1
|
Wang Y, Yu Y, Gu L, Sun Y, Yan J, Zhang H, Zhang Y. Radiomics feature is a risk factor for locally advanced cervical cancer treated using concurrent chemoradiotherapy based on magnetic resonance imaging: a retrospective study. BMC Cancer 2025; 25:230. [PMID: 39930343 PMCID: PMC11809009 DOI: 10.1186/s12885-025-13625-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 02/03/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Although concurrent chemoradiotherapy (CCRT) is the standard treatment strategy for locally advanced cervical squamous carcinoma (LACSC), there are still individual differences. It is of vital importance to establish a radiomics-based model for predicting overall survival (OS) of LACSC patients treated using CCRT, and evaluating the feasibility of adjuvant chemotherapy (ACT). METHODS 122 LACSC patients were retrospectively analyzed who underwent pelvic MRI before standard CCRT between January 2013 and September 2016, including 85 patients in training set and 37 patients in testing set. 3D Slicer was used to segment images and extract features. IPMs software was used to select features and construct radscore. We selected the group with the largest area under the curves as the best result from 150 feature subsets and corresponding radscore. A nomogram was established using univariate and multivariate Cox analyses. We used Shapley Additive Explanations (SHAP) for further interpretation of the nomogram. Kaplan-Meier curves demonstrated the associations of radscore and clinical characteristics with OS and ACT. RESULTS Radscore was a prognostic factor (P = 0.001) which constructed using 10 radiomics features influencing the OS of patients with LACSC treated using CCRT. The radiomics-clinical model estimated OS (training, C-index: 0.761; testing, C-index: 0.718) more accurately than the clinical (training, C-index: 0.745; testing, C-index: 0.708) and radiomics models (training, C-index: 0.702; testing, C-index: 0.671). Radscore has the greatest impact on the prognosis of LACSC patients. We combined radscore and clinical factors to obtain risk scores. There was a better OS rate among low-risk patients than among high-risk patients (training, P = 0.034; testing, P = 0.003). Compared with CCRT, ACT + CCRT did not improve prognosis (high-risk patients, P = 0.703; all patients, P = 0.425). CONCLUSIONS Radscore independently predicted OS in LACSC. The radiomics-clinical nomogram improved individualized OS estimation. Patients did not benefit from ACT.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150081, Heilongjiang Province, China
| | - Yanyan Yu
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Lina Gu
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150081, Heilongjiang Province, China
| | - Yunfeng Sun
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jiazhuo Yan
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150081, Heilongjiang Province, China
| | - Hongxia Zhang
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yunyan Zhang
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150081, Heilongjiang Province, China.
| |
Collapse
|
2
|
Li H, Dong D, Fang M, He B, Liu S, Hu C, Liu Z, Wang H, Tang L, Tian J. ContraSurv: Enhancing Prognostic Assessment of Medical Images via Data-Efficient Weakly Supervised Contrastive Learning. IEEE J Biomed Health Inform 2025; 29:1232-1242. [PMID: 39437290 DOI: 10.1109/jbhi.2024.3484991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Prognostic assessment remains a critical challenge in medical research, often limited by the lack of well-labeled data. In this work, we introduce ContraSurv, a weakly-supervised learning framework based on contrastive learning, designed to enhance prognostic predictions in 3D medical images. ContraSurv utilizes both the self-supervised information inherent in unlabeled data and the weakly-supervised cues present in censored data, refining its capacity to extract prognostic representations. For this purpose, we establish a Vision Transformer architecture optimized for our medical image datasets and introduce novel methodologies for both self-supervised and supervised contrastive learning for prognostic assessment. Additionally, we propose a specialized supervised contrastive loss function and introduce SurvMix, a novel data augmentation technique for survival analysis. Evaluations were conducted across three cancer types and two imaging modalities on three real-world datasets. The results confirmed the enhanced performance of ContraSurv over competing methods, particularly in data with a high censoring rate.
Collapse
|
3
|
Bruixola G, Dualde-Beltrán D, Jimenez-Pastor A, Nogué A, Bellvís F, Fuster-Matanzo A, Alfaro-Cervelló C, Grimalt N, Salhab-Ibáñez N, Escorihuela V, Iglesias ME, Maroñas M, Alberich-Bayarri Á, Cervantes A, Tarazona N. CT-based clinical-radiomics model to predict progression and drive clinical applicability in locally advanced head and neck cancer. Eur Radiol 2024:10.1007/s00330-024-11301-6. [PMID: 39706922 DOI: 10.1007/s00330-024-11301-6] [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: 06/30/2024] [Revised: 10/19/2024] [Accepted: 11/17/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND Definitive chemoradiation is the primary treatment for locally advanced head and neck carcinoma (LAHNSCC). Optimising outcome predictions requires validated biomarkers, since TNM8 and HPV could have limitations. Radiomics may enhance risk stratification. METHODS This single-centre observational study collected clinical data and baseline CT scans from 171 LAHNSCC patients treated with chemoradiation. The dataset was divided into training (80%) and test (20%) sets, with a 5-fold cross-validation on the training set. Researchers extracted 108 radiomics features from each primary tumour and applied survival analysis and classification models to predict progression-free survival (PFS) and 5-year progression, respectively. Performance was evaluated using inverse probability of censoring weights and c-index for the PFS model and AUC, sensitivity, specificity, and accuracy for the 5-year progression model. Feature importance was measured by the SHapley Additive exPlanations (SHAP) method and patient stratification was assessed through Kaplan-Meier curves. RESULTS The final dataset included 171 LAHNSCC patients, with 53% experiencing disease progression at 5 years. The random survival forest model best predicted PFS, with an AUC of 0.64 and CI of 0.66 on the test set, highlighting 4 radiomics features and TNM8 as significant contributors. It successfully stratified patients into low and high-risk groups (log-rank p < 0.005). The extreme gradient boosting model most effectively predicted a 5-year progression, incorporating 12 radiomics features and four clinical variables, achieving an AUC of 0.74, sensitivity of 0.53, specificity of 0.81, and accuracy of 0.66 on the test set. CONCLUSION The combined clinical-radiomics model improved the standard TNM8 and clinical variables in predicting 5-year progression though further validation is necessary. KEY POINTS Question There is an unmet need for non-invasive biomarkers to guide treatment in locally advanced head and neck cancer. Findings Clinical data (TNM8 staging, primary tumour site, age, and smoking) plus radiomics improved 5-year progression prediction compared with the clinical comprehensive model or TNM staging alone. Clinical relevance SHAP simplifies complex machine learning radiomics models for clinicians by using easy-to-understand graphical representations, promoting explainability.
Collapse
Affiliation(s)
- Gema Bruixola
- Medical Oncology Department, Hospital Clinico Universitario de Valencia-INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - Delfina Dualde-Beltrán
- Radiology Department, Hospital Clinico Universitario de Valencia, University of Valencia, Valencia, Spain
| | | | - Anna Nogué
- Quibim-Quantitative Imaging Biomarkers in Medicine, Valencia, Spain
| | | | | | - Clara Alfaro-Cervelló
- Pathology Department, Hospital Clinico Universitario de Valencia-INCLIVA Instituto de Investigación Sanitaria, University of Valencia, Valencia, Spain
| | - Nuria Grimalt
- Medical Oncology Department, Hospital Clinico Universitario de Valencia-INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - Nader Salhab-Ibáñez
- Radiology Department, Hospital Clinico Universitario de Valencia, University of Valencia, Valencia, Spain
| | - Vicente Escorihuela
- Otorhinolaryngology Department, Hospital Clinico Universitario de Valencia, Valencia, Spain
| | - María Eugenia Iglesias
- Oral and Maxillary Surgery Department, Hospital Clinico Universitario de Valencia, Valencia, Spain
| | - María Maroñas
- Radiation Oncology Department, Hospital Clinico Universitario de Valencia, Valencia, Spain
| | | | - Andrés Cervantes
- Medical Oncology Department, Hospital Clinico Universitario de Valencia-INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain.
- Instituto de Salud Carlos III, CIBERONC, Madrid, Spain.
| | - Noelia Tarazona
- Medical Oncology Department, Hospital Clinico Universitario de Valencia-INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain.
- Instituto de Salud Carlos III, CIBERONC, Madrid, Spain.
| |
Collapse
|
4
|
Wang B, Liu J, Xie J, Zhang X, Wang Z, Cao Z, Wen D, Wan Hasan WZ, Harun Ramli HR, Dong X. Systematic review and meta-analysis of the prognostic value of 18F-Fluorodeoxyglucose ( 18F-FDG) positron emission tomography (PET) and/or computed tomography (CT)-based radiomics in head and neck cancer. Clin Radiol 2024; 79:757-772. [PMID: 38944542 DOI: 10.1016/j.crad.2024.05.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 05/16/2024] [Accepted: 05/24/2024] [Indexed: 07/01/2024]
Abstract
AIM Radiomics involves the extraction of quantitative data from medical images to facilitate the diagnosis, prognosis, and staging of tumors. This study provides a comprehensive overview of the efficacy of radiomics in prognostic applications for head and neck cancer (HNC) in recent years. It undertakes a systematic review of prognostic models specific to HNC and conducts a meta-analysis to evaluate their predictive performance. MATERIALS AND METHODS This study adhered rigorously to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines for literature searches. The literature databases, including PubMed, Embase, Cochrane, and Scopus were systematically searched individually. The methodological quality of the incorporated studies underwent assessment utilizing the radiomics quality score (RQS) tool. A random-effects meta-analysis employing the Harrell concordance index (C-index) was conducted to evaluate the performance of all radiomics models. RESULTS Among the 388 studies retrieved, 24 studies encompassing a total of 6,978 cases were incorporated into the systematic review. Furthermore, eight studies, focusing on overall survival as an endpoint, were included in the meta-analysis. The meta-analysis revealed that the estimated random effect of the C-index for all studies utilizing radiomics alone was 0.77 (0.71-0.82), with a substantial degree of heterogeneity indicated by an I2 of 80.17%. CONCLUSIONS Based on this review, prognostic modeling utilizing radiomics has demonstrated enhanced efficacy for head and neck cancers; however, there remains room for improvement in this approach. In the future, advancements are warranted in the integration of clinical parameters and multimodal features, balancing multicenter data, as well as in feature screening and model construction within this field.
Collapse
Affiliation(s)
- B Wang
- Department of Electrical and Electronic Engineering, Faculty of Engineering, Universiti Putra Malaysia, Serdang, Malaysia; Department of Biomedical Engineering, Chengde Medical University, Chengde City, Hebei Province, China.
| | - J Liu
- Department of Nursing, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia; Department of Nursing, Chengde Central Hospital, Chengde city, Hebei Province, China.
| | - J Xie
- Department of Automatic, Tsinghua University, Beijing, China.
| | - X Zhang
- Department of Biomedical Engineering, Chengde Medical University, Chengde City, Hebei Province, China.
| | - Z Wang
- Department of Biomedical Engineering, Chengde Medical University, Chengde City, Hebei Province, China.
| | - Z Cao
- Department of Radiology, The Affiliated Hospital of Chengde Medical University, Chengde City, Hebei Province, China.
| | - D Wen
- Institute of Artificial Intelligence, University of Science and Technology Beijing, Beijing, China.
| | - W Z Wan Hasan
- Department of Electrical and Electronic Engineering, Faculty of Engineering, Universiti Putra Malaysia, Serdang, Malaysia.
| | - H R Harun Ramli
- Department of Electrical and Electronic Engineering, Faculty of Engineering, Universiti Putra Malaysia, Serdang, Malaysia.
| | - X Dong
- Department of Biomedical Engineering, Chengde Medical University, Chengde City, Hebei Province, China; Hebei Key Laboratory of Nerve Injury and Repair, Chengde Medical University, Chengde City, Hebei, China; Hebei International Research Center of Medical Engineering, Chengde Medical University, Hebei, China.
| |
Collapse
|
5
|
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
|
6
|
Liu Q, Liu S, Mao Y, Kang X, Yu M, Chen G. Machine learning model to preoperatively predict T2/T3 staging of laryngeal and hypopharyngeal cancer based on the CT radiomic signature. Eur Radiol 2024; 34:5349-5359. [PMID: 38206403 DOI: 10.1007/s00330-023-10557-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 11/28/2023] [Accepted: 12/06/2023] [Indexed: 01/12/2024]
Abstract
OBJECTIVES To develop and assess a radiomics-based prediction model for distinguishing T2/T3 staging of laryngeal and hypopharyngeal squamous cell carcinoma (LHSCC) METHODS: A total of 118 patients with pathologically proven LHSCC were enrolled in this retrospective study. We performed feature processing based on 851 radiomic features derived from contrast-enhanced CT images and established multiple radiomic models by combining three feature selection methods and seven machine learning classifiers. The area under the receiver operating characteristic curve (AUC), accuracy, sensitivity, and specificity were used to assess the performance of the models. The radiomic signature obtained from the optimal model and statistically significant morphological image characteristics were incorporated into the predictive nomogram. The performance of the nomogram was assessed by calibration curve and decision curve analysis. RESULTS Using analysis of variance (ANOVA) feature selection and logistic regression (LR) classifier produced the best model. The AUCs of the training, validation, and test sets were 0.919, 0.857, and 0.817, respectively. A nomogram based on the model integrating the radiomic signature and a morphological imaging characteristic (suspicious thyroid cartilage invasion) exhibited C-indexes of 0.899 (95% confidence interval (CI) 0.843-0.955), fitting well in calibration curves (p > 0.05). Decision curve analysis further confirmed the clinical usefulness of the nomogram. CONCLUSIONS The nomogram based on the radiomics model derived from contrast-enhanced CT images had good diagnostic performance for distinguishing T2/T3 staging of LHSCC. CLINICAL RELEVANCE STATEMENT Accurate T2/T3 staging assessment of LHSCC aids in determining whether laryngectomy or laryngeal preservation therapy should be performed. The nomogram based on the radiomics model derived from contrast-enhanced CT images has the potential to predict the T2/T3 staging of LHSCC, which can provide a non-invasive and robust approach for guiding the optimization of clinical decision-making. KEY POINTS • Combining analysis of variance with logistic regression yielded the optimal radiomic model. • A nomogram based on the CT-radiomic signature has good performance for differentiating T2 from T3 staging of laryngeal and hypopharyngeal squamous cell carcinoma. • It provides a non-invasive and robust approach for guiding the optimization of clinical decision-making.
Collapse
Affiliation(s)
- Qianhan Liu
- Department of Radiology, The Affiliated Hospital of Southwest Medical University, No. 23 Tai Ping Street, Luzhou, 646000, Sichuan, China
| | - Shengdan Liu
- Department of Radiology, The Affiliated Hospital of Southwest Medical University, No. 23 Tai Ping Street, Luzhou, 646000, Sichuan, China
| | - Yu Mao
- Department of Radiology, The Affiliated Hospital of Southwest Medical University, No. 23 Tai Ping Street, Luzhou, 646000, Sichuan, China
| | - Xuefeng Kang
- Department of Radiology, The Affiliated Hospital of Southwest Medical University, No. 23 Tai Ping Street, Luzhou, 646000, Sichuan, China
| | - Mingling Yu
- Department of Radiology, The Affiliated Hospital of Southwest Medical University, No. 23 Tai Ping Street, Luzhou, 646000, Sichuan, China
| | - Guangxiang Chen
- Department of Radiology, The Affiliated Hospital of Southwest Medical University, No. 23 Tai Ping Street, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
7
|
Li H, Fang M, He B, Dong D, Tian J. Enhancing Prognostic Prediction of Gastrointestinal Stromal Tumors Using Semi-Supervised Regression Based on CT Imaging Data. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2024; 2024:1-4. [PMID: 40039356 DOI: 10.1109/embc53108.2024.10782508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
The construction of prognostic prediction models based on follow-up data is crucial for devising individualized treatment plans for patients. However, the performance of current supervised survival analysis methods is constrained due to the prevalence of weakly supervised censored samples during follow-up. To address this limitation, this study introduces the Prognostic Co-Training Regression (PCTR) algorithm, a semi-supervised prognostic prediction model developed through the co-training of two KNN regressors. By integrating the prior information of censored data, PCTR harnesses the prior information embedded in censored data, effectively extracting latent prognostic insights, thereby constructing machine learning models with enhanced prognostic accuracy. Validating this approach, we extracted and selected radiomic features from CT imaging data of 523 patients with gastrointestinal stromal tumors. The PCTR algorithm demonstrated superior performance over commonly used Cox Proportional Hazards and Random Survival Forest algorithms in external test cohort, offering clinical researchers a more effective method for prognostic model development.
Collapse
|
8
|
Li Z, Wang R, Wang L, Tan C, Xu J, Fang J, Xian J. Machine Learning-Based MRI Radiogenomics for Evaluation of Response to Induction Chemotherapy in Head and Neck Squamous Cell Carcinoma. Acad Radiol 2024; 31:2464-2475. [PMID: 37985290 DOI: 10.1016/j.acra.2023.10.054] [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: 09/20/2023] [Revised: 10/16/2023] [Accepted: 10/28/2023] [Indexed: 11/22/2023]
Abstract
RATIONALE AND OBJECTIVES To develop and validate a radiogenomics model integrating clinical data, radiomics-based machine learning (RBML) classifiers, and transcriptomics data for predicting the response to induction chemotherapy (IC) in patients with head and neck squamous cell carcinoma (HNSCC). MATERIALS AND METHODS Radiomics features derived from T2-weighted, pre- and post-contrast-enhanced T1-weighted MRI sequences, clinical data, and RNA sequencing data of 150 patients with HNSCC were included in the study. Analysis of variance or recursive feature elimination was used to reduce radiomics features. Three RBML classifiers were developed to distinguish non-responders from responders. Weighted correlation network analysis (WGCNA) was performed to identify the correlation between clinical data or radiomics features and molecular features; subsequently, protein interaction and functional enrichment analyses were performed. The predictive performance of the radiogenomics model integrating significant clinical variables, RBML classifiers, and molecular features was evaluated using receiver operating characteristic curve analysis. RESULTS Five radiomics features and two conventional MRI findings significantly stratified HNSCC patients into responders and non-responders. On WGCNA analysis, 809 genes showed a significant correlation with two radiomics features. Functional enrichment analysis suggested that our proposed radiomics features could reflect the T cell-mediated immune response and immune infiltration of HNSCC. The radiogenomics model showed the highest area under the curve (0.88[95%CI 0.75-0.96]) for predicting IC response, which was better than MRI findings(p = 0.0407) or molecular features(p = 0.004) alone, but showed no significant difference with that of RBML model (p = 0.2254) in test cohort. CONCLUSION Merging imaging phenotypes with transcriptomic data improved the prediction of IC response in HNSCC.
Collapse
Affiliation(s)
- Zheng Li
- Department of Radiology, Beijing Tongren Hospital, Capital Medical University, Beijing, China (Z.L., J.X.).
| | - Ru Wang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China (R.W., L.W., C.T., J.X., J.F.).
| | - Lingwa Wang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China (R.W., L.W., C.T., J.X., J.F.).
| | - Chen Tan
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China (R.W., L.W., C.T., J.X., J.F.).
| | - Jiaqi Xu
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China (R.W., L.W., C.T., J.X., J.F.).
| | - Jugao Fang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China (R.W., L.W., C.T., J.X., J.F.).
| | - Junfang Xian
- Department of Radiology, Beijing Tongren Hospital, Capital Medical University, Beijing, China (Z.L., J.X.).
| |
Collapse
|
9
|
Wu TC, Liu YL, Chen JH, Chen TY, Ko CC, Lin CY, Kao CY, Yeh LR, Su MY. Radiomics analysis for the prediction of locoregional recurrence of locally advanced oropharyngeal cancer and hypopharyngeal cancer. Eur Arch Otorhinolaryngol 2024; 281:1473-1481. [PMID: 38127096 DOI: 10.1007/s00405-023-08380-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023]
Abstract
PURPOSE By radiomic analysis of the postcontrast CT images, this study aimed to predict locoregional recurrence (LR) of locally advanced oropharyngeal cancer (OPC) and hypopharyngeal cancer (HPC). METHODS A total of 192 patients with stage III-IV OPC or HPC from two independent cohort were randomly split into a training cohort with 153 cases and a testing cohort with 39 cases. Only primary tumor mass was manually segmented. Radiomic features were extracted using PyRadiomics, and then the support vector machine was used to build the radiomic model with fivefold cross-validation process in the training data set. For each case, a radiomics score was generated to indicate the probability of LR. RESULTS There were 94 patients with LR assigned in the progression group and 98 patients without LR assigned in the stable group. There was no significant difference of TNM staging, treatment strategies and common risk factors between these two groups. For the training data set, the radiomics model to predict LR showed 83.7% accuracy and 0.832 (95% CI 0.72, 0.87) area under the ROC curve (AUC). For the test data set, the accuracy and AUC slightly declined to 79.5% and 0.770 (95% CI 0.64, 0.80), respectively. The sensitivity/specificity of training and test data set for LR prediction were 77.6%/89.6%, and 66.7%/90.5%, respectively. CONCLUSIONS The image-based radiomic approach could provide a reliable LR prediction model in locally advanced OPC and HPC. Early identification of those prone to post-treatment recurrence would be helpful for appropriate adjustments to treatment strategies and post-treatment surveillance.
Collapse
Affiliation(s)
- Te-Chang Wu
- Department of Medical Imaging, Chi-Mei Medical Center, Tainan, Taiwan
- Department of Medical Sciences Industry, Chang Jung Christian University, Tainan, Taiwan
| | - Yan-Lin Liu
- Department of Radiological Sciences, University of California, Irvine, Irvine, CA, USA
| | - Jeon-Hor Chen
- Department of Radiological Sciences, University of California, Irvine, Irvine, CA, USA
- Department of Medical Imaging, E-DA Hospital, Kaohsiung, Taiwan
| | - Tai-Yuan Chen
- Department of Medical Imaging, Chi-Mei Medical Center, Tainan, Taiwan
- Graduate Institute of Medical Sciences, Chang Jung Christian University, Tainan, Taiwan
| | - Ching-Chung Ko
- Department of Medical Imaging, Chi-Mei Medical Center, Tainan, Taiwan
- Center of General Education, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Chiao-Yun Lin
- Department of Medical Imaging, E-DA Hospital, Kaohsiung, Taiwan
- College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Cheng-Yi Kao
- College of Medicine, I-Shou University, Kaohsiung, Taiwan
- Division of Medical Radiology, E-DA Cancer Hospital, Kaohsiung, Taiwan
| | - Lee-Ren Yeh
- Department of Medical Imaging, E-DA Hospital, Kaohsiung, Taiwan.
- Department of Medical Imaging and Radiological Sciences, College of Medicine, I-Shou University, No. 1 Yida Road, Jiaosu Village, Yanchao District, Kaohsiung, 824, Taiwan.
| | - Min-Ying Su
- Department of Radiological Sciences, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
10
|
Zhao Z, Li W, Liu P, Zhang A, Sun J, Xu LX. Survival Analysis for Multimode Ablation Using Self-Adapted Deep Learning Network Based on Multisource Features. IEEE J Biomed Health Inform 2024; 28:19-30. [PMID: 37015120 DOI: 10.1109/jbhi.2023.3260776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
Novel multimode thermal therapy by freezing before radio-frequency heating has achieved a desirable therapeutic effect in liver cancer. Compared with surgical resection, ablation treatment has a relatively high risk of tumor recurrence. To monitor tumor progression after ablation, we developed a novel survival analysis framework for survival prediction and efficacy assessment. We extracted preoperative and postoperative MRI radiomics features and vision transformer-based deep learning features. We also combined the immune features extracted from peripheral blood immune responses using flow cytometry and routine blood tests before and after treatment. We selected features using random survival forest and improved the deep Cox mixture (DCM) for survival analysis. To properly accommodate multitype input features, we proposed a self-adapted fully connected layer for locally and globally representing features. We evaluated the method using our clinical dataset. Of note, the immune features rank the highest feature importance and contribute significantly to the prediction accuracy. The results showed a promising C$^{\mathit{td}}$-index of 0.885 $\pm$ 0.040 and an integrated Brier score of 0.041 $\pm$ 0.014, which outperformed state-of-the-art method combinations of survival prediction. For each patient, individual survival probability was accurately predicted over time, which provided clinicians with trustable prognosis suggestions.
Collapse
|
11
|
Qi M, Sha Y, Zhang D, Ren J. An MRI-based radiomics nomogram for detecting cervical esophagus invasion in hypopharyngeal squamous cell carcinoma. Cancer Imaging 2023; 23:120. [PMID: 38102719 PMCID: PMC10724942 DOI: 10.1186/s40644-023-00642-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 12/04/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Accurate detection of cervical esophagus invasion (CEI) in HPSCC is challenging but crucial. We aimed to investigate the value of magnetic resonance imaging (MRI)-based radiomics for detecting CEI in patients with HPSCC. METHODS This retrospective study included 151 HPSCC patients with or without CEI, which were randomly assigned into a training (n = 101) or validation (n = 50) cohort. A total of 750 radiomics features were extracted from T2-weighted imaging (T2WI) and contrast-enhanced T1-weighted imaging (ceT1WI), respectively. A radiomics signature was constructed using the least absolute shrinkage and selection operator method. Multivariable logistic regression analyses were adopted to establish a clinical model and a radiomics nomogram. Two experienced radiologists evaluated the CEI status based on morphological findings. Areas under the curve (AUCs) of the models and readers were compared using the DeLong method. The performance of the nomogram was also assessed by its calibration and clinical usefulness. RESULTS The radiomics signature, consisting of five T2WI and six ceT1WI radiomics features, was significantly associated with CEI in both cohorts (all p < 0.001). The radiomics nomogram combining the radiomics signature and clinical T stage achieved significantly higher predictive value than the clinical model and pooled readers in the training (AUC 0.923 vs. 0.723 and 0.621, all p < 0.001) and validation (AUC 0.888 vs. 0.754 and 0.647, all p < 0.05) cohorts. The radiomics nomogram showed favorable calibration in both cohorts and provided better net benefit than the clinical model. CONCLUSIONS The MRI-based radiomics nomogram is a promising method for detecting CEI in HPSCC.
Collapse
Affiliation(s)
- Meng Qi
- Department of Radiology, Eye & ENT Hospital, Fudan University, No.83 Fenyang Road, Shanghai, 200030, China
| | - Yan Sha
- Department of Radiology, Eye & ENT Hospital, Fudan University, No.83 Fenyang Road, Shanghai, 200030, China
| | - Duo Zhang
- Department of Otolaryngology-HNS, Eye & ENT Hospital, Fudan University, No.83 Fenyang Road, Shanghai, 200030, China.
| | - Jiliang Ren
- Department of Radiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.639 Zhizaoju Road, Shanghai, 200010, China.
| |
Collapse
|
12
|
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: 1.0] [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
|
13
|
Small C, Prior P, Nasief H, Zeitlin R, Saeed H, Paulson E, Morrow N, Rownd J, Erickson B, Bedi M. A general framework to develop a radiomic fingerprint for progression-free survival in cervical cancer. Brachytherapy 2023; 22:728-735. [PMID: 37574352 DOI: 10.1016/j.brachy.2023.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/11/2023] [Accepted: 06/06/2023] [Indexed: 08/15/2023]
Abstract
PURPOSE Treatment of locally advanced cervical cancer patients includes chemoradiation followed by brachytherapy. Our aim is to develop a delta radiomics (DRF) model from MRI-based brachytherapy treatment and assess its association with progression free survival (PFS). MATERIALS AND METHODS A retrospective analysis of FIGO stage IB- IV cervical cancer patients between 2012 and 2018 who were treated with definitive chemoradiation followed by MRI-based intracavitary brachytherapy was performed. Clinical factors together with 18 radiomic features extracted from different radiomics matrices were analyzed. The delta radiomic features (DRFs) were extracted from MRI on the first and last brachytherapy fractions. Support Vector Machine (SVM) models were fitted to combinations of 2-3 DRFs found significant after Spearman correlation and Wilcoxon rank sum test statistics. Additional models were tested that included clinical factors together with DRFs. RESULTS A total of 39 patients were included in the analysis with a median patient age of 52 years. Progression occurred in 20% of patients (8/39). The significant DRFs using two DRF feature combinations was a model using auto correlation (AC) and sum variance (SV). The best performing three feature model combined mean, AC & SV. Additionally, the inclusion of FIGO stages with the 2- and 3 DRF combination model(s) improved performance compared to models with only DRFs. However, all the clinical factor + DRF models were not significantly different from one another (all AUCs were 0.77). CONCLUSIONS Our study shows promising evidence that radiomics metrics are associated with progression free survival in cervical cancer.
Collapse
Affiliation(s)
- Christina Small
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI.
| | - Phillip Prior
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI
| | - Haidy Nasief
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI
| | - Ross Zeitlin
- Department of Radiation Oncology, John H Stroger, Jr. Hospital of Cook County, Chicago, IL
| | - Hina Saeed
- Department of Radiation Oncology, Lynn Cancer Institute, Baptist Health South Florida, Boynton Beach, FL
| | - Eric Paulson
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI
| | - Natalya Morrow
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI
| | - Jason Rownd
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI
| | - Beth Erickson
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI
| | - Meena Bedi
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
14
|
Le VH, Kha QH, Minh TNT, Nguyen VH, Le VL, Le NQK. Development and Validation of CT-Based Radiomics Signature for Overall Survival Prediction in Multi-organ Cancer. J Digit Imaging 2023; 36:911-922. [PMID: 36717518 PMCID: PMC10287593 DOI: 10.1007/s10278-023-00778-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 02/01/2023] Open
Abstract
The malignant tumors in nature share some common morphological characteristics. Radiomics is not only images but also data; we think that a probability exists in a set of radiomics signatures extracted from CT scan images of one cancer tumor in one specific organ also be utilized for overall survival prediction in different types of cancers in different organs. The retrospective study enrolled four data sets of cancer patients in three different organs (420, 157, 137, and 191 patients for lung 1 training, lung 2 testing, and two external validation set: kidney and head and neck, respectively). In the training set, radiomics features were obtained from CT scan images, and essential features were chosen by LASSO algorithm. Univariable and multivariable analyses were then conducted to find a radiomics signature via Cox proportional hazard regression. The Kaplan-Meier curve was performed based on the risk score. The integrated time-dependent area under the ROC curve (iAUC) was calculated for each predictive model. In the training set, Kaplan-Meier curve classified patients as high or low-risk groups (p-value < 0.001; log-rank test). The risk score of radiomics signature was locked and independently evaluated in the testing set, and two external validation sets showed significant differences (p-value < 0.05; log-rank test). A combined model (radiomics + clinical) showed improved iAUC in lung 1, lung 2, head and neck, and kidney data set are 0.621 (95% CI 0.588, 0.654), 0.736 (95% CI 0.654, 0.819), 0.732 (95% CI 0.655, 0.809), and 0.834 (95% CI 0.722, 0.946), respectively. We believe that CT-based radiomics signatures for predicting overall survival in various cancer sites may exist.
Collapse
Affiliation(s)
- Viet Huan Le
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
- Department of Thoracic Surgery, Khanh Hoa General Hospital, Nha Trang, 65000, Vietnam
| | - Quang Hien Kha
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Tran Nguyen Tuan Minh
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Van Hiep Nguyen
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
- Oncology Center, Bai Chay Hospital, Quang Ninh, 20000, Vietnam
| | - Van Long Le
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
- Department of Anesthesiology and Critical Care, Hue University of Medicine and Pharmacy, Hue University, Hue City, 52000, Vietnam
| | - Nguyen Quoc Khanh Le
- Professional Master Program in Artificial Intelligence in Medicine, College of Medicine, Taipei Medical University, Taipei, 106, Taiwan.
- Research Center for Artificial Intelligence in Medicine, Taipei Medical University, Taipei, 106, Taiwan.
- Translational Imaging Research Center, Taipei Medical University Hospital, Taipei, 110, Taiwan.
| |
Collapse
|
15
|
Chiesa-Estomba CM, Mayo-Yanez M, Guntinas-Lichius O, Vander-Poorten V, Takes RP, de Bree R, Halmos GB, Saba NF, Nuyts S, Ferlito A. Radiomics in Hypopharyngeal Cancer Management: A State-of-the-Art Review. Biomedicines 2023; 11:805. [PMID: 36979783 PMCID: PMC10045560 DOI: 10.3390/biomedicines11030805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 02/25/2023] [Accepted: 03/04/2023] [Indexed: 03/09/2023] Open
Abstract
(1) Background: Hypopharyngeal squamous cell carcinomas usually present with locally advanced disease and a correspondingly poor prognosis. Currently, efforts are being made to improve tumor characterization and provide insightful information for outcome prediction. Radiomics is an emerging area of study that involves the conversion of medical images into mineable data; these data are then used to extract quantitative features based on shape, intensity, texture, and other parameters; (2) Methods: A systematic review of the peer-reviewed literature was conducted; (3) Results: A total of 437 manuscripts were identified. Fifteen manuscripts met the inclusion criteria. The main targets described were the evaluation of textural features to determine tumor-programmed death-ligand 1 expression; a surrogate for microvessel density and heterogeneity of perfusion; patient stratification into groups at high and low risk of progression; prediction of early recurrence, 1-year locoregional failure and survival outcome, including progression-free survival and overall survival, in patients with locally advanced HPSCC; thyroid cartilage invasion, early disease progression, recurrence, induction chemotherapy response, treatment response, and prognosis; and (4) Conclusions: our findings suggest that radiomics represents a potentially useful tool in the diagnostic workup as well as during the treatment and follow-up of patients with HPSCC. Large prospective studies are essential to validate this technology in these patients.
Collapse
Affiliation(s)
- Carlos M. Chiesa-Estomba
- Otorhinolaryngology-Head & Neck Surgery Department, Hospital Universitario Donostia, Biodonostia Research Institute, Faculty of Medicine, Deusto University, 20014 San Sebastian, Spain
| | - Miguel Mayo-Yanez
- Otorhinolaryngology-Head and Neck Surgery Department, Complexo Hospitalario Universitario A Coruña (CHUAC), 15006 A Coruña, Spain
| | | | - Vincent Vander-Poorten
- Section Head and Neck Oncology, Department of Oncology, KU Leuven—University of Leuven, 3000 Leuven, Belgium
| | - Robert P. Takes
- Department of Otolaryngology/Head and Neck Surgery, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Remco de Bree
- Department of Head and Neck Surgical Oncology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Gyorgy B. Halmos
- Department of Otorhinolaryngology/Head and Neck Surgery, University Medical Center Groningen, University of Groningen, 9712 CP Groningen, The Netherlands
| | - Nabil F. Saba
- Department of Hematology and Medical Oncology, The Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | - Sandra Nuyts
- Department of Radiation Oncology, University Hospitals Leuven, KU Leuven—University of Leuven, 3000 Leuven, Belgium
| | - Alfio Ferlito
- Coordinator of the International Head and Neck Scientific Group, 35125 Padua, Italy
| |
Collapse
|
16
|
Liu H, Zhao D, Huang Y, Li C, Dong Z, Tian H, Sun Y, Lu Y, Chen C, Wu H, Zhang Y. Comprehensive prognostic modeling of locoregional recurrence after radiotherapy for patients with locoregionally advanced hypopharyngeal squamous cell carcinoma. Front Oncol 2023; 13:1129918. [PMID: 37025592 PMCID: PMC10072214 DOI: 10.3389/fonc.2023.1129918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 03/13/2023] [Indexed: 04/08/2023] Open
Abstract
Purpose To propose and evaluate a comprehensive modeling approach combing radiomics, dosiomics and clinical components, for more accurate prediction of locoregional recurrence risk after radiotherapy for patients with locoregionally advanced HPSCC. Materials and methods Clinical data of 77 HPSCC patients were retrospectively investigated, whose median follow-up duration was 23.27 (4.83-81.40) months. From the planning CT and dose distribution, 1321 radiomics and dosiomics features were extracted respectively from planning gross tumor volume (PGTV) region each patient. After stability test, feature dimension was further reduced by Principal Component Analysis (PCA), yielding Radiomic and Dosiomic Principal Components (RPCs and DPCs) respectively. Multiple Cox regression models were constructed using various combinations of RPC, DPC and clinical variables as the predictors. Akaike information criterion (AIC) and C-index were used to evaluate the performance of Cox regression models. Results PCA was performed on 338 radiomic and 873 dosiomic features that were tested as stable (ICC1 > 0.7 and ICC2 > 0.95), yielding 5 RPCs and DPCs respectively. Three comprehensive features (RPC0, P<0.01, DPC0, P<0.01 and DPC3, P<0.05) were found to be significant in the individual Radiomic or Dosiomic Cox regression models. The model combining the above features and clinical variable (total stage IVB) provided best risk stratification of locoregional recurrence (C-index, 0.815; 95%CI, 0.770-0.859) and prevailing balance between predictive accuracy and complexity (AIC, 143.65) than any other investigated models using either single factors or two combined components. Conclusion This study provided quantitative tools and additional evidence for the personalized treatment selection and protocol optimization for HPSCC, a relatively rare cancer. By combining complementary information from radiomics, dosiomics, and clinical variables, the proposed comprehensive model provided more accurate prediction of locoregional recurrence risk after radiotherapy.
Collapse
Affiliation(s)
- Hongjia Liu
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Dan Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yuliang Huang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital & Institute, Beijing, China
- Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - Chenguang Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhengkun Dong
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hongbo Tian
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yijie Sun
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yanye Lu
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China
| | - Chen Chen
- School of Electronics Engineering and Computer Science, Peking University, Beijing, China
| | - Hao Wu
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yibao Zhang
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital & Institute, Beijing, China
- *Correspondence: Yibao Zhang,
| |
Collapse
|
17
|
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: 1.7] [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
|
18
|
Liu X, Long M, Sun C, Yang Y, Lin P, Shen Z, Xia S, Shen W. CT-based radiomics signature analysis for evaluation of response to induction chemotherapy and progression-free survival in locally advanced hypopharyngeal carcinoma. Eur Radiol 2022; 32:7755-7766. [PMID: 35608663 DOI: 10.1007/s00330-022-08859-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/28/2022] [Accepted: 05/01/2022] [Indexed: 01/03/2023]
Abstract
OBJECTIVES To establish and validate a CT radiomics model for prediction of induction chemotherapy (IC) response and progression-free survival (PFS) among patients with locally advanced hypopharyngeal carcinoma (LAHC). METHODS One hundred twelve patients with LAHC (78 in training cohort and 34 in validation cohort) who underwent contrast-enhanced CT (CECT) scans prior to IC were enrolled. Least absolute shrinkage and selection operator (LASSO) was used to select the crucial radiomic features in the training cohort. Radiomics signature and clinical data were used to build a radiomics nomogram to predict individual response to IC. Kaplan-Meier analysis and log-rank test were used to evaluate ability of radiomics signature in progression-free survival risk stratification. RESULTS The radiomics signature consisted of 6 selected features from the arterial and venous phases of CECT images and demonstrated good performance in predicting the IC response in both two cohorts. The radiomics nomogram showed good discriminative performance, and the C-index of nomogram was 0.899 (95% confidence interval (CI), 0.831-0.967) and 0.775 (95% CI, 0.591-0.959) in the training and validation cohorts, respectively. Survival analysis indicated that low-risk and high-risk groups defined by the value of radiomics signature had significant difference in PFS (3-year PFS 66.4% vs 29.7%, p < 0.001). CONCLUSIONS Multiparametric CT-based radiomics model could be useful for predicting treatment response and PFS in patients with LAHC who underwent IC. KEY POINTS • CT radiomics can predict IC response and progression-free survival in hypopharyngeal carcinoma. • We combined significant radiomics signature with clinical predictors to establish a nomogram to predict individual response to IC. • Radiomics signature could divide patients into the high-risk and low-risk groups based on the PFS.
Collapse
Affiliation(s)
- Xiaobin Liu
- Department of Radiology, Tianjin Medical Imaging Institute, Tianjin First Central Hospital, School of Medicine, Nankai University, Fukang Road No. 24, Nankai District, 300192, Tianjin, China
| | - Miaomiao Long
- Department of Radiology, Tianjin Medical Imaging Institute, Tianjin First Central Hospital, School of Medicine, Nankai University, Fukang Road No. 24, Nankai District, 300192, Tianjin, China
| | - Chuanqi Sun
- Department of Biomedical Engineering, Guangzhou Medical University, Xinzao Road No. 1, Panyu District, Guangzhou, 511436, China
| | - Yining Yang
- Department of Radiotherapy, Tianjin First Central Hospital, School of Medicine, Nankai University, Fukang Road No. 24, Nankai District, Tianjin, 300192, China
| | - Peng Lin
- Department of Otorhinolaryngology Head and Neck Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Fukang Road No. 24, Nankai District, Tianjin, 300192, China
| | - Zhiwei Shen
- Philips Healthcare, World Profit Centre, 100125, Tianze Road No. 16, Chaoyang District, Beijing, China
| | - Shuang Xia
- Department of Radiology, Tianjin Medical Imaging Institute, Tianjin First Central Hospital, School of Medicine, Nankai University, Fukang Road No. 24, Nankai District, 300192, Tianjin, China.
| | - Wen Shen
- Department of Radiology, Tianjin Medical Imaging Institute, Tianjin First Central Hospital, School of Medicine, Nankai University, Fukang Road No. 24, Nankai District, 300192, Tianjin, China.
| |
Collapse
|
19
|
Liu L, Pei W, Liao H, Wang Q, Gu D, Liu L, Su D, Jin G. A Clinical-Radiomics Nomogram Based on Magnetic Resonance Imaging for Predicting Progression-Free Survival After Induction Chemotherapy in Nasopharyngeal Carcinoma. Front Oncol 2022; 12:792535. [PMID: 35814380 PMCID: PMC9256909 DOI: 10.3389/fonc.2022.792535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 04/07/2022] [Indexed: 11/13/2022] Open
Abstract
PurposeThis paper aimed to establish and verify a radiomics model based on magnetic resonance imaging (MRI) for predicting the progression-free survival of nasopharyngeal carcinoma (NPC) after induction chemotherapy (IC).Materials and MethodsThis cohort consists of 288 patients with clinical pathologically confirmed NPC, which was collected from January 2015 to December 2018. All NPC patients were randomly divided into two cohorts: training (n=202) and validation (n=86). Radiomics features from the MRI images of NPC patients were extracted and selected before IC. The patients were classified into high- and low-risk groups according to the median of Radscores. The significant imaging features and clinical variables in the univariate analysis were constructed for progression-free survival (PFS) using the multivariate Cox regression model. A survival analysis was performed using Kaplan–Meier with log-rank test and then each model’s stratification ability was evaluated.ResultsEpstein–Barr virus (EBV) DNA before treatment was an independent predictor for PFS (p < 0.05). Based on the pyradiomic platform, we extracted 1,316 texture parameters in total. Finally, 16 texture features were used to build the model. The clinical radiomics-based model had good prediction capability for PFS, with a C-index of 0.827. The survival curve revealed that the PFS of the high-risk group was poorer than that of the low-risk group.ConclusionThis research presents a nomogram that merges the radiomics signature and the clinical feature of the plasma EBV DNA load, which may improve the ability of preoperative prediction of progression-free survival and facilitate individualization of treatment in NPC patients before IC.
Collapse
Affiliation(s)
- Lu Liu
- Department of Radiology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Wei Pei
- Department of Radiology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Hai Liao
- Department of Radiology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Qiang Wang
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Donglian Gu
- Department of Radiology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Lijuan Liu
- Department of Radiology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Danke Su
- Department of Radiology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Guanqiao Jin
- Department of Radiology, Guangxi Medical University Cancer Hospital, Nanning, China
- *Correspondence: Guanqiao Jin,
| |
Collapse
|
20
|
Wang X, Zhang H, Ye J, Gao M, Jiang Q, Zhao T, Wang S, Mao W, Wang K, Wang Q, Chen X, Hou X, Han D. Genome Instability-Associated Long Non-Coding RNAs Reveal Biomarkers for Glioma Immunotherapy and Prognosis. Front Genet 2022; 13:850888. [PMID: 35571034 PMCID: PMC9094631 DOI: 10.3389/fgene.2022.850888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Genome instability is a hallmark of tumors and is involved in proliferation, invasion, migration, and treatment resistance of many tumors. However, the relationship of genome instability with gliomas remains unclear. Here, we constructed genome instability-derived long non-coding RNA (lncRNA)-based gene signatures (GILncSig) using genome instability-related lncRNAs derived from somatic mutations. Multiple platforms were used to confirm that the GILncSig were closely related to patient prognosis and clinical characteristics. We found that GILncSig, the glioma microenvironment, and glioma cell DNA methylation-based stemness index (mDNAsi) interacted with each other to form a complex regulatory network. In summary, this study confirmed that GILncSig was an independent prognostic indicator for patients, distinguished high-risk and low-risk groups, and affected immune-cell infiltration and tumor-cell stemness indicators (mDNAsi) in the tumor microenvironment, resulting in tumor heterogeneity and immunotherapy resistance. GILncSig are expected to provide new molecular targets for the clinical treatment of patients with gliomas.
Collapse
Affiliation(s)
- Xinzhuang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hong Zhang
- Department of Hematology, Liaocheng People's Hospital, Liaocheng, China
| | - Junyi Ye
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ming Gao
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qiuyi Jiang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tingting Zhao
- Biochip Laboratory, Yantai Yu-Huang-Ding Hospital, Qingdao University, Yantai, China
| | - Shengtao Wang
- The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenbin Mao
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kaili Wang
- The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qi Wang
- The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xin Chen
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xu Hou
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dayong Han
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
21
|
Chen J, Lu S, Mao Y, Tan L, Li G, Gao Y, Tan P, Huang D, Zhang X, Qiu Y, Liu Y. An MRI-based radiomics-clinical nomogram for the overall survival prediction in patients with hypopharyngeal squamous cell carcinoma: a multi-cohort study. Eur Radiol 2022; 32:1548-1557. [PMID: 34665315 DOI: 10.1007/s00330-021-08292-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 07/01/2021] [Accepted: 07/12/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To explore whether radiomics features extracted from pre-treatment magnetic resonance imaging (MRI) can predict the overall survival (OS) in patients with hypopharyngeal squamous cell carcinoma. METHODS A total of 190 patients with hypopharyngeal squamous cell carcinoma were eligibly enrolled from two institutions. Radiomics features were extracted from contrast-enhanced axial T1-weighted (CE-T1WI) sequence. The least absolute shrinkage selection operator (LASSO) algorithm was applied to establish a radiomics score correlated with OS. Multivariate logistic regression analysis was applied to determine the independent risk factors, which was combined with radiomics score to build the final radiomics nomogram. RESULTS A radiomics score with 6 CE-T1WI features for OS prediction was constructed and validated; its integration with specific clinicopathologic factors (N stage) showed a better prediction performance in the training, internal validation, and external validation cohorts (C-index 0.78, 0.75, and 0.75). Calibration curves determined a good agreement between the predicted and actual overall survival. CONCLUSIONS The radiomics-clinical nomogram and radiomics score might be non-invasive and reliable methods for the risk stratification in patients with hypopharyngeal squamous cell carcinoma. KEY POINTS • An MRI-based radiomics model was constructed to evaluate of OS in patients with hypopharyngeal squamous cell carcinoma. • A radiomics-clinical nomogram that combined radiomics features and clinical characteristics was established. • Multi-cohort study validated the predictive performance of the radiomics-clinical nomogram to stratify patients with high risk in clinical practice.
Collapse
Affiliation(s)
- Juan Chen
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, 410008, Hunan, China
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, 410008, Hunan, China
| | - Shanhong Lu
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, 410008, Hunan, China
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, 410008, Hunan, China
| | - Yitao Mao
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Lei Tan
- College of Computer and Information Engineering, Hunan University of Technology and Business, Changsha, 410205, Hunan, China
| | - Guo Li
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, 410008, Hunan, China
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, 410008, Hunan, China
| | - Yan Gao
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, 410008, Hunan, China
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, 410008, Hunan, China
| | - Pingqing Tan
- Department of Head and Neck Surgery, The Affiliated Tumor Hospital of Xiangya Medical School, Hunan Cancer Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Donghai Huang
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, 410008, Hunan, China
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Xiangya Road, Changsha, 410008, Hunan, China
| | - Xin Zhang
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, 410008, Hunan, China
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Xiangya Road, Changsha, 410008, Hunan, China
| | - Yuanzheng Qiu
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, 410008, Hunan, China.
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Xiangya Road, Changsha, 410008, Hunan, China.
| | - Yong Liu
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Changsha, 410008, Hunan, China.
- Clinical Research Center for Pharyngolaryngeal Diseases and Voice Disorders in Hunan Province, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Xiangya Road, Changsha, 410008, Hunan, China.
| |
Collapse
|
22
|
Cho HH, Kim CK, Park H. Overview of radiomics in prostate imaging and future directions. Br J Radiol 2022; 95:20210539. [PMID: 34797688 PMCID: PMC8978251 DOI: 10.1259/bjr.20210539] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Recent advancements in imaging technology and analysis methods have led to an analytic framework known as radiomics. This framework extracts comprehensive high-dimensional features from imaging data and performs data mining to build analytical models for improved decision-support. Its features include many categories spanning texture and shape; thus, it can provide abundant information for precision medicine. Many studies of prostate radiomics have shown promising results in the assessment of pathological features, prediction of treatment response, and stratification of risk groups. Herein, we aimed to provide a general overview of radiomics procedures, discuss technical issues, explain various clinical applications, and suggest future research directions, especially for prostate imaging.
Collapse
Affiliation(s)
- Hwan-Ho Cho
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Korea.,Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Korea
| | - Chan Kyo Kim
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hyunjin Park
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Korea.,School of Electronic and Electrical Engineering, Sungkyunkwan University, Suwon, Korea
| |
Collapse
|
23
|
Liu X, Sun C, Long M, Yang Y, Lin P, Xia S, Shen W. Computed tomography-based radiomics signature as a pretreatment predictor of progression-free survival in locally advanced hypopharyngeal carcinoma with a different response to induction chemotherapy. Eur Arch Otorhinolaryngol 2022; 279:3551-3562. [PMID: 35212776 DOI: 10.1007/s00405-022-07306-w] [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: 12/12/2021] [Accepted: 02/07/2022] [Indexed: 11/27/2022]
Abstract
PURPOSE To establish and validate a radiomics signature for stratifying the risk of progression-free survival (PFS) in patients with locally advanced hypopharyngeal carcinoma (LAHC) undergoing induction chemotherapy (IC). METHODS We extracted radiomics features from baseline contrast-enhanced computed tomography (CECT) images. We enrolled 112 LAHC patients (78 in the training cohort and 34 in the validation cohort). We used cox regression model and random survival forests variable hunting (RSFVH) algorithm for feature selection and radiomics signature building. The radiomics signature was established in the training cohort and tested in the validation cohort. We used the Kaplan-Meier analysis and log-rank test to evaluate the ability of radiomics signature in PFS risk stratification among patients with different IC responses and constructed a radiomics nomogram to predict individual PFS risk. RESULTS The radiomics signature performed well in stratifying patients into highrisk and low-risk groups of progression in both the training and validation cohorts. The radiomics nomogram showed good discriminative ability for predicting PFS. Survival outcome analysis of subsets indicated that the radiomics signature performed well in stratifying the risk of PFS in patients with LAHC with different IC responses. CONCLUSIONS The radiomics signature was a pretreatment predictor for PFS in patients with LAHC who exhibited different responses to IC.
Collapse
Affiliation(s)
- Xiaobin Liu
- Department of Radiology, First Central Clinical College, Tianjin Medical University, Qixiangtai Road No. 22, Heping District, Tianjin, 300070, China
- Department of Radiology, Tianjin Medical Imaging Institute, Tianjin First Central Hospital, School of Medicine, Nankai University, Fukang Road No. 24, Nankai District, Tianjin, 300192, China
| | - Chuanqi Sun
- Department of Biomedical Engineering, Guangzhou Medical University, Xinzao Road No. 1, Panyu District, Guangzhou, 511436, China
| | - Miaomiao Long
- Department of Radiology, Tianjin Medical Imaging Institute, Tianjin First Central Hospital, School of Medicine, Nankai University, Fukang Road No. 24, Nankai District, Tianjin, 300192, China
| | - Yining Yang
- Department of Radiotherapy, Tianjin First Central Hospital, School of Medicine, Nankai University, Fukang Road No. 24, Nankai District, Tianjin, 300192, China
| | - Peng Lin
- Department of Otorhinolaryngology Head and Neck Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Fukang Road No. 24, Nankai District, Tianjin, 300192, China
| | - Shuang Xia
- Department of Radiology, Tianjin Medical Imaging Institute, Tianjin First Central Hospital, School of Medicine, Nankai University, Fukang Road No. 24, Nankai District, Tianjin, 300192, China
| | - Wen Shen
- Department of Radiology, Tianjin Medical Imaging Institute, Tianjin First Central Hospital, School of Medicine, Nankai University, Fukang Road No. 24, Nankai District, Tianjin, 300192, China.
| |
Collapse
|
24
|
Luo C, Li S, Zhao Q, Ou Q, Huang W, Ruan G, Liang S, Liu L, Zhang Y, Li H. RuleFit-Based Nomogram Using Inflammatory Indicators for Predicting Survival in Nasopharyngeal Carcinoma, a Bi-Center Study. J Inflamm Res 2022; 15:4803-4815. [PMID: 36042867 PMCID: PMC9420437 DOI: 10.2147/jir.s366922] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 08/11/2022] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Traditional prognostic studies utilized different cut-off values, without evaluating potential information contained in inflammation-related hematological indicators. Using the interpretable machine-learning algorithm RuleFit, this study aimed to explore valuable inflammatory rules reflecting prognosis in nasopharyngeal carcinoma (NPC) patients. PATIENTS AND METHODS In total, 1706 biopsy-proven NPC patients treated in two independent hospitals (1320 and 386) between January 2010 and March 2014 were included. RuleFit was used to develop risk-predictive rules using hematological indicators with no distributive difference between the two centers. Time-event-dependent hematological rules were further selected by stepwise multivariate Cox analysis. Combining high-efficiency hematological rules and clinical predictors, a final model was established. Models based on other algorithms (AutoML, Lasso) and clinical predictors were built for comparison, as well as a reported nomogram. Area under the receiver operating characteristic curve (AUROC) and concordance index (C-index) were used to verify the predictive precision of different models. A site-based app was established for convenience. RESULTS RuleFit identified 22 combined baseline hematological rules, achieving AUROCs of 0.69 and 0.64 in the training and validation cohorts, respectively. By contrast, the AUROCs of the optimal contrast model based on AutoML were 1.00 and 0.58. For overall survival, the final model had a much higher C-index than the base model using TN staging in two cohorts (0.769 vs 0.717, P<0.001; 0.752 vs 0.688, P<0.001), and showing great generalizability in training and validation cohorts. The two models based on RuleFit rules performed best, compared with other models. As for other endpoints, the final model showed a similar trend. Kaplan-Meier curve exhibited 22.9% (390/1706) patients were "misclassified" by AJCC staging, but the final model could assess risk classification accurately. CONCLUSION The proposed final models based on inflammation-related rules based on RuleFit showed significantly elevated predictive performance.
Collapse
Affiliation(s)
- Chao Luo
- Department of Radiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, Guangdong, People’s Republic of China
| | - Shuqi Li
- Department of Radiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, Guangdong, People’s Republic of China
| | - Qin Zhao
- Department of Radiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, Guangdong, People’s Republic of China
| | - Qiaowen Ou
- Department of Clinical Nutrition, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, People’s Republic of China
| | - Wenjie Huang
- Department of Radiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, Guangdong, People’s Republic of China
| | - Guangying Ruan
- Department of Radiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, Guangdong, People’s Republic of China
| | - Shaobo Liang
- Department of Radiotherapy, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, People’s Republic of China
| | - Lizhi Liu
- Department of Radiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, Guangdong, People’s Republic of China
- Department of Radiology, The Third People’s Hospital of Shenzhen, Shenzhen, Guangdong, People’s Republic of China
| | - Yu Zhang
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, People’s Republic of China
- Yu Zhang, Department of Pathology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, People’s Republic of China, Email
| | - Haojiang Li
- Department of Radiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, Guangdong, People’s Republic of China
- Correspondence: Haojiang Li, Department of Radiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, 651 Dongfeng Road East, Guangzhou, Guangdong, 510060, People’s Republic of China, Tel +86-20-87342135, Fax +86-20-87342125, Email
| |
Collapse
|
25
|
Bruixola G, Remacha E, Jiménez-Pastor A, Dualde D, Viala A, Montón JV, Ibarrola-Villava M, Alberich-Bayarri Á, Cervantes A. Radiomics and radiogenomics in head and neck squamous cell carcinoma: Potential contribution to patient management and challenges. Cancer Treat Rev 2021; 99:102263. [PMID: 34343892 DOI: 10.1016/j.ctrv.2021.102263] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/06/2021] [Accepted: 07/23/2021] [Indexed: 12/12/2022]
Abstract
The application of imaging biomarkers in oncology is still in its infancy, but with the expansion of radiomics and radiogenomics a revolution is expected in this field. This may be of special interest in head and neck cancer, since it can promote precision medicine and personalization of treatment by overcoming several intrinsic obstacles in this pathology. Our goal is to provide the medical oncologist with the basis to approach these disciplines and appreciate their main uses in clinical research and clinical practice in the medium term. Aligned with this objective we analyzed the most relevant studies in the field, also highlighting novel opportunities and current challenges.
Collapse
Affiliation(s)
- Gema Bruixola
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - Elena Remacha
- Quantitative Imaging Biomarkers in Medicine (QUIBIM SL), Valencia, Spain
| | - Ana Jiménez-Pastor
- Quantitative Imaging Biomarkers in Medicine (QUIBIM SL), Valencia, Spain
| | - Delfina Dualde
- Department of Radiology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - Alba Viala
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - Jose Vicente Montón
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - Maider Ibarrola-Villava
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Andrés Cervantes
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain; CIBERONC, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
26
|
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: 0.8] [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
|
27
|
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: 0.5] [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
|
28
|
Wu LL, Wang JL, Huang W, Liu X, Huang YY, Zeng J, Cui CY, Lu JB, Lin P, Long H, Zhang LJ, Wei J, Lu Y, Ma GW. Prognostic Modeling of Patients Undergoing Surgery Alone for Esophageal Squamous Cell Carcinoma: A Histopathological and Computed Tomography Based Quantitative Analysis. Front Oncol 2021; 11:565755. [PMID: 33912439 PMCID: PMC8072145 DOI: 10.3389/fonc.2021.565755] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 03/17/2021] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE To evaluate the effectiveness of a novel computerized quantitative analysis based on histopathological and computed tomography (CT) images for predicting the postoperative prognosis of esophageal squamous cell carcinoma (ESCC) patients. METHODS We retrospectively reviewed the medical records of 153 ESCC patients who underwent esophagectomy alone and quantitatively analyzed digital histological specimens and diagnostic CT images. We cut pathological images (6000 × 6000) into 50 × 50 patches; each patient had 14,400 patches. Cluster analysis was used to process these patches. We used the pathological clusters to all patches ratio (PCPR) of each case for pathological features and we obtained 20 PCPR quantitative features. Totally, 125 computerized quantitative (20 PCPR and 105 CT) features were extracted. We used a recursive feature elimination approach to select features. A Cox hazard model with L1 penalization was used for prognostic indexing. We compared the following prognostic models: Model A: clinical features; Model B: quantitative CT and clinical features; Model C: quantitative histopathological and clinical features; and Model D: combined information of clinical, CT, and histopathology. Indices of concordance (C-index) and leave-one-out cross-validation (LOOCV) were used to assess prognostic model accuracy. RESULTS Five PCPR and eight CT features were treated as significant indicators in ESCC prognosis. C-indices adjusted for LOOCV were comparable among four models, 0.596 (Model A) vs. 0.658 (Model B) vs. 0.651 (Model C), and improved to 0.711with Model D combining information of clinical, CT, and histopathology (all p<0.05). Using Model D, we stratified patients into low- and high-risk groups. The 3-year overall survival rates of low- and high-risk patients were 38.0% and 25.0%, respectively (p<0.001). CONCLUSION Quantitative prognostic modeling using a combination of clinical data, histopathological, and CT images can stratify ESCC patients with surgery alone into high-risk and low-risk groups.
Collapse
Affiliation(s)
- Lei-Lei Wu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jin-Long Wang
- School of Data and Computer Science, Sun Yat-sen University, Guangzhou, China
| | - Wei Huang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xuan Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yang-Yu Huang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jing Zeng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Chun-Yan Cui
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jia-Bin Lu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Peng Lin
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Hao Long
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Lan-Jun Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jun Wei
- Department of Radiology, University of Michigan, Ann Arbor, MI, United States
| | - Yao Lu
- School of Data and Computer Science, Sun Yat-sen University, Guangzhou, China
| | - Guo-Wei Ma
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
29
|
Nazari M, Shiri I, Zaidi H. Radiomics-based machine learning model to predict risk of death within 5-years in clear cell renal cell carcinoma patients. Comput Biol Med 2020; 129:104135. [PMID: 33254045 DOI: 10.1016/j.compbiomed.2020.104135] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 10/21/2020] [Accepted: 11/11/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE The aim of this study was to develop radiomics-based machine learning models based on extracted radiomic features and clinical information to predict the risk of death within 5 years for prognosis of clear cell renal cell carcinoma (ccRCC) patients. METHODS According to image quality and clinical data availability, we eventually selected 70 ccRCC patients that underwent CT scans. Manual volume-of-interest (VOI) segmentation of each image was performed by an experienced radiologist using the 3D slicer software package. Prior to feature extraction, image pre-processing was performed on CT images to extract different image features, including wavelet, Laplacian of Gaussian, and resampling of the intensity values to 32, 64 and 128 bin levels. Overall, 2544 3D radiomics features were extracted from each VOI for each patient. Minimum Redundancy Maximum Relevance (MRMR) algorithm was used as feature selector. Four classification algorithms were used, including Generalized Linear Model (GLM), Support Vector Machine (SVM), K-nearest Neighbor (KNN) and XGBoost. We used the Bootstrap resampling method to create validation sets. Area under the receiver operating characteristic (ROC) curve (AUROC), accuracy, sensitivity, and specificity were used to assess the performance of the classification models. RESULTS The best single performance among 8 different models was achieved by the XGBoost model using a combination of radiomic features and clinical information (AUROC, accuracy, sensitivity, and specificity with 95% confidence interval were 0.95-0.98, 0.93-0.98, 0.93-0.96 and ~1.0, respectively). CONCLUSIONS We developed a robust radiomics-based classifier that is capable of accurately predicting overall survival of RCC patients for prognosis of ccRCC patients. This signature may help identifying high-risk patients who require additional treatment and follow up regimens.
Collapse
Affiliation(s)
- Mostafa Nazari
- Department of Biomedical Engineering and Medical Physics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Isaac Shiri
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211, Geneva 4, Switzerland
| | - Habib Zaidi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, CH-1211, Geneva 4, Switzerland; Geneva University Neurocenter, Geneva University, Geneva, Switzerland; Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, Netherlands; Department of Nuclear Medicine, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
30
|
Li Q, Xiao Q, Li J, Duan S, Wang H, Gu Y. MRI-Based Radiomic Signature as a Prognostic Biomarker for HER2-Positive Invasive Breast Cancer Treated with NAC. Cancer Manag Res 2020; 12:10603-10613. [PMID: 33149669 PMCID: PMC7602910 DOI: 10.2147/cmar.s271876] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/19/2020] [Indexed: 12/15/2022] Open
Abstract
Purpose To identify MRI-based radiomics signature (Rad-score) as a biomarker of risk stratification for disease-free survival (DFS) in patients with HER2-positive invasive breast cancer treated with trastuzumab-based neoadjuvant chemotherapy (NAC) and establish a radiomics-clinicoradiologic-based nomogram that combines Rad-score, MRI findings, and clinicopathological variables for DFS estimation. Patients and Methods A total of 127 patients were divided into a training set and testing set according to the ratio of 7:3. Radiomic features were extracted from multiphase CE-MRI (CEm). Rad-score was calculated using the LASSO (least absolute shrinkage and selection operator) regression analysis. The cutoff point of Rad-score to divide the patients into high- and low-risk groups was determined by receiver operating characteristic curve analysis. A Kaplan–Meier survival curves and the Log rank test were used to investigate the association of the Rad-score with DFS. Univariate and multivariate Cox proportional hazards model were used to determine the association of Rad-score, MRI features, and clinicopathological variables with DFS. A radiomics-clinicoradiologic-based nomogram combining the Rad-score, MRI features, and clinicopathological findings was plotted to validate the radiomic signatures for DFS estimation. Results The Rad-score stratified patients into high- and low-risk groups for DFS in the training set (P<0.0001) and was validated in the testing set (P=0.002). The radiomics-clinicoradiologic-based nomogram estimated DFS (training set: C-index=0.974, 95% confidence interval (CI)=0.954–0.994; testing set: C-index=0.917, 95% CI=0.842–0.991) better than the clinicoradiologic-based nomogram (training set: C-index=0.855, 95% CI=0.739–0.971; testing set: C-index=0.831, 95% CI=0.643–0.999). Conclusion The Rad-score is an independent biomarker for the estimation of DFS in invasive HER2-positive breast cancer with NAC and the radiomics-clinicoradiologic-based nomogram improved individualized DFS estimation.
Collapse
Affiliation(s)
- Qin Li
- Shanghai Institute of Medical Imaging, Shanghai, China.,Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Qin Xiao
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jianwei Li
- Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | | | - He Wang
- Shanghai Center for Mathematical Sciences, Fudan University, Shanghai, China
| | - Yajia Gu
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| |
Collapse
|
31
|
Shiyam Sundar LK, Muzik O, Buvat I, Bidaut L, Beyer T. Potentials and caveats of AI in hybrid imaging. Methods 2020; 188:4-19. [PMID: 33068741 DOI: 10.1016/j.ymeth.2020.10.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 12/18/2022] Open
Abstract
State-of-the-art patient management frequently mandates the investigation of both anatomy and physiology of the patients. Hybrid imaging modalities such as the PET/MRI, PET/CT and SPECT/CT have the ability to provide both structural and functional information of the investigated tissues in a single examination. With the introduction of such advanced hardware fusion, new problems arise such as the exceedingly large amount of multi-modality data that requires novel approaches of how to extract a maximum of clinical information from large sets of multi-dimensional imaging data. Artificial intelligence (AI) has emerged as one of the leading technologies that has shown promise in facilitating highly integrative analysis of multi-parametric data. Specifically, the usefulness of AI algorithms in the medical imaging field has been heavily investigated in the realms of (1) image acquisition and reconstruction, (2) post-processing and (3) data mining and modelling. Here, we aim to provide an overview of the challenges encountered in hybrid imaging and discuss how AI algorithms can facilitate potential solutions. In addition, we highlight the pitfalls and challenges in using advanced AI algorithms in the context of hybrid imaging and provide suggestions for building robust AI solutions that enable reproducible and transparent research.
Collapse
Affiliation(s)
- Lalith Kumar Shiyam Sundar
- QIMP Team, Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | | | - Irène Buvat
- Laboratoire d'Imagerie Translationnelle en Oncologie, Inserm, Institut Curie, Orsay, France
| | - Luc Bidaut
- College of Science, University of Lincoln, Lincoln, UK
| | - Thomas Beyer
- QIMP Team, Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
32
|
Discovery and Validation of a CT-Based Radiomic Signature for Preoperative Prediction of Early Recurrence in Hypopharyngeal Carcinoma. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4340521. [PMID: 32851071 PMCID: PMC7436349 DOI: 10.1155/2020/4340521] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/28/2020] [Accepted: 06/22/2020] [Indexed: 12/16/2022]
Abstract
Purpose In the clinical management of hypopharyngeal squamous cell carcinoma (HSCC), preoperative identification of early recurrence (≤2 years) after curative resection is essential. Thus, we aimed to develop a CT-based radiomic signature to predict early recurrence in HSCC patients preoperatively. Methods In total, 167 HSCC patients who underwent partial surgery were enrolled in this retrospective study and divided into two groups, i.e., the training cohort (n = 133) and the validation cohort (n = 34). Each individual was followed up for at least for 2 years. Radiomic features were extracted from CT images, and the radiomic signature was built with the least absolute shrinkage and selection operator (LASSO) logistic regression (LR) model. The associations of preoperative clinical factors with early recurrence were evaluated. A radiomic signature-combined model was built, and the area under the curve (AUC) was used to explore their performance in discriminating early recurrence. Results Among the 1415 features, 335 of them were selected using the variance threshold method. Then, the SelectKBest method was further used for the selection of 31 candidate features. Finally, 11 out of 31 optimal features were identified with the LASSO algorithm. In the LR classifier, the AUCs of the training and validation sets in discriminating early recurrence were 0.83 (95% CI: 0.76-0.90) (sensitivity 0.8 and specificity 0.83) and 0.83 (95% CI: 0.67-0.99) (sensitivity 0.69 and specificity 0.71), respectively. Conclusions Using the radiomic signature, we developed a radiomic signature to preoperatively predict early recurrence in patients with HSCC, which may serve as a potential noninvasive tool to guide personalized treatment.
Collapse
|
33
|
Shen H, Wang Y, Liu D, Lv R, Huang Y, Peng C, Jiang S, Wang Y, He Y, Lan X, Huang H, Sun J, Zhang J. Predicting Progression-Free Survival Using MRI-Based Radiomics for Patients With Nonmetastatic Nasopharyngeal Carcinoma. Front Oncol 2020; 10:618. [PMID: 32477932 PMCID: PMC7235342 DOI: 10.3389/fonc.2020.00618] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 04/03/2020] [Indexed: 12/14/2022] Open
Abstract
Objectives: This study aimed to explore the predictive value of MRI-based radiomic model for progression-free survival (PFS) in nonmetastatic nasopharyngeal carcinoma (NPC). Methods: A total of 327 nonmetastatic NPC patients [training cohort (n = 230) and validation cohort (n = 97)] were enrolled. The clinical and MRI data were collected. The least absolute shrinkage selection operator (LASSO) and recursive feature elimination (RFE) were used to select radiomic features. Five models [Model 1: clinical data, Model 2: overall stage, Model 3: radiomics, Model 4: radiomics + overall stage, Model 5: radiomics + overall stage + Epstein–Barr virus (EBV) DNA] were constructed. The prognostic performances of these models were evaluated by Harrell's concordance index (C-index). The Kaplan–Meier method was applied for the survival analysis. Results: Model 5 incorporating radiomics, overall stage, and EBV DNA yielded the highest C-indices for predicting PFS in comparison with Model 1, Model 2, Model 3, and Model 4 (training cohorts: 0.805 vs. 0.766 vs. 0.749 vs. 0.641 vs. 0.563, validation cohorts: 0.874 vs. 0.839 vs. 836 vs. 0.689 vs. 0.456). The survival curve showed that the high-risk group yielded a lower PFS than the low-risk group. Conclusions: The model incorporating radiomics, overall stage, and EBV DNA showed better performance for predicting PFS in nonmetastatic NPC patients.
Collapse
Affiliation(s)
- Hesong Shen
- Department of Radiology, Chongqing University Cancer Hospital and Chongqing Cancer Institute and Chongqing Cancer Hospital, Chongqing, China.,Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), 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.,Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), 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.,Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), Chongqing University Cancer Hospital and Chongqing Cancer Institute and Chongqing Cancer Hospital, Chongqing, China
| | - Rongfei Lv
- Key Laboratory of Optoelectronic Technology and Systems of the Education Ministry of China, Chongqing University, Chongqing, China
| | - Yuanying Huang
- Department of Oncology and Hematology, Chongqing General Hospital, Chongqing, China
| | - Chao Peng
- Key Laboratory of Optoelectronic Technology and Systems of the Education Ministry of China, Chongqing University, Chongqing, China
| | - Shixi Jiang
- Department of Radiology, Chongqing University Cancer Hospital and Chongqing Cancer Institute and Chongqing Cancer Hospital, Chongqing, China
| | - Ying Wang
- Department of Radiotherapy, Chongqing University Cancer Hospital and Chongqing Cancer Institute and Chongqing Cancer Hospital, Chongqing, China
| | - Yongpeng He
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital and Chongqing Cancer Institute and Chongqing Cancer Hospital, Chongqing, China
| | - Xiaosong Lan
- Department of Radiology, Chongqing University Cancer Hospital and Chongqing Cancer Institute and Chongqing Cancer Hospital, Chongqing, China
| | - Hong Huang
- Key Laboratory of Optoelectronic Technology and Systems of the Education Ministry of China, Chongqing University, Chongqing, China
| | - Jianqing Sun
- Clinical Science, Philips Healthcare, Shanghai, China
| | - Jiuquan Zhang
- Department of Radiology, Chongqing University Cancer Hospital and Chongqing Cancer Institute and Chongqing Cancer Hospital, Chongqing, China.,Key Laboratory for Biorheological Science and Technology of Ministry of Education (Chongqing University), Chongqing University Cancer Hospital and Chongqing Cancer Institute and Chongqing Cancer Hospital, Chongqing, China
| |
Collapse
|
34
|
Haider SP, Burtness B, Yarbrough WG, Payabvash S. Applications of radiomics in precision diagnosis, prognostication and treatment planning of head and neck squamous cell carcinomas. CANCERS OF THE HEAD & NECK 2020; 5:6. [PMID: 32391171 PMCID: PMC7197186 DOI: 10.1186/s41199-020-00053-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 03/09/2020] [Indexed: 12/15/2022]
Abstract
Recent advancements in computational power, machine learning, and artificial intelligence technology have enabled automated evaluation of medical images to generate quantitative diagnostic and prognostic biomarkers. Such objective biomarkers are readily available and have the potential to improve personalized treatment, precision medicine, and patient selection for clinical trials. In this article, we explore the merits of the most recent addition to the “-omics” concept for the broader field of head and neck cancer – “Radiomics”. This review discusses radiomics studies focused on (molecular) characterization, classification, prognostication and treatment guidance for head and neck squamous cell carcinomas (HNSCC). We review the underlying hypothesis, general concept and typical workflow of radiomic analysis, and elaborate on current and future challenges to be addressed before routine clinical application.
Collapse
Affiliation(s)
- Stefan P Haider
- 1Department of Radiology and Biomedical Imaging, Division of Neuroradiology, Yale School of Medicine, New Haven, CT USA.,2Department of Otorhinolaryngology, University Hospital of Ludwig Maximilians University of Munich, Munich, Germany
| | - Barbara Burtness
- 3Department of Internal Medicine, Division of Medical Oncology, Yale School of Medicine, New Haven, CT USA
| | - Wendell G Yarbrough
- 4Department of Otolaryngology/Head and Neck Surgery, University of North Carolina School of Medicine, Chapel Hill, NC USA
| | - Seyedmehdi Payabvash
- 1Department of Radiology and Biomedical Imaging, Division of Neuroradiology, Yale School of Medicine, New Haven, CT USA
| |
Collapse
|