1
|
Vos D, Yaffe N, Cabrera CI, Fowler NM, D'Anza BD. Diagnostic Performance of Radiomics Modeling in Predicting the Human Papillomavirus Status of Oropharyngeal Cancer: A Systematic Review and Meta-Analysis. Cureus 2025; 17:e82085. [PMID: 40351986 PMCID: PMC12066096 DOI: 10.7759/cureus.82085] [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] [Accepted: 04/11/2025] [Indexed: 05/14/2025] Open
Abstract
In this review, we sought to assess the diagnostic performance and methodological quality of studies utilizing radiomics for the prediction of human papillomavirus (HPV) status in patients with oropharyngeal squamous cell carcinoma. A comprehensive literature search of PubMed, Ovid, Cochrane, Web of Science, and Scopus from inception until June 7, 2022, was performed to identify eligible studies. Strict inclusion and exclusion criteria were applied to the identified studies. Data collection was performed by two independent reviewers with disagreements resolved by consensus review with a third reviewer. In total, 14 articles were chosen, with a total of 15 radiomics models. Of the included studies, 12 models reported sensitivity, with a mean of 0.778 (standard deviation (SD) = 0.073). Similarly, 12 models reported specificity, with a mean of 0.751 (SD = 0.111). The area under the curve (AUC) was reported by all 15 models, with a mean of 0.814 (SD = 0.081). Finally, accuracy was reported by eight models, with a mean of 0.768 (SD = 0.044). A meta-analysis was performed on eight studies that reported AUCs with confidence intervals (CIs), returning a pooled AUC of 0.764 (95% CI = 0.758 to 0.770). The Radiomics Quality Score (RQS) was applied to each included study as a measure of quality. RQS ranged from -1 to 22, with a mean of 13.4 and an intraclass coefficient of 0.874. Radiomics modeling has shown promise in serving as a diagnostic indicator for HPV status in patients with oropharyngeal cancer. Nevertheless, the quality of research methodologies in this area is a limiting factor for its broader clinical application and highlights the need for enhanced funding to support further research efforts.
Collapse
Affiliation(s)
- Derek Vos
- Otolaryngology, Case Western Reserve University School of Medicine, Cleveland, USA
| | - Noah Yaffe
- Otolaryngology, Case Western Reserve University School of Medicine, Cleveland, USA
| | - Claudia I Cabrera
- Otolaryngology - Head and Neck Surgery, University Hospitals Cleveland Medical Center, Cleveland, USA
| | - Nicole M Fowler
- Otolaryngology - Head and Neck Surgery, University Hospitals Cleveland Medical Center, Cleveland, USA
| | - Brian D D'Anza
- Otolaryngology - Head and Neck Surgery, University Hospitals Cleveland Medical Center, Cleveland, USA
| |
Collapse
|
2
|
Alapati R, Renslo B, Wagoner SF, Karadaghy O, Serpedin A, Kim YE, Feucht M, Wang N, Ramesh U, Bon Nieves A, Lawrence A, Virgen C, Sawaf T, Rameau A, Bur AM. Assessing the Reporting Quality of Machine Learning Algorithms in Head and Neck Oncology. Laryngoscope 2025; 135:687-694. [PMID: 39258420 DOI: 10.1002/lary.31756] [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: 02/07/2024] [Revised: 07/25/2024] [Accepted: 08/23/2024] [Indexed: 09/12/2024]
Abstract
OBJECTIVE This study aimed to assess reporting quality of machine learning (ML) algorithms in the head and neck oncology literature using the TRIPOD-AI criteria. DATA SOURCES A comprehensive search was conducted using PubMed, Scopus, Embase, and Cochrane Database of Systematic Reviews, incorporating search terms related to "artificial intelligence," "machine learning," "deep learning," "neural network," and various head and neck neoplasms. REVIEW METHODS Two independent reviewers analyzed each published study for adherence to the 65-point TRIPOD-AI criteria. Items were classified as "Yes," "No," or "NA" for each publication. The proportion of studies satisfying each TRIPOD-AI criterion was calculated. Additionally, the evidence level for each study was evaluated independently by two reviewers using the Oxford Centre for Evidence-Based Medicine (OCEBM) Levels of Evidence. Discrepancies were reconciled through discussion until consensus was reached. RESULTS The study highlights the need for improvements in ML algorithm reporting in head and neck oncology. This includes more comprehensive descriptions of datasets, standardization of model performance reporting, and increased sharing of ML models, data, and code with the research community. Adoption of TRIPOD-AI is necessary for achieving standardized ML research reporting in head and neck oncology. CONCLUSION Current reporting of ML algorithms hinders clinical application, reproducibility, and understanding of the data used for model training. To overcome these limitations and improve patient and clinician trust, ML developers should provide open access to models, code, and source data, fostering iterative progress through community critique, thus enhancing model accuracy and mitigating biases. LEVEL OF EVIDENCE NA Laryngoscope, 135:687-694, 2025.
Collapse
Affiliation(s)
- Rahul Alapati
- Department of Otolaryngology-Head & Neck Surgery, University of Kansas Medical Center, Kansas City, Kansas, U.S.A
| | - Bryan Renslo
- Department of Otolaryngology-Head & Neck Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, U.S.A
| | - Sarah F Wagoner
- Department of Otolaryngology-Head & Neck Surgery, University of Kansas Medical Center, Kansas City, Kansas, U.S.A
| | - Omar Karadaghy
- Department of Otolaryngology-Head & Neck Surgery, University of Kansas Medical Center, Kansas City, Kansas, U.S.A
| | - Aisha Serpedin
- Department of Otolaryngology-Head & Neck Surgery, Weill Cornell, New York City, New York, U.S.A
| | - Yeo Eun Kim
- Department of Otolaryngology-Head & Neck Surgery, Weill Cornell, New York City, New York, U.S.A
| | - Maria Feucht
- Department of Otolaryngology-Head & Neck Surgery, University of Kansas Medical Center, Kansas City, Kansas, U.S.A
| | - Naomi Wang
- Department of Otolaryngology-Head & Neck Surgery, University of Kansas Medical Center, Kansas City, Kansas, U.S.A
| | - Uma Ramesh
- Department of Otolaryngology-Head & Neck Surgery, University of Kansas Medical Center, Kansas City, Kansas, U.S.A
| | - Antonio Bon Nieves
- Department of Otolaryngology-Head & Neck Surgery, University of Kansas Medical Center, Kansas City, Kansas, U.S.A
| | - Amelia Lawrence
- Department of Otolaryngology-Head & Neck Surgery, University of Kansas Medical Center, Kansas City, Kansas, U.S.A
| | - Celina Virgen
- Department of Otolaryngology-Head & Neck Surgery, University of Kansas Medical Center, Kansas City, Kansas, U.S.A
| | - Tuleen Sawaf
- Department of Otolaryngology-Head & Neck Surgery, University of Maryland, Baltimore, Maryland, U.S.A
| | - Anaïs Rameau
- Department of Otolaryngology-Head & Neck Surgery, Weill Cornell, New York City, New York, U.S.A
| | - Andrés M Bur
- Department of Otolaryngology-Head & Neck Surgery, University of Kansas Medical Center, Kansas City, Kansas, U.S.A
| |
Collapse
|
3
|
Nakajo M, Hirahara D, Jinguji M, Hirahara M, Tani A, Nagano H, Takumi K, Kamimura K, Kanzaki F, Yamashita M, Yoshiura T. Applying deep learning-based ensemble model to [ 18F]-FDG-PET-radiomic features for differentiating benign from malignant parotid gland diseases. Jpn J Radiol 2025; 43:91-100. [PMID: 39254903 PMCID: PMC11717794 DOI: 10.1007/s11604-024-01649-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 08/26/2024] [Indexed: 09/11/2024]
Abstract
OBJECTIVES To develop and identify machine learning (ML) models using pretreatment 2-deoxy-2-[18F]fluoro-D-glucose ([18F]-FDG)-positron emission tomography (PET)-based radiomic features to differentiate benign from malignant parotid gland diseases (PGDs). MATERIALS AND METHODS This retrospective study included 62 patients with 63 PGDs who underwent pretreatment [18F]-FDG-PET/computed tomography (CT). The lesions were assigned to the training (n = 44) and testing (n = 19) cohorts. In total, 49 [18F]-FDG-PET-based radiomic features were utilized to differentiate benign from malignant PGDs using five different conventional ML algorithmic models (random forest, neural network, k-nearest neighbors, logistic regression, and support vector machine) and the deep learning (DL)-based ensemble ML model. In the training cohort, each conventional ML model was constructed using the five most important features selected by the recursive feature elimination method with the tenfold cross-validation and synthetic minority oversampling technique. The DL-based ensemble ML model was constructed using the five most important features of the bagging and multilayer stacking methods. The area under the receiver operating characteristic curves (AUCs) and accuracies were used to compare predictive performances. RESULTS In total, 24 benign and 39 malignant PGDs were identified. Metabolic tumor volume and four GLSZM features (GLSZM_ZSE, GLSZM_SZE, GLSZM_GLNU, and GLSZM_ZSNU) were the five most important radiomic features. All five features except GLSZM_SZE were significantly higher in malignant PGDs than in benign ones (each p < 0.05). The DL-based ensemble ML model had the best performing classifier in the training and testing cohorts (AUC = 1.000, accuracy = 1.000 vs AUC = 0.976, accuracy = 0.947). CONCLUSIONS The DL-based ensemble ML model using [18F]-FDG-PET-based radiomic features can be useful for differentiating benign from malignant PGDs. The DL-based ensemble ML model using [18F]-FDG-PET-based radiomic features can overcome the previously reported limitation of [18F]-FDG-PET/CT scan for differentiating benign from malignant PGDs. The DL-based ensemble ML approach using [18F]-FDG-PET-based radiomic features can provide useful information for managing PGD.
Collapse
Affiliation(s)
- Masatoyo Nakajo
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan.
| | - Daisuke Hirahara
- Department of Management Planning Division, Harada Academy, 2-54-4 Higashitaniyama, Kagoshima, 890-0113, Japan
| | - Megumi Jinguji
- Department of Radiology, Nanpuh Hospital, 14-3 Nagata, Kagoshima, 892-8512, Japan
| | - Mitsuho Hirahara
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Atsushi Tani
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Hiromi Nagano
- Department of Otolaryngology Head and Neck Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Koji Takumi
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Kiyohisa Kamimura
- Department of Advanced Radiological Imaging, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Fumiko Kanzaki
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Masaru Yamashita
- Department of Otolaryngology Head and Neck Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Takashi Yoshiura
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| |
Collapse
|
4
|
Lyu W, Gong J, Zhu L, Xu T, Huang S, Shen C, Wang C, He X, Ying H, Hu C, Wang Y, Ji Q, Gu Y, Zhou X, Lu X. MR radiomics unveils neoadjuvant chemo-responsiveness with insights into selective treatment de-intensification in HPV-positive oropharyngeal carcinoma. Oral Oncol 2024; 159:107049. [PMID: 39341091 DOI: 10.1016/j.oraloncology.2024.107049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 08/26/2024] [Accepted: 09/21/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND Accurate prediction of neoadjuvant chemotherapy (NAC) response allows for NAC-guided personalized treatment de-intensification in HPV-positive oropharyngeal squamous cell carcinoma (OPSCC). In this study, we aimed to apply baseline MR radiomic features to predict NAC response to help select NAC-guided de-intensification candidates, and to explore biological underpinnings of response-oriented radiomics. METHODS Pre-treatment MR images and clinical data of 131 patients with HPV-positive OPSCC were retrieved from Fudan University Shanghai Cancer Center. Patients were divided into training cohort (n = 47), validation cohort 1 (n = 49) from NAC response-adapted de-intensification trial (IChoice-01, NCT04012502) and real-world validation cohort 2 (n = 35). NAC prediction model using linear support vector machine (SVM) was built and validated. Subsequent nomograms combined radiomics and clinical characteristics were established to predict survival outcomes. RNA-seq and proteomic data were compared to interpret the molecular features underlying radiomic signatures with differential NAC response. FINDINGS For NAC response prediction, the fusion model with both oropharyngeal and nodal signatures achieved encouraging performance to predict good responders in the training cohort (AUC 0·89, 95% CI, 0·79-0·95) and validation cohort 1 (AUC 0·71, 95% CI, 0·59-0·83). For prognosis prediction, radiomics-based nomograms exhibited satisfactory discriminative ability between low-risk and high-risk patients (PFS, C-index 0·85, 0·76 and 0·83; OS, C-index 0·79, 0·76 and 0·87, respectively) in three cohorts. Expression analysis unveiled NAC poor responders had predominantly enhanced keratinization while good responders were featured by upregulated immune response and oxidative stress. INTERPRETATION The MR-based radiomic models and prognostic models efficiently discriminate among patients with different NAC response and survival risk, which help candidate selection in HPV-positive OPSCC with regard to personalized treatment de-intensification.
Collapse
Affiliation(s)
- Wenjiao Lyu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
| | - Jing Gong
- Department of Radiology, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
| | - Lin Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
| | - Tingting Xu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
| | - Shenglin Huang
- The Shanghai Key Laboratory of Medical Epigenetics, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
| | - Chunying Shen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
| | - Cuihong Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
| | - Xiayun He
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
| | - Hongmei Ying
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
| | - Chaosu Hu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
| | - Yu Wang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
| | - Qinghai Ji
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, 200032 Shanghai, China
| | - Yajia Gu
- Department of Radiology, Fudan University Shanghai Cancer Center, 200032 Shanghai, China.
| | - Xin Zhou
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 200032 Shanghai, China.
| | - Xueguan Lu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 200032 Shanghai, China.
| |
Collapse
|
5
|
Oliver J, Alapati R, Lee J, Bur A. Artificial Intelligence in Head and Neck Surgery. Otolaryngol Clin North Am 2024; 57:803-820. [PMID: 38910064 PMCID: PMC11374486 DOI: 10.1016/j.otc.2024.05.001] [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] [Indexed: 06/25/2024]
Abstract
This article explores artificial intelligence's (AI's) role in otolaryngology for head and neck cancer diagnosis and management. It highlights AI's potential in pattern recognition for early cancer detection, prognostication, and treatment planning, primarily through image analysis using clinical, endoscopic, and histopathologic images. Radiomics is also discussed at length, as well as the many ways that radiologic image analysis can be utilized, including for diagnosis, lymph node metastasis prediction, and evaluation of treatment response. The study highlights AI's promise and limitations, underlining the need for clinician-data scientist collaboration to enhance head and neck cancer care.
Collapse
Affiliation(s)
- Jamie Oliver
- Department of Otolaryngology-Head and Neck Surgery, University of Kansas School of Medicine, 3901 Rainbow Boulevard M.S. 3010, Kansas City, KS, USA
| | - Rahul Alapati
- Department of Otolaryngology-Head and Neck Surgery, University of Kansas School of Medicine, 3901 Rainbow Boulevard M.S. 3010, Kansas City, KS, USA
| | - Jason Lee
- Department of Otolaryngology-Head and Neck Surgery, University of Kansas School of Medicine, 3901 Rainbow Boulevard M.S. 3010, Kansas City, KS, USA
| | - Andrés Bur
- Department of Otolaryngology-Head and Neck Surgery, University of Kansas School of Medicine, 3901 Rainbow Boulevard M.S. 3010, Kansas City, KS, USA.
| |
Collapse
|
6
|
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
|
7
|
Alabi RO, Elmusrati M, Leivo I, Almangush A, Mäkitie AA. Artificial Intelligence-Driven Radiomics in Head and Neck Cancer: Current Status and Future Prospects. Int J Med Inform 2024; 188:105464. [PMID: 38728812 DOI: 10.1016/j.ijmedinf.2024.105464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 04/20/2024] [Accepted: 04/22/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND Radiomics is a rapidly growing field used to leverage medical radiological images by extracting quantitative features. These are supposed to characterize a patient's phenotype, and when combined with artificial intelligence techniques, to improve the accuracy of diagnostic models and clinical outcome prediction. OBJECTIVES This review aims at examining the application areas of artificial intelligence-based radiomics (AI-based radiomics) for the management of head and neck cancer (HNC). It further explores the workflow of AI-based radiomics for personalized and precision oncology in HNC. Finally, it examines the current challenges of AI-based radiomics in daily clinical oncology and offers possible solutions to these challenges. METHODS Comprehensive electronic databases (PubMed, Medline via Ovid, Scopus, Web of Science, CINAHL, and Cochrane Library) were searched following the Preferred Reporting Items for Systematic Review and Meta-Analysis (PRISMA) guidelines. The quality of included studies and their risk of biases were evaluated using the Transparent Reporting of a Multivariable Prediction Model for Individual Prognosis or Diagnosis (TRIPOD)and Prediction Model Risk of Bias Assessment Tool (PROBAST). RESULTS Out of the 659 search hits retrieved, 45 fulfilled the inclusion criteria. Our review revealed that the application of AI-based radiomics model as an ancillary tool for improved decision-making in HNC management includes radiomics-based cancer diagnosis and radiomics-based cancer prognosis. The radiomics-based cancer diagnosis includes tumor staging, tumor grading, and classification of malignant and benign tumors. Similarly, radiomics-based cancer prognosis includes prediction for treatment response, recurrence, metastasis, and survival. In addition, the challenges in the implementation of these models for clinical evaluations include data imbalance, feature engineering (extraction and selection), model generalizability, multi-modal fusion, and model interpretability. CONCLUSION Considering the highly subjective and interobserver variability that is peculiar to the interpretation of medical images by expert clinicians, AI-based radiomics seeks to offer potentially useful quantitative information, which is not visible to the human eye or unintentionally often remain ignored during clinical imaging practice. By enabling the extraction of this type of information, AI-based radiomics has the potential to revolutionize HNC oncology, providing a platform for more personalized, higher quality, and cost-effective care for HNC patients.
Collapse
Affiliation(s)
- Rasheed Omobolaji Alabi
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Industrial Digitalization, School of Technology and Innovations, University of Vaasa, Vaasa, Finland.
| | - Mohammed Elmusrati
- Department of Industrial Digitalization, School of Technology and Innovations, University of Vaasa, Vaasa, Finland
| | - Ilmo Leivo
- University of Turku, Institute of Biomedicine, Pathology, Turku, Finland
| | - Alhadi Almangush
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland; University of Turku, Institute of Biomedicine, Pathology, Turku, Finland; Department of Pathology, University of Helsinki, Helsinki, Finland; Faculty of Dentistry, Misurata University, Misurata, Libya
| | - Antti A Mäkitie
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Otorhinolaryngology - Head and Neck Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Division of Ear, Nose and Throat Diseases, Department of Clinical Sciences, Intervention and Technology, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
8
|
Bicci E, Calamandrei L, Di Finizio A, Pietragalla M, Paolucci S, Busoni S, Mungai F, Nardi C, Bonasera L, Miele V. Predicting Response to Exclusive Combined Radio-Chemotherapy in Naso-Oropharyngeal Cancer: The Role of Texture Analysis. Diagnostics (Basel) 2024; 14:1036. [PMID: 38786334 PMCID: PMC11120575 DOI: 10.3390/diagnostics14101036] [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: 03/31/2024] [Revised: 04/28/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024] Open
Abstract
The aim of this work is to identify MRI texture features able to predict the response to radio-chemotherapy (RT-CHT) in patients with naso-oropharyngeal carcinoma (NPC-OPC) before treatment in order to help clinical decision making. Textural features were derived from ADC maps and post-gadolinium T1-images on a single MRI machine for 37 patients with NPC-OPC. Patients were divided into two groups (responders/non-responders) according to results from MRI scans and 18F-FDG-PET/CT performed at follow-up 3-4 and 12 months after therapy and biopsy. Pre-RT-CHT lesions were segmented, and radiomic features were extracted. A non-parametric Mann-Whitney test was performed. A p-value < 0.05 was considered significant. Receiver operating characteristic curves and area-under-the-curve values were generated; a 95% confidence interval (CI) was reported. A radiomic model was constructed using the LASSO algorithm. After feature selection on MRI T1 post-contrast sequences, six features were statistically significant: gldm_DependenceEntropy and DependenceNonUniformity, glrlm_RunEntropy and RunLengthNonUniformity, and glszm_SizeZoneNonUniformity and ZoneEntropy, with significant cut-off values between responder and non-responder group. With the LASSO algorithm, the radiomic model showed an AUC of 0.89 and 95% CI: 0.78-0.99. In ADC, five features were selected with an AUC of 0.84 and 95% CI: 0.68-1. Texture analysis on post-gadolinium T1-images and ADC maps could potentially predict response to therapy in patients with NPC-OPC who will undergo exclusive treatment with RT-CHT, being, therefore, a useful tool in therapeutical-clinical decision making.
Collapse
Affiliation(s)
- Eleonora Bicci
- Department of Radiology, Azienda Ospedaliero-Universitaria Careggi, 50134 Florence, Italy; (F.M.); (L.B.); (V.M.)
| | - Leonardo Calamandrei
- Department of Experimental and Clinical Biomedical Sciences, Radiodiagnostic Unit n. 2, University of Florence, Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla 3, 50134 Florence, Italy; (L.C.); (A.D.F.) (C.N.)
| | - Antonio Di Finizio
- Department of Experimental and Clinical Biomedical Sciences, Radiodiagnostic Unit n. 2, University of Florence, Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla 3, 50134 Florence, Italy; (L.C.); (A.D.F.) (C.N.)
| | - Michele Pietragalla
- Department of Radiology, Ospedale San Jacopo, Via Ciliegiole 97, 51100 Pistoia, Italy;
| | - Sebastiano Paolucci
- Department of Health Physics, L.Go Brambilla, Careggi University Hospital, 50134 Florence, Italy; (S.P.); (S.B.)
| | - Simone Busoni
- Department of Health Physics, L.Go Brambilla, Careggi University Hospital, 50134 Florence, Italy; (S.P.); (S.B.)
| | - Francesco Mungai
- Department of Radiology, Azienda Ospedaliero-Universitaria Careggi, 50134 Florence, Italy; (F.M.); (L.B.); (V.M.)
| | - Cosimo Nardi
- Department of Experimental and Clinical Biomedical Sciences, Radiodiagnostic Unit n. 2, University of Florence, Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla 3, 50134 Florence, Italy; (L.C.); (A.D.F.) (C.N.)
| | - Luigi Bonasera
- Department of Radiology, Azienda Ospedaliero-Universitaria Careggi, 50134 Florence, Italy; (F.M.); (L.B.); (V.M.)
| | - Vittorio Miele
- Department of Radiology, Azienda Ospedaliero-Universitaria Careggi, 50134 Florence, Italy; (F.M.); (L.B.); (V.M.)
| |
Collapse
|
9
|
Sim Y, Kim M, Kim J, Lee SK, Han K, Sohn B. Multiparametric MRI-based radiomics model for predicting human papillomavirus status in oropharyngeal squamous cell carcinoma: optimization using oversampling and machine learning techniques. Eur Radiol 2024; 34:3102-3112. [PMID: 37848774 DOI: 10.1007/s00330-023-10338-3] [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: 08/29/2022] [Revised: 07/08/2023] [Accepted: 08/20/2023] [Indexed: 10/19/2023]
Abstract
OBJECTIVES To develop and validate a multiparametric MRI-based radiomics model with optimal oversampling and machine learning techniques for predicting human papillomavirus (HPV) status in oropharyngeal squamous cell carcinoma (OPSCC). METHODS This retrospective, multicenter study included consecutive patients with newly diagnosed and pathologically confirmed OPSCC between January 2017 and December 2020 (110 patients in the training set, 44 patients in the external validation set). A total of 293 radiomics features were extracted from three sequences (T2-weighted images [T2WI], contrast-enhanced T1-weighted images [CE-T1WI], and ADC). Combinations of three feature selection, five oversampling, and 12 machine learning techniques were evaluated to optimize its diagnostic performance. The area under the receiver operating characteristic curve (AUC) of the top five models was validated in the external validation set. RESULTS A total of 154 patients (59.2 ± 9.1 years; 132 men [85.7%]) were included, and oversampling was employed to account for data imbalance between HPV-positive and HPV-negative OPSCC (86.4% [133/154] vs. 13.6% [21/154]). For the ADC radiomics model, the combination of random oversampling and ridge showed the highest diagnostic performance in the external validation set (AUC, 0.791; 95% CI, 0.775-0.808). The ADC radiomics model showed a higher trend in diagnostic performance compared to the radiomics model using CE-T1WI (AUC, 0.604; 95% CI, 0.590-0.618), T2WI (AUC, 0.695; 95% CI, 0.673-0.717), and a combination of both (AUC, 0.642; 95% CI, 0.626-0.657). CONCLUSIONS The ADC radiomics model using random oversampling and ridge showed the highest diagnostic performance in predicting the HPV status of OPSCC in the external validation set. CLINICAL RELEVANCE STATEMENT Among multiple sequences, the ADC radiomics model has a potential for generalizability and applicability in clinical practice. Exploring multiple oversampling and machine learning techniques was a valuable strategy for optimizing radiomics model performance. KEY POINTS • Previous radiomics studies using multiparametric MRI were conducted at single centers without external validation and had unresolved data imbalances. • Among the ADC, CE-T1WI, and T2WI radiomics models and the ADC histogram models, the ADC radiomics model was the best-performing model for predicting human papillomavirus status in oropharyngeal squamous cell carcinoma. • The ADC radiomics model with the combination of random oversampling and ridge showed the highest diagnostic performance.
Collapse
Affiliation(s)
- Yongsik Sim
- Department of Radiology, Research Institute of Radiological Science, Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Minjae Kim
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Jinna Kim
- Department of Radiology, Research Institute of Radiological Science, Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Seung-Koo Lee
- Department of Radiology, Research Institute of Radiological Science, Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Kyunghwa Han
- Department of Radiology, Research Institute of Radiological Science, Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Beomseok Sohn
- Department of Radiology, Research Institute of Radiological Science, Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, South Korea.
- Department of Radiology and Center for Imaging Sciences, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| |
Collapse
|
10
|
Haider SP, Zeevi T, Sharaf K, Gross M, Mahajan A, Kann BH, Judson BL, Prasad ML, Burtness B, Aboian M, Canis M, Reichel CA, Baumeister P, Payabvash S. Impact of 18F-FDG PET Intensity Normalization on Radiomic Features of Oropharyngeal Squamous Cell Carcinomas and Machine Learning-Generated Biomarkers. J Nucl Med 2024; 65:803-809. [PMID: 38514087 PMCID: PMC11927063 DOI: 10.2967/jnumed.123.266637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 02/13/2024] [Indexed: 03/23/2024] Open
Abstract
We aimed to investigate the effects of 18F-FDG PET voxel intensity normalization on radiomic features of oropharyngeal squamous cell carcinoma (OPSCC) and machine learning-generated radiomic biomarkers. Methods: We extracted 1,037 18F-FDG PET radiomic features quantifying the shape, intensity, and texture of 430 OPSCC primary tumors. The reproducibility of individual features across 3 intensity-normalized images (body-weight SUV, reference tissue activity ratio to lentiform nucleus of brain and cerebellum) and the raw PET data was assessed using an intraclass correlation coefficient (ICC). We investigated the effects of intensity normalization on the features' utility in predicting the human papillomavirus (HPV) status of OPSCCs in univariate logistic regression, receiver-operating-characteristic analysis, and extreme-gradient-boosting (XGBoost) machine-learning classifiers. Results: Of 1,037 features, a high (ICC ≥ 0.90), medium (0.90 > ICC ≥ 0.75), and low (ICC < 0.75) degree of reproducibility across normalization methods was attained in 356 (34.3%), 608 (58.6%), and 73 (7%) features, respectively. In univariate analysis, features from the PET normalized to the lentiform nucleus had the strongest association with HPV status, with 865 of 1,037 (83.4%) significant features after multiple testing corrections and a median area under the receiver-operating-characteristic curve (AUC) of 0.65 (interquartile range, 0.62-0.68). Similar tendencies were observed in XGBoost models, with the lentiform nucleus-normalized model achieving the numerically highest average AUC of 0.72 (SD, 0.07) in the cross validation within the training cohort. The model generalized well to the validation cohorts, attaining an AUC of 0.73 (95% CI, 0.60-0.85) in independent validation and 0.76 (95% CI, 0.58-0.95) in external validation. The AUCs of the XGBoost models were not significantly different. Conclusion: Only one third of the features demonstrated a high degree of reproducibility across intensity-normalization techniques, making uniform normalization a prerequisite for interindividual comparability of radiomic markers. The choice of normalization technique may affect the radiomic features' predictive value with respect to HPV. Our results show trends that normalization to the lentiform nucleus may improve model performance, although more evidence is needed to draw a firm conclusion.
Collapse
Affiliation(s)
- Stefan P Haider
- Department of Otorhinolaryngology, LMU Clinic of Ludwig Maximilians University of Munich, Munich, Germany;
- Section of Neuroradiology, Yale School of Medicine, New Haven, Connecticut
| | - Tal Zeevi
- Section of Neuroradiology, Yale School of Medicine, New Haven, Connecticut
| | - Kariem Sharaf
- Department of Otorhinolaryngology, LMU Clinic of Ludwig Maximilians University of Munich, Munich, Germany
| | - Moritz Gross
- Section of Neuroradiology, Yale School of Medicine, New Haven, Connecticut
- Charité Center for Diagnostic and Interventional Radiology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Amit Mahajan
- Section of Neuroradiology, Yale School of Medicine, New Haven, Connecticut
| | - Benjamin H Kann
- Department of Radiation Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Benjamin L Judson
- Division of Otolaryngology, Yale School of Medicine, New Haven, Connecticut
| | - Manju L Prasad
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut; and
| | - Barbara Burtness
- Section of Medical Oncology, Yale School of Medicine, New Haven, Connecticut
| | - Mariam Aboian
- Section of Neuroradiology, Yale School of Medicine, New Haven, Connecticut
| | - Martin Canis
- Department of Otorhinolaryngology, LMU Clinic of Ludwig Maximilians University of Munich, Munich, Germany
| | - Christoph A Reichel
- Department of Otorhinolaryngology, LMU Clinic of Ludwig Maximilians University of Munich, Munich, Germany
| | - Philipp Baumeister
- Department of Otorhinolaryngology, LMU Clinic of Ludwig Maximilians University of Munich, Munich, Germany
| | | |
Collapse
|
11
|
Ammirabile A, Mastroleo F, Marvaso G, Alterio D, Franzese C, Scorsetti M, Franco P, Giannitto C, Jereczek-Fossa BA. Mapping the research landscape of HPV-positive oropharyngeal cancer: a bibliometric analysis. Crit Rev Oncol Hematol 2024; 196:104318. [PMID: 38431241 DOI: 10.1016/j.critrevonc.2024.104318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 02/25/2024] [Accepted: 02/27/2024] [Indexed: 03/05/2024] Open
Abstract
OBJECTIVE The aim of the study is to evaluate the scientific interest, the collaboration patterns and the emerging trends regarding HPV+ OPSCC diagnosis and treatment. MATERIALS AND METHODS A cross-sectional bibliometric analysis of articles reporting on HPV+ OPSCC within Scopus database was performed and all documents published up to December 31th, 2022 were eligible for analysis. Outcomes included the exploration of key characteristics (number of manuscripts published per year, growth rate, top productive countries, most highly cited papers, and the most well-represented journals), collaboration parameters (international collaboration ratio and networks, co-occurrence networks), keywords analysis (trend topics, factorial analysis). RESULTS A total of 5200 documents were found, published from March, 1987 to December, 2022. The number of publications increased annually with an average growth rate of 19.94%, reaching a peak of 680 documents published in 2021. The 10 most cited documents (range 1105-4645) were published from 2000 to 2012. The keywords factorial analysis revealed two main clusters: one on epidemiology, diagnosis, prevention and association with other HPV tumors; the other one about the therapeutic options. According to the frequency of keywords, new items are emerging in the last three years regarding the application of Artifical Intelligence (machine learning and radiomics) and the diagnostic biomarkers (circulating tumor DNA). CONCLUSIONS This bibliometric analysis highlights the importance of research efforts in prevention, diagnostics, and treatment strategies for this disease. Given the urgency of optimizing treatment and improving clinical outcomes, further clinical trials are needed to bridge unaddressed gaps in the management of HPV+ OPSCC patients.
Collapse
Affiliation(s)
- Angela Ammirabile
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Milan, Pieve Emanuele 20090, Italy; Department of Diagnostic and Interventional Radiology, IRCCS Humanitas Research Hospital, Via Manzoni 56, Milan, Rozzano 20089, Italy
| | - Federico Mastroleo
- Department of Translational Medicine (DIMET), University of Eastern Piedmont and 'Maggiore della Carità' University Hospital, Novara, Italy; Division of Radiation Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Giulia Marvaso
- Division of Radiation Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.
| | - Daniela Alterio
- Division of Radiation Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Ciro Franzese
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Milan, Pieve Emanuele 20090, Italy; Radiotherapy and Radiosurgery Department, IRCSS Humanitas Research Hospital, Milan, Rozzano, Italy
| | - Marta Scorsetti
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Milan, Pieve Emanuele 20090, Italy; Radiotherapy and Radiosurgery Department, IRCSS Humanitas Research Hospital, Milan, Rozzano, Italy
| | - Pierfrancesco Franco
- Department of Translational Medicine (DIMET), University of Eastern Piedmont and 'Maggiore della Carità' University Hospital, Novara, Italy
| | - Caterina Giannitto
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Milan, Pieve Emanuele 20090, Italy; Department of Diagnostic and Interventional Radiology, IRCCS Humanitas Research Hospital, Via Manzoni 56, Milan, Rozzano 20089, Italy
| | - Barbara Alicja Jereczek-Fossa
- Division of Radiation Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
12
|
Ansari G, Mirza-Aghazadeh-Attari M, Mosier KM, Fakhry C, Yousem DM. Radiomics Features in Predicting Human Papillomavirus Status in Oropharyngeal Squamous Cell Carcinoma: A Systematic Review, Quality Appraisal, and Meta-Analysis. Diagnostics (Basel) 2024; 14:737. [PMID: 38611650 PMCID: PMC11011663 DOI: 10.3390/diagnostics14070737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
We sought to determine the diagnostic accuracy of radiomics features in predicting HPV status in oropharyngeal squamous cell carcinoma (SCC) compared to routine paraclinical measures used in clinical practice. Twenty-six articles were included in the systematic review, and thirteen were used for the meta-analysis. The overall sensitivity of the included studies was 0.78, the overall specificity was 0.76, and the overall area under the ROC curve was 0.84. The diagnostic odds ratio (DOR) equaled 12 (8, 17). Subgroup analysis showed no significant difference between radiomics features extracted from CT or MR images. Overall, the studies were of low quality in regard to radiomics quality score, although most had a low risk of bias based on the QUADAS-2 tool. Radiomics features showed good overall sensitivity and specificity in determining HPV status in OPSCC, though the low quality of the included studies poses problems for generalizability.
Collapse
Affiliation(s)
- Golnoosh Ansari
- Department of Radiology, Northwestern Hospital, Northwestern School of Medicine, Chicago, IL 60611, USA;
| | - Mohammad Mirza-Aghazadeh-Attari
- Division of Interventional Radiology, Department of Radiology and Radiological Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Kristine M. Mosier
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Carole Fakhry
- Department of Otolaryngology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA;
| | - David M. Yousem
- Division of Neuroradiology, Department of Radiology and Radiological Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA;
| |
Collapse
|
13
|
Avery EW, Abou-Karam A, Abi-Fadel S, Behland J, Mak A, Haider SP, Zeevi T, Sanelli PC, Filippi CG, Malhotra A, Matouk CC, Falcone GJ, Petersen N, Sansing LH, Sheth KN, Payabvash S. Radiomics-Based Prediction of Collateral Status from CT Angiography of Patients Following a Large Vessel Occlusion Stroke. Diagnostics (Basel) 2024; 14:485. [PMID: 38472957 PMCID: PMC10930945 DOI: 10.3390/diagnostics14050485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/01/2024] [Accepted: 02/15/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND A major driver of individual variation in long-term outcomes following a large vessel occlusion (LVO) stroke is the degree of collateral arterial circulation. We aimed to develop and evaluate machine-learning models that quantify LVO collateral status using admission computed tomography angiography (CTA) radiomics. METHODS We extracted 1116 radiomic features from the anterior circulation territories from admission CTAs of 600 patients experiencing an acute LVO stroke. We trained and validated multiple machine-learning models for the prediction of collateral status based on consensus from two neuroradiologists as ground truth. Models were first trained to predict (1) good vs. intermediate or poor, or (2) good vs. intermediate or poor collateral status. Then, model predictions were combined to determine a three-tier collateral score (good, intermediate, or poor). We used the receiver operating characteristics area under the curve (AUC) to evaluate prediction accuracy. RESULTS We included 499 patients in training and 101 in an independent test cohort. The best-performing models achieved an averaged cross-validation AUC of 0.80 ± 0.05 for poor vs. intermediate/good collateral and 0.69 ± 0.05 for good vs. intermediate/poor, and AUC = 0.77 (0.67-0.87) and AUC = 0.78 (0.70-0.90) in the independent test cohort, respectively. The collateral scores predicted by the radiomics model were correlated with (rho = 0.45, p = 0.002) and were independent predictors of 3-month clinical outcome (p = 0.018) in the independent test cohort. CONCLUSIONS Automated tools for the assessment of collateral status from admission CTA-such as the radiomics models described here-can generate clinically relevant and reproducible collateral scores to facilitate a timely treatment triage in patients experiencing an acute LVO stroke.
Collapse
Affiliation(s)
- Emily W. Avery
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT 06520, USA; (E.W.A.); (A.M.)
| | - Anthony Abou-Karam
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT 06520, USA; (E.W.A.); (A.M.)
| | - Sandra Abi-Fadel
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT 06520, USA; (E.W.A.); (A.M.)
| | - Jonas Behland
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT 06520, USA; (E.W.A.); (A.M.)
- CLAIM—Charité Lab for Artificial Intelligence in Medicine, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Adrian Mak
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT 06520, USA; (E.W.A.); (A.M.)
- CLAIM—Charité Lab for Artificial Intelligence in Medicine, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Stefan P. Haider
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT 06520, USA; (E.W.A.); (A.M.)
- Department of Otorhinolaryngology, University Hospital of Ludwig Maximilians Universität München, 81377 Munich, Germany
| | - Tal Zeevi
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT 06520, USA; (E.W.A.); (A.M.)
| | - Pina C. Sanelli
- Section of Neuroradiology, Department of Radiology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, Manhasset, NY 11030, USA
| | - Christopher G. Filippi
- Section of Neuroradiology, Department of Radiology, Tufts School of Medicine, Boston, MA 02111, USA
| | - Ajay Malhotra
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT 06520, USA; (E.W.A.); (A.M.)
| | - Charles C. Matouk
- Division of Neurovascular Surgery, Department of Neurosurgery, Yale School of Medicine, New Haven, CT 06520, USA
| | - Guido J. Falcone
- Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Nils Petersen
- Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Lauren H. Sansing
- Division of Stroke and Vascular Neurology, Department of Neurology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Kevin N. Sheth
- Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Seyedmehdi Payabvash
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT 06520, USA; (E.W.A.); (A.M.)
| |
Collapse
|
14
|
Nakajo M, Jinguji M, Ito S, Tani A, Hirahara M, Yoshiura T. Clinical application of 18F-fluorodeoxyglucose positron emission tomography/computed tomography radiomics-based machine learning analyses in the field of oncology. Jpn J Radiol 2024; 42:28-55. [PMID: 37526865 PMCID: PMC10764437 DOI: 10.1007/s11604-023-01476-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 07/18/2023] [Indexed: 08/02/2023]
Abstract
Machine learning (ML) analyses using 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography (PET)/computed tomography (CT) radiomics features have been applied in the field of oncology. The current review aimed to summarize the current clinical articles about 18F-FDG PET/CT radiomics-based ML analyses to solve issues in classifying or constructing prediction models for several types of tumors. In these studies, lung and mediastinal tumors were the most commonly evaluated lesions, followed by lymphatic, abdominal, head and neck, breast, gynecological, and other types of tumors. Previous studies have commonly shown that 18F-FDG PET radiomics-based ML analysis has good performance in differentiating benign from malignant tumors, predicting tumor characteristics and stage, therapeutic response, and prognosis by examining significant differences in the area under the receiver operating characteristic curves, accuracies, or concordance indices (> 0.70). However, these studies have reported several ML algorithms. Moreover, different ML models have been applied for the same purpose. Thus, various procedures were used in 18F-FDG PET/CT radiomics-based ML analysis in oncology, and 18F-FDG PET/CT radiomics-based ML models, which are easy and universally applied in clinical practice, would be expected to be established.
Collapse
Affiliation(s)
- Masatoyo Nakajo
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan.
| | - Megumi Jinguji
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Soichiro Ito
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Atushi Tani
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Mitsuho Hirahara
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Takashi Yoshiura
- Department of Radiology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| |
Collapse
|
15
|
Jo KH, Kim J, Cho H, Kang WJ, Lee SK, Sohn B. 18F-FDG PET/CT Parameters Enhance MRI Radiomics for Predicting Human Papilloma Virus Status in Oropharyngeal Squamous Cell Carcinoma. Yonsei Med J 2023; 64:738-744. [PMID: 37992746 PMCID: PMC10681825 DOI: 10.3349/ymj.2023.0187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/26/2023] [Accepted: 08/17/2023] [Indexed: 11/24/2023] Open
Abstract
PURPOSE Predicting human papillomavirus (HPV) status is critical in oropharyngeal squamous cell carcinoma (OPSCC) radiomics. In this study, we developed a model for HPV status prediction using magnetic resonance imaging (MRI) radiomics and 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography (PET)/computed tomography (CT) parameters in patients with OPSCC. MATERIALS AND METHODS Patients with OPSCC who underwent 18F-FDG PET/CT and contrast-enhanced MRI before treatment between January 2012 and February 2020 were enrolled. Training and test sets (3:2) were randomly selected. 18F-FDG PET/CT parameters and MRI radiomics feature were extracted. We developed three light-gradient boosting machine prediction models using the training set: Model 1, MRI radiomics features; Model 2, 18F-FDG PET/CT parameters; and Model 3, combination of MRI radiomics features and 18F-FDG PET/CT parameters. Area under the receiver operating characteristic curve (AUROC) values were used to analyze the performance of the models in predicting HPV status in the test set. RESULTS A total of 126 patients (118 male and 8 female; mean age: 60 years) were included. Of these, 103 patients (81.7%) were HPV-positive, and 23 patients (18.3%) were HPV-negative. AUROC values in the test set were 0.762 [95% confidence interval (CI), 0.564-0.959], 0.638 (95% CI, 0.404-0.871), and 0.823 (95% CI, 0.668-0.978) for Models 1, 2, and 3, respectively. The net reclassification improvement of Model 3, compared with that of Model 1, in the test set was 0.119. CONCLUSION When combined with an MRI radiomics model, 18F-FDG PET/CT exhibits incremental value in predicting HPV status in patients with OPSCC.
Collapse
Affiliation(s)
- Kwan Hyeong Jo
- Department of Nuclear Medicine, Korea University Guro Hospital, Seoul, Korea
| | - Jinna Kim
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hojin Cho
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Won Jun Kang
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Seung-Koo Lee
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Beomseok Sohn
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| |
Collapse
|
16
|
Bicci E, Calamandrei L, Mungai F, Granata V, Fusco R, De Muzio F, Bonasera L, Miele V. Imaging of human papilloma virus (HPV) related oropharynx tumour: what we know to date. Infect Agent Cancer 2023; 18:58. [PMID: 37814320 PMCID: PMC10563217 DOI: 10.1186/s13027-023-00530-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/11/2023] [Indexed: 10/11/2023] Open
Abstract
The tumours of head and neck district are around 3% of all malignancies and squamous cell carcinoma is the most frequent histotype, with rapid increase during the last two decades because of the increment of the infection due to human papilloma virus (HPV). Even if the gold standard for the diagnosis is histological examination, including the detection of viral DNA and transcription products, imaging plays a fundamental role in the detection and staging of HPV + tumours, in order to assess the primary tumour, to establish the extent of disease and for follow-up. The main diagnostic tools are Computed Tomography (CT), Positron Emission Tomography-Computed Tomography (PET-CT) and Magnetic Resonance Imaging (MRI), but also Ultrasound (US) and the use of innovative techniques such as Radiomics have an important role. Aim of our review is to illustrate the main imaging features of HPV + tumours of the oropharynx, in US, CT and MRI imaging. In particular, we will outline the main limitations and strengths of the various imaging techniques, the main uses in the diagnosis, staging and follow-up of disease and the fundamental differential diagnoses of this type of tumour. Finally, we will focus on the innovative technique of texture analysis, which is increasingly gaining importance as a diagnostic tool in aid of the radiologist.
Collapse
Affiliation(s)
- Eleonora Bicci
- Department of Radiology, University of Florence - Azienda Ospedaliero-Universitaria Careggi, Florence, 50134, Italy.
| | - Leonardo Calamandrei
- Department of Experimental and Clinical Biomedical Sciences, Radiodiagnostic Unit n. 2, University of Florence - Azienda Ospedaliero-Universitaria Careggi, Florence, 50134, Italy
| | - Francesco Mungai
- Department of Radiology, University of Florence - Azienda Ospedaliero-Universitaria Careggi, Florence, 50134, Italy
| | - Vincenza Granata
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale-IRCCS di Napoli, Naples, 80131, Italy
| | - Roberta Fusco
- Medical Oncology Division, Igea SpA, Naples, 80013, Italy
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Milan, 20122, Italy
| | - Federica De Muzio
- Department of Medicine and Health Sciences V. Tiberio, University of Molise, Campobasso, 86100, Italy
| | - Luigi Bonasera
- Department of Radiology, University of Florence - Azienda Ospedaliero-Universitaria Careggi, Florence, 50134, Italy
| | - Vittorio Miele
- Department of Radiology, University of Florence - Azienda Ospedaliero-Universitaria Careggi, Florence, 50134, Italy
| |
Collapse
|
17
|
Song C, Chen X, Tang C, Xue P, Jiang Y, Qiao Y. Artificial intelligence for HPV status prediction based on disease-specific images in head and neck cancer: A systematic review and meta-analysis. J Med Virol 2023; 95:e29080. [PMID: 37691329 DOI: 10.1002/jmv.29080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 07/14/2023] [Accepted: 08/03/2023] [Indexed: 09/12/2023]
Abstract
Accurate early detection of the human papillomavirus (HPV) status in head and neck cancer (HNC) is crucial to identify at-risk populations, stratify patients, personalized treatment options, and predict prognosis. Artificial intelligence (AI) is an emerging tool to dissect imaging features. This systematic review and meta-analysis aimed to evaluate the performance of AI to predict the HPV positivity through the HPV-associated diseased images in HNC patients. A systematic literature search was conducted in databases including Ovid-MEDLINE, Embase, and Web of Science Core Collection for studies continuously published from inception up to October 30, 2022. Search strategies included keywords such as "artificial intelligence," "head and neck cancer," "HPV," and "sensitivity & specificity." Duplicates, articles without HPV predictions, letters, scientific reports, conference abstracts, or reviews were excluded. Binary diagnostic data were then extracted to generate contingency tables and then used to calculate the pooled sensitivity (SE), specificity (SP), area under the curve (AUC), and their 95% confidence interval (CI). A random-effects model was used for meta-analysis, four subgroup analyses were further explored. Totally, 22 original studies were included in the systematic review, 15 of which were eligible to generate 33 contingency tables for meta-analysis. The pooled SE and SP for all studies were 79% (95% CI: 75-82%) and 74% (95% CI: 69-78%) respectively, with an AUC of 0.83 (95% CI: 0.79-0.86). When only selecting one contingency table with the highest accuracy from each study, our analysis revealed a pooled SE of 79% (95% CI: 75-83%), SP of 75% (95% CI: 69-79%), and an AUC of 0.84 (95% CI: 0.81-0.87). The respective heterogeneities were moderate (I2 for SE and SP were 51.70% and 51.01%) and only low (35.99% and 21.44%). This evidence-based study showed an acceptable and promising performance for AI algorithms to predict HPV status in HNC but was not comparable to the routine p16 immunohistochemistry. The exploitation and optimization of AI algorithms warrant further research. Compared with previous studies, future studies anticipate to make progress in the selection of databases, improvement of international reporting guidelines, and application of high-quality deep learning algorithms.
Collapse
Affiliation(s)
- Cheng Song
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xu Chen
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chao Tang
- Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Peng Xue
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Jiang
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Youlin Qiao
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
18
|
Haider SP, Qureshi AI, Jain A, Tharmaseelan H, Berson ER, Zeevi T, Werring DJ, Gross M, Mak A, Malhotra A, Sansing LH, Falcone GJ, Sheth KN, Payabvash S. Radiomic markers of intracerebral hemorrhage expansion on non-contrast CT: independent validation and comparison with visual markers. Front Neurosci 2023; 17:1225342. [PMID: 37655013 PMCID: PMC10467422 DOI: 10.3389/fnins.2023.1225342] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 07/10/2023] [Indexed: 09/02/2023] Open
Abstract
Objective To devise and validate radiomic signatures of impending hematoma expansion (HE) based on admission non-contrast head computed tomography (CT) of patients with intracerebral hemorrhage (ICH). Methods Utilizing a large multicentric clinical trial dataset of hypertensive patients with spontaneous supratentorial ICH, we developed signatures predictive of HE in a discovery cohort (n = 449) and confirmed their performance in an independent validation cohort (n = 448). In addition to n = 1,130 radiomic features, n = 6 clinical variables associated with HE, n = 8 previously defined visual markers of HE, the BAT score, and combinations thereof served as candidate variable sets for signatures. The area under the receiver operating characteristic curve (AUC) quantified signatures' performance. Results A signature combining select radiomic features and clinical variables attained the highest AUC (95% confidence interval) of 0.67 (0.61-0.72) and 0.64 (0.59-0.70) in the discovery and independent validation cohort, respectively, significantly outperforming the clinical (pdiscovery = 0.02, pvalidation = 0.01) and visual signature (pdiscovery = 0.03, pvalidation = 0.01) as well as the BAT score (pdiscovery < 0.001, pvalidation < 0.001). Adding visual markers to radiomic features failed to improve prediction performance. All signatures were significantly (p < 0.001) correlated with functional outcome at 3-months, underlining their prognostic relevance. Conclusion Radiomic features of ICH on admission non-contrast head CT can predict impending HE with stable generalizability; and combining radiomic with clinical predictors yielded the highest predictive value. By enabling selective anti-expansion treatment of patients at elevated risk of HE in future clinical trials, the proposed markers may increase therapeutic efficacy, and ultimately improve outcomes.
Collapse
Affiliation(s)
- Stefan P. Haider
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States
- Department of Otorhinolaryngology, University Hospital of Ludwig-Maximilians-Universität München, Munich, Germany
| | - Adnan I. Qureshi
- Zeenat Qureshi Stroke Institute and Department of Neurology, University of Missouri, Columbia, MO, United States
| | - Abhi Jain
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States
| | - Hishan Tharmaseelan
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States
| | - Elisa R. Berson
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States
| | - Tal Zeevi
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States
| | - David J. Werring
- Stroke Research Centre, University College London, Queen Square Institute of Neurology, London, United Kingdom
| | - Moritz Gross
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States
| | - Adrian Mak
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States
| | - Ajay Malhotra
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States
| | - Lauren H. Sansing
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
| | - Guido J. Falcone
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
| | - Kevin N. Sheth
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
| | - Seyedmehdi Payabvash
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
19
|
Yao H, Zhang X. A comprehensive review for machine learning based human papillomavirus detection in forensic identification with multiple medical samples. Front Microbiol 2023; 14:1232295. [PMID: 37529327 PMCID: PMC10387549 DOI: 10.3389/fmicb.2023.1232295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 06/30/2023] [Indexed: 08/03/2023] Open
Abstract
Human papillomavirus (HPV) is a sexually transmitted virus. Cervical cancer is one of the highest incidences of cancer, almost all patients are accompanied by HPV infection. In addition, the occurrence of a variety of cancers is also associated with HPV infection. HPV vaccination has gained widespread popularity in recent years with the increase in public health awareness. In this context, HPV testing not only needs to be sensitive and specific but also needs to trace the source of HPV infection. Through machine learning and deep learning, information from medical examinations can be used more effectively. In this review, we discuss recent advances in HPV testing in combination with machine learning and deep learning.
Collapse
Affiliation(s)
- Huanchun Yao
- Department of Cancer, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xinglong Zhang
- Department of Hematology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
20
|
Zhong H, Huang D, Wu J, Chen X, Chen Y, Huang C. 18F‑FDG PET/CT based radiomics features improve prediction of prognosis: multiple machine learning algorithms and multimodality applications for multiple myeloma. BMC Med Imaging 2023; 23:87. [PMID: 37370013 DOI: 10.1186/s12880-023-01033-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
PURPOSE Multiple myeloma (MM), the second most hematological malignancy, have been studied extensively in the prognosis of the clinical parameters, however there are only a few studies have discussed the role of dual modalities and multiple algorithms of 18F-FDG (18F-fluorodeoxyglucose) PET/CT based radiomics signatures for prognosis in MM patients. We hope to deeply mine the utility of raiomics data in the prognosis of MM. METHODS We extensively explored the predictive ability and clinical decision-making ability of different combination image data of PET, CT, clinical parameters and six machine learning algorithms, Cox proportional hazards model (Cox), linear gradient boosting models based on Cox's partial likelihood (GB-Cox), Cox model by likelihood based boosting (CoxBoost), generalized boosted regression modelling (GBM), random forests for survival model (RFS) and support vector regression for censored data model (SVCR). And the model evaluation methods include Harrell concordance index, time dependent receiver operating characteristic (ROC) curve, and decision curve analysis (DCA). RESULTS We finally confirmed 5 PET based features, and 4 CT based features, as well as 6 clinical derived features significantly related to progression free survival (PFS) and we included them in the model construction. In various modalities combinations, RSF and GBM algorithms significantly improved the accuracy and clinical net benefit of predicting prognosis compared with other algorithms. For all combinations of various modalities based models, single-modality PET based prognostic models' performance was outperformed baseline clinical parameters based models, while the performance of models of PET and CT combined with clinical parameters was significantly improved in various algorithms. CONCLUSION 18F‑FDG PET/CT based radiomics models implemented with machine learning algorithms can significantly improve the clinical prediction of progress and increased clinical benefits providing prospects for clinical prognostic stratification for precision treatment as well as new research areas.
Collapse
Affiliation(s)
- Haoshu Zhong
- Department of Hematology, the Affiliated Hospital of Southwest Medical University, Luzhou City, Sichuan, China
- Stem Cell Laboratory, The Clinical Research Institute, Affiliated Hospital of Southwest Medical University, Luzhou City, Sichuan, China
| | - Delong Huang
- Southwest Medical University, Luzhou City, Sichuan, China
| | - Junhao Wu
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Xiaomin Chen
- Department of Hematology, the Affiliated Hospital of Southwest Medical University, Luzhou City, Sichuan, China
- Stem Cell Laboratory, The Clinical Research Institute, Affiliated Hospital of Southwest Medical University, Luzhou City, Sichuan, China
| | - Yue Chen
- Department of Nuclear Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou City, Sichuan, China
| | - Chunlan Huang
- Department of Hematology, the Affiliated Hospital of Southwest Medical University, Luzhou City, Sichuan, China.
- Stem Cell Laboratory, The Clinical Research Institute, Affiliated Hospital of Southwest Medical University, Luzhou City, Sichuan, China.
| |
Collapse
|
21
|
Chen K, Wang J, Li S, Zhou W, Xu W. Predictive value of 18F-FDG PET/CT-based radiomics model for neoadjuvant chemotherapy efficacy in breast cancer: a multi-scanner/center study with external validation. Eur J Nucl Med Mol Imaging 2023; 50:1869-1880. [PMID: 36808002 DOI: 10.1007/s00259-023-06150-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 02/12/2023] [Indexed: 02/23/2023]
Abstract
PURPOSE To develop and validate the predictive value of an 18F-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) model for breast cancer neoadjuvant chemotherapy (NAC) efficacy based on the tumor-to-liver ratio (TLR) radiomic features and multiple data pre-processing methods. METHODS One hundred and ninety-three breast cancer patients from multiple centers were retrospectively included in this study. According to the endpoint of NAC, we divided the patients into pathological complete remission (pCR) and non-pCR groups. All patients underwent 18F-FDG PET/CT imaging before NAC treatment, and CT and PET images volume of interest (VOI) segmentation by manual segmentation and semi-automated absolute threshold segmentation, respectively. Then, feature extraction of VOI was performed with the pyradiomics package. A total of 630 models were created based on the source of radiomic features, the elimination of the batch effect approach, and the discretization method. The differences in data pre-processing approaches were compared and analyzed to identify the best-performing model, which was further tested by the permutation test. RESULTS A variety of data pre-processing methods contributed in varying degrees to the improvement of model effects. Among them, TLR radiomic features and Combat and Limma methods that eliminate batch effects could enhance the model prediction overall, and data discretization could be used as a potential method that can further optimize the model. A total of seven excellent models were selected and then based on the AUC of each model in the four test sets and their standard deviations, we selected the optimal model. The optimal model predicted AUC between 0.7 and 0.77 for the four test groups, with p-values less than 0.05 for the permutation test. CONCLUSION It is necessary to enhance the predictive effect of the model by eliminating confounding factors through data pre-processing. The model developed in this way is effective in predicting the efficacy of NAC for breast cancer.
Collapse
Affiliation(s)
- Kun Chen
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhuxi Road, Hexi Distinct, 300060, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Jian Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhuxi Road, Hexi Distinct, 300060, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Shuai Li
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Wen Zhou
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, People's Republic of China.
| | - Wengui Xu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhuxi Road, Hexi Distinct, 300060, Tianjin, China.
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China.
| |
Collapse
|
22
|
Avery EW, Joshi K, Mehra S, Mahajan A. Role of PET/CT in Oropharyngeal Cancers. Cancers (Basel) 2023; 15:2651. [PMID: 37174116 PMCID: PMC10177278 DOI: 10.3390/cancers15092651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 04/03/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
Oropharyngeal squamous cell carcinoma (OPSCC) comprises cancers of the tonsils, tongue base, soft palate, and uvula. The staging of oropharyngeal cancers varies depending upon the presence or absence of human papillomavirus (HPV)-directed pathogenesis. The incidence of HPV-associated oropharyngeal cancer (HPV + OPSCC) is expected to continue to rise over the coming decades. PET/CT is a useful modality for the diagnosis, staging, and follow up of patients with oropharyngeal cancers undergoing treatment and surveillance.
Collapse
Affiliation(s)
- Emily W. Avery
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Kavita Joshi
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Saral Mehra
- Department of Otolaryngology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Amit Mahajan
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
23
|
Lin H, Haider SP, Kaltenhauser S, Mozayan A, Malhotra A, Constable RT, Scheinost D, Ment LR, Konrad K, Payabvash S. Population level multimodal neuroimaging correlates of attention-deficit hyperactivity disorder among children. Front Neurosci 2023; 17:1138670. [PMID: 36908780 PMCID: PMC9992191 DOI: 10.3389/fnins.2023.1138670] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 02/07/2023] [Indexed: 02/24/2023] Open
Abstract
Objectives Leveraging a large population-level morphologic, microstructural, and functional neuroimaging dataset, we aimed to elucidate the underlying neurobiology of attention-deficit hyperactivity disorder (ADHD) in children. In addition, we evaluated the applicability of machine learning classifiers to predict ADHD diagnosis based on imaging and clinical information. Methods From the Adolescents Behavior Cognitive Development (ABCD) database, we included 1,798 children with ADHD diagnosis and 6,007 without ADHD. In multivariate logistic regression adjusted for age and sex, we examined the association of ADHD with different neuroimaging metrics. The neuroimaging metrics included fractional anisotropy (FA), neurite density (ND), mean-(MD), radial-(RD), and axial diffusivity (AD) of white matter (WM) tracts, cortical region thickness and surface areas from T1-MPRAGE series, and functional network connectivity correlations from resting-state fMRI. Results Children with ADHD showed markers of pervasive reduced microstructural integrity in white matter (WM) with diminished neural density and fiber-tracks volumes - most notable in the frontal and parietal lobes. In addition, ADHD diagnosis was associated with reduced cortical volume and surface area, especially in the temporal and frontal regions. In functional MRI studies, ADHD children had reduced connectivity among default-mode network and the central and dorsal attention networks, which are implicated in concentration and attention function. The best performing combination of feature selection and machine learning classifier could achieve a receiver operating characteristics area under curve of 0.613 (95% confidence interval = 0.580-0.645) to predict ADHD diagnosis in independent validation, using a combination of multimodal imaging metrics and clinical variables. Conclusion Our study highlights the neurobiological implication of frontal lobe cortex and associate WM tracts in pathogenesis of childhood ADHD. We also demonstrated possible potentials and limitations of machine learning models to assist with ADHD diagnosis in a general population cohort based on multimodal neuroimaging metrics.
Collapse
Affiliation(s)
- Huang Lin
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States
- Child Neuropsychology Section, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital RWTH Aachen, Aachen, Germany
| | - Stefan P. Haider
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States
| | - Simone Kaltenhauser
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States
| | - Ali Mozayan
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States
| | - Ajay Malhotra
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States
| | - R. Todd Constable
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States
| | - Dustin Scheinost
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States
| | - Laura R. Ment
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
| | - Kerstin Konrad
- Child Neuropsychology Section, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital RWTH Aachen, Aachen, Germany
- Jülich Research Centre, JARA Brain Institute II, Molecular Neuroscience and Neuroimaging (INM-11), Jülich, Germany
| | - Seyedmehdi Payabvash
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
24
|
Woo C, Jo KH, Sohn B, Park K, Cho H, Kang WJ, Kim J, Lee SK. Development and Testing of a Machine Learning Model Using 18F-Fluorodeoxyglucose PET/CT-Derived Metabolic Parameters to Classify Human Papillomavirus Status in Oropharyngeal Squamous Carcinoma. Korean J Radiol 2023; 24:51-61. [PMID: 36606620 PMCID: PMC9830147 DOI: 10.3348/kjr.2022.0397] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 09/28/2022] [Accepted: 10/31/2022] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVE To develop and test a machine learning model for classifying human papillomavirus (HPV) status of patients with oropharyngeal squamous cell carcinoma (OPSCC) using 18F-fluorodeoxyglucose (18F-FDG) PET-derived parameters in derived parameters and an appropriate combination of machine learning methods in patients with OPSCC. MATERIALS AND METHODS This retrospective study enrolled 126 patients (118 male; mean age, 60 years) with newly diagnosed, pathologically confirmed OPSCC, that underwent 18F-FDG PET-computed tomography (CT) between January 2012 and February 2020. Patients were randomly assigned to training and internal validation sets in a 7:3 ratio. An external test set of 19 patients (16 male; mean age, 65.3 years) was recruited sequentially from two other tertiary hospitals. Model 1 used only PET parameters, Model 2 used only clinical features, and Model 3 used both PET and clinical parameters. Multiple feature transforms, feature selection, oversampling, and training models are all investigated. The external test set was used to test the three models that performed best in the internal validation set. The values for area under the receiver operating characteristic curve (AUC) were compared between models. RESULTS In the external test set, ExtraTrees-based Model 3, which uses two PET-derived parameters and three clinical features, with a combination of MinMaxScaler, mutual information selection, and adaptive synthetic sampling approach, showed the best performance (AUC = 0.78; 95% confidence interval, 0.46-1). Model 3 outperformed Model 1 using PET parameters alone (AUC = 0.48, p = 0.047) and Model 2 using clinical parameters alone (AUC = 0.52, p = 0.142) in predicting HPV status. CONCLUSION Using oversampling and mutual information selection, an ExtraTree-based HPV status classifier was developed by combining metabolic parameters derived from 18F-FDG PET/CT and clinical parameters in OPSCC, which exhibited higher performance than the models using either PET or clinical parameters alone.
Collapse
Affiliation(s)
- Changsoo Woo
- Department of Radiology and Research Institute of Radiological Science and Center for Clinical Imaging Data Science, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Kwan Hyeong Jo
- Department of Nuclear Medicine, Korea University Guro Hospital, Seoul, Korea.
| | - Beomseok Sohn
- Department of Radiology and Research Institute of Radiological Science and Center for Clinical Imaging Data Science, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.
| | - Kisung Park
- Department of Radiology and Research Institute of Radiological Science and Center for Clinical Imaging Data Science, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.,Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, Korea
| | - Hojin Cho
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Won Jun Kang
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jinna Kim
- Department of Radiology and Research Institute of Radiological Science and Center for Clinical Imaging Data Science, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Seung-Koo Lee
- Department of Radiology and Research Institute of Radiological Science and Center for Clinical Imaging Data Science, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
25
|
Avery EW, Behland J, Mak A, Haider SP, Zeevi T, Sanelli PC, Filippi CG, Malhotra A, Matouk CC, Griessenauer CJ, Zand R, Hendrix P, Abedi V, Falcone GJ, Petersen N, Sansing LH, Sheth KN, Payabvash S. Dataset on acute stroke risk stratification from CT angiographic radiomics. Data Brief 2022; 44:108542. [PMID: 36060820 PMCID: PMC9428796 DOI: 10.1016/j.dib.2022.108542] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/02/2022] [Accepted: 08/10/2022] [Indexed: 01/05/2023] Open
Abstract
With advances in high-throughput image processing technologies and increasing availability of medical mega-data, the growing field of radiomics opened the door for quantitative analysis of medical images for prediction of clinically relevant information. One clinical area in which radiomics have proven useful is stroke neuroimaging, where rapid treatment triage is vital for patient outcomes and automated decision assistance tools have potential for significant clinical impact. Recent research, for example, has applied radiomics features extracted from CT angiography (CTA) images and a machine learning framework to facilitate risk-stratification in acute stroke. We here provide methodological guidelines and radiomics data supporting the referenced article "CT angiographic radiomics signature for risk-stratification in anterior large vessel occlusion stroke." The data were extracted from the stroke center registry at Yale New Haven Hospital between 1/1/2014 and 10/31/2020; and Geisinger Medical Center between 1/1/2016 and 12/31/2019. It includes detailed radiomics features of the anterior circulation territories on admission CTA scans in stroke patients with large vessel occlusion stroke who underwent thrombectomy. We also provide the methodological details of the analysis framework utilized for training, optimization, validation and external testing of the machine learning and feature selection algorithms. With the goal of advancing the feasibility and quality of radiomics-based analyses to improve patient care within and beyond the field of stroke, the provided data and methodological support can serve as a baseline for future studies applying radiomics algorithms to machine-learning frameworks, and allow for analysis and utilization of radiomics features extracted in this study.
Collapse
Affiliation(s)
- Emily W. Avery
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar St, New Haven, CT 06510, USA
| | - Jonas Behland
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar St, New Haven, CT 06510, USA
- CLAIM - Charité Lab for Artificial Intelligence in Medicine, Charité Universitätsmedizin Berlin, Charitépl.1, Berlin 10117, Germany
| | - Adrian Mak
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar St, New Haven, CT 06510, USA
- CLAIM - Charité Lab for Artificial Intelligence in Medicine, Charité Universitätsmedizin Berlin, Charitépl.1, Berlin 10117, Germany
| | - Stefan P. Haider
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar St, New Haven, CT 06510, USA
- Department of Otorhinolaryngology, University Hospital of Ludwig Maximilians Universität München, Ziemssenstraße 1, München 80336, Germany
| | - Tal Zeevi
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar St, New Haven, CT 06510, USA
| | - Pina C. Sanelli
- Section of Neuroradiology, Department of Radiology, Northwell Health, 300 Community Dr, Manhasset, NY 11030, USA
| | - Christopher G. Filippi
- Section of Neuroradiology, Department of Radiology, Tufts School of Medicine, 1 Washington St, Boston, MA 02111, USA
| | - Ajay Malhotra
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar St, New Haven, CT 06510, USA
| | - Charles C. Matouk
- Division of Neurovascular Surgery, Department of Neurosurgery, Yale University School of Medicine, 333 Cedar St, New Haven, CT 06510, USA
| | - Christoph J. Griessenauer
- Department of Neurosurgery, Geisinger Medical Center, 100N Academy Ave, Danville, PA 17822, USA
- Research Institute of Neurointervention, Paracelsus Medical University, Strubergasse 21, Salzburg 5020, Austria
- Department of Neurosurgery, Paracelsus Medical University, Strubergasse 21, Salzburg 5020, Austria
| | - Ramin Zand
- Department of Neurology, Geisinger Medical Center, 100N Academy Ave, Danville, PA 17822, USA
| | - Philipp Hendrix
- Department of Neurosurgery, Geisinger Medical Center, 100N Academy Ave, Danville, PA 17822, USA
- Department of Neurosurgery, Saarland University Medical Center, Kirrberger Str 100, Homburg 66421, Germany
| | - Vida Abedi
- Department of Molecular and Functional Genomics, Geisinger Medical Center, 100N Academy Ave, Danville, PA 17822, USA
- Biocomplexity Institute, Virginia Tech, 1015 Life Science Cir, Blacksburg, VA 24061, USA
| | - Guido J. Falcone
- Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Yale University School of Medicine, 333 Cedar St, New Haven, CT 06510, USA
| | - Nils Petersen
- Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Yale University School of Medicine, 333 Cedar St, New Haven, CT 06510, USA
| | - Lauren H. Sansing
- Division of Stroke and Vascular Neurology, Department of Neurology, Yale University School of Medicine, 333 Cedar St, New Haven, CT 06510, USA
| | - Kevin N. Sheth
- Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Yale University School of Medicine, 333 Cedar St, New Haven, CT 06510, USA
| | - Seyedmehdi Payabvash
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar St, New Haven, CT 06510, USA
- Corresponding author. @SamPayabvash
| |
Collapse
|
26
|
Milara E, Gómez-Grande A, Tomás-Soler S, Seiffert AP, Alonso R, Gómez EJ, Martínez-López J, Sánchez-González P. Bone marrow segmentation and radiomics analysis of [ 18F]FDG PET/CT images for measurable residual disease assessment in multiple myeloma. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2022; 225:107083. [PMID: 36044803 DOI: 10.1016/j.cmpb.2022.107083] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/07/2022] [Accepted: 08/22/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND AND OBJECTIVES The last few years have been crucial in defining the most appropriate way to quantitatively assess [18F]FDG PET images in Multiple Myeloma (MM) patients to detect persistent tumor burden. The visual evaluation of images complements the assessment of Measurable Residual Disease (MRD) in bone marrow samples by multiparameter flow cytometry (MFC) or next-generation sequencing (NGS). The aim of this study was to quantify MRD by analyzing quantitative and texture [18F]FDG PET features. METHODS Whole body [18F]FDG PET of 39 patients with newly diagnosed MM were included in the database, and visually evaluated by experts in nuclear medicine. A segmentation methodology of the skeleton from CT images and an additional manual segmentation tool were proposed, implemented in a software solution including a graphical user interface. Both the compact bone and the spinal canal were removed from the segmentation to obtain only the bone marrow mask. SUV metrics, GLCM, GLRLM, and NGTDM parameters were extracted from the PET images and evaluated by Mann-Whitney U-tests and Spearman ρ rank correlation as valuable features differentiating PET+/PET- and MFC+/MFC- groups. Seven machine learning algorithms were applied for evaluating the classification performance of the extracted features. RESULTS Quantitative analysis for PET+/PET- differentiating demonstrated to be significant for most of the variables assessed with Mann-Whitney U-test such as Variance, Energy, and Entropy (p-value = 0.001). Moreover, the quantitative analysis with a balanced database evaluated by Mann-Whitney U-test revealed in even better results with 19 features with p-values < 0.001. On the other hand, radiomics analysis for MFC+/MFC- differentiating demonstrated the necessity of combining MFC evaluation with [18F]FDG PET assessment in the MRD diagnosis. Machine learning algorithms using the image features for the PET+/PET- classification demonstrated high performance metrics but decreasing for the MFC+/MFC- classification. CONCLUSIONS A proof-of-concept for the extraction and evaluation of bone marrow radiomics features of [18F]FDG PET images was proposed and implemented. The validation showed the possible use of these features for the image-based assessment of MRD.
Collapse
Affiliation(s)
- Eva Milara
- Biomedical Engineering and Telemedicine Centre, ETSI Telecomunicación, Center for Biomedical Technology, Universidad Politécnica de Madrid, Avenida Complutense 30, Madrid 28040, Spain.
| | - Adolfo Gómez-Grande
- Department of Nuclear Medicine, Hospital Universitario 12 de Octubre, Madrid, Spain; Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Sebastián Tomás-Soler
- Biomedical Engineering and Telemedicine Centre, ETSI Telecomunicación, Center for Biomedical Technology, Universidad Politécnica de Madrid, Avenida Complutense 30, Madrid 28040, Spain
| | - Alexander P Seiffert
- Biomedical Engineering and Telemedicine Centre, ETSI Telecomunicación, Center for Biomedical Technology, Universidad Politécnica de Madrid, Avenida Complutense 30, Madrid 28040, Spain
| | - Rafael Alonso
- Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain; Department of Hematology and Instituto de Investigación Sanitaria (imas12), Hospital Universitario 12 de Octubre, Madrid, Spain; Clinical Research Hematology Unit, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - Enrique J Gómez
- Biomedical Engineering and Telemedicine Centre, ETSI Telecomunicación, Center for Biomedical Technology, Universidad Politécnica de Madrid, Avenida Complutense 30, Madrid 28040, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
| | - Joaquín Martínez-López
- Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain; Department of Hematology and Instituto de Investigación Sanitaria (imas12), Hospital Universitario 12 de Octubre, Madrid, Spain; Clinical Research Hematology Unit, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - Patricia Sánchez-González
- Biomedical Engineering and Telemedicine Centre, ETSI Telecomunicación, Center for Biomedical Technology, Universidad Politécnica de Madrid, Avenida Complutense 30, Madrid 28040, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain.
| |
Collapse
|
27
|
Weber CF, Lake EMR, Haider SP, Mozayan A, Mukherjee P, Scheinost D, Bamford NS, Ment L, Constable T, Payabvash S. Age-dependent white matter microstructural disintegrity in autism spectrum disorder. Front Neurosci 2022; 16:957018. [PMID: 36161157 PMCID: PMC9490315 DOI: 10.3389/fnins.2022.957018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
There has been increasing evidence of White Matter (WM) microstructural disintegrity and connectome disruption in Autism Spectrum Disorder (ASD). We evaluated the effects of age on WM microstructure by examining Diffusion Tensor Imaging (DTI) metrics and connectome Edge Density (ED) in a large dataset of ASD and control patients from different age cohorts. N = 583 subjects from four studies from the National Database of Autism Research were included, representing four different age groups: (1) A Longitudinal MRI Study of Infants at Risk of Autism [infants, median age: 7 (interquartile range 1) months, n = 155], (2) Biomarkers of Autism at 12 months [toddlers, 32 (11)m, n = 102], (3) Multimodal Developmental Neurogenetics of Females with ASD [adolescents, 13.1 (5.3) years, n = 230], (4) Atypical Late Neurodevelopment in Autism [young adults, 19.1 (10.7)y, n = 96]. For each subject, we created Fractional Anisotropy (FA), Mean- (MD), Radial- (RD), and Axial Diffusivity (AD) maps as well as ED maps. We performed voxel-wise and tract-based analyses to assess the effects of age, ASD diagnosis and sex on DTI metrics and connectome ED. We also optimized, trained, tested, and validated different combinations of machine learning classifiers and dimensionality reduction algorithms for prediction of ASD diagnoses based on tract-based DTI and ED metrics. There is an age-dependent increase in FA and a decline in MD and RD across WM tracts in all four age cohorts, as well as an ED increase in toddlers and adolescents. After correction for age and sex, we found an ASD-related decrease in FA and ED only in adolescents and young adults, but not in infants or toddlers. While DTI abnormalities were mostly limited to the corpus callosum, connectomes showed a more widespread ASD-related decrease in ED. Finally, the best performing machine-leaning classification model achieved an area under the receiver operating curve of 0.70 in an independent validation cohort. Our results suggest that ASD-related WM microstructural disintegrity becomes evident in adolescents and young adults-but not in infants and toddlers. The ASD-related decrease in ED demonstrates a more widespread involvement of the connectome than DTI metrics, with the most striking differences being localized in the corpus callosum.
Collapse
Affiliation(s)
- Clara F. Weber
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States,Social Neuroscience Lab, Department of Psychiatry and Psychotherapy, Lübeck University, Lübeck, Germany
| | - Evelyn M. R. Lake
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States
| | - Stefan P. Haider
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States,Department of Otorhinolaryngology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Ali Mozayan
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States
| | - Pratik Mukherjee
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
| | - Dustin Scheinost
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States
| | - Nigel S. Bamford
- Departments of Pediatrics, Neurology, Cellular and Molecular Physiology, Yale University, New Haven, CT, United States
| | - Laura Ment
- Departments of Pediatrics, Neurology, Cellular and Molecular Physiology, Yale University, New Haven, CT, United States
| | - Todd Constable
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States
| | - Seyedmehdi Payabvash
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States,*Correspondence: Seyedmehdi Payabvash,
| |
Collapse
|
28
|
Haider SP, Qureshi AI, Jain A, Tharmaseelan H, Berson ER, Majidi S, Filippi CG, Mak A, Werring DJ, Acosta JN, Malhotra A, Kim JA, Sansing LH, Falcone GJ, Sheth KN, Payabvash S. The coronal plane maximum diameter of deep intracerebral hemorrhage predicts functional outcome more accurately than hematoma volume. Int J Stroke 2022; 17:777-784. [PMID: 34569877 PMCID: PMC9005571 DOI: 10.1177/17474930211050749] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Among prognostic imaging variables, the hematoma volume on admission computed tomography (CT) has long been considered the strongest predictor of outcome and mortality in intracerebral hemorrhage. AIMS To examine whether different features of hematoma shape are associated with functional outcome in deep intracerebral hemorrhage. METHODS We analyzed 790 patients from the ATACH-2 trial, and 14 shape features were quantified. We calculated Spearman's Rho to assess the correlation between shape features and three-month modified Rankin scale (mRS) score, and the area under the receiver operating characteristic curve (AUC) to quantify the association between shape features and poor outcome defined as mRS>2 as well as mRS > 3. RESULTS Among 14 shape features, the maximum intracerebral hemorrhage diameter in the coronal plane was the strongest predictor of functional outcome, with a maximum coronal diameter >∼3.5 cm indicating higher three-month mRS scores. The maximum coronal diameter versus hematoma volume yielded a Rho of 0.40 versus 0.35 (p = 0.006), an AUC[mRS>2] of 0.71 versus 0.68 (p = 0.004), and an AUC[mRS>3] of 0.71 versus 0.69 (p = 0.029). In multiple regression analysis adjusted for known outcome predictors, the maximum coronal diameter was independently associated with three-month mRS (p < 0.001). CONCLUSIONS A coronal-plane maximum diameter measurement offers greater prognostic value in deep intracerebral hemorrhage than hematoma volume. This simple shape metric may expedite assessment of admission head CTs, offer a potential biomarker for hematoma size eligibility criteria in clinical trials, and may substitute volume in prognostic intracerebral hemorrhage scoring systems.
Collapse
Affiliation(s)
- Stefan P Haider
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
- Department of Otorhinolaryngology, University Hospital of Ludwig Maximilians Universität München, Munich, Germany
| | - Adnan I Qureshi
- Zeenat Qureshi Stroke Institute and Department of Neurology, University of Missouri, Columbia, MO, USA
| | - Abhi Jain
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Hishan Tharmaseelan
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Elisa R Berson
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Shahram Majidi
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Adrian Mak
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
- Charité Lab for Artificial Intelligence in Medicine (CLAIM), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - David J Werring
- Stroke Research Centre, University College London, Queen Square Institute of Neurology, London, UK
| | - Julian N Acosta
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Ajay Malhotra
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Jennifer A Kim
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Lauren H Sansing
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Guido J Falcone
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Kevin N Sheth
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Seyedmehdi Payabvash
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
29
|
Zhao M, Kluge K, Papp L, Grahovac M, Yang S, Jiang C, Krajnc D, Spielvogel CP, Ecsedi B, Haug A, Wang S, Hacker M, Zhang W, Li X. Multi-lesion radiomics of PET/CT for non-invasive survival stratification and histologic tumor risk profiling in patients with lung adenocarcinoma. Eur Radiol 2022; 32:7056-7067. [PMID: 35896836 DOI: 10.1007/s00330-022-08999-7] [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: 05/11/2022] [Revised: 06/15/2022] [Accepted: 06/27/2022] [Indexed: 12/01/2022]
Abstract
OBJECTIVES This study investigates the ability of machine learning (ML) models trained on clinical data and 2-deoxy-2-[18F]fluoro-D-glucose(FDG) positron emission tomography/computed tomography (PET/CT) radiomics to predict overall survival (OS), tumor grade (TG), and histologic growth pattern risk (GPR) in lung adenocarcinoma (LUAD) patients. METHODS A total of 421 treatment-naive patients with histologically-proven LUAD and available FDG PET/CT imaging were retrospectively included. Four cohorts were assessed for predicting 4-year OS (n = 276), 3-year OS (n = 280), TG (n = 298), and GPR (n = 265). FDG-avid lesions were delineated, and 2082 radiomics features were extracted and combined with endpoint-specific clinical parameters. ML models were built for the prediction of 4-year OS (M4OS), 3-year OS (M3OS), tumor grading (MTG), and histologic growth pattern risk (MGPR). A 100-fold Monte Carlo cross-validation with 80:20 training to validation split was employed as a performance evaluation for all models. The association between the M4OS and M3OS predictions with OS was assessed by the Kaplan-Meier survival analysis. RESULTS The area under the receiver operator characteristics curve (AUC) was the highest for M4OS (AUC 0.88, 95% confidence interval (CI) 86.7-88.7), followed by M3OS (AUC 0.84, CI 82.9-84.9), while MTG and MGPR performed equally well (AUC 0.76, CI 74.4-77.9, CI 74.6-78, respectively). Predictions of M4OS (hazard ratio (HR) -2.4, CI -2.47 to -1.64, p < 0.05) and M3OS (HR -2.36, CI -2.79 to -1.93, p < 0.05) were independently associated with OS. CONCLUSION ML models are able to predict long-term survival outcomes in LUAD patients with high accuracy. Furthermore, histologic grade and predominant growth pattern risk can be predicted with satisfactory accuracy. KEY POINTS • Machine learning models trained on pre-therapeutic PET/CT radiomics enable highly accurate long-term survival prediction of patients with lung adenocarcinoma. • Highly accurate survival predictions are achieved in lung adenocarcinoma patients despite heterogenous histologies and treatment regimens. • Radiomic machine learning models are able to predict lung adenocarcinoma tumor grade and histologic growth pattern risk with satisfactory accuracy.
Collapse
Affiliation(s)
- Meixin Zhao
- Department of Nuclear Medicine, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing, 100191, People's Republic of China
| | - Kilian Kluge
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, Floor 3L, 1090, Vienna, Austria.,Christian Doppler Laboratory for Applied Metabolomics (CDLAM), Vienna, Austria
| | - Laszlo Papp
- QIMP Team, Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Marko Grahovac
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, Floor 3L, 1090, Vienna, Austria
| | - Shaomin Yang
- Department of Pathology, Peking University Health Science Center, Beijing, China
| | - Chunting Jiang
- Department of Nuclear Medicine, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing, 100191, People's Republic of China
| | - Denis Krajnc
- QIMP Team, Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Clemens P Spielvogel
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, Floor 3L, 1090, Vienna, Austria.,Christian Doppler Laboratory for Applied Metabolomics (CDLAM), Vienna, Austria
| | - Boglarka Ecsedi
- QIMP Team, Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Alexander Haug
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, Floor 3L, 1090, Vienna, Austria.,Christian Doppler Laboratory for Applied Metabolomics (CDLAM), Vienna, Austria
| | - Shiwei Wang
- Evomics Medical Technology Co., Ltd., Shanghai, China
| | - Marcus Hacker
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, Floor 3L, 1090, Vienna, Austria
| | - Weifang Zhang
- Department of Nuclear Medicine, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing, 100191, People's Republic of China.
| | - Xiang Li
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, Floor 3L, 1090, Vienna, Austria.
| |
Collapse
|
30
|
Multifactorial Model Based on DWI-Radiomics to Determine HPV Status in Oropharyngeal Squamous Cell Carcinoma. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12147244] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
Background: Oropharyngeal squamous cell carcinoma (OPSCC) associated with human papillomavirus (HPV) has higher rates of locoregional control and a better prognosis than HPV-negative OPSCC. These differences are due to some unique biological characteristics that are also visible through advanced imaging modalities. We investigated the ability of a multifactorial model based on both clinical factors and diffusion-weighted imaging (DWI) to determine the HPV status in OPSCC. Methods: The apparent diffusion coefficient (ADC) and the perfusion-free tissue diffusion coefficient D were derived from DWI, both in the primary tumor (PT) and lymph node (LN). First- and second-order radiomic features were extracted from ADC and D maps. Different families of machine learning (ML) algorithms were trained on our dataset using five-fold cross-validation. Results: A cohort of 144 patients was evaluated retrospectively, which was divided into a training set (n = 95) and a validation set (n = 49). The 50th percentile of DPT, the inverse difference moment of ADCLN, smoke habits, and tumor subsite (tonsil versus base of the tongue) were the most relevant predictors. Conclusions: DWI-based radiomics, together with patient-related parameters, allowed us to obtain good diagnostic accuracies in differentiating HPV-positive from HPV-negative patients. A substantial decrease in predictive power was observed in the validation cohort, underscoring the need for further analyses on a larger sample size.
Collapse
|
31
|
Morland D, Triumbari EKA, Boldrini L, Gatta R, Pizzuto D, Annunziata S. Radiomics in Oncological PET Imaging: A Systematic Review-Part 1, Supradiaphragmatic Cancers. Diagnostics (Basel) 2022; 12:1329. [PMID: 35741138 PMCID: PMC9221970 DOI: 10.3390/diagnostics12061329] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 12/10/2022] Open
Abstract
Radiomics is an upcoming field in nuclear oncology, both promising and technically challenging. To summarize the already undertaken work on supradiaphragmatic neoplasia and assess its quality, we performed a literature search in the PubMed database up to 18 February 2022. Inclusion criteria were: studies based on human data; at least one specified tumor type; supradiaphragmatic malignancy; performing radiomics on PET imaging. Exclusion criteria were: studies only based on phantom or animal data; technical articles without a clinically oriented question; fewer than 30 patients in the training cohort. A review database containing PMID, year of publication, cancer type, and quality criteria (number of patients, retrospective or prospective nature, independent validation cohort) was constructed. A total of 220 studies met the inclusion criteria. Among them, 119 (54.1%) studies included more than 100 patients, 21 studies (9.5%) were based on prospectively acquired data, and 91 (41.4%) used an independent validation set. Most studies focused on prognostic and treatment response objectives. Because the textural parameters and methods employed are very different from one article to another, it is complicated to aggregate and compare articles. New contributions and radiomics guidelines tend to help improving quality of the reported studies over the years.
Collapse
Affiliation(s)
- David Morland
- Nuclear Medicine Unit, TracerGLab, Department of Radiology, Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (E.K.A.T.); (D.P.); (S.A.)
- Service de Médecine Nucléaire, Institut Godinot, 51100 Reims, France
- Laboratoire de Biophysique, UFR de Médecine, Université de Reims Champagne-Ardenne, 51100 Reims, France
- CReSTIC (Centre de Recherche en Sciences et Technologies de l’Information et de la Communication), EA 3804, Université de Reims Champagne-Ardenne, 51100 Reims, France
| | - Elizabeth Katherine Anna Triumbari
- Nuclear Medicine Unit, TracerGLab, Department of Radiology, Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (E.K.A.T.); (D.P.); (S.A.)
| | - Luca Boldrini
- Radiotherapy Unit, Radiomics, Department of Radiology, Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (L.B.); (R.G.)
| | - Roberto Gatta
- Radiotherapy Unit, Radiomics, Department of Radiology, Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (L.B.); (R.G.)
- Department of Clinical and Experimental Sciences, University of Brescia, 25121 Brescia, Italy
- Department of Oncology, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Daniele Pizzuto
- Nuclear Medicine Unit, TracerGLab, Department of Radiology, Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (E.K.A.T.); (D.P.); (S.A.)
| | - Salvatore Annunziata
- Nuclear Medicine Unit, TracerGLab, Department of Radiology, Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (E.K.A.T.); (D.P.); (S.A.)
| |
Collapse
|
32
|
Role of Texture Analysis in Oropharyngeal Carcinoma: A Systematic Review of the Literature. Cancers (Basel) 2022; 14:cancers14102445. [PMID: 35626048 PMCID: PMC9139172 DOI: 10.3390/cancers14102445] [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: 03/29/2022] [Revised: 05/02/2022] [Accepted: 05/10/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary The incidence of squamous cell carcinomas of the oropharynx has rapidly increased in the last two decades due to human papilloma virus infection (HPV). HPV-positive and HPV-negative squamous cell tumours differ in radiological imaging, treatment, and prognosis; therefore, differential diagnosis is mandatory. Radiomics with texture analysis is an innovative technique that has been used increasingly in recent years to characterise the tissue heterogeneity of certain structures such as neoplasms or organs by measuring the spatial distribution of pixel values on radiological imaging. This review delineates the application of texture analysis in oropharyngeal tumours and explores how radiomics may potentially improve clinical decision-making. Abstract Human papilloma virus infection (HPV) is associated with the development of lingual and palatine tonsil carcinomas. Diagnosing, differentiating HPV-positive from HPV-negative cancers, and assessing the presence of lymph node metastases or recurrences by the visual interpretation of images is not easy. Texture analysis can provide structural information not perceptible to human eyes. A systematic literature search was performed on 16 February 2022 for studies with a focus on texture analysis in oropharyngeal cancers. We conducted the research on PubMed, Scopus, and Web of Science platforms. Studies were screened for inclusion according to the preferred reporting items for systematic reviews. Twenty-six studies were included in our review. Nineteen articles related specifically to the oropharynx and seven articles analysed the head and neck area with sections dedicated to the oropharynx. Six, thirteen, and seven articles used MRI, CT, and PET, respectively, as the imaging techniques by which texture analysis was performed. Regarding oropharyngeal tumours, this review delineates the applications of texture analysis in (1) the diagnosis, prognosis, and assessment of disease recurrence or persistence after therapy, (2) early differentiation of HPV-positive versus HPV-negative cancers, (3) the detection of cancers not visualised by imaging alone, and (4) the assessment of lymph node metastases from unknown primary carcinomas.
Collapse
|
33
|
Avery EW, Behland J, Mak A, Haider SP, Zeevi T, Sanelli PC, Filippi CG, Malhotra A, Matouk CC, Griessenauer CJ, Zand R, Hendrix P, Abedi V, Falcone GJ, Petersen N, Sansing LH, Sheth KN, Payabvash S. CT angiographic radiomics signature for risk stratification in anterior large vessel occlusion stroke. Neuroimage Clin 2022; 34:103034. [PMID: 35550243 PMCID: PMC9108990 DOI: 10.1016/j.nicl.2022.103034] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 03/27/2022] [Accepted: 05/03/2022] [Indexed: 01/05/2023]
Abstract
BACKGROUND AND PURPOSE As "time is brain" in acute stroke triage, the need for automated prognostication tools continues to increase, particularly in rapidly expanding tele-stroke settings. We aimed to create an automated prognostication tool for anterior circulation large vessel occlusion (LVO) stroke based on admission CTA radiomics. METHODS We automatically extracted 1116 radiomics features from the anterior circulation territory on admission CTAs of 829 acute LVO stroke patients who underwent mechanical thrombectomy in two academic centers. We trained, optimized, validated, and compared different machine-learning models to predict favorable outcome (modified Rankin Scale ≤ 2) at discharge and 3-month follow-up using four different input sets: "Radiomics", "Radiomics + Treatment" (radiomics, post-thrombectomy reperfusion grade, and intravenous thrombolysis), "Clinical + Treatment" (baseline clinical variables and treatment), and "Combined" (radiomics, treatment, and baseline clinical variables). RESULTS For discharge outcome prediction, models were optimized/trained on n = 494 and tested on an independent cohort of n = 100 patients from Yale. Receiver operating characteristic analysis of the independent cohort showed no significant difference between best-performing Combined input models (area under the curve, AUC = 0.77) versus Radiomics + Treatment (AUC = 0.78, p = 0.78), Radiomics (AUC = 0.78, p = 0.55), or Clinical + Treatment (AUC = 0.77, p = 0.87) models. For 3-month outcome prediction, models were optimized/trained on n = 373 and tested on an independent cohort from Yale (n = 72), and an external cohort from Geisinger Medical Center (n = 232). In the independent cohort, there was no significant difference between Combined input models (AUC = 0.76) versus Radiomics + Treatment (AUC = 0.72, p = 0.39), Radiomics (AUC = 0.72, p = 0.39), or Clinical + Treatment (AUC = 76, p = 0.90) models; however, in the external cohort, the Combined model (AUC = 0.74) outperformed Radiomics + Treatment (AUC = 0.66, p < 0.001) and Radiomics (AUC = 0.68, p = 0.005) models for 3-month prediction. CONCLUSION Machine-learning signatures of admission CTA radiomics can provide prognostic information in acute LVO stroke candidates for mechanical thrombectomy. Such objective and time-sensitive risk stratification can guide treatment decisions and facilitate tele-stroke assessment of patients. Particularly in the absence of reliable clinical information at the time of admission, models solely using radiomics features can provide a useful prognostication tool.
Collapse
Affiliation(s)
- Emily W Avery
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States
| | - Jonas Behland
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States; CLAIM - Charité Lab for Artificial Intelligence in Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Adrian Mak
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States; CLAIM - Charité Lab for Artificial Intelligence in Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Stefan P Haider
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States; Department of Otorhinolaryngology, University Hospital of Ludwig Maximilians Universität München, Munich, Germany
| | - Tal Zeevi
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States
| | - Pina C Sanelli
- Section of Neuroradiology, Department of Radiology, Northwell Health, Manhasset, NY, United States
| | - Christopher G Filippi
- Section of Neuroradiology, Department of Radiology, Tufts School of Medicine, Boston, MA, United States
| | - Ajay Malhotra
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States
| | - Charles C Matouk
- Division of Neurovascular Surgery, Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, United States
| | - Christoph J Griessenauer
- Department of Neurosurgery, Geisinger Medical Center, Danville, PA, United States; Research Institute of Neurointervention, Paracelsus Medical University, Salzburg, Austria; Department of Neurosurgery, Paracelsus Medical University, Salzburg, Austria
| | - Ramin Zand
- Department of Neurology, Geisinger, Danville, PA, United States
| | - Philipp Hendrix
- Department of Neurosurgery, Geisinger Medical Center, Danville, PA, United States; Department of Neurosurgery, Saarland University Medical Center, Homburg, Germany
| | - Vida Abedi
- Department of Molecular and Functional Genomics, Geisinger, Danville, PA, United States; Biocomplexity Institute, Virginia Tech, Blacksburg, VA, USA
| | - Guido J Falcone
- Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Yale University School of Medicine, New Haven, CT, United States
| | - Nils Petersen
- Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Yale University School of Medicine, New Haven, CT, United States
| | - Lauren H Sansing
- Division of Stroke and Vascular Neurology, Department of Neurology, Yale University School of Medicine, New Haven, CT, United States
| | - Kevin N Sheth
- Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Yale University School of Medicine, New Haven, CT, United States
| | - Seyedmehdi Payabvash
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States.
| |
Collapse
|
34
|
The impact of radiomics for human papillomavirus status prediction in oropharyngeal cancer: systematic review and radiomics quality score assessment. Neuroradiology 2022; 64:1639-1647. [PMID: 35459957 PMCID: PMC9271107 DOI: 10.1007/s00234-022-02959-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 04/07/2022] [Indexed: 11/19/2022]
Abstract
Purpose
Human papillomavirus (HPV) status assessment is crucial for decision making in oropharyngeal cancer patients. In last years, several articles have been published investigating the possible role of radiomics in distinguishing HPV-positive from HPV-negative neoplasms. Aim of this review was to perform a systematic quality assessment of radiomic studies published on this topic. Methods Radiomics studies on HPV status prediction in oropharyngeal cancer patients were selected. The Radiomic Quality Score (RQS) was assessed by three readers to evaluate their methodological quality. In addition, possible correlations between RQS% and journal type, year of publication, impact factor, and journal rank were investigated. Results After the literature search, 19 articles were selected whose RQS median was 33% (range 0–42%). Overall, 16/19 studies included a well-documented imaging protocol, 13/19 demonstrated phenotypic differences, and all were compared with the current gold standard. No study included a public protocol, phantom study, or imaging at multiple time points. More than half (13/19) included feature selection and only 2 were comprehensive of non-radiomic features. Mean RQS was significantly higher in clinical journals. Conclusion Radiomics has been proposed for oropharyngeal cancer HPV status assessment, with promising results. However, these are supported by low methodological quality investigations. Further studies with higher methodological quality, appropriate standardization, and greater attention to validation are necessary prior to clinical adoption. Supplementary Information The online version contains supplementary material available at 10.1007/s00234-022-02959-0.
Collapse
|
35
|
Lv W, Xu H, Han X, Zhang H, Ma J, Rahmim A, Lu L. Context-Aware Saliency Guided Radiomics: Application to Prediction of Outcome and HPV-Status from Multi-Center PET/CT Images of Head and Neck Cancer. Cancers (Basel) 2022; 14:cancers14071674. [PMID: 35406449 PMCID: PMC8996849 DOI: 10.3390/cancers14071674] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/19/2022] [Accepted: 03/21/2022] [Indexed: 12/15/2022] Open
Abstract
Simple Summary This study investigated the ability of context-aware saliency-guided PET/CT radiomics in the prediction of outcome and HPV status for head and neck cancer. In total, 806 HNC patients (training vs. validation vs. external testing: 500 vs. 97 vs. 209) from 9 centers were collected from The Cancer Imaging Archive (TCIA). Saliency-guided radiomics showed enhanced performance for both outcome and HPV-status predictions relative to conventional radiomics. The radiomics-predicted HPV status also showed complementary prognostic value. This multi-center study highlights the feasibility of saliency-guided PET/CT radiomics in outcome predictions of head and neck cancer, confirming that certain regions are more relevant to tumor aggressiveness and prognosis. Abstract Purpose: This multi-center study aims to investigate the prognostic value of context-aware saliency-guided radiomics in 18F-FDG PET/CT images of head and neck cancer (HNC). Methods: 806 HNC patients (training vs. validation vs. external testing: 500 vs. 97 vs. 209) from 9 centers were collected from The Cancer Imaging Archive (TCIA). There were 100/384 and 60/123 oropharyngeal carcinoma (OPC) patients with human papillomavirus (HPV) status in training and testing cohorts, respectively. Six types of images were used for radiomics feature extraction and further model construction, namely (i) the original image (Origin), (ii) a context-aware saliency map (SalMap), (iii, iv) high- or low-saliency regions in the original image (highSal or lowSal), (v) a saliency-weighted image (SalxImg), and finally, (vi) a fused PET-CT image (FusedImg). Four outcomes were evaluated, i.e., recurrence-free survival (RFS), metastasis-free survival (MFS), overall survival (OS), and disease-free survival (DFS), respectively. Multivariate Cox analysis and logistic regression were adopted to construct radiomics scores for the prediction of outcome (Rad_Ocm) and HPV-status (Rad_HPV), respectively. Besides, the prognostic value of their integration (Rad_Ocm_HPV) was also investigated. Results: In the external testing cohort, compared with the Origin model, SalMap and SalxImg achieved the highest C-indices for RFS (0.621 vs. 0.559) and MFS (0.785 vs. 0.739) predictions, respectively, while FusedImg performed the best for both OS (0.685 vs. 0.659) and DFS (0.641 vs. 0.582) predictions. In the OPC HPV testing cohort, FusedImg showed higher AUC for HPV-status prediction compared with the Origin model (0.653 vs. 0.484). In the OPC testing cohort, compared with Rad_Ocm or Rad_HPV alone, Rad_Ocm_HPV performed the best for OS and DFS predictions with C-indices of 0.702 (p = 0.002) and 0.684 (p = 0.006), respectively. Conclusion: Saliency-guided radiomics showed enhanced performance for both outcome and HPV-status predictions relative to conventional radiomics. The radiomics-predicted HPV status also showed complementary prognostic value.
Collapse
Affiliation(s)
- Wenbing Lv
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai Road, Guangzhou 510515, China; (W.L.); (H.X.); (X.H.); (J.M.)
- Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, 1023 Shatai Road, Guangzhou 510515, China
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, 1023 Shatai Road, Guangzhou 510515, China
- Pazhou Lab, Guangzhou 510330, China
| | - Hui Xu
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai Road, Guangzhou 510515, China; (W.L.); (H.X.); (X.H.); (J.M.)
- Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, 1023 Shatai Road, Guangzhou 510515, China
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, 1023 Shatai Road, Guangzhou 510515, China
- Pazhou Lab, Guangzhou 510330, China
| | - Xu Han
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai Road, Guangzhou 510515, China; (W.L.); (H.X.); (X.H.); (J.M.)
- Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, 1023 Shatai Road, Guangzhou 510515, China
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, 1023 Shatai Road, Guangzhou 510515, China
- Pazhou Lab, Guangzhou 510330, China
| | - Hao Zhang
- Department of Medical Imaging, Nanfang Hospital, Southern Medical University, 1023 Shatai Road, Guangzhou 510515, China;
| | - Jianhua Ma
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai Road, Guangzhou 510515, China; (W.L.); (H.X.); (X.H.); (J.M.)
- Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, 1023 Shatai Road, Guangzhou 510515, China
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, 1023 Shatai Road, Guangzhou 510515, China
- Pazhou Lab, Guangzhou 510330, China
| | - Arman Rahmim
- Department of Integrative Oncology, BC Cancer Research Institute, 675 West 10th Avenue, Vancouver, BC V5Z 1L3, Canada;
- Department of Radiology, University of British Columbia, 2211 Wesbrook Mall, Vancouver, BC V6T 1Z1, Canada
- Department of Physics, University of British Columbia, 6224 Agricultural Road, Vancouver, BC V6T 1Z1, Canada
| | - Lijun Lu
- School of Biomedical Engineering, Southern Medical University, 1023 Shatai Road, Guangzhou 510515, China; (W.L.); (H.X.); (X.H.); (J.M.)
- Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, 1023 Shatai Road, Guangzhou 510515, China
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, 1023 Shatai Road, Guangzhou 510515, China
- Pazhou Lab, Guangzhou 510330, China
- Correspondence: ; Tel.: +86-020-62789116
| |
Collapse
|
36
|
La Greca Saint-Esteven A, Bogowicz M, Konukoglu E, Riesterer O, Balermpas P, Guckenberger M, Tanadini-Lang S, van Timmeren JE. A 2.5D convolutional neural network for HPV prediction in advanced oropharyngeal cancer. Comput Biol Med 2022; 142:105215. [PMID: 34999414 DOI: 10.1016/j.compbiomed.2022.105215] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/22/2021] [Accepted: 01/02/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Infection with human papilloma virus (HPV) is one of the most relevant prognostic factors in advanced oropharyngeal cancer (OPC) treatment. In this study we aimed to assess the diagnostic accuracy of a deep learning-based method for HPV status prediction in computed tomography (CT) images of advanced OPC. METHOD An internal dataset and three public collections were employed (internal: n = 151, HNC1: n = 451; HNC2: n = 80; HNC3: n = 110). Internal and HNC1 datasets were used for training, whereas HNC2 and HNC3 collections were used as external test cohorts. All CT scans were resampled to a 2 mm3 resolution and a sub-volume of 72x72x72 pixels was cropped on each scan, centered around the tumor. Then, a 2.5D input of size 72x72x3 pixels was assembled by selecting the 2D slice containing the largest tumor area along the axial, sagittal and coronal planes, respectively. The convolutional neural network employed consisted of the first 5 modules of the Xception model and a small classification network. Ten-fold cross-validation was applied to evaluate training performance. At test time, soft majority voting was used to predict HPV status. RESULTS A final training mean [range] area under the curve (AUC) of 0.84 [0.76-0.89], accuracy of 0.76 [0.64-0.83] and F1-score of 0.74 [0.62-0.83] were achieved. AUC/accuracy/F1-score values of 0.83/0.75/0.69 and 0.88/0.79/0.68 were achieved on the HNC2 and HNC3 test sets, respectively. CONCLUSION Deep learning was successfully applied and validated in two external cohorts to predict HPV status in CT images of advanced OPC, proving its potential as a support tool in cancer precision medicine.
Collapse
Affiliation(s)
- Agustina La Greca Saint-Esteven
- Department of Radiation Oncology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland; Computer Vision Laboratory, ETH Zurich, Zurich, Switzerland.
| | - Marta Bogowicz
- Department of Radiation Oncology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | | | - Oliver Riesterer
- Department of Radiation Oncology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland; Center for Radiation Oncology KSA-KSB, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Panagiotis Balermpas
- Department of Radiation Oncology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Matthias Guckenberger
- Department of Radiation Oncology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Stephanie Tanadini-Lang
- Department of Radiation Oncology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Janita E van Timmeren
- Department of Radiation Oncology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
37
|
Gharavi SMH, Faghihimehr A. Clinical Application of Artificial Intelligence in PET Imaging of Head and Neck Cancer. PET Clin 2021; 17:65-76. [PMID: 34809871 DOI: 10.1016/j.cpet.2021.09.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Applications of "artificial intelligence" (AI) have been exponentially expanding in health care. Readily accessible archives of enormous digital data in medical imaging have made radiology a leader in exploring and taking advantage of this technology. AI-assisted radiology has paved the way toward another level of precision in medicine. In this article, the authors aim to review current AI applications in PET imaging of head and neck cancers, beginning with radiomics and followed by deep learning in each section.
Collapse
Affiliation(s)
- Seyed Mohammad H Gharavi
- Virginia Commonwealth University, VCU School of Medicine, Department of Radiology, West Hospital, 1200 East Broad Street, North Wing, Room 2-013, Box 980470, Richmond, VA 23298-0470, USA.
| | - Armaghan Faghihimehr
- Virginia Commonwealth University, VCU School of Medicine, Department of Radiology, West Hospital, 1200 East Broad Street, North Wing, Room 2-013, Box 980470, Richmond, VA 23298-0470, USA
| |
Collapse
|
38
|
Maleki F, Le WT, Sananmuang T, Kadoury S, Forghani R. Machine Learning Applications for Head and Neck Imaging. Neuroimaging Clin N Am 2021; 30:517-529. [PMID: 33039001 DOI: 10.1016/j.nic.2020.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The head and neck (HN) consists of a large number of vital anatomic structures within a compact area. Imaging plays a central role in the diagnosis and management of major disorders affecting the HN. This article reviews the recent applications of machine learning (ML) in HN imaging with a focus on deep learning approaches. It categorizes ML applications in HN imaging into deep learning and traditional ML applications and provides examples of each category. It also discusses the main challenges facing the successful deployment of ML-based applications in the clinical setting and provides suggestions for addressing these challenges.
Collapse
Affiliation(s)
- Farhad Maleki
- Augmented Intelligence & Precision Health Laboratory (AIPHL), Department of Radiology & Research Institute of the McGill University Health Centre, 5252 Boulevard de Maisonneuve Ouest, Montreal, Quebec H4A 3S5, Canada
| | - William Trung Le
- Polytechnique Montreal, PO Box 6079, succ. Centre-ville, Montreal, Quebec H3C 3A7, Canada
| | - Thiparom Sananmuang
- Department of Diagnostic and Therapeutic Radiology and Research, Faculty of Medicine Ramathibodi Hospital, Ratchathewi, Bangkok 10400, Thailand
| | - Samuel Kadoury
- Polytechnique Montreal, PO Box 6079, succ. Centre-ville, Montreal, Quebec H3C 3A7, Canada; CHUM Research Center, 900 St Denis Street, Montreal, Quebec H2X 0A9, Canada
| | - Reza Forghani
- Augmented Intelligence & Precision Health Laboratory (AIPHL), Department of Radiology & Research Institute of the McGill University Health Centre, 5252 Boulevard de Maisonneuve Ouest, Montreal, Quebec H4A 3S5, Canada; Department of Radiology, McGill University, 1650 Cedar Avenue, Montreal, Quebec H3G1A4, Canada; Segal Cancer Centre, Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Cote Ste-Catherine Road, Montreal, Quebec H3T 1E2, Canada; Gerald Bronfman Department of Oncology, McGill University, Suite 720, 5100 Maisonneuve Boulevard West, Montreal, Quebec H4A3T2, Canada; Department of Otolaryngology, Head and Neck Surgery, Royal Victoria Hospital, McGill University Health Centre, 1001 boul. Decarie Boulevard, Montreal, Quebec H3A 3J1, Canada.
| |
Collapse
|
39
|
[Artificial intelligence in otorhinolaryngology]. HNO 2021; 70:87-93. [PMID: 34374811 PMCID: PMC8353610 DOI: 10.1007/s00106-021-01095-0] [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] [Accepted: 05/26/2021] [Indexed: 11/24/2022]
Abstract
Hintergrund Die fortschreitende Digitalisierung ermöglicht zunehmend den Einsatz von künstlicher Intelligenz (KI). Sie wird Gesellschaft und Medizin in den nächsten Jahren maßgeblich beeinflussen. Ziel der Arbeit Darstellung des gegenwärtigen Einsatzspektrums von KI in der Hals-Nasen-Ohren-Heilkunde und Skizzierung zukünftiger Entwicklungen bei der Anwendung dieser Technologie. Material und Methoden Es erfolgte die Auswertung und Diskussion wissenschaftlicher Studien und Expertenanalysen. Ergebnisse Durch die Verwendung von KI kann der Nutzen herkömmlicher diagnostischer Werkzeuge in der Hals-Nasen-Ohren-Heilkunde gesteigert werden. Zudem kann der Einsatz dieser Technologie die chirurgische Präzision in der Kopf-Hals-Chirurgie weiter erhöhen. Schlussfolgerungen KI besitzt ein großes Potenzial zur weiteren Verbesserung diagnostischer und therapeutischer Verfahren in der Hals-Nasen-Ohren-Heilkunde. Allerdings ist die Anwendung dieser Technologie auch mit Herausforderungen verbunden, beispielsweise im Bereich des Datenschutzes.
Collapse
|
40
|
Haider SP, Qureshi AI, Jain A, Tharmaseelan H, Berson ER, Zeevi T, Majidi S, Filippi CG, Iseke S, Gross M, Acosta JN, Malhotra A, Kim JA, Sansing LH, Falcone GJ, Sheth KN, Payabvash S. Admission computed tomography radiomic signatures outperform hematoma volume in predicting baseline clinical severity and functional outcome in the ATACH-2 trial intracerebral hemorrhage population. Eur J Neurol 2021; 28:2989-3000. [PMID: 34189814 DOI: 10.1111/ene.15000] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/24/2021] [Accepted: 06/27/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND PURPOSE Radiomics provides a framework for automated extraction of high-dimensional feature sets from medical images. We aimed to determine radiomics signature correlates of admission clinical severity and medium-term outcome from intracerebral hemorrhage (ICH) lesions on baseline head computed tomography (CT). METHODS We used the ATACH-2 (Antihypertensive Treatment of Acute Cerebral Hemorrhage II) trial dataset. Patients included in this analysis (n = 895) were randomly allocated to discovery (n = 448) and independent validation (n = 447) cohorts. We extracted 1130 radiomics features from hematoma lesions on baseline noncontrast head CT scans and generated radiomics signatures associated with admission Glasgow Coma Scale (GCS), admission National Institutes of Health Stroke Scale (NIHSS), and 3-month modified Rankin Scale (mRS) scores. Spearman's correlation between radiomics signatures and corresponding target variables was compared with hematoma volume. RESULTS In the discovery cohort, radiomics signatures, compared to ICH volume, had a significantly stronger association with admission GCS (0.47 vs. 0.44, p = 0.008), admission NIHSS (0.69 vs. 0.57, p < 0.001), and 3-month mRS scores (0.44 vs. 0.32, p < 0.001). Similarly, in independent validation, radiomics signatures, compared to ICH volume, had a significantly stronger association with admission GCS (0.43 vs. 0.41, p = 0.02), NIHSS (0.64 vs. 0.56, p < 0.001), and 3-month mRS scores (0.43 vs. 0.33, p < 0.001). In multiple regression analysis adjusted for known predictors of ICH outcome, the radiomics signature was an independent predictor of 3-month mRS in both cohorts. CONCLUSIONS Limited by the enrollment criteria of the ATACH-2 trial, we showed that radiomics features quantifying hematoma texture, density, and shape on baseline CT can provide imaging correlates for clinical presentation and 3-month outcome. These findings couldtrigger a paradigm shift where imaging biomarkers may improve current modelsfor prognostication, risk-stratification, and treatment triage of ICH patients.
Collapse
Affiliation(s)
- Stefan P Haider
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA.,Department of Otorhinolaryngology, University Hospital of Ludwig Maximilians Universität München, Munich, Germany
| | - Adnan I Qureshi
- Zeenat Qureshi Stroke Institute and Department of Neurology, University of Missouri, Columbia, MO, USA
| | - Abhi Jain
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Hishan Tharmaseelan
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Elisa R Berson
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Tal Zeevi
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Shahram Majidi
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Simon Iseke
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Moritz Gross
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Julian N Acosta
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Ajay Malhotra
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Jennifer A Kim
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Lauren H Sansing
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Guido J Falcone
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Kevin N Sheth
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Seyedmehdi Payabvash
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
41
|
A Systematic Review of PET Textural Analysis and Radiomics in Cancer. Diagnostics (Basel) 2021; 11:diagnostics11020380. [PMID: 33672285 PMCID: PMC7926413 DOI: 10.3390/diagnostics11020380] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/10/2021] [Accepted: 02/19/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Although many works have supported the utility of PET radiomics, several authors have raised concerns over the robustness and replicability of the results. This study aimed to perform a systematic review on the topic of PET radiomics and the used methodologies. Methods: PubMed was searched up to 15 October 2020. Original research articles based on human data specifying at least one tumor type and PET image were included, excluding those that apply only first-order statistics and those including fewer than 20 patients. Each publication, cancer type, objective and several methodological parameters (number of patients and features, validation approach, among other things) were extracted. Results: A total of 290 studies were included. Lung (28%) and head and neck (24%) were the most studied cancers. The most common objective was prognosis/treatment response (46%), followed by diagnosis/staging (21%), tumor characterization (18%) and technical evaluations (15%). The average number of patients included was 114 (median = 71; range 20–1419), and the average number of high-order features calculated per study was 31 (median = 26, range 1–286). Conclusions: PET radiomics is a promising field, but the number of patients in most publications is insufficient, and very few papers perform in-depth validations. The role of standardization initiatives will be crucial in the upcoming years.
Collapse
|
42
|
Haider SP, Sharaf K, Zeevi T, Baumeister P, Reichel C, Forghani R, Kann BH, Petukhova A, Judson BL, Prasad ML, Liu C, Burtness B, Mahajan A, Payabvash S. Prediction of post-radiotherapy locoregional progression in HPV-associated oropharyngeal squamous cell carcinoma using machine-learning analysis of baseline PET/CT radiomics. Transl Oncol 2020; 14:100906. [PMID: 33075658 PMCID: PMC7568193 DOI: 10.1016/j.tranon.2020.100906] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/27/2020] [Accepted: 09/28/2020] [Indexed: 12/15/2022] Open
Abstract
Radiomics quantitatively captures visually inappreciable imaging features. PET/CT radiomics provides wholistic metabolic and structural tumor characterization. Machine-learning algorithms can generate radiomics-based biomarkers for OPSCC. PET/CT radiomics can predict post-radiotherapy locoregional progression in HPV-associated OPSCC. Such biomarkers may improve patient selection for treatment de-intensification trials.
Locoregional failure remains a therapeutic challenge in oropharyngeal squamous cell carcinoma (OPSCC). We aimed to devise novel objective imaging biomarkers for prediction of locoregional progression in HPV-associated OPSCC. Following manual lesion delineation, 1037 PET and 1037 CT radiomic features were extracted from each primary tumor and metastatic cervical lymph node on baseline PET/CT scans. Applying random forest machine-learning algorithms, we generated radiomic models for censoring-aware locoregional progression prognostication (evaluated by Harrell's C-index) and risk stratification (evaluated in Kaplan-Meier analysis). A total of 190 patients were included; an optimized model yielded a median (interquartile range) C-index of 0.76 (0.66-0.81; p = 0.01) in prognostication of locoregional progression, using combined PET/CT radiomic features from primary tumors. Radiomics-based risk stratification reliably identified patients at risk for locoregional progression within 2-, 3-, 4-, and 5-year follow-up intervals, with log-rank p-values of p = 0.003, p = 0.001, p = 0.02, p = 0.006 in Kaplan-Meier analysis, respectively. Our results suggest PET/CT radiomic biomarkers can predict post-radiotherapy locoregional progression in HPV-associated OPSCC. Pending validation in large, independent cohorts, such objective biomarkers may improve patient selection for treatment de-intensification trials in this prognostically favorable OPSCC entity, and eventually facilitate personalized therapy.
Collapse
Affiliation(s)
- Stefan P Haider
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, 789 Howard Ave, PO Box 208042, New Haven, CT 06519, United States; Department of Otorhinolaryngology, University Hospital of Ludwig Maximilians Universität München, Marchioninistrasse 15, 81377 Munich, Germany
| | - Kariem Sharaf
- Department of Otorhinolaryngology, University Hospital of Ludwig Maximilians Universität München, Marchioninistrasse 15, 81377 Munich, Germany
| | - Tal Zeevi
- Center for Translational Imaging Analysis and Machine Learning, Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06510, United States
| | - Philipp Baumeister
- Department of Otorhinolaryngology, University Hospital of Ludwig Maximilians Universität München, Marchioninistrasse 15, 81377 Munich, Germany
| | - Christoph Reichel
- Department of Otorhinolaryngology, University Hospital of Ludwig Maximilians Universität München, Marchioninistrasse 15, 81377 Munich, Germany
| | - Reza Forghani
- Department of Diagnostic Radiology and Augmented Intelligence & Precision Health Laboratory (AIPHL), McGill University Health Centre & Research Institute, 1650 Cedar Avenue, Montreal, Quebec QC H3G 1A4, Canada
| | - Benjamin H Kann
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, United States
| | - Alexandra Petukhova
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, 789 Howard Ave, PO Box 208042, New Haven, CT 06519, United States
| | - Benjamin L Judson
- Division of Otolaryngology, Department of Surgery, Yale School of Medicine, 330 Cedar Street, New Haven, CT 06520, United States
| | - Manju L Prasad
- Department of Pathology, Yale School of Medicine, 310 Cedar Street, New Haven, CT 06520, United States
| | - Chi Liu
- Division of Bioimaging Sciences, Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06510, United States
| | - Barbara Burtness
- Section of Medical Oncology, Department of Internal Medicine, Yale School of Medicine and Yale Cancer Center, 25 York Street, New Haven, CT 06520, United States
| | - Amit Mahajan
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, 789 Howard Ave, PO Box 208042, New Haven, CT 06519, United States
| | - Seyedmehdi Payabvash
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, 789 Howard Ave, PO Box 208042, New Haven, CT 06519, United States.
| |
Collapse
|
43
|
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
|
44
|
Haider SP, Zeevi T, Baumeister P, Reichel C, Sharaf K, Forghani R, Kann BH, Judson BL, Prasad ML, Burtness B, Mahajan A, Payabvash S. Potential Added Value of PET/CT Radiomics for Survival Prognostication beyond AJCC 8th Edition Staging in Oropharyngeal Squamous Cell Carcinoma. Cancers (Basel) 2020; 12:cancers12071778. [PMID: 32635216 PMCID: PMC7407414 DOI: 10.3390/cancers12071778] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 12/18/2022] Open
Abstract
Accurate risk-stratification can facilitate precision therapy in oropharyngeal squamous cell carcinoma (OPSCC). We explored the potential added value of baseline positron emission tomography (PET)/computed tomography (CT) radiomic features for prognostication and risk stratification of OPSCC beyond the American Joint Committee on Cancer (AJCC) 8th edition staging scheme. Using institutional and publicly available datasets, we included OPSCC patients with known human papillomavirus (HPV) status, without baseline distant metastasis and treated with curative intent. We extracted 1037 PET and 1037 CT radiomic features quantifying lesion shape, imaging intensity, and texture patterns from primary tumors and metastatic cervical lymph nodes. Utilizing random forest algorithms, we devised novel machine-learning models for OPSCC progression-free survival (PFS) and overall survival (OS) using “radiomics” features, “AJCC” variables, and the “combined” set as input. We designed both single- (PET or CT) and combined-modality (PET/CT) models. Harrell’s C-index quantified survival model performance; risk stratification was evaluated in Kaplan–Meier analysis. A total of 311 patients were included. In HPV-associated OPSCC, the best “radiomics” model achieved an average C-index ± standard deviation of 0.62 ± 0.05 (p = 0.02) for PFS prediction, compared to 0.54 ± 0.06 (p = 0.32) utilizing “AJCC” variables. Radiomics-based risk-stratification of HPV-associated OPSCC was significant for PFS and OS. Similar trends were observed in HPV-negative OPSCC. In conclusion, radiomics imaging features extracted from pre-treatment PET/CT may provide complimentary information to the current AJCC staging scheme for survival prognostication and risk-stratification of HPV-associated OPSCC.
Collapse
Affiliation(s)
- Stefan P. Haider
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, 789 Howard Ave, New Haven, CT 06519, USA; (S.P.H.); (A.M.)
- Department of Otorhinolaryngology, University Hospital of Ludwig Maximilians Universität München, Marchioninistrasse 15, 81377 Munich, Germany; (P.B.); (C.R.); (K.S.)
| | - Tal Zeevi
- Center for Translational Imaging Analysis and Machine Learning, Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA;
| | - Philipp Baumeister
- Department of Otorhinolaryngology, University Hospital of Ludwig Maximilians Universität München, Marchioninistrasse 15, 81377 Munich, Germany; (P.B.); (C.R.); (K.S.)
| | - Christoph Reichel
- Department of Otorhinolaryngology, University Hospital of Ludwig Maximilians Universität München, Marchioninistrasse 15, 81377 Munich, Germany; (P.B.); (C.R.); (K.S.)
| | - Kariem Sharaf
- Department of Otorhinolaryngology, University Hospital of Ludwig Maximilians Universität München, Marchioninistrasse 15, 81377 Munich, Germany; (P.B.); (C.R.); (K.S.)
| | - Reza Forghani
- Department of Diagnostic Radiology and Augmented Intelligence & Precision Health Laboratory, McGill University Health Centre & Research Institute, 1650 Cedar Avenue, Montreal, QC H3G 1A4, Canada;
| | - Benjamin H. Kann
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA;
| | - Benjamin L. Judson
- Division of Otolaryngology, Department of Surgery, Yale School of Medicine, 330 Cedar Street, New Haven, CT 06520, USA;
| | - Manju L. Prasad
- Department of Pathology, Yale School of Medicine, 310 Cedar Street, New Haven, CT 06520, USA;
| | - Barbara Burtness
- Section of Medical Oncology, Department of Internal Medicine, Yale School of Medicine, 25 York Street, New Haven, CT 06520, USA;
| | - Amit Mahajan
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, 789 Howard Ave, New Haven, CT 06519, USA; (S.P.H.); (A.M.)
| | - Seyedmehdi Payabvash
- Section of Neuroradiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, 789 Howard Ave, New Haven, CT 06519, USA; (S.P.H.); (A.M.)
- Correspondence: ; Tel.: +1-(203)-214-4650
| |
Collapse
|