C.L.)
| | - Andre Luiz Ferreira Costa
- Department of Anesthesiology, Oncology and Radiology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas 13084-971, SP, Brazil;
- Postgraduate Program in Dentistry, Dentomaxillofacial Radiology and Imaging Laboratory, Department of Dentistry, Cruzeiro do Sul University (UNICSUL), São Paulo 01506-000, SP, Brazil;
- School of Dentistry of Paulista Association of Dentists (FAOA), São Paulo 02010-000, SP, Brazil;
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Zheng YM, Che JY, Yuan MG, Wu ZJ, Pang J, Zhou RZ, Li XL, Dong C. A CT-Based Deep Learning Radiomics Nomogram to Predict Histological Grades of Head and Neck Squamous Cell Carcinoma. Acad Radiol 2023; 30:1591-1599. [PMID: 36460582 DOI: 10.1016/j.acra.2022.11.007] [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: 10/03/2022] [Revised: 11/01/2022] [Accepted: 11/10/2022] [Indexed: 12/03/2022]
Abstract
RATIONALE AND OBJECTIVES Accurate pretreatment assessment of histological differentiation grade of head and neck squamous cell carcinoma (HNSCC) is crucial for prognosis evaluation. This study aimed to construct and validate a contrast-enhanced computed tomography (CECT)-based deep learning radiomics nomogram (DLRN) to predict histological differentiation grades of HNSCC. MATERIALS AND METHODS A total of 204 patients with HNSCC who underwent CECT scans were enrolled in this study. The participants recruited from two hospitals were split into a training set (n=124, 74 well/moderately differentiated and 50 poorly differentiated) of patients from one hospital and an external test set of patients from the other hospital (n=80, 49 well/moderately differentiated and 31 poorly differentiated). CECT-based manually-extracted radiomics (MER) features and deep learning (DL) features were extracted and selected. The selected MER features and DL features were then combined to construct a DLRN via multivariate logistic regression. The predictive performance of the DLRN was assessed using ROCs and decision curve analysis (DCA). RESULTS Three MER features and seven DL features were finally selected. The DLRN incorporating the selected MER and DL features showed good predictive value for the histological differentiation grades of HNSCC (well/moderately differentiated vs. poorly differentiated) in both the training (AUC, 0.878) and test (AUC, 0.822) sets. DCA demonstrated that the DLRN was clinically useful for predicting histological differentiation grades of HNSCC. CONCLUSION A CECT-based DLRN was constructed to predict histological differentiation grades of HNSCC. The DLRN showed good predictive efficacy and might be useful for prognostic evaluation of patients with HNSCC.
Collapse
Affiliation(s)
- Ying-Mei Zheng
- Health Management Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jun-Yi Che
- Department of Radiology, Qingdao Municipal Hospital, Qingdao, China
| | - Ming-Gang Yuan
- Department of Nuclear Medicine, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, China
| | - Zeng-Jie Wu
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jing Pang
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Rui-Zhi Zhou
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiao-Li Li
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Cheng Dong
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
9
|
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
|
10
|
Committeri U, Barone S, Salzano G, Arena A, Borriello G, Giovacchini F, Fusco R, Vaira LA, Scarpa A, Abbate V, Ugga L, Piombino P, Ionna F, Califano L, Orabona GD. Support Tools in the Differential Diagnosis of Salivary Gland Tumors through Inflammatory Biomarkers and Radiomics Metrics: A Preliminary Study. Cancers (Basel) 2023; 15:cancers15061876. [PMID: 36980760 PMCID: PMC10047378 DOI: 10.3390/cancers15061876] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/03/2023] [Accepted: 03/12/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND The purpose of this study was to investigate how the systemic inflammation response index (SIRI), systemic immune-inflammation index (SII), neutrophil/lymphocyte ratio (NLR) and platelet/lymphocyte ratio (PLR), and radiomic metrics (quantitative descriptors of image content) extracted from MRI sequences by machine learning increase the efficacy of proper presurgical differentiation between benign and malignant salivary gland tumors. METHODS A retrospective study of 117 patients with salivary gland tumors was conducted between January 2015 and November 2022. Univariate analyses with nonparametric tests and multivariate analyses with machine learning approaches were used. RESULTS Inflammatory biomarkers showed statistically significant differences (p < 0.05) in the Kruskal-Wallis test based on median values in discriminating Warthin tumors from pleomorphic adenoma and malignancies. The accuracy of NLR, PLR, SII, and SIRI was 0.88, 0.74, 0.76, and 0.83, respectively. Analysis of radiomic metrics to discriminate Warthin tumors from pleomorphic adenoma and malignancies showed statistically significant differences (p < 0.05) in nine radiomic features. The best multivariate analysis result was obtained from an SVM model with 86% accuracy, 68% sensitivity, and 91% specificity for six features. CONCLUSIONS Inflammatory biomarkers and radiomic features can comparably support a pre-surgical differential diagnosis.
Collapse
Affiliation(s)
- Umberto Committeri
- Maxillofacial Surgery Operative Unit, Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University of Naples, 80131 Naples, Italy
| | - Simona Barone
- Maxillofacial Surgery Operative Unit, Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University of Naples, 80131 Naples, Italy
| | - Giovanni Salzano
- Maxillofacial Surgery Operative Unit, Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University of Naples, 80131 Naples, Italy
| | - Antonio Arena
- Maxillofacial Surgery Operative Unit, Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University of Naples, 80131 Naples, Italy
| | - Gerardo Borriello
- Maxillofacial Surgery Operative Unit, Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University of Naples, 80131 Naples, Italy
| | - Francesco Giovacchini
- Department of Maxillo-Facial Medicine Surgery, Hospital of Perugia, 06132 Perugia, Italy
| | - Roberta Fusco
- Medical Oncology Division, Igea SpA, 80013 Naples, Italy
| | - Luigi Angelo Vaira
- Maxillofacial Surgery Operative Unit, Department of Medical, Surgical and Experimental Sciences, University of Sassari, 07100 Sassari, Italy
| | - Alfonso Scarpa
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84084 Salerno, Italy
| | - Vincenzo Abbate
- Maxillofacial Surgery Operative Unit, Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University of Naples, 80131 Naples, Italy
| | - Lorenzo Ugga
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Via S. Pansini, 5, 80131 Naples, Italy
| | - Pasquale Piombino
- Maxillofacial Surgery Operative Unit, Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University of Naples, 80131 Naples, Italy
| | - Franco Ionna
- Otolaryngology and Maxillo-Facial Surgery Unit, Istituto Nazionale Tumori-IRCCS Fondazione G. Pascale, 80131 Naples, Italy
| | - Luigi Califano
- Maxillofacial Surgery Operative Unit, Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University of Naples, 80131 Naples, Italy
| | - Giovanni Dell'Aversana Orabona
- Maxillofacial Surgery Operative Unit, Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University of Naples, 80131 Naples, Italy
| |
Collapse
|
11
|
Xie Y, Wang M, Xia H, Sun H, Yuan Y, Jia J, Chen L. Development and validation of a CECT-based radiomics model for predicting IL1B expression and prognosis of head and neck squamous cell carcinoma. Front Oncol 2023; 13:1121485. [PMID: 36969073 PMCID: PMC10036854 DOI: 10.3389/fonc.2023.1121485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/23/2023] [Indexed: 03/12/2023] Open
Abstract
IntroductionIt is necessary to explore a noninvasive method to stratify head and neck squamous cell carcinoma (HNSCC)’s prognosis and to seek new indicators for individualized precision treatment. As a vital inflammatory cytokine, IL1B might drive a new tumor subtype that could be reflected in overall survival (OS) and predicted using the radiomics method.MethodsA total of 139 patients with RNA-Seq data from The Cancer Genome Atlas (TCGA) and matched CECT data from The Cancer Image Archive (TCIA) were included in the analysis. The prognostic value of IL1B expression in patients with HNSCC was analyzed using Kaplan-Meier analysis, Cox regression analysis and subgroup analysis. Furthermore, the molecular function of IL1B on HNSCC was explored using function enrichment and immunocytes infiltration analyses. Radiomic features were extracted with PyRadiomics and processed using max-relevance minredundancy, recursive feature elimination, and gradient boosting machine algorithm to construct aradiomics model for predicting IL1B expression. The area under the receiver operating characteristic curve (AUC), calibration curve, precision recall (PR) curve, and decision curve analysis (DCA) curve were used to examine the performance of the model.ResultsIncreased IL1B expression in patients with HNSCC indicated a poor prognosis (hazard ratio [HR] = 1.56, P = 0.003) and was harmful in patients who underwent radiotherapy (HR = 1.87, P = 0.007) or chemotherapy (HR = 2.514, P < 0.001). Shape_Sphericity, glszm_SmallAreaEmphasis, and firstorder_Kurtosis were included in the radiomics model (AUC: training cohort, 0.861; validation cohort, 0.703). The calibration curves, PR curves and DCA showed good diagnostic effect of the model. The rad-score was close related to IL1B (P = 4.490*10-9), and shared the same corelated trend to EMT-related genes with IL1B. A higher rad-score was associated with worse overall survival (P = 0.041).DiscussionThe CECT-based radiomics model provides preoperative IL1B expression predictionand offers non-invasive instructions for the prognosis and individualized treatment of patients withHNSCC.
Collapse
Affiliation(s)
- Yang Xie
- The State Key Laboratory Breeding Base of Basic Science of Stomatology, Hubei Province and Key Laboratory of Oral Biomedicine (Hubei-MOST and KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Min Wang
- Hubei-MOST and KLOBM, Department of Oral Implantology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Haibin Xia
- Hubei-MOST and KLOBM, Department of Oral Implantology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Huifang Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology, Hubei Province and Key Laboratory of Oral Biomedicine (Hubei-MOST and KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yi Yuan
- Department of Oral Radiology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jun Jia
- Department of Oral Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Liangwen Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology, Hubei Province and Key Laboratory of Oral Biomedicine (Hubei-MOST and KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
- *Correspondence: Liangwen Chen,
| |
Collapse
|
12
|
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
|
13
|
Gangil T, Sharan K, Rao BD, Palanisamy K, Chakrabarti B, Kadavigere R. Utility of adding Radiomics to clinical features in predicting the outcomes of radiotherapy for head and neck cancer using machine learning. PLoS One 2022; 17:e0277168. [PMID: 36520945 PMCID: PMC9754241 DOI: 10.1371/journal.pone.0277168] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 10/24/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Radiomics involves the extraction of quantitative information from annotated Computed-Tomography (CT) images, and has been used to predict outcomes in Head and Neck Squamous Cell Carcinoma (HNSCC). Subjecting combined Radiomics and Clinical features to Machine Learning (ML) could offer better predictions of clinical outcomes. This study is a comparative performance analysis of ML models with Clinical, Radiomics, and Clinico-Radiomic datasets for predicting four outcomes of HNSCC treated with Curative Radiation Therapy (RT): Distant Metastases, Locoregional Recurrence, New Primary, and Residual Disease. METHODOLOGY The study used retrospective data of 311 HNSCC patients treated with radiotherapy between 2013-2018 at our centre. Binary prediction models were developed for the four outcomes with Clinical-only, Clinico-Radiomic, and Radiomics-only datasets, using three different ML classification algorithms namely, Random Forest (RF), Kernel Support Vector Machine (KSVM), and XGBoost. The best-performing ML algorithms of the three dataset groups was then compared. RESULTS The Clinico-Radiomic dataset using KSVM classifier provided the best prediction. Predicted mean testing accuracy for Distant Metastases, Locoregional Recurrence, New Primary, and Residual Disease was 97%, 72%, 99%, and 96%, respectively. The mean area under the receiver operating curve (AUC) was calculated and displayed for all the models using three dataset groups. CONCLUSION Clinico-Radiomic dataset improved the predictive ability of ML models over clinical features alone, while models built using Radiomics performed poorly. Radiomics data could therefore effectively supplement clinical data in predicting outcomes.
Collapse
Affiliation(s)
- Tarun Gangil
- Department of Radiotherapy and Oncology, Kasturba Medical College-Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Krishna Sharan
- Department of Radiotherapy and Oncology, Kasturba Medical College-Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - B. Dinesh Rao
- Manipal School of Information Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | | | | | - Rajagopal Kadavigere
- Department of Radiology, Kasturba Medical College-Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
- * E-mail:
| |
Collapse
|
14
|
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
|
15
|
Zheng YM, Yuan MG, Zhou RQ, Hou F, Zhan JF, Liu ND, Hao DP, Dong C. A computed tomography-based radiomics signature for predicting expression of programmed death ligand 1 in head and neck squamous cell carcinoma. Eur Radiol 2022; 32:5362-5370. [PMID: 35298679 DOI: 10.1007/s00330-022-08651-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 02/02/2022] [Accepted: 02/13/2022] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Accurate prediction of the expression of programmed death ligand 1 (PD-L1) in head and neck squamous cell carcinoma (HNSCC) before immunotherapy is crucial. This study was performed to construct and validate a contrast-enhanced computed tomography (CECT)-based radiomics signature to predict the expression of PD-L1 in HNSCC. METHODS In total, 157 patients with confirmed HNSCC who underwent CECT scans and immunohistochemical examination of tumor PD-L1 expression were enrolled in this study. The patients were divided into a training set (n = 104; 62 PD-L1-positive and 42 PD-L1-negative) and an external validation set (n = 53; 34 PD-L1-positive and 19 PD-L1-negative). A radiomics signature was constructed from radiomics features extracted from the CECT images, and a radiomics score was calculated. Performance of the radiomics signature was assessed using receiver operating characteristics analysis. RESULTS Nine features were finally selected to construct the radiomics signature. The performance of the radiomics signature to distinguish between a PD-L1-positive and PD-L1-negative status in both the training and validation sets was good, with an area under the receiver operating characteristics curve of 0.852 and 0.802 for the training and validation sets, respectively. CONCLUSIONS A CECT-based radiomics signature was constructed to predict the expression of PD-L1 in HNSCC. This model showed favorable predictive efficacy and might be useful for identifying patients with HNSCC who can benefit from anti-PD-L1 immunotherapy. KEY POINTS • Accurate prediction of the expression of PD-L1 in HNSCC before immunotherapy is crucial. • A CECT-based radiomics signature showed favorable predictive efficacy in estimation of the PD-L1 expression status in patients with HNSCC.
Collapse
Affiliation(s)
- Ying-Mei Zheng
- Health Management Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ming-Gang Yuan
- Department of Nuclear Medicine, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, China
| | - Rui-Qing Zhou
- Department of Radiology, Jiaozhou Hospital of Traditional Chinese Medicine, Jiaozhou, China
| | - Feng Hou
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jin-Feng Zhan
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Nai-Dong Liu
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Da-Peng Hao
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Cheng Dong
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
16
|
Zhu Z, Gong G, Wang L, Su Y, Lu J, Yin Y. Three-Dimensional Arterial Spin Labeling-Guided Sub-Volume Segmentation of Radiotherapy in Adult Non-Enhancing Low-Grade Gliomas. Front Oncol 2022; 12:914507. [PMID: 35860561 PMCID: PMC9291222 DOI: 10.3389/fonc.2022.914507] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Objective The present study aimed to evaluate the feasibility of sub-volume segmentation for radiotherapy planning of adult non-enhancing low-grade gliomas (NE-LGGs) guided by three-dimensional arterial spin labeling (3D-ASL). The differences in high- and low-perfusion areas of NE-LGGs were analyzed using multi-sequence magnetic resonance imaging (MRI) radiomics. Methods Fifteen adult patients with NE-LGGs were included in the study. MR images, including T1-weighted imaging (T1WI), T2 Propeller, T2 fluid-attenuated inversion recovery (T2 Flair), 3D-ASL, and contrast-enhanced T1WI (CE-T1WI), were obtained. The gross tumor volume (GTV) was delineated according to the hyperintensity on T2 Flair. The GTV was divided into high- and low-perfusion areas, namely GTV-ASL and GTV-SUB, respectively, based on the differences in cerebral blood flow (CBF) value. The volumes and CBF values of high- and low-perfusion areas were measured and compared. The least absolute shrinkage and selection operator (LASSO) regression was used to select the optimal features of all MR maps. Receiver operating characteristic (ROC) curve analysis was used to evaluate the diagnostic accuracy of the absolute CBFmean (aCBFmean), relative CBFmean (rCBFmean, normalized by the CBF value of the normal gray matter), and screened features in differentiating high- and low-perfusion areas. Results Among the enrolled patients, three (20%) patients with NE-LGGs showed focal intra- and post-radiotherapy contrast enhancement within a prior high-perfusion area of 3D-ASL. The volume ratio of the GTV-ASL to the GTV was (37.08% ± 17.88)% (46.26 ± 44.51 vs. 167.46 ± 209.64 cm3, P = 0.000). The CBFmean in the high-perfusion area was approximately two times of that in the edema area or normal gray matter (66.98 ± 18.03 vs. 35.19 ± 7.75 or 33.92 ± 8.48 ml/100g/min, P = 0.000). Thirteen features were screened, seven of which were extracted from 3D-ASL. The area undercurve (AUC) values of aCBFmean, rCBFmean, and firstorder_10Percentile from 3D-ASL were more than 0.9, of which firstorder_10Percentile was the highest. Their cut-off values were 44.16 ml/100 g/min, 1.49 and 31, respectively. Conclusion The difference in blood perfusion in the GTV can be quantified and analyzed based on 3D-ASL images for NE-LGGs, which could guide the sub-volume segmentation of the GTV. 3D-ASL should become a routine method for NE-LGGs during simulation and radiotherapy.
Collapse
Affiliation(s)
- Zihong Zhu
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Radiation Oncology Physics and Technology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Guanzhong Gong
- Department of Radiation Oncology Physics and Technology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Lizhen Wang
- Department of Radiation Oncology Physics and Technology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Ya Su
- Department of Radiation Oncology Physics and Technology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jie Lu
- Department of Radiation Oncology Physics and Technology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yong Yin
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Radiation Oncology Physics and Technology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- *Correspondence: Yong Yin,
| |
Collapse
|
17
|
CT-based radiomics analysis of different machine learning models for differentiating benign and malignant parotid tumors. Eur Radiol 2022; 32:6953-6964. [PMID: 35484339 DOI: 10.1007/s00330-022-08830-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 04/03/2022] [Accepted: 04/20/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVES This study aimed to explore and validate the value of different radiomics models for differentiating benign and malignant parotid tumors preoperatively. METHODS This study enrolled 388 patients with pathologically confirmed parotid tumors (training cohort: n = 272; test cohort: n = 116). Radiomics features were extracted from CT images of the non-enhanced, arterial, and venous phases. After dimensionality reduction and selection, radiomics models were constructed by logistic regression (LR), support vector machine (SVM), and random forest (RF). The best radiomic model was selected by using ROC curve analysis. Univariate and multivariable logistic regression was applied to analyze clinical-radiological characteristics and identify variables for developing a clinical model. A combined model was constructed by incorporating radiomics and clinical features. Model performances were assessed by ROC curve analysis, and decision curve analysis (DCA) was used to estimate the models' clinical values. RESULTS In total, 2874 radiomic features were extracted from CT images. Ten radiomics features were deemed valuable by dimensionality reduction and selection. Among radiomics models, the SVM model showed greater predictive efficiency and robustness, with AUCs of 0.844 in the training cohort; and 0.840 in the test cohort. Ultimate clinical features constructed a clinical model. The discriminatory capability of the combined model was the best (AUC, training cohort: 0.904; test cohort: 0.854). Combined model DCA revealed optimal clinical efficacy. CONCLUSIONS The combined model incorporating radiomics and clinical features exhibited excellent ability to distinguish benign and malignant parotid tumors, which may provide a noninvasive and efficient method for clinical decision making. KEY POINTS The current study is the first to compare the value of different radiomics models (LR, SVM, and RF) for preoperative differentiation of benign and malignant parotid tumors. A CT-based combined model, integrating clinical-radiological and radiomics features, is conducive to distinguishing benign and malignant parotid tumors, thereby improving diagnostic performance and aiding treatment.
Collapse
|
18
|
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
|
19
|
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]
|
20
|
Dong C, Zheng YM, Li J, Wu ZJ, Yang ZT, Li XL, Xu WJ, Hao DP. A CT-based radiomics nomogram for differentiation of squamous cell carcinoma and non-Hodgkin's lymphoma of the palatine tonsil. Eur Radiol 2022; 32:243-253. [PMID: 34236464 DOI: 10.1007/s00330-021-08153-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 06/07/2021] [Accepted: 06/14/2021] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Accurate preoperative differentiation between squamous cell carcinoma (SCC) and non-Hodgkin's lymphoma (NHL) in the palatine tonsil is crucial because of their different treatment. This study aimed to construct and validate a contrast-enhanced CT (CECT)-based radiomics nomogram for preoperative differentiation of SCC and NHL in the palatine tonsil. METHODS This study enrolled 135 patients with a pathological diagnosis of SCC or NHL from two clinical centers, who were divided into training (n = 94; SCC = 50, NHL = 44) and external validation sets (n = 41; SCC = 22, NHL = 19). A radiomics signature was constructed from radiomics features extracted from routine CECT images and a radiomics score (Rad-score) was calculated. A clinical model was established using demographic features and CT findings. The independent clinical factors and Rad-score were combined to construct a radiomics nomogram. Performance of the clinical model, radiomics signature, and nomogram was assessed using receiver operating characteristics analysis and decision curve analysis. RESULTS Eleven features were finally selected to construct the radiomics signature. The radiomics nomogram incorporating gender, mean CECT value, and radiomics signature showed better predictive value for differentiating SCC from NHL than the clinical model for training (AUC, 0.919 vs. 0.801, p = 0.004) and validation (AUC, 0.876 vs. 0.703, p = 0.029) sets. Decision curve analysis demonstrated that the radiomics nomogram was more clinically useful than the clinical model. CONCLUSIONS A CECT-based radiomics nomogram was constructed incorporating gender, mean CECT value, and radiomics signature. This nomogram showed favorable predictive efficacy for differentiating SCC from NHL in the palatine tonsil, and might be useful for clinical decision-making. KEY POINTS • Differential diagnosis between SCC and NHL in the palatine tonsil is difficult by conventional imaging modalities. • A radiomics nomogram integrated with the radiomics signature, gender, and mean contrast-enhanced CT value facilitates differentiation of SCC from NHL with improved diagnostic efficacy.
Collapse
Affiliation(s)
- Cheng Dong
- Department of Radiology, The Affiliated Hospital of Qingdao University, NO. 16, Jiangsu Road, Qingdao, 266000, China
| | - Ying-Mei Zheng
- Health Management Center, The Affiliated Hospital of Qingdao University, NO. 16, Jiangsu Road, Qingdao, 266000, China
| | - Jian Li
- Department of Radiology, The University of Hong Kong - Shenzhen Hospital, 1, Haiyuan Road, Futian District, Shenzhen, 518000, NO, China
| | - Zeng-Jie Wu
- Department of Radiology, The Affiliated Hospital of Qingdao University, NO. 16, Jiangsu Road, Qingdao, 266000, China
| | - Zhi-Tao Yang
- Department of Radiology, The Affiliated Hospital of Qingdao University, NO. 16, Jiangsu Road, Qingdao, 266000, China
| | - Xiao-Li Li
- Department of Radiology, The Affiliated Hospital of Qingdao University, NO. 16, Jiangsu Road, Qingdao, 266000, China
| | - Wen-Jian Xu
- Department of Radiology, The Affiliated Hospital of Qingdao University, NO. 16, Jiangsu Road, Qingdao, 266000, China
| | - Da-Peng Hao
- Department of Radiology, The Affiliated Hospital of Qingdao University, NO. 16, Jiangsu Road, Qingdao, 266000, China.
| |
Collapse
|
21
|
Zheng YM, Zhan JF, Yuan MG, Hou F, Jiang G, Wu ZJ, Dong C. A CT-based radiomics signature for preoperative discrimination between high and low expression of programmed death ligand 1 in head and neck squamous cell carcinoma. Eur J Radiol 2022; 146:110093. [PMID: 34890937 DOI: 10.1016/j.ejrad.2021.110093] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/19/2021] [Accepted: 11/30/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE Accurate prediction of the expression level of programmed death ligand 1 (PD-L1) in head and neck squamous cell carcinoma (HNSCC) is crucial before immunotherapy. The purpose of this study was to construct and validate a contrast-enhanced computed tomography (CECT)-based radiomics signature to discriminate between high and low expression status of PD-L1. METHODS A total of 179 HNSCC patients who underwent immunohistochemical examination of tumor PD-L1 expression at one of two centers were enrolled in this study and divided into a training set (n = 122; 55 high PD-L1 expression and 67 low PD-L1 expression) and an external validation set (n = 57; 26 high PD-L1 expression and 31 low PD-L1 expression). The least absolute shrinkage and selection operator method was used to select the key features for a CECT-image-based radiomics signature. The performance of the radiomics signature was assessed using receiver operating characteristics analysis. RESULTS Six features were finally selected to construct the radiomics signature. The performance of the radiomics signature in the discrimination between high and low PD-L1 expression status was good in both the training and validation sets, with areas under the receiver operating characteristics curve of 0.889 and 0.834 for the training and validation sets, respectively. CONCLUSIONS The constructed CECT-based radiomics signature model showed favorable performance for discriminating between high and low PD-L1 expression status in HNSCC patients. It may be useful for screening out those patients with HNSCC who can best benefit from anti-PD-L1 immunotherapy.
Collapse
Affiliation(s)
- Ying-Mei Zheng
- Health Management Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jin-Feng Zhan
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ming-Gang Yuan
- Department of Nuclear Medicine, Affiliated Qingdao Central Hospital, Qingdao University, Qingdao, China
| | - Feng Hou
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Gang Jiang
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zeng-Jie Wu
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Cheng Dong
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
22
|
Li Z, Liu Z, Guo Y, Wang S, Qu X, Li Y, Pan Y, Zhang L, Su D, Yang Q, Tao X, Yue Q, Xian J. Dual-energy CT-based radiomics nomogram in predicting histological differentiation of head and neck squamous carcinoma: a multicenter study. Neuroradiology 2021; 64:361-369. [PMID: 34860278 DOI: 10.1007/s00234-021-02860-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/09/2021] [Indexed: 02/05/2023]
Abstract
PURPOSE To develop and validate a dual-energy CT (DECT)-based radiomics nomogram from multicenter trials for predicting the histological differentiation of head and neck squamous cell carcinoma (HNSCC). METHODS A total of 178 patients (112 in the training and 66 in the validation cohorts) from eight institutions with histologically proven HNSCCs were included in this retrospective study. Radiomics-signature models were constructed from features extracted from virtual monoenergetic images (VMI) and iodine-based material decomposition images (IMDI), reconstructed from venous-phase DECT images. Clinical factors were also assessed to build a clinical model. Multivariate logistic regression analysis was used to develop a nomogram combining the radiomics signature models and clinical model for predicting poorly differentiated HNSCC and moderately well-differentiated HNSCC. The predictive performance of the clinical model, radiomics signature models, and nomogram was compared. The calibration degree of the nomogram was also assessed. RESULTS The tumor location, VMI-signature, and IMDI-signature were associated with the degree of HNSCC differentiation, and areas under the ROC curves (AUCs) were 0.729, 0.890, and 0.833 in the training cohort and 0.627, 0.859, and 0.843 in the validation cohort, respectively. The nomogram incorporating tumor location and two radiomics-signature models yielded the best performance in training (AUC = 0.987) and validation (AUC = 0.968) cohorts with a good calibration degree. CONCLUSION The nomogram that integrated the DECT-based radiomics-signature models and tumor location showed good performance in predicting histological differentiation degree of HNSCC, providing a novel combination for predicting HNSCC differentiation.
Collapse
Affiliation(s)
- Zheng Li
- Department of Radiology, Beijing Tongren Hospital, Capital Medical University, No. 1, DongJiaoMinXiang Street, Dongcheng District, Beijing, 100730, China
| | - Zhaohui Liu
- Department of Radiology, Beijing Tongren Hospital, Capital Medical University, No. 1, DongJiaoMinXiang Street, Dongcheng District, Beijing, 100730, China
| | - Yan Guo
- Pharmaceutical Diagnostics, Precision Health Institute, GE Healthcare China, Beijing, 100176, China
| | - Sicong Wang
- Pharmaceutical Diagnostics, Precision Health Institute, GE Healthcare China, Beijing, 100176, China
| | - Xiaoxia Qu
- Department of Radiology, Beijing Tongren Hospital, Capital Medical University, No. 1, DongJiaoMinXiang Street, Dongcheng District, Beijing, 100730, China
| | - Yajun Li
- Department of Radiology, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Yucheng Pan
- Department of Radiology, Eye Ear Nose and Throat Hospital of Fudan University, Shanghai, 200031, China
| | - Longjiang Zhang
- Department of Diagnostic Radiology, General Hospital of Eastern Theater Command/Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Danke Su
- Imaging Center, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Qian Yang
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China
| | - Xiaofeng Tao
- Department of Radiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Qiang Yue
- Department of Radiology, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Junfang Xian
- Department of Radiology, Beijing Tongren Hospital, Capital Medical University, No. 1, DongJiaoMinXiang Street, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
23
|
Bao H, Chen T, Zhu J, Xie H, Chen F. CEUS-Based Radiomics Can Show Changes in Protein Levels in Liver Metastases After Incomplete Thermal Ablation. Front Oncol 2021; 11:694102. [PMID: 34513676 PMCID: PMC8427517 DOI: 10.3389/fonc.2021.694102] [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: 04/12/2021] [Accepted: 08/03/2021] [Indexed: 01/04/2023] Open
Abstract
Objective To investigate the ability of contrast-enhanced ultrasound (CEUS)-based radiomics combined with machine learning to detect early protein changes after incomplete thermal ablation. Methods HCT-26 colorectal adenoma cells were engrafted into the livers of 80 mice, which were randomly divided into 4 groups for palliative laser ablation. Changes in heat shock protein (HSP) and apoptosis-related protein expression in the tumors were assessed. SCID mice subjected to CEUS and ultrasonography were divided into training (n=56) and test (n=24) datasets. Then, 102 features from seven feature groups were extracted. We use the least absolute shrinkage and selection operator (LASSO) feature selection method to fit the machine learning classifiers. The feature selection methods and four classifiers were combined to determine the best prediction model. Results The areas under the receiver-operating characteristic curves (AUCs) of the classifiers in the test dataset ranged from 0.450 to 0.932 (median: 0.721). The best score was obtained from the model in which the omics data of CEUS was analyzed in the arterial phase by random forest (RF) classification. Conclusions A machine learning model, in which radiomics characteristics are extracted by multimodal ultrasonography, can accurately, rapidly and noninvasively identify protein changes after ablation.
Collapse
Affiliation(s)
- Haiwei Bao
- Department of Ultrasound, The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ting Chen
- Department of Ultrasound, The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Junyan Zhu
- Department of Ultrasound, The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Haiyang Xie
- Key Laboratory of Combined Multi-organ Transplantation, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Fen Chen
- Department of Ultrasound, The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
24
|
Liu X, Maleki F, Muthukrishnan N, Ovens K, Huang SH, Pérez-Lara A, Romero-Sanchez G, Bhatnagar SR, Chatterjee A, Pusztaszeri MP, Spatz A, Batist G, Payabvash S, Haider SP, Mahajan A, Reinhold C, Forghani B, O’Sullivan B, Yu E, Forghani R. Site-Specific Variation in Radiomic Features of Head and Neck Squamous Cell Carcinoma and Its Impact on Machine Learning Models. Cancers (Basel) 2021; 13:cancers13153723. [PMID: 34359623 PMCID: PMC8345201 DOI: 10.3390/cancers13153723] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 07/07/2021] [Accepted: 07/20/2021] [Indexed: 12/17/2022] Open
Abstract
Simple Summary Head and neck squamous cell carcinoma (HNSCC) is the most common mucosal malignancy of the head and neck and a leading cause of cancer death. HNSCC arises from different primary anatomical locations that are typically combined during radiomic analyses assuming that the radiomic features, i.e., quantitative image-based features, are similar based on histopathologic characteristics. However, whether these quantitative features are comparable across tumor sites remains unknown. The aim of our retrospective study was to assess if systematic differences exist between radiomic features based on different tumor sites in HNSCC and how they might affect machine learning model performance in endpoint prediction. Using a population of 605 HNSCC patients, we observed significant differences in radiomic features of tumors from different locations and showed that these differences can impact machine learning model performance. This suggests that tumor site should be considered when developing and evaluating radiomics-based models. Abstract Current radiomic studies of head and neck squamous cell carcinomas (HNSCC) are typically based on datasets combining tumors from different locations, assuming that the radiomic features are similar based on histopathologic characteristics. However, molecular pathogenesis and treatment in HNSCC substantially vary across different tumor sites. It is not known if a statistical difference exists between radiomic features from different tumor sites and how they affect machine learning model performance in endpoint prediction. To answer these questions, we extracted radiomic features from contrast-enhanced neck computed tomography scans (CTs) of 605 patients with HNSCC originating from the oral cavity, oropharynx, and hypopharynx/larynx. The difference in radiomic features of tumors from these sites was assessed using statistical analyses and Random Forest classifiers on the radiomic features with 10-fold cross-validation to predict tumor sites, nodal metastasis, and HPV status. We found statistically significant differences (p-value ≤ 0.05) between the radiomic features of HNSCC depending on tumor location. We also observed that differences in quantitative features among HNSCC from different locations impact the performance of machine learning models. This suggests that radiomic features may reveal biologic heterogeneity complementary to current gold standard histopathologic evaluation. We recommend considering tumor site in radiomic studies of HNSCC.
Collapse
Affiliation(s)
- Xiaoyang Liu
- Princess Margaret Hospital, University of Toronto, University Health Network, Toronto, ON M5G 2C1, Canada; (X.L.); (S.H.H.); (B.O.)
- Department of Radiology, Brigham and Women’s Hospital, Harvard University, Cambridge, MA 02115, USA
- Department of Medical Imaging, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Farhad Maleki
- Augmented Intelligence & Precision Health Laboratory (AIPHL), Department of Radiology and the Research Institute of the McGill University Health Centre, McGill University, Montreal, QC H4A 3J1, Canada; (F.M.); (N.M.); (K.O.); (S.R.B.); (C.R.); (B.F.)
| | - Nikesh Muthukrishnan
- Augmented Intelligence & Precision Health Laboratory (AIPHL), Department of Radiology and the Research Institute of the McGill University Health Centre, McGill University, Montreal, QC H4A 3J1, Canada; (F.M.); (N.M.); (K.O.); (S.R.B.); (C.R.); (B.F.)
| | - Katie Ovens
- Augmented Intelligence & Precision Health Laboratory (AIPHL), Department of Radiology and the Research Institute of the McGill University Health Centre, McGill University, Montreal, QC H4A 3J1, Canada; (F.M.); (N.M.); (K.O.); (S.R.B.); (C.R.); (B.F.)
| | - Shao Hui Huang
- Princess Margaret Hospital, University of Toronto, University Health Network, Toronto, ON M5G 2C1, Canada; (X.L.); (S.H.H.); (B.O.)
- Princess Margaret Cancer Centre, Department of Radiation Oncology, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Almudena Pérez-Lara
- Segal Cancer Centre & Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada; (A.P.-L.); (G.R.-S.); (G.B.)
| | - Griselda Romero-Sanchez
- Segal Cancer Centre & Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada; (A.P.-L.); (G.R.-S.); (G.B.)
| | - Sahir Rai Bhatnagar
- Augmented Intelligence & Precision Health Laboratory (AIPHL), Department of Radiology and the Research Institute of the McGill University Health Centre, McGill University, Montreal, QC H4A 3J1, Canada; (F.M.); (N.M.); (K.O.); (S.R.B.); (C.R.); (B.F.)
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC H3A 1A2, Canada
| | | | | | - Alan Spatz
- Division of Pathology, Jewish General Hospital, Montreal, QC H3Y 1E2, Canada; (M.P.P.); (A.S.)
| | - Gerald Batist
- Segal Cancer Centre & Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada; (A.P.-L.); (G.R.-S.); (G.B.)
| | - Seyedmehdi Payabvash
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT 06520, USA; (S.P.); (S.P.H.); (A.M.)
| | - Stefan P. Haider
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT 06520, USA; (S.P.); (S.P.H.); (A.M.)
| | - Amit Mahajan
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT 06520, USA; (S.P.); (S.P.H.); (A.M.)
| | - Caroline Reinhold
- Augmented Intelligence & Precision Health Laboratory (AIPHL), Department of Radiology and the Research Institute of the McGill University Health Centre, McGill University, Montreal, QC H4A 3J1, Canada; (F.M.); (N.M.); (K.O.); (S.R.B.); (C.R.); (B.F.)
| | - Behzad Forghani
- Augmented Intelligence & Precision Health Laboratory (AIPHL), Department of Radiology and the Research Institute of the McGill University Health Centre, McGill University, Montreal, QC H4A 3J1, Canada; (F.M.); (N.M.); (K.O.); (S.R.B.); (C.R.); (B.F.)
- Segal Cancer Centre & Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada; (A.P.-L.); (G.R.-S.); (G.B.)
| | - Brian O’Sullivan
- Princess Margaret Hospital, University of Toronto, University Health Network, Toronto, ON M5G 2C1, Canada; (X.L.); (S.H.H.); (B.O.)
- Princess Margaret Cancer Centre, Department of Radiation Oncology, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Eugene Yu
- Princess Margaret Hospital, University of Toronto, University Health Network, Toronto, ON M5G 2C1, Canada; (X.L.); (S.H.H.); (B.O.)
- Department of Medical Imaging, University of Toronto, Toronto, ON M5S 1A1, Canada
- Correspondence: (E.Y.); (R.F.)
| | - Reza Forghani
- Augmented Intelligence & Precision Health Laboratory (AIPHL), Department of Radiology and the Research Institute of the McGill University Health Centre, McGill University, Montreal, QC H4A 3J1, Canada; (F.M.); (N.M.); (K.O.); (S.R.B.); (C.R.); (B.F.)
- Segal Cancer Centre & Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada; (A.P.-L.); (G.R.-S.); (G.B.)
- Correspondence: (E.Y.); (R.F.)
| |
Collapse
|
25
|
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
|
26
|
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
|
27
|
Li Q, Dong F, Jiang B, Zhang M. Exploring MRI Characteristics of Brain Diffuse Midline Gliomas With the H3 K27M Mutation Using Radiomics. Front Oncol 2021; 11:646267. [PMID: 34109112 PMCID: PMC8182051 DOI: 10.3389/fonc.2021.646267] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 04/26/2021] [Indexed: 01/01/2023] Open
Abstract
Objectives To explore the magnetic resonance imaging (MRI) characteristics of brain diffuse midline gliomas with the H3 K27M mutation (DMG-M) using radiomics. Materials and Methods Thirty patients with diffuse midline gliomas, including 16 with the H3 K27M mutant and 14 with wild type tumors, were retrospectively included in this study. A total of 272 radiomic features were initially extracted from MR images of each tumor. Principal component analysis, univariate analysis, and three other feature selection methods, including variance thresholding, recursive feature elimination, and the elastic net, were used to analyze the radiomic features. Based on the results, related visually accessible features of the tumors were further evaluated. Results Patients with DMG-M were younger than those with diffuse midline gliomas with H3 K27M wild (DMG-W) (median, 25.5 and 48 years old, respectively; p=0.005). Principal component analysis showed that there were obvious overlaps in the first two principal components for both DMG-M and DMG-W tumors. The feature selection results showed that few features from T2-weighted images (T2WI) were useful for differentiating DMG-M and DMG-W tumors. Thereafter, four visually accessible features related to T2WI were further extracted and analyzed. Among these features, only cystic formation showed a significant difference between the two types of tumors (OR=7.800, 95% CI 1.476-41.214, p=0.024). Conclusions DMGs with and without the H3 K27M mutation shared similar MRI characteristics. T2W sequences may be valuable, and cystic formation a useful MRI biomarker, for diagnosing brain DMG-M.
Collapse
Affiliation(s)
- Qian Li
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fei Dong
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Biao Jiang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Minming Zhang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
28
|
Khodrog OA, Cui F, Xu N, Han Q, Liu J, Gong T, Yuan Q. Prediction of squamous cell carcinoma cases from squamous cell hyperplasia in throat lesions using CT radiomics model. Saudi Med J 2021; 42:284-292. [PMID: 33632907 PMCID: PMC7989270 DOI: 10.15537/smj.2021.42.3.20200617] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 02/01/2021] [Indexed: 12/23/2022] Open
Abstract
Objectives: To differentiate squamous cell hyperplasia (SCH) (benign) from squamous cell carcinoma (SCC) malignant) using textural features extracted from CT images and thereby, facilitate the preoperative medical diagnosis and treatment of throat cancers without the need for sample biopsies. Methods: In total, 100 throat cancer patients were selected for this retrospective study. The cases were collected from the Second Hospital of Jilin University, Changchun, China, from June 2017 to January 2019. The patients were separated into a training and validation cohort consisting of 70 and 30 cases, respectively. The Artificial Intelligence Kit software (A.K. software) was used to extract the radiomics features from the CT images. These features were further processed using the minimum redundancy maximum relevance (mRMR) and least absolute shrinkage and selection operator (LASSO) methods to obtain a subset of optimal features. The radiomics model was validated based on area-under-the-curve (AUC) values, accuracy, specificity, and sensitivity using the R-studio software. Results: The diagnostic accuracy, specificity, PPV, NPV, and AUC values obtained for the training cohort was 0.91, 0.9, 0.93, 0.9, and 0.96 CT angiography (CTA), 0.93, 0.93, 0.95, 0.90, and 0.96 computed tomography normal (CTN), and 0.92, 0.87, 0.91, 0.96, and 0.96 CT venogram (CTV). These values were subsequently confirmed in the validation cohort. Conclusion: The radiomics-based prediction model proposed in this study successfully differentiated between SCH and SCC throat cancers using CT imaging, thereby facilitating the development of accurate preoperative diagnosis based on specific biomarkers and cancer phenotypes.
Collapse
Affiliation(s)
- Osama A. Khodrog
- From the Department of Radiology (Khodrog, Cui, Xu, Han, Liu, Gong, Yuan), the Second Hospital of Jilin University, Changchun, China and from the Department of Medical Imaging (Khodrog), Faculty of Applied Medical Health, Palestine Ahliya University, Bethlehem, Palestine.
| | - Fengzhi Cui
- From the Department of Radiology (Khodrog, Cui, Xu, Han, Liu, Gong, Yuan), the Second Hospital of Jilin University, Changchun, China and from the Department of Medical Imaging (Khodrog), Faculty of Applied Medical Health, Palestine Ahliya University, Bethlehem, Palestine.
| | - Nannan Xu
- From the Department of Radiology (Khodrog, Cui, Xu, Han, Liu, Gong, Yuan), the Second Hospital of Jilin University, Changchun, China and from the Department of Medical Imaging (Khodrog), Faculty of Applied Medical Health, Palestine Ahliya University, Bethlehem, Palestine.
| | - Qinghe Han
- From the Department of Radiology (Khodrog, Cui, Xu, Han, Liu, Gong, Yuan), the Second Hospital of Jilin University, Changchun, China and from the Department of Medical Imaging (Khodrog), Faculty of Applied Medical Health, Palestine Ahliya University, Bethlehem, Palestine.
| | - Jianhua Liu
- From the Department of Radiology (Khodrog, Cui, Xu, Han, Liu, Gong, Yuan), the Second Hospital of Jilin University, Changchun, China and from the Department of Medical Imaging (Khodrog), Faculty of Applied Medical Health, Palestine Ahliya University, Bethlehem, Palestine.
| | - Tingting Gong
- From the Department of Radiology (Khodrog, Cui, Xu, Han, Liu, Gong, Yuan), the Second Hospital of Jilin University, Changchun, China and from the Department of Medical Imaging (Khodrog), Faculty of Applied Medical Health, Palestine Ahliya University, Bethlehem, Palestine.
| | - Qinghai Yuan
- From the Department of Radiology (Khodrog, Cui, Xu, Han, Liu, Gong, Yuan), the Second Hospital of Jilin University, Changchun, China and from the Department of Medical Imaging (Khodrog), Faculty of Applied Medical Health, Palestine Ahliya University, Bethlehem, Palestine.
- Address correspondence and reprint request to: Dr. Qinghai Yuan, Department of Radiology, Norman Bethune College of Medicine, The Second Hospital of Jilin University, Changchun, China. E-mail: ORCID ID: http://orcid.org/0000-0002-5337-5354
| |
Collapse
|
29
|
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
|
30
|
Song LL, Chen SJ, Chen W, Shi Z, Wang XD, Song LN, Chen DS. Radiomic model for differentiating parotid pleomorphic adenoma from parotid adenolymphoma based on MRI images. BMC Med Imaging 2021; 21:54. [PMID: 33743615 PMCID: PMC7981906 DOI: 10.1186/s12880-021-00581-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 03/07/2021] [Indexed: 01/04/2023] Open
Abstract
Background Distinguishing parotid pleomorphic adenoma (PPA) from parotid adenolymphoma (PA) is important for precision treatment, but there is a lack of readily available diagnostic methods. In this study, we aimed to explore the diagnostic value of radiomic signatures based on magnetic resonance imaging (MRI) for PPA and PA. Methods The clinical characteristic and imaging data were retrospectively collected from 252 cases (126 cases in the training cohort and 76 patients in the validation cohort) in this study. Radiomic features were extracted from MRI scans, including T1-weighted imaging (T1WI) sequences and T2-weighted imaging (T2WI) sequences. The radiomic features from three sequences (T1WI, T2WI and T1WI combined with T2WI) were selected using univariate analysis, LASSO correlation and Spearman correlation. Then, we built six quantitative radiomic models using the selected features through two machine learning methods (multivariable logistic regression, MLR, and support vector machine, SVM). The performances of the six radiomic models were assessed and the diagnostic efficacies of the ideal T1-2WI radiomic model and the clinical model were compared. Results The T1-2WI radiomic model using MLR showed optimal discriminatory ability (accuracy = 0.87 and 0.86, F-1 score = 0.88 and 0.86, sensitivity = 0.90 and 0.88, specificity = 0.82 and 0.80, positive predictive value = 0.86 and 0.84, negative predictive value = 0.86 and 0.84 in the training and validation cohorts, respectively) and its calibration was observed to be good (p > 0.05). The area under the curve (AUC) of the T1-2WI radiomic model was significantly better than that of the clinical model for both the training (0.95 vs. 0.67, p < 0.001) and validation (0.90 vs. 0.68, p = 0.001) cohorts. Conclusions The T1-2WI radiomic model in our study is complementary to the current knowledge of differential diagnosis for PPA and PA. Supplementary Information The online version contains supplementary material available at 10.1186/s12880-021-00581-9.
Collapse
Affiliation(s)
- Le-le Song
- The Department of Radiology, the First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan, China
| | - Shun-Jun Chen
- The Department of Ultrasound, the First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan, China
| | - Wang Chen
- The Department of Radiology, the First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan, China
| | - Zhan Shi
- The Department of Radiology, the First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan, China
| | - Xiao-Dong Wang
- The Department of Radiology, the First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan, China
| | - Li-Na Song
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dian-Sen Chen
- The Department of Radiology, the First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan, China.
| |
Collapse
|
31
|
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
|
32
|
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
|
33
|
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
|
34
|
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]
|
35
|
Kalendralis P, Shi Z, Traverso A, Choudhury A, Sloep M, Zhovannik I, Starmans MPA, Grittner D, Feltens P, Monshouwer R, Klein S, Fijten R, Aerts H, Dekker A, van Soest J, Wee L. FAIR-compliant clinical, radiomics and DICOM metadata of RIDER, interobserver, Lung1 and head-Neck1 TCIA collections. Med Phys 2020; 47:5931-5940. [PMID: 32521049 PMCID: PMC7754296 DOI: 10.1002/mp.14322] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/19/2020] [Accepted: 06/02/2020] [Indexed: 12/17/2022] Open
Abstract
Purpose One of the most frequently cited radiomics investigations showed that features automatically extracted from routine clinical images could be used in prognostic modeling. These images have been made publicly accessible via The Cancer Imaging Archive (TCIA). There have been numerous requests for additional explanatory metadata on the following datasets — RIDER, Interobserver, Lung1, and Head–Neck1. To support repeatability, reproducibility, generalizability, and transparency in radiomics research, we publish the subjects’ clinical data, extracted radiomics features, and digital imaging and communications in medicine (DICOM) headers of these four datasets with descriptive metadata, in order to be more compliant with findable, accessible, interoperable, and reusable (FAIR) data management principles. Acquisition and validation methods Overall survival time intervals were updated using a national citizens registry after internal ethics board approval. Spatial offsets of the primary gross tumor volume (GTV) regions of interest (ROIs) associated with the Lung1 CT series were improved on the TCIA. GTV radiomics features were extracted using the open‐source Ontology‐Guided Radiomics Analysis Workflow (O‐RAW). We reshaped the output of O‐RAW to map features and extraction settings to the latest version of Radiomics Ontology, so as to be consistent with the Image Biomarker Standardization Initiative (IBSI). Digital imaging and communications in medicine metadata was extracted using a research version of Semantic DICOM (SOHARD, GmbH, Fuerth; Germany). Subjects’ clinical data were described with metadata using the Radiation Oncology Ontology. All of the above were published in Resource Descriptor Format (RDF), that is, triples. Example SPARQL queries are shared with the reader to use on the online triples archive, which are intended to illustrate how to exploit this data submission. Data format The accumulated RDF data are publicly accessible through a SPARQL endpoint where the triples are archived. The endpoint is remotely queried through a graph database web application at http://sparql.cancerdata.org. SPARQL queries are intrinsically federated, such that we can efficiently cross‐reference clinical, DICOM, and radiomics data within a single query, while being agnostic to the original data format and coding system. The federated queries work in the same way even if the RDF data were partitioned across multiple servers and dispersed physical locations. Potential applications The public availability of these data resources is intended to support radiomics features replication, repeatability, and reproducibility studies by the academic community. The example SPARQL queries may be freely used and modified by readers depending on their research question. Data interoperability and reusability are supported by referencing existing public ontologies. The RDF data are readily findable and accessible through the aforementioned link. Scripts used to create the RDF are made available at a code repository linked to this submission: https://gitlab.com/UM‐CDS/FAIR‐compliant_clinical_radiomics_and_DICOM_metadata.
Collapse
Affiliation(s)
- Petros Kalendralis
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, Maastricht, 6229 ET, The Netherlands
| | - Zhenwei Shi
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, Maastricht, 6229 ET, The Netherlands
| | - Alberto Traverso
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, Maastricht, 6229 ET, The Netherlands
| | - Ananya Choudhury
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, Maastricht, 6229 ET, The Netherlands
| | - Matthijs Sloep
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, Maastricht, 6229 ET, The Netherlands
| | - Ivan Zhovannik
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, Maastricht, 6229 ET, The Netherlands.,Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, 6525 GC, The Netherlands
| | - Martijn P A Starmans
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, 3015 GD, The Netherlands.,Department of Medical Informatics, Erasmus Medical Center, Rotterdam, 3015 GD, The Netherlands
| | | | | | - Rene Monshouwer
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, 6525 GC, The Netherlands
| | - Stefan Klein
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, 3015 GD, The Netherlands.,Department of Medical Informatics, Erasmus Medical Center, Rotterdam, 3015 GD, The Netherlands
| | - Rianne Fijten
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, Maastricht, 6229 ET, The Netherlands
| | - Hugo Aerts
- Artificial Intelligence in Medicine (AIM) Program, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, United States.,Radiology and Nuclear Medicine, CARIM & GROW School for Oncology, Maastricht University, Maastricht, 6211 LK, The Netherlands
| | - Andre Dekker
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, Maastricht, 6229 ET, The Netherlands
| | - Johan van Soest
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, Maastricht, 6229 ET, The Netherlands
| | - Leonard Wee
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Centre+, Maastricht, 6229 ET, The Netherlands
| |
Collapse
|