1
|
Awais M, Rehman A, Bukhari SS. Advances in liquid biopsy and virtual biopsy for care of patients with glioma: a narrative review. Expert Rev Anticancer Ther 2025; 25:529-550. [PMID: 40183671 DOI: 10.1080/14737140.2025.2489629] [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: 01/19/2025] [Accepted: 04/02/2025] [Indexed: 04/05/2025]
Abstract
INTRODUCTION The World Health Organization's 2021 classification of central nervous system neoplasms incorporated molecular and genetic features for classifying gliomas. Classification of gliomas located in deep-seated structures became a clinical conundrum given the absence of crucial pathological and molecular data. Advances in noninvasive imaging modalities offered virtual biopsy as a novel solution to this problem by identifying surrogate radiomic signatures. Liquid biopsies of blood or cerebrospinal fluid provided another enormous opportunity for identifying genomic, metabolomic and proteomic signatures. AREAS COVERED We summarize and appraise the current state of evidence with regards to virtual biopsy and liquid biopsy in the care of patients with gliomas. PubMed, Embase and Google Scholar were searched on 7/30/2024 for relevant articles published after the year 2013 in the English language. EXPERT OPINION A large body of preclinical and preliminary clinical evidence suggests that virtual biopsy is possible with the combined use of multiple novel imaging modalities in conjunction with machine learning and radiomics. Likewise, liquid biopsy in conjunction with focused ultrasound may be a valuable tool to obtain proteomic and genomic data regarding glioma in a minimally invasive manner. These modalities will likely become an integral part of care for patients with glioma in the future.
Collapse
Affiliation(s)
- Muhammad Awais
- Department of Radiology, The Aga Khan University, Karachi, Pakistan
| | - Abdul Rehman
- Department of Medicine, Tidal Health Peninsula Regional, Salisbury, MD, USA
| | - Syed Sarmad Bukhari
- Department of Neurosurgery, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
2
|
Chen Y, Fan Z, Luo Z, Kang X, Wan R, Li F, Lin W, Han Z, Qi B, Lin J, Sun Y, Huang J, Xu Y, Chen S. Impacts of Nutlin-3a and exercise on murine double minute 2-enriched glioma treatment. Neural Regen Res 2025; 20:1135-1152. [PMID: 38989952 PMCID: PMC11438351 DOI: 10.4103/nrr.nrr-d-23-00875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 12/21/2023] [Indexed: 07/12/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202504000-00029/figure1/v/2024-07-06T104127Z/r/image-tiff Recent research has demonstrated the impact of physical activity on the prognosis of glioma patients, with evidence suggesting exercise may reduce mortality risks and aid neural regeneration. The role of the small ubiquitin-like modifier (SUMO) protein, especially post-exercise, in cancer progression, is gaining attention, as are the potential anti-cancer effects of SUMOylation. We used machine learning to create the exercise and SUMO-related gene signature (ESLRS). This signature shows how physical activity might help improve the outlook for low-grade glioma and other cancers. We demonstrated the prognostic and immunotherapeutic significance of ESLRS markers, specifically highlighting how murine double minute 2 (MDM2), a component of the ESLRS, can be targeted by nutlin-3. This underscores the intricate relationship between natural compounds such as nutlin-3 and immune regulation. Using comprehensive CRISPR screening, we validated the effects of specific ESLRS genes on low-grade glioma progression. We also revealed insights into the effectiveness of Nutlin-3a as a potent MDM2 inhibitor through molecular docking and dynamic simulation. Nutlin-3a inhibited glioma cell proliferation and activated the p53 pathway. Its efficacy decreased with MDM2 overexpression, and this was reversed by Nutlin-3a or exercise. Experiments using a low-grade glioma mouse model highlighted the effect of physical activity on oxidative stress and molecular pathway regulation. Notably, both physical exercise and Nutlin-3a administration improved physical function in mice bearing tumors derived from MDM2-overexpressing cells. These results suggest the potential for Nutlin-3a, an MDM2 inhibitor, with physical exercise as a therapeutic approach for glioma management. Our research also supports the use of natural products for therapy and sheds light on the interaction of exercise, natural products, and immune regulation in cancer treatment.
Collapse
Affiliation(s)
- Yisheng Chen
- Department of Sport Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhongcheng Fan
- Department of Orthopedic Surgery, Hainan Province Clinical Medical Center, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, Hainan Province, China
| | - Zhiwen Luo
- Department of Sport Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Xueran Kang
- Department of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Renwen Wan
- Department of Sport Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Fangqi Li
- Department of Sport Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Weiwei Lin
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Zhihua Han
- Department of Orthopedics, Shanghai General Hospital, School of Medicine Shanghai Jiao Tong University, Shanghai, China
| | - Beijie Qi
- Department of Sport Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jinrong Lin
- Department of Sport Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yaying Sun
- Department of Sport Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiebin Huang
- Department of Infectious Diseases, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Yuzhen Xu
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| | - Shiyi Chen
- Department of Sport Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
3
|
Wang Z, Wang L, Wang Y. Radiomics in glioma: emerging trends and challenges. Ann Clin Transl Neurol 2025; 12:460-477. [PMID: 39901654 PMCID: PMC11920724 DOI: 10.1002/acn3.52306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/11/2024] [Accepted: 12/31/2024] [Indexed: 02/05/2025] Open
Abstract
Radiomics is a promising neuroimaging technique for extracting and analyzing quantitative glioma features. This review discusses the application, emerging trends, and challenges associated with using radiomics in glioma. Integrating deep learning algorithms enhances various radiomics components, including image normalization, region of interest segmentation, feature extraction, feature selection, and model construction and can potentially improve model accuracy and performance. Moreover, investigating specific tumor habitats of glioblastomas aids in a better understanding of glioblastoma aggressiveness and the development of effective treatment strategies. Additionally, advanced imaging techniques, such as diffusion-weighted imaging, perfusion-weighted imaging, magnetic resonance spectroscopy, magnetic resonance fingerprinting, functional MRI, and positron emission tomography, can provide supplementary information for tumor characterization and classification. Furthermore, radiomics analysis helps understand the glioma immune microenvironment by predicting immune-related biomarkers and characterizing immune responses within tumors. Integrating multi-omics data, such as genomics, transcriptomics, proteomics, and pathomics, with radiomics, aids the understanding of the biological significance of the underlying radiomics features and improves the prediction of genetic mutations, prognosis, and treatment response in patients with glioma. Addressing challenges, such as model reproducibility, model generalizability, model interpretability, and multi-omics data integration, is crucial for the clinical translation of radiomics in glioma.
Collapse
Affiliation(s)
- Zihan Wang
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Lei Wang
- Department of NeurosurgeryGuiqian International General HospitalGuiyangGuizhouChina
| | - Yinyan Wang
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
4
|
Li X, Zhang H, Hu C, Hu L, Guo H, Chen H, Li G, Huang Q, Jiang S, Zhang S, Xing Z, Wang X. Prognostic significance of collagen content in solitary fibrous tumors of the central nervous system. Front Oncol 2024; 14:1450813. [PMID: 39600632 PMCID: PMC11588704 DOI: 10.3389/fonc.2024.1450813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024] Open
Abstract
Purpose We aimed to explore the prognostic significance of collagen content in solitary fibrous tumors (SFTs) of the central nervous system (CNS) and preliminarily investigate its relationship with magnetic resonance imaging (MRI) features of SFTs. Methods Collagen content was identified using Masson's trichrome staining, and quantitatively assessed. Radiomic methods were applied to extract quantitative MRI features of SFTs, which were then analyzed in relation to collagen content. Results The collagen content in CNS SFTs was categorized into high- and low-content groups, with a cutoff value of 6%. Survival analysis indicated a positive correlation between collagen content and overall survival (OS). In multivariate Cox regression analysis, incorporating factors such as mitosis, necrosis, Ki67, and collagen content and other indicators, collagen content emerged as an independent prognostic factor. Collagen content demonstrated a negative correlation with tumor histological phenotype, Ki67, WHO grade, mitosis, necrosis, and brain invasion. Additionally, the signal intensity of SFTs on T2-weighted imaging (T2WI) decreased with increasing collagen content. Radiomics analysis identified 1,702 features from each patient's region of interest, with 12 features showing significant differences between the high and low collagen content groups. Among the quantitative parameters and radiomic models, the combined T1- and T2WI models exhibited the highest diagnostic performance. Conclusion These findings suggest that collagen content is an independent prognostic risk factor for OS. Furthermore, combined radiomic models based on T1-and T2WI sequences may offer a more comprehensive, objective, and accurate assessment of collagen content in CNS SFTs.
Collapse
Affiliation(s)
- Xiaoling Li
- Department of Pathology, The Second Hospital of Longyan, Longyan, Fujian, China
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Pathology, First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, Fujian, China
| | - Hua Zhang
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Radiology, First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, Fujian, China
| | - Chengcong Hu
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Pathology, First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, Fujian, China
| | - Liwen Hu
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Pathology, First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, Fujian, China
| | - Huibin Guo
- Department of Pathology, The Second Hospital of Longyan, Longyan, Fujian, China
| | - Hongbao Chen
- Department of Pathology, The Second Hospital of Longyan, Longyan, Fujian, China
| | - Guoping Li
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Pathology, First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, Fujian, China
| | - Qian Huang
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Pathology, First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, Fujian, China
| | - Shuie Jiang
- Department of Pathology, Jianning General Hospital, Sanming, Fujian, China
| | - Sheng Zhang
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Pathology, First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhen Xing
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Radiology, First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, Fujian, China
| | - Xingfu Wang
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Pathology, First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, Fujian, China
- Department of Pathology, Jianning General Hospital, Sanming, Fujian, China
| |
Collapse
|
5
|
Singh G, Singh A, Bae J, Manjila S, Spektor V, Prasanna P, Lignelli A. -New frontiers in domain-inspired radiomics and radiogenomics: increasing role of molecular diagnostics in CNS tumor classification and grading following WHO CNS-5 updates. Cancer Imaging 2024; 24:133. [PMID: 39375809 PMCID: PMC11460168 DOI: 10.1186/s40644-024-00769-6] [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: 07/28/2024] [Accepted: 08/31/2024] [Indexed: 10/09/2024] Open
Abstract
Gliomas and Glioblastomas represent a significant portion of central nervous system (CNS) tumors associated with high mortality rates and variable prognosis. In 2021, the World Health Organization (WHO) updated its Glioma classification criteria, most notably incorporating molecular markers including CDKN2A/B homozygous deletion, TERT promoter mutation, EGFR amplification, + 7/-10 chromosome copy number changes, and others into the grading and classification of adult and pediatric Gliomas. The inclusion of these markers and the corresponding introduction of new Glioma subtypes has allowed for more specific tailoring of clinical interventions and has inspired a new wave of Radiogenomic studies seeking to leverage medical imaging information to explore the diagnostic and prognostic implications of these new biomarkers. Radiomics, deep learning, and combined approaches have enabled the development of powerful computational tools for MRI analysis correlating imaging characteristics with various molecular biomarkers integrated into the updated WHO CNS-5 guidelines. Recent studies have leveraged these methods to accurately classify Gliomas in accordance with these updated molecular-based criteria based solely on non-invasive MRI, demonstrating the great promise of Radiogenomic tools. In this review, we explore the relative benefits and drawbacks of these computational frameworks and highlight the technical and clinical innovations presented by recent studies in the landscape of fast evolving molecular-based Glioma subtyping. Furthermore, the potential benefits and challenges of incorporating these tools into routine radiological workflows, aiming to enhance patient care and optimize clinical outcomes in the evolving field of CNS tumor management, have been highlighted.
Collapse
Affiliation(s)
- Gagandeep Singh
- Neuroradiology Division, Columbia University Irving Medical Center, New York, NY, USA.
| | - Annie Singh
- Atal Bihari Vajpayee Institute of Medical Sciences, New Delhi, India
| | - Joseph Bae
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, USA
| | - Sunil Manjila
- Department of Neurological Surgery, Garden City Hospital, Garden City, MI, USA
| | - Vadim Spektor
- Neuroradiology Division, Columbia University Irving Medical Center, New York, NY, USA
| | - Prateek Prasanna
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, USA
| | - Angela Lignelli
- Neuroradiology Division, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
6
|
Agarwal A, Edgar MA, Desai A, Gupta V, Soni N, Bathla G. Molecular GBM versus Histopathological GBM: Radiology-Pathology-Genetic Correlation and the New WHO 2021 Definition of Glioblastoma. AJNR Am J Neuroradiol 2024; 45:1006-1012. [PMID: 38438167 PMCID: PMC11383408 DOI: 10.3174/ajnr.a8225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 02/29/2024] [Indexed: 03/06/2024]
Abstract
Given the recent advances in molecular pathogenesis of tumors, with better correlation with tumor behavior and prognosis, major changes were made to the new 2021 World Health Organization (WHO) classification of CNS tumors, including updated criteria for diagnosis of glioblastoma (GBM). Diagnosis of GBM now requires absence of isocitrate dehydrogenase and histone 3 mutations (IDH-wild-type and H3-wild-type) as the basic cornerstone, with elimination of the IDH-mutant category. The requirements for diagnosis were conventionally histopathological, based on the presence of pathognomonic features such as microvascular proliferation and necrosis. However, even if these histologic features are absent, many lower-grade (WHO grade 2/3) diffuse astrocytic gliomas behave clinically similar to GBM (grade 4). The 2021 WHO classification introduced new molecular criteria that can be used to upgrade the diagnosis of such histologically lower-grade, IDH-wild-type, astrocytomas to GBM. The 3 molecular criteria include: concurrent gain of whole chromosome 7 and loss of whole chromosome 10 (+7/-10); telomerase reverse transcriptase promoter mutation; and epidermal growth factor receptor amplification. Given these changes, it is now strongly recommended to have molecular analysis of WHO grade 2/3 diffuse astrocytic, IDH-wild-type, gliomas in adult patients, as identification of any of the above mutations allows for upgrading the tumor to WHO grade 4 ("molecular GBM") with important prognostic implications. Despite an early stage, there is active ongoing research on the unique MR imaging features of molecular GBM. This paper highlights the differences between "molecular" and "histopathological" GBM, with the aim of providing a basic understanding about these changes.
Collapse
Affiliation(s)
- Amit Agarwal
- From the Department of Radiology (A.A., A.D., V.G., N.S.), Mayo Clinic, Jacksonville, Florida
| | - Mark A Edgar
- Department of Laboratory Medicine and Pathology (Neuropathology) (M.A.E.), Mayo Clinic, Jacksonville, Florida
| | - Amit Desai
- From the Department of Radiology (A.A., A.D., V.G., N.S.), Mayo Clinic, Jacksonville, Florida
| | - Vivek Gupta
- From the Department of Radiology (A.A., A.D., V.G., N.S.), Mayo Clinic, Jacksonville, Florida
| | - Neetu Soni
- From the Department of Radiology (A.A., A.D., V.G., N.S.), Mayo Clinic, Jacksonville, Florida
| | - Girish Bathla
- Department of Radiology (G.B.), Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
7
|
Li Y, Liu X, Gu M, Xu T, Ge C, Chang P. Significance of MRI-based radiomics in predicting pathological complete response to neoadjuvant chemoradiotherapy of locally advanced rectal cancer: A narrative review. Cancer Radiother 2024; 28:390-401. [PMID: 39174361 DOI: 10.1016/j.canrad.2024.04.003] [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] [Received: 03/27/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 08/24/2024]
Abstract
Neoadjuvant chemoradiotherapy is the standard treatment for patients with locally advanced rectal cancers owing to its ability to downstage primary tumours. Some patients can achieve pathological complete response after neoadjuvant therapy, and can adopt a "watch and wait" treatment strategy to avoid overtreatment. Therefore, it is essential to develop strategies for predicting responses to neoadjuvant therapy. Radiomics has shown great potential in extracting tumour features from high-throughput medical images for the construction of mathematics models for predicting the effects of anticancerous therapies. Herein, we explored MRI-based radiomics and found that it can predict responses of locally advanced rectal cancers to chemoradiation. Efficient radiomics model allow early-stage prediction of the effect of neoadjuvant chemoradiotherapy on locally advanced rectal cancers. It helps clinicians to make informed therapeutic decisions. In this review, we discuss the workflow of radiomics, and summarize the clinical application of MRI-based radiomics in predicting pathological complete response to neoadjuvant chemoradiotherapy of locally advanced rectal cancer.
Collapse
Affiliation(s)
- Y Li
- Department of Radiation Oncology and Therapy, The First Hospital of Jilin University, Changchun, China
| | - X Liu
- Department of Radiation Oncology and Therapy, The First Hospital of Jilin University, Changchun, China
| | - M Gu
- Department of Radiation Oncology and Therapy, The First Hospital of Jilin University, Changchun, China
| | - T Xu
- Department of Radiation Oncology and Therapy, The First Hospital of Jilin University, Changchun, China
| | - C Ge
- Department of Radiation Oncology and Therapy, The First Hospital of Jilin University, Changchun, China
| | - P Chang
- Department of Radiation Oncology and Therapy, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
8
|
Kersch CN, Kim M, Stoller J, Barajas RF, Park JE. Imaging Genomics of Glioma Revisited: Analytic Methods to Understand Spatial and Temporal Heterogeneity. AJNR Am J Neuroradiol 2024; 45:537-548. [PMID: 38548303 PMCID: PMC11288537 DOI: 10.3174/ajnr.a8148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 11/09/2023] [Indexed: 04/12/2024]
Abstract
An improved understanding of the cellular and molecular biologic processes responsible for brain tumor development, growth, and resistance to therapy is fundamental to improving clinical outcomes. Imaging genomics is the study of the relationships between microscopic, genetic, and molecular biologic features and macroscopic imaging features. Imaging genomics is beginning to shift clinical paradigms for diagnosing and treating brain tumors. This article provides an overview of imaging genomics in gliomas, in which imaging data including hallmarks such as IDH-mutation, MGMT methylation, and EGFR-mutation status can provide critical insights into the pretreatment and posttreatment stages. This article will accomplish the following: 1) review the methods used in imaging genomics, including visual analysis, quantitative analysis, and radiomics analysis; 2) recommend suitable analytic methods for imaging genomics according to biologic characteristics; 3) discuss the clinical applicability of imaging genomics; and 4) introduce subregional tumor habitat analysis with the goal of guiding future radiogenetics research endeavors toward translation into critically needed clinical applications.
Collapse
Affiliation(s)
- Cymon N Kersch
- From the Department of Radiation Medicine (C.N.K.), Oregon Health and Science University, Portland, Oregon
| | - Minjae Kim
- Department of Radiology and Research Institute of Radiology (M.K., J.E.P.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jared Stoller
- Department of Diagnostic Radiology (J.S., R.F.B.), Oregon Health and Science University, Portland, Oregon
| | - Ramon F Barajas
- Department of Diagnostic Radiology (J.S., R.F.B.), Oregon Health and Science University, Portland, Oregon
- Knight Cancer Institute (R.F.B.), Oregon Health and Science University, Portland, Oregon
- Advanced Imaging Research Center (R.F.B.), Oregon Health and Science University, Portland, Oregon
| | - Ji Eun Park
- Department of Radiology and Research Institute of Radiology (M.K., J.E.P.), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
9
|
Liang Q, Jing H, Shao Y, Wang Y, Zhang H. Artificial Intelligence Imaging for Predicting High-risk Molecular Markers of Gliomas. Clin Neuroradiol 2024; 34:33-43. [PMID: 38277059 DOI: 10.1007/s00062-023-01375-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 12/20/2023] [Indexed: 01/27/2024]
Abstract
Gliomas, the most prevalent primary malignant tumors of the central nervous system, present significant challenges in diagnosis and prognosis. The fifth edition of the World Health Organization Classification of Tumors of the Central Nervous System (WHO CNS5) published in 2021, has emphasized the role of high-risk molecular markers in gliomas. These markers are crucial for enhancing glioma grading and influencing survival and prognosis. Noninvasive prediction of these high-risk molecular markers is vital. Genetic testing after biopsy, the current standard for determining molecular type, is invasive and time-consuming. Magnetic resonance imaging (MRI) offers a non-invasive alternative, providing structural and functional insights into gliomas. Advanced MRI methods can potentially reflect the pathological characteristics associated with glioma molecular markers; however, they struggle to fully represent gliomas' high heterogeneity. Artificial intelligence (AI) imaging, capable of processing vast medical image datasets, can extract critical molecular information. AI imaging thus emerges as a noninvasive and efficient method for identifying high-risk molecular markers in gliomas, a recent focus of research. This review presents a comprehensive analysis of AI imaging's role in predicting glioma high-risk molecular markers, highlighting challenges and future directions.
Collapse
Affiliation(s)
- Qian Liang
- Department of Radiology, First Hospital of Shanxi Medical University, 030001, Taiyuan, Shanxi Province, China
- College of Medical Imaging, Shanxi Medical University, 030001, Taiyuan, Shanxi Province, China
| | - Hui Jing
- Department of MRI, The Sixth Hospital, Shanxi Medical University, 030008, Taiyuan, Shanxi Province, China
| | - Yingbo Shao
- Department of Radiology, First Hospital of Shanxi Medical University, 030001, Taiyuan, Shanxi Province, China
- College of Medical Imaging, Shanxi Medical University, 030001, Taiyuan, Shanxi Province, China
| | - Yinhua Wang
- Department of Radiology, First Hospital of Shanxi Medical University, 030001, Taiyuan, Shanxi Province, China
- College of Medical Imaging, Shanxi Medical University, 030001, Taiyuan, Shanxi Province, China
| | - Hui Zhang
- Department of Radiology, First Hospital of Shanxi Medical University, 030001, Taiyuan, Shanxi Province, China.
- College of Medical Imaging, Shanxi Medical University, 030001, Taiyuan, Shanxi Province, China.
- Shanxi Key Laboratory of Intelligent Imaging and Nanomedicine, First Hospital of Shanxi Medical University, 030001, Taiyuan, Shanxi Province, China.
- Intelligent Imaging Big Data and Functional Nano-imaging Engineering Research Center of Shanxi Province, First Hospital of Shanxi Medical University, 030001, Taiyuan, Shanxi Province, China.
| |
Collapse
|
10
|
Li X, Cheng Y, Han X, Cui B, Li J, Yang H, Xu G, Lin Q, Xiao X, Tang J, Lu J. Exploring the association of glioma tumor residuals from incongruent [ 18F]FET PET/MR imaging with tumor proliferation using a multiparametric MRI radiomics nomogram. Eur J Nucl Med Mol Imaging 2024; 51:779-796. [PMID: 37864593 DOI: 10.1007/s00259-023-06468-x] [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: 06/05/2023] [Accepted: 09/28/2023] [Indexed: 10/23/2023]
Abstract
PURPOSE The study aimed to using multiparametric MRI radiomics to predict glioma tumor residuals (TRFET over MR) derived from incongruent [18F]fluoroethyl-L-tyrosine ([18F]FET) PET/MR imaging. METHODS One hundred ten patients with gliomas who underwent [18F]FET PET/MR scanning were retrospectively analyzed. The TRFET over MR was identified by the discrepancy-PET that the extent of resection (EOR) based on MRI subtracted the biological tumor volume on PET images. The MRI parameters and radiomics features were extracted based on EOR and selected by the least absolute shrinkage and selection operator to construct radiomics score (Rad-score). The correlation network analysis of all features was analyzed by Spearman's correlation tests. The methods for evaluating the clinical usefulness consisted of the receiver operating characteristic curve, the calibration curve, and decision curve analysis. RESULTS The Rad-score of the patients with the TRFET over MR was significantly higher than those with the non TRFET over MR (p < 0.001). The Rad-score was significantly correlated with the discrepancy-PET (r = 0.72, p < 0.001), Ki-67 level (r = 0.76, p < 0.001), and epidermal growth factor receptor (EGFR) of gliomas (r = 0.75, p < 0.001), respectively. Moreover, there was a difference of the correlation network analysis between the TRPET over MR group and non TRFET over MR group. The nomogram combing Rad-score and clinical features had the greatest performance in predicting TRFET over MR (AUC = 0.90/0.87, training/testing). There was a significant difference in prognosis (median OS, 17 m vs. 43 m) between patients with TRFET over MR and non TRFET over MR based on nomogram prediction (p < 0.001). CONCLUSION The nomogram based on MRI radiomics would predict gliomas tumor residuals caused by the absence of 18F-PET PET examination and adjust EOR to improve prognosis.
Collapse
Affiliation(s)
- Xiaoran Li
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Capital Medical University, Beijing, China
| | - Ye Cheng
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xin Han
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Capital Medical University, Beijing, China
| | - Bixiao Cui
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Capital Medical University, Beijing, China
| | - Jing Li
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Capital Medical University, Beijing, China
| | - Hongwei Yang
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Capital Medical University, Beijing, China
| | - Geng Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Qingtang Lin
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xinru Xiao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jie Tang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Jie Lu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Capital Medical University, Beijing, China.
| |
Collapse
|
11
|
Huo X, Wang Y, Ma S, Zhu S, Wang K, Ji Q, Chen F, Wang L, Wu Z, Li W. Multimodal MRI-based radiomic nomogram for predicting telomerase reverse transcriptase promoter mutation in IDH-wildtype histological lower-grade gliomas. Medicine (Baltimore) 2023; 102:e36581. [PMID: 38134061 PMCID: PMC10735121 DOI: 10.1097/md.0000000000036581] [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: 06/29/2023] [Accepted: 11/17/2023] [Indexed: 12/24/2023] Open
Abstract
The presence of TERTp mutation in isocitrate dehydrogenase-wildtype (IDHwt) histologically lower-grade glioma (LGA) has been linked to a poor prognosis. In this study, we aimed to develop and validate a radiomic nomogram based on multimodal MRI for predicting TERTp mutations in IDHwt LGA. One hundred and nine IDH wildtype glioma patients (TERTp-mutant, 78; TERTp-wildtype, 31) with clinical, radiomic, and molecular information were collected and randomly divided into training and validation set. Clinical model, fusion radiomic model, and combined radiomic nomogram were constructed for the discrimination. Radiomic features were screened with 3 algorithms (Wilcoxon rank sum test, elastic net, and the recursive feature elimination) and the clinical characteristics of combined radiomic nomogram were screened by the Akaike information criterion. Finally, receiver operating characteristic curve, calibration curve, Hosmer-Lemeshow test, and decision curve analysis were utilized to assess these models. Fusion radiomic model with 4 radiomic features achieved an area under the curve value of 0.876 and 0.845 in the training and validation set. And, the combined radiomic nomogram achieved area under the curve value of 0.897 (training set) and 0.882 (validation set). Above that, calibration curve and Hosmer-Lemeshow test showed that the radiomic model and combined radiomic nomogram had good agreement between observations and predictions in the training set and the validation set. Finally, the decision curve analysis revealed that the 2 models had good clinical usefulness for the prediction of TERTp mutation status in IDHwt LGA. The combined radiomics nomogram performed great performance and high sensitivity in prediction of TERTp mutation status in IDHwt LGA, and has good clinical application.
Collapse
Affiliation(s)
- Xulei Huo
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yali Wang
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Sihan Ma
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Sipeng Zhu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ke Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Qiang Ji
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Feng Chen
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Liang Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhen Wu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wenbin Li
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
12
|
Lan L, Feng K, Wu Y, Zhang W, Wei L, Che H, Xue L, Gao Y, Tao J, Qian S, Cao W, Zhang J, Wang C, Tian M. Phenomic Imaging. PHENOMICS (CHAM, SWITZERLAND) 2023; 3:597-612. [PMID: 38223684 PMCID: PMC10781914 DOI: 10.1007/s43657-023-00128-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 08/13/2023] [Accepted: 08/17/2023] [Indexed: 01/16/2024]
Abstract
Human phenomics is defined as the comprehensive collection of observable phenotypes and characteristics influenced by a complex interplay among factors at multiple scales. These factors include genes, epigenetics at the microscopic level, organs, microbiome at the mesoscopic level, and diet and environmental exposures at the macroscopic level. "Phenomic imaging" utilizes various imaging techniques to visualize and measure anatomical structures, biological functions, metabolic processes, and biochemical activities across different scales, both in vivo and ex vivo. Unlike conventional medical imaging focused on disease diagnosis, phenomic imaging captures both normal and abnormal traits, facilitating detailed correlations between macro- and micro-phenotypes. This approach plays a crucial role in deciphering phenomes. This review provides an overview of different phenomic imaging modalities and their applications in human phenomics. Additionally, it explores the associations between phenomic imaging and other omics disciplines, including genomics, transcriptomics, proteomics, immunomics, and metabolomics. By integrating phenomic imaging with other omics data, such as genomics and metabolomics, a comprehensive understanding of biological systems can be achieved. This integration paves the way for the development of new therapeutic approaches and diagnostic tools.
Collapse
Affiliation(s)
- Lizhen Lan
- Human Phenome Institute, Fudan University, 825 Zhangheng Road, Pudong New District, Shanghai, 201203 China
| | - Kai Feng
- Human Phenome Institute, Fudan University, 825 Zhangheng Road, Pudong New District, Shanghai, 201203 China
| | - Yudan Wu
- Human Phenome Institute, Fudan University, 825 Zhangheng Road, Pudong New District, Shanghai, 201203 China
| | - Wenbo Zhang
- Human Phenome Institute, Fudan University, 825 Zhangheng Road, Pudong New District, Shanghai, 201203 China
| | - Ling Wei
- Human Phenome Institute, Fudan University, 825 Zhangheng Road, Pudong New District, Shanghai, 201203 China
| | - Huiting Che
- Human Phenome Institute, Fudan University, 825 Zhangheng Road, Pudong New District, Shanghai, 201203 China
| | - Le Xue
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009 Zhejiang China
| | - Yidan Gao
- Human Phenome Institute, Fudan University, 825 Zhangheng Road, Pudong New District, Shanghai, 201203 China
| | - Ji Tao
- Human Phenome Institute, Fudan University, 825 Zhangheng Road, Pudong New District, Shanghai, 201203 China
| | - Shufang Qian
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009 Zhejiang China
| | - Wenzhao Cao
- Human Phenome Institute, Fudan University, 825 Zhangheng Road, Pudong New District, Shanghai, 201203 China
| | - Jun Zhang
- Department of Radiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, National Center for Neurological Disorders, Fudan University, Shanghai, 200040 China
| | - Chengyan Wang
- Human Phenome Institute, Fudan University, 825 Zhangheng Road, Pudong New District, Shanghai, 201203 China
| | - Mei Tian
- Human Phenome Institute, Fudan University, 825 Zhangheng Road, Pudong New District, Shanghai, 201203 China
| |
Collapse
|
13
|
Noor J, Chaudhry A, Batool S. Microfluidic Technology, Artificial Intelligence, and Biosensors As Advanced Technologies in Cancer Screening: A Review Article. Cureus 2023; 15:e39634. [PMID: 37388583 PMCID: PMC10305590 DOI: 10.7759/cureus.39634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/29/2023] [Indexed: 07/01/2023] Open
Abstract
Cancer screening techniques aim to detect premalignant lesions and enable early intervention to delay the onset of cancer while keeping incidence constant. Technology advancements have led to the development of powerful tools such as microfluidic technology, artificial intelligence, machine learning algorithms, and electrochemical biosensors to aid in early cancer detection. Non-invasive cancer screening methods like virtual colonoscopy and endoscopic ultrasonography have also been developed to provide comprehensive pictures of organs and detect cancer early. This review article provides an overview of recent advances in cancer screening in microfluidic technology, artificial intelligence, and biomarkers through a narrative literature search. Microfluidic devices enable easy handling of sub-microliter volumes and have become a promising tool for cancer detection, drug screening, and modeling angiogenesis and metastasis in cancer research. Machine learning and artificial intelligence have shown high accuracy in oncology-related diagnostic imaging, reducing the manual steps in lesion detection and providing standardized and accurate results, with potential for global standardization in areas like colon polyps, breast cancer, and primary and metastatic brain cancer. A biomarker-based cancer diagnosis is promising for early detection and effective therapy, and electrochemical biosensors integrated with nanoparticles offer multiplexing and amplification capabilities. Understanding these advanced technologies' basics, achievements, and challenges is crucial for advancing their use in oncology.
Collapse
Affiliation(s)
- Jawad Noor
- Internal Medicine, St. Dominic Hospital, Jackson, USA
| | | | - Saima Batool
- Pathology, Nishtar Medical University, Multan, PAK
| |
Collapse
|
14
|
Wu Q, Yu YX, Zhang T, Zhu WJ, Fan YF, Wang XM, Hu CH. Preoperative Diagnosis of Dual-Phenotype Hepatocellular Carcinoma Using Enhanced MRI Radiomics Models. J Magn Reson Imaging 2023; 57:1185-1196. [PMID: 36190656 DOI: 10.1002/jmri.28391] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Dual-phenotype hepatocellular carcinoma (DPHCC) is highly aggressive and difficult to distinguish from hepatocellular carcinoma (HCC). PURPOSE To develop and validate clinical and radiomics models based on contrast-enhanced MRI for the preoperative diagnosis of DPHCC. STUDY TYPE Retrospective. POPULATION A total of 87 patients with DPHCC and 92 patients with non-DPHCC randomly divided into a training cohort (n = 125: 64 non-DPHCC; 61 DPHCC) and a validation cohort (n = 54: 28 non-DPHCC; 26 DPHCC). FIELD STRENGTH/SEQUENCE A 3.0 T; dynamic contrast-enhanced MRI with time-resolved T1-weighted imaging sequence. ASSESSMENT In the clinical model, the maximum tumor diameter and hepatitis B virus (HBV) were independent risk factors of DPHCC. In the radiomics model, a total of 1781 radiomics features were extracted from tumor volumes of interest (VOIs) in the arterial phase (AP) and portal venous phase (PP) images. For feature reduction and selection, Pearson correlation coefficient (PCC) and recursive feature elimination (RFE) were used. Clinical, AP, PP, and combined radiomics models were established using machine learning algorithms (support vector machine [SVM], logistic regression [LR], and logistic regression-least absolute shrinkage and selection operator [LR-LASSO]) and their discriminatory efficacy assessed and compared. STATISTICAL TESTS The independent sample t test, Mann-Whitney U test, Chi-square test, regression analysis, receiver operating characteristic curve (ROC) analysis, Pearson correlation analysis, the Delong test. A P value < 0.05 was considered statistically significant. RESULTS In the validation cohort, the combined radiomics model (area under the curve [AUC] = 0.908, 95% confidence interval [CI]: 0.831-0.985) showed the highest diagnostic performance. The AUCs of the PP (AUC = 0.879, 95% CI: 0.779-0.979) and combined radiomics models were significantly higher than that of clinical model (AUC = 0.685, 95% CI: 0.526-0.844). There were no significant differences in AUC between AP or PP radiomics model and combined radiomics model (P = 0.286, 0.180 and 0.543). CONCLUSION MRI radiomics models may be useful for discriminating DPHCC from non-DPHCC before surgery. EVIDENCE LEVEL 4 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Qian Wu
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yi-Xing Yu
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Tao Zhang
- Department of Radiology, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Wen-Jing Zhu
- Department of Radiology, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Yan-Fen Fan
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xi-Ming Wang
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chun-Hong Hu
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
15
|
Jiang J, Wei J, Zhu Y, Wei L, Wei X, Tian H, Zhang L, Wang T, Cheng Y, Zhao Q, Sun Z, Du H, Huang Y, Liu H, Li Y. Clot-based radiomics model for cardioembolic stroke prediction with CT imaging before recanalization: a multicenter study. Eur Radiol 2023; 33:970-980. [PMID: 36066731 DOI: 10.1007/s00330-022-09116-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 07/11/2022] [Accepted: 08/12/2022] [Indexed: 02/03/2023]
Abstract
OBJECTIVES To develop a clot-based radiomics model using CT imaging radiomic features and machine learning to identify cardioembolic (CE) stroke before mechanical thrombectomy (MTB) in patients with acute ischemic stroke (AIS). MATERIALS AND METHODS This retrospective four-center study consecutively included 403 patients with AIS who sequentially underwent CT and MTB between April 2016 and July 2021. These were grouped into training, testing, and external validation cohorts. Thrombus-extracted radiomic features and basic information were gathered to construct a machine learning model to predict CE stroke. The radiological characteristics and basic information were used to build a routine radiological model. A combined radiomics and radiological features model was also developed. The performances of all models were evaluated and compared in the validation cohort. A histological analysis helped further assess the proposed model in all patients. RESULTS The radiomics model yielded an area under the curve (AUC) of 0.838 (95% confidence interval [CI], 0.771-0.891) for predicting CE stroke in the validation cohort, significantly higher than the radiological model (AUC, 0.713; 95% CI, 0.636-0.781; p = 0.007) but similar to the combined model (AUC, 0.855; 95% CI, 0.791-0.906; p = 0.14). The thrombus radiomic features achieved stronger correlations with red blood cells (|rmax|, 0.74 vs. 0.32) and fibrin and platelet (|rmax|, 0.68 vs. 0.18) than radiological characteristics. CONCLUSION The proposed CT-based radiomics model could reliably predict CE stroke in AIS, performing better than the routine radiological method. KEY POINTS • Admission CT imaging could offer valuable information to identify the acute ischemic stroke source by radiomics analysis. • The proposed CT imaging-based radiomics model yielded a higher area under the curve (0.838) than the routine radiological method (0.713; p = 0.007). • Several radiomic features showed significantly stronger correlations with two main thrombus constituents (red blood cells, |rmax|, 0.74; fibrin and platelet, |rmax|, 0.68) than routine radiological characteristics.
Collapse
Affiliation(s)
- Jingxuan Jiang
- Institute of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China.,Department of Radiology, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Jianyong Wei
- Clinical Research Center, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Yueqi Zhu
- Institute of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Liming Wei
- Institute of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Xiaoer Wei
- Institute of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Hao Tian
- Department of Radiology, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Lei Zhang
- Department of Radiology, Wuxi Second People's Hospital, Wuxi, 214000, China
| | - Tianle Wang
- Department of Radiology, Affiliated No. 1 People's Hospital of Nantong University, Nantong, 226001, China
| | - Yue Cheng
- Department of Radiology, Wuxi Second People's Hospital, Wuxi, 214000, China
| | - Qianqian Zhao
- Institute of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Zheng Sun
- Institute of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Haiyan Du
- Institute of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Yu Huang
- Institute of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Hui Liu
- Institute of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Yuehua Li
- Institute of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China.
| |
Collapse
|
16
|
Deng Y, Li Y, Wu JL, Zhou T, Tang MY, Chen Y, Zuo HD, Tang W, Chen TW, Zhang XM. Radiomics models based on multi-sequence MRI for preoperative evaluation of MUC4 status in pancreatic ductal adenocarcinoma: a preliminary study. Quant Imaging Med Surg 2022; 12:5129-5139. [PMID: 36330180 PMCID: PMC9622441 DOI: 10.21037/qims-22-112] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 08/24/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Mucin 4 (MUC4) overexpression promotes tumorigenesis and increases the aggressiveness of pancreatic ductal adenocarcinoma (PDAC). To date, no study has reported the association between radiomics and MUC4 expression in PDAC. Thus, we aimed to explore the utility of radiomics based on multi-sequence magnetic resonance imaging (MRI) to predict the status of MUC4 expression in PDAC preoperatively. METHODS This retrospective study included 52 patients with PDAC who underwent MRI. The patients were divided into two groups based on MUC4 expression status. Two feature sets were extracted from the arterial and portal phases (PPs) of dynamic contrast-enhanced MRI (DCE-MRI). Univariate analysis, minimum redundancy maximum relevance (MRMR), and principal component analysis (PCA) were performed for the feature selection of each dataset, and features with a cumulative variance of 90% were selected to develop radiomics models. Clinical characteristics were gathered to develop a clinical model. The selected radiomics features and clinical characteristics were modeled by multivariable logistic regression. The combined model integrated radiomics features from different selected data sets and clinical characteristics. The classification metrics were applied to assess the discriminatory power of the models. RESULTS There were 22 PDACs with a high expression of MUC4 and 30 PDACs with a low expression of MUC4. The area under the receiver operating characteristic (ROC) curve (AUC) values of the arterial phase (AP) model, the PP model, and the combined model were 0.732 (0.591-0.872), 0.709 (0.569-0.849), and 0.861 (0.760-0.961), respectively. The AUC of the clinical model was 0.666 (0.600-0.682). The combined model that was constructed outperformed the AP, the PP, and the clinical models (P<0.05, although no statistical significance was observed in the combined model vs. AP model). CONCLUSIONS Radiomics models based on multi-sequence MRI have the potential to predict MUC4 expression levels in PDAC.
Collapse
Affiliation(s)
- Yan Deng
- Medical Imaging Key Laboratory of Sichuan Province and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yong Li
- Medical Imaging Key Laboratory of Sichuan Province and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Jia-Long Wu
- Department of Radiology, The Second Clinical Medical College of North Sichuan Medical College, Nanchong Central Hospital, Nanchong, China
| | - Ting Zhou
- Department of Radiology, Sichuan Cancer Hospital, Chengdu, China
| | - Meng-Yue Tang
- Medical Imaging Key Laboratory of Sichuan Province and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yong Chen
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hou-Dong Zuo
- Medical Imaging Key Laboratory of Sichuan Province and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Wei Tang
- Medical Imaging Key Laboratory of Sichuan Province and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Tian-Wu Chen
- Medical Imaging Key Laboratory of Sichuan Province and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xiao-Ming Zhang
- Medical Imaging Key Laboratory of Sichuan Province and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| |
Collapse
|
17
|
Hong ZL, Chen S, Peng XR, Li JW, Yang JC, Wu SS. Nomograms for prediction of breast cancer in breast imaging reporting and data system (BI-RADS) ultrasound category 4 or 5 lesions: A single-center retrospective study based on radiomics features. Front Oncol 2022; 12:894476. [PMID: 36212503 PMCID: PMC9538156 DOI: 10.3389/fonc.2022.894476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 08/29/2022] [Indexed: 11/24/2022] Open
Abstract
Purpose To develop nomograms for predicting breast malignancy in BI-RADS ultrasound (US) category 4 or 5 lesions based on radiomics features. Methods Between January 2020 and January 2022, we prospectively collected and retrospectively analyzed the medical records of 496 patients pathologically proven breast lesions in our hospital. The data set was divided into model training group and validation testing group with a 75/25 split. Radiomics features were obtained using the PyRadiomics package, and the radiomics score was established by least absolute shrinkage and selection operator regression. A nomogram was developed for BI-RADS US category 4 or 5 lesions according to the results of multivariate regression analysis from the training group. Result The AUCs of radiomics score consisting of 31 US features was 0.886. The AUC of the model constructed with radiomics score, patient age, lesion diameter identified by US and BI-RADS category involved was 0.956 (95% CI, 0.910–0.972) for the training group and 0.937 (95% CI, 0.893–0.965) for the validation cohort. The calibration curves showed good agreement between the predictions and observations. Conclusions Both nomogram and radiomics score can be used as methods to assist radiologists and clinicians in predicting breast malignancy in BI-RADS US category 4 or 5 lesions.
Collapse
Affiliation(s)
- Zhi-Liang Hong
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- Department of Ultrasound, Fujian Provincial Hospital, Fuzhou, China
| | - Sheng Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- Department of Ultrasound, Fujian Provincial Hospital, Fuzhou, China
| | - Xiao-Rui Peng
- Clinical Skills Teaching Center, Fujian Medical University, Fuzhou, China
| | - Jian-Wei Li
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- Department of Ultrasound, Fujian Provincial Hospital, Fuzhou, China
| | - Jian-Chuan Yang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- Department of Ultrasound, Fujian Provincial Hospital, Fuzhou, China
| | - Song-Song Wu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- Department of Ultrasound, Fujian Provincial Hospital, Fuzhou, China
- *Correspondence: Song-Song Wu,
| |
Collapse
|
18
|
Kihira S, Mei X, Mahmoudi K, Liu Z, Dogra S, Belani P, Tsankova N, Hormigo A, Fayad ZA, Doshi A, Nael K. U-Net Based Segmentation and Characterization of Gliomas. Cancers (Basel) 2022; 14:4457. [PMID: 36139616 PMCID: PMC9496685 DOI: 10.3390/cancers14184457] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/18/2022] Open
Abstract
(1) Background: Gliomas are the most common primary brain neoplasms accounting for roughly 40−50% of all malignant primary central nervous system tumors. We aim to develop a deep learning-based framework for automated segmentation and prediction of biomarkers and prognosis in patients with gliomas. (2) Methods: In this retrospective two center study, patients were included if they (1) had a diagnosis of glioma with known surgical histopathology and (2) had preoperative MRI with FLAIR sequence. The entire tumor volume including FLAIR hyperintense infiltrative component and necrotic and cystic components was segmented. Deep learning-based U-Net framework was developed based on symmetric architecture from the 512 × 512 segmented maps from FLAIR as the ground truth mask. (3) Results: The final cohort consisted of 208 patients with mean ± standard deviation of age (years) of 56 ± 15 with M/F of 130/78. DSC of the generated mask was 0.93. Prediction for IDH-1 and MGMT status had a performance of AUC 0.88 and 0.62, respectively. Survival prediction of <18 months demonstrated AUC of 0.75. (4) Conclusions: Our deep learning-based framework can detect and segment gliomas with excellent performance for the prediction of IDH-1 biomarker status and survival.
Collapse
Affiliation(s)
- Shingo Kihira
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Radiological Sciences, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA 90033, USA
| | - Xueyan Mei
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Keon Mahmoudi
- Department of Radiological Sciences, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA 90033, USA
| | - Zelong Liu
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Siddhant Dogra
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Puneet Belani
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Nadejda Tsankova
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Adilia Hormigo
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Zahi A. Fayad
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Amish Doshi
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kambiz Nael
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Radiological Sciences, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA 90033, USA
| |
Collapse
|
19
|
Abdollahi H, Chin E, Clark H, Hyde DE, Thomas S, Wu J, Uribe CF, Rahmim A. Radiomics-guided radiation therapy: opportunities and challenges. Phys Med Biol 2022; 67. [DOI: 10.1088/1361-6560/ac6fab] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 05/13/2022] [Indexed: 11/11/2022]
Abstract
Abstract
Radiomics is an advanced image-processing framework, which extracts image features and considers them as biomarkers towards personalized medicine. Applications include disease detection, diagnosis, prognosis, and therapy response assessment/prediction. As radiation therapy aims for further individualized treatments, radiomics could play a critical role in various steps before, during and after treatment. Elucidation of the concept of radiomics-guided radiation therapy (RGRT) is the aim of this review, attempting to highlight opportunities and challenges underlying the use of radiomics to guide clinicians and physicists towards more effective radiation treatments. This work identifies the value of RGRT in various steps of radiotherapy from patient selection to follow-up, and subsequently provides recommendations to improve future radiotherapy using quantitative imaging features.
Collapse
|
20
|
Varriano G, Guerriero P, Santone A, Mercaldo F, Brunese L. Explainability of radiomics through formal methods. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2022; 220:106824. [PMID: 35483269 DOI: 10.1016/j.cmpb.2022.106824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 04/12/2022] [Accepted: 04/18/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND AND OBJECTIVE Artificial Intelligence has proven to be effective in radiomics. The main problem in using Artificial Intelligence is that researchers and practitioners are not able to know how the predictions are generated. This is currently an open issue because results' explainability is advantageous in understanding the reasoning behind the model, both for patients than for implementing a feedback mechanism for medical specialists using decision support systems. METHODS Addressing transparency issues related to the Artificial Intelligence field, the innovative technique of Formal methods use a mathematical logic reasoning to produce an automatic, quick and reliable diagnosis. In this paper we analyze results given by the adoption of Formal methods for the diagnosis of the Coronavirus disease: specifically, we want to analyse and understand, in a more medical way, the meaning of some radiomic features to connect them with clinical or radiological evidences. RESULTS In particular, the usage of Formal methods allows the authors to do statistical analysis on the feature value distributions, to do pattern recognition on disease models, to generalize the model of a disease and to reach high performances of results and interpretation of them. A further step for explainability can be accounted by the localization and selection of the most important slices in a multi-slice approach. CONCLUSIONS In conclusion, we confirmed the clinical significance of some First order features as Skewness and Kurtosis. On the other hand, we suggest to decline the use of the Minimum feature because of its intrinsic connection with the Computational Tomography exam of the lung.
Collapse
Affiliation(s)
- Giulia Varriano
- Department of Medicine and Health Sciences V. Tiberio, University of Molise, 86100 Campobasso, Italy.
| | - Pasquale Guerriero
- Department of Medicine and Health Sciences V. Tiberio, University of Molise, 86100 Campobasso, Italy.
| | - Antonella Santone
- Department of Medicine and Health Sciences V. Tiberio, University of Molise, 86100 Campobasso, Italy.
| | - Francesco Mercaldo
- Department of Medicine and Health Sciences V. Tiberio, University of Molise, 86100 Campobasso, Italy.
| | - Luca Brunese
- Department of Medicine and Health Sciences V. Tiberio, University of Molise, 86100 Campobasso, Italy.
| |
Collapse
|
21
|
Nakajo M, Takeda A, Katsuki A, Jinguji M, Ohmura K, Tani A, Sato M, Yoshiura T. The efficacy of 18F-FDG-PET-based radiomic and deep-learning features using a machine-learning approach to predict the pathological risk subtypes of thymic epithelial tumors. Br J Radiol 2022; 95:20211050. [PMID: 35312337 PMCID: PMC10996420 DOI: 10.1259/bjr.20211050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 02/28/2022] [Accepted: 03/14/2022] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE To examine whether the machine-learning approach using 18-fludeoxyglucose positron emission tomography (18F-FDG-PET)-based radiomic and deep-learning features is useful for predicting the pathological risk subtypes of thymic epithelial tumors (TETs). METHODS This retrospective study included 79 TET [27 low-risk thymomas (types A, AB and B1), 31 high-risk thymomas (types B2 and B3) and 21 thymic carcinomas] patients who underwent pre-therapeutic 18F-FDG-PET/CT. High-risk TETs (high-risk thymomas and thymic carcinomas) were 52 patients. The 107 PET-based radiomic features, including SUV-related parameters [maximum SUV (SUVmax), metabolic tumor volume (MTV), and total lesion glycolysis (TLG)] and 1024 deep-learning features extracted from the convolutional neural network were used to predict the pathological risk subtypes of TETs using six different machine-learning algorithms. The area under the curves (AUCs) were calculated to compare the predictive performances. RESULTS SUV-related parameters yielded the following AUCs for predicting thymic carcinomas: SUVmax 0.713, MTV 0.442, and TLG 0.479 or high-risk TETs: SUVmax 0.673, MTV 0.533, and TLG 0.539. The best-performing algorithm was the logistic regression model for predicting thymic carcinomas (AUC 0.900, accuracy 81.0%), and the random forest (RF) model for high-risk TETs (AUC 0.744, accuracy 72.2%). The AUC was significantly higher in the logistic regression model than three SUV-related parameters for predicting thymic carcinomas, and in the RF model than MTV and TLG for predicting high-risk TETs (each; p < 0.05). CONCLUSION 18F-FDG-PET-based radiomic analysis using a machine-learning approach may be useful for predicting the pathological risk subtypes of TETs. ADVANCES IN KNOWLEDGE Machine-learning approach using 18F-FDG-PET-based radiomic features has the potential to predict the pathological risk subtypes of TETs.
Collapse
Affiliation(s)
- Masatoyo Nakajo
- Department of Radiology, Kagoshima University, Graduate School
of Medical and Dental Sciences,
Kagoshima, Japan
| | - Aya Takeda
- Department of General Thoracic Surgery, Kagoshima University,
Graduate School of Medical and Dental Sciences,
Kagoshima, Japan
| | - Akie Katsuki
- Research and Development Department, GE Healthcare
Japan, Tokyo,
Japan
| | - Megumi Jinguji
- Department of Radiology, Kagoshima University, Graduate School
of Medical and Dental Sciences,
Kagoshima, Japan
| | - Kazuyuki Ohmura
- Research and Development Department, GE Healthcare
Japan, Tokyo,
Japan
| | - Atsushi Tani
- Department of Radiology, Kagoshima University, Graduate School
of Medical and Dental Sciences,
Kagoshima, Japan
| | - Masami Sato
- Department of General Thoracic Surgery, Kagoshima University,
Graduate School of Medical and Dental Sciences,
Kagoshima, Japan
| | - Takashi Yoshiura
- Department of Radiology, Kagoshima University, Graduate School
of Medical and Dental Sciences,
Kagoshima, Japan
| |
Collapse
|
22
|
Balana C, Castañer S, Carrato C, Moran T, Lopez-Paradís A, Domenech M, Hernandez A, Puig J. Preoperative Diagnosis and Molecular Characterization of Gliomas With Liquid Biopsy and Radiogenomics. Front Neurol 2022; 13:865171. [PMID: 35693015 PMCID: PMC9177999 DOI: 10.3389/fneur.2022.865171] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 05/05/2022] [Indexed: 12/13/2022] Open
Abstract
Gliomas are a heterogenous group of central nervous system tumors with different outcomes and different therapeutic needs. Glioblastoma, the most common subtype in adults, has a very poor prognosis and disabling consequences. The World Health Organization (WHO) classification specifies that the typing and grading of gliomas should include molecular markers. The molecular characterization of gliomas has implications for prognosis, treatment planning, and prediction of treatment response. At present, gliomas are diagnosed via tumor resection or biopsy, which are always invasive and frequently risky methods. In recent years, however, substantial advances have been made in developing different methods for the molecular characterization of tumors through the analysis of products shed in body fluids. Known as liquid biopsies, these analyses can potentially provide diagnostic and prognostic information, guidance on choice of treatment, and real-time information on tumor status. In addition, magnetic resonance imaging (MRI) is another good source of tumor data; radiomics and radiogenomics can link the imaging phenotypes to gene expression patterns and provide insights to tumor biology and underlying molecular signatures. Machine and deep learning and computational techniques can also use quantitative imaging features to non-invasively detect genetic mutations. The key molecular information obtained with liquid biopsies and radiogenomics can be useful not only in the diagnosis of gliomas but can also help predict response to specific treatments and provide guidelines for personalized medicine. In this article, we review the available data on the molecular characterization of gliomas using the non-invasive methods of liquid biopsy and MRI and suggest that these tools could be used in the future for the preoperative diagnosis of gliomas.
Collapse
Affiliation(s)
- Carmen Balana
- Medical Oncology Service, Institut Català d'Oncologia Badalona (ICO), Badalona Applied Research Group in Oncology (B-ARGO Group), Institut Investigació Germans Trias i Pujol (IGTP), Barcelona, Spain
- *Correspondence: Carmen Balana
| | - Sara Castañer
- Diagnostic Imaging Institute (IDI), Hospital Universitari Germans Trias I Pujol, Institut Investigació Germans Trias i Pujol (IGTP), Barcelona, Spain
| | - Cristina Carrato
- Department of Pathology, Hospital Universitari Germans Trias I Pujol, Institut Investigació Germans Trias i Pujol (IGTP), Barcelona, Spain
| | - Teresa Moran
- Medical Oncology Service, Institut Català d'Oncologia Badalona (ICO), Badalona Applied Research Group in Oncology (B-ARGO Group), Institut Investigació Germans Trias i Pujol (IGTP), Barcelona, Spain
| | - Assumpció Lopez-Paradís
- Medical Oncology Service, Institut Català d'Oncologia Badalona (ICO), Badalona Applied Research Group in Oncology (B-ARGO Group), Institut Investigació Germans Trias i Pujol (IGTP), Barcelona, Spain
| | - Marta Domenech
- Medical Oncology Service, Institut Català d'Oncologia Badalona (ICO), Badalona Applied Research Group in Oncology (B-ARGO Group), Institut Investigació Germans Trias i Pujol (IGTP), Barcelona, Spain
| | - Ainhoa Hernandez
- Medical Oncology Service, Institut Català d'Oncologia Badalona (ICO), Badalona Applied Research Group in Oncology (B-ARGO Group), Institut Investigació Germans Trias i Pujol (IGTP), Barcelona, Spain
| | - Josep Puig
- Department of Radiology IDI [Girona Biomedical Research Institute] IDIBGI, Hospital Universitari Dr Josep Trueta, Girona, Spain
- Department of Medical Sciences, School of Medicine, University of Girona, Girona, Spain
- Comparative Medicine and Bioimage of Catalonia, Institut Investigació Germans Trias i Pujol (IGTP), Barcelona, Spain
| |
Collapse
|
23
|
Texture Analysis of Enhanced MRI and Pathological Slides Predicts EGFR Mutation Status in Breast Cancer. BIOMED RESEARCH INTERNATIONAL 2022; 2022:1376659. [PMID: 35663041 PMCID: PMC9162871 DOI: 10.1155/2022/1376659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/25/2022] [Accepted: 04/29/2022] [Indexed: 12/02/2022]
Abstract
Objective Image texture information was extracted from enhanced magnetic resonance imaging (MRI) and pathological hematoxylin and eosin- (HE-) stained images of female breast cancer patients. We established models individually, and then, we combine the two kinds of data to establish model. Through this method, we verified whether sufficient information could be obtained from enhanced MRI and pathological slides to assist in the determination of epidermal growth factor receptor (EGFR) mutation status in patients. Methods We obtained enhanced MRI data from patients with breast cancer before treatment and selected diffusion-weighted imaging (DWI), T1 fast-spin echo (T1 FSE), and T2 fast-spin echo (T2 FSE) as the data sources for extracting texture information. Imaging physicians manually outlined the 3D regions of interest (ROIs) and extracted texture features according to the gray level cooccurrence matrix (GLCM) of the images. For the HE staining images of the patients, we adopted a specific normalization algorithm to simulate the images dyed with only hematoxylin or eosin and extracted textures. We extracted texture features to predict the expression of EGFR. After evaluating the predictive power of each model, the models from the two data sources were combined for remodeling. Results For enhanced MRI data, the modeling of texture information of T1 FSE had a good predictive effect for EGFR mutation status. For pathological images, eosin-stained images can achieve a better prediction effect. We selected these two classifiers as the weak classifiers of the final model and obtained good results (training group: AUC, 0.983; 95% CI, 0.95-1.00; accuracy, 0.962; specificity, 0.936; and sensitivity, 0.979; test group: AUC, 0.983; 95% CI, 0.94-1.00; accuracy, 0.943; specificity, 1.00; and sensitivity, 0.905). Conclusion The EGFR mutation status of patients with breast cancer can be well predicted based on enhanced MRI data and pathological data. This helps hospitals that do not test the EGFR mutation status of patients with breast cancer. The technology gives clinicians more information about breast cancer, which helps them make accurate diagnoses and select suitable treatments.
Collapse
|
24
|
Huang W, Wang J, Wang H, Zhang Y, Zhao F, Li K, Su L, Kang F, Cao X. PET/CT Based EGFR Mutation Status Classification of NSCLC Using Deep Learning Features and Radiomics Features. Front Pharmacol 2022; 13:898529. [PMID: 35571081 PMCID: PMC9092283 DOI: 10.3389/fphar.2022.898529] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 04/11/2022] [Indexed: 12/24/2022] Open
Abstract
Purpose: This study aimed to compare the performance of radiomics and deep learning in predicting EGFR mutation status in patients with lung cancer based on PET/CT images, and tried to explore a model with excellent prediction performance to accurately predict EGFR mutation status in patients with non-small cell lung cancer (NSCLC). Method: PET/CT images of 194 NSCLC patients from Xijing Hospital were collected and divided into a training set and a validation set according to the ratio of 7:3. Statistics were made on patients' clinical characteristics, and a large number of features were extracted based on their PET/CT images (4306 radiomics features and 2048 deep learning features per person) with the pyradiomics toolkit and 3D convolutional neural network. Then a radiomics model (RM), a deep learning model (DLM), and a hybrid model (HM) were established. The performance of the three models was compared by receiver operating characteristic (ROC) curves, sensitivity, specificity, accuracy, calibration curves, and decision curves. In addition, a nomogram based on a deep learning score (DS) and the most significant clinical characteristic was plotted. Result: In the training set composed of 138 patients (64 with EGFR mutation and 74 without EGFR mutation), the area under the ROC curve (AUC) of HM (0.91, 95% CI: 0.86-0.96) was higher than that of RM (0.82, 95% CI: 0.75-0.89) and DLM (0.90, 95% CI: 0.85-0.95). In the validation set composed of 57 patients (32 with EGFR mutation and 25 without EGFR mutation), the AUC of HM (0.85, 95% CI: 0.77-0.93) was also higher than that of RM (0.68, 95% CI: 0.52-0.84) and DLM (0.79, 95% CI: 0.67-0.91). In all, HM achieved better diagnostic performance in predicting EGFR mutation status in NSCLC patients than two other models. Conclusion: Our study showed that the deep learning model based on PET/CT images had better performance than radiomics model in diagnosing EGFR mutation status of NSCLC patients based on PET/CT images. Combined with the most statistically significant clinical characteristic (smoking) and deep learning features, our hybrid model had better performance in predicting EGFR mutation types of patients than two other models, which could enable NSCLC patients to choose more personalized treatment schemes.
Collapse
Affiliation(s)
- Weicheng Huang
- School of Information Science and Technology, Northwest University, Xi'an, China.,National and Local Joint Engineering Research Center for Cultural Heritage Digitization, Xi'an, China
| | - Jingyi Wang
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Haolin Wang
- School of Information Science and Technology, Northwest University, Xi'an, China.,National and Local Joint Engineering Research Center for Cultural Heritage Digitization, Xi'an, China
| | - Yuxiang Zhang
- School of Information Science and Technology, Northwest University, Xi'an, China.,National and Local Joint Engineering Research Center for Cultural Heritage Digitization, Xi'an, China
| | - Fengjun Zhao
- School of Information Science and Technology, Northwest University, Xi'an, China.,National and Local Joint Engineering Research Center for Cultural Heritage Digitization, Xi'an, China
| | - Kang Li
- School of Information Science and Technology, Northwest University, Xi'an, China.,National and Local Joint Engineering Research Center for Cultural Heritage Digitization, Xi'an, China
| | - Linzhi Su
- School of Information Science and Technology, Northwest University, Xi'an, China.,National and Local Joint Engineering Research Center for Cultural Heritage Digitization, Xi'an, China
| | - Fei Kang
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xin Cao
- School of Information Science and Technology, Northwest University, Xi'an, China.,National and Local Joint Engineering Research Center for Cultural Heritage Digitization, Xi'an, China
| |
Collapse
|
25
|
Shaffer A, Kwok SS, Naik A, Anderson AT, Lam F, Wszalek T, Arnold PM, Hassaneen W. Ultra-High-Field MRI in the Diagnosis and Management of Gliomas: A Systematic Review. Front Neurol 2022; 13:857825. [PMID: 35449515 PMCID: PMC9016277 DOI: 10.3389/fneur.2022.857825] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 03/02/2022] [Indexed: 12/03/2022] Open
Abstract
Importance: Gliomas, tumors of the central nervous system, are classically diagnosed through invasive surgical biopsy and subsequent histopathological study. Innovations in ultra-high field (UHF) imaging, namely 7-Tesla magnetic resonance imaging (7T MRI) are advancing preoperative tumor grading, visualization of intratumoral structures, and appreciation of small brain structures and lesions. Objective Summarize current innovative uses of UHF imaging techniques in glioma diagnostics and treatment. Methods A systematic review in accordance with PRISMA guidelines was performed utilizing PubMed. Case reports and series, observational clinical trials, and randomized clinical trials written in English were included. After removing unrelated studies and those with non-human subjects, only those related to 7T MRI were independently reviewed and summarized for data extraction. Some preclinical animal models are briefly described to demonstrate future usages of ultra-high-field imaging. Results We reviewed 46 studies (43 human and 3 animal models) which reported clinical usages of UHF MRI in the diagnosis and management of gliomas. Current literature generally supports greater resolution imaging from 7T compared to 1.5T or 3T MRI, improving visualization of cerebral microbleeds and white and gray matter, and providing more precise localization for radiotherapy targeting. Additionally, studies found that diffusion or susceptibility-weighted imaging techniques applied to 7T MRI, may be used to predict tumor grade, reveal intratumoral structures such as neovasculature and microstructures like axons, and indicate isocitrate dehydrogenase 1 mutation status in preoperative imaging. Similarly, newer imaging techniques such as magnetic resonance spectroscopy and chemical exchange saturation transfer imaging can be performed on 7T MRI to predict tumor grading and treatment efficacy. Geometrical distortion, a known challenge of 7T MRI, was at a tolerable level in all included studies. Conclusion UHF imaging has the potential to preoperatively and non-invasively grade gliomas, provide precise therapy target areas, and visualize lesions not seen on conventional MRI.
Collapse
Affiliation(s)
- Annabelle Shaffer
- Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | - Susanna S Kwok
- Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | - Anant Naik
- Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | - Aaron T Anderson
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Champaign, IL, United States.,Carle Illinois Advanced Imaging Center, University of Illinois and Carle Health, Urbana, IL, United States
| | - Fan Lam
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Champaign, IL, United States.,Department of Bioengineering, University of Illinois at Urbana-Champaign, Champaign, IL, United States.,Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | - Tracey Wszalek
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Champaign, IL, United States.,Carle Illinois Advanced Imaging Center, University of Illinois and Carle Health, Urbana, IL, United States
| | - Paul M Arnold
- Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, Champaign, IL, United States.,Carle Department of Neurosurgery, Carle Foundation Hospital, Urbana, IL, United States
| | - Wael Hassaneen
- Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, Champaign, IL, United States.,Carle Department of Neurosurgery, Carle Foundation Hospital, Urbana, IL, United States
| |
Collapse
|
26
|
Cui Z, Ren G, Cai R, Wu C, Shi H, Wang X, Zhu M. MRI-based texture analysis for differentiate between pediatric posterior fossa ependymoma type A and B. Eur J Radiol 2022; 152:110288. [DOI: 10.1016/j.ejrad.2022.110288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/01/2022] [Accepted: 03/28/2022] [Indexed: 11/03/2022]
|
27
|
Vobugari N, Raja V, Sethi U, Gandhi K, Raja K, Surani SR. Advancements in Oncology with Artificial Intelligence-A Review Article. Cancers (Basel) 2022; 14:1349. [PMID: 35267657 PMCID: PMC8909088 DOI: 10.3390/cancers14051349] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 02/26/2022] [Accepted: 02/28/2022] [Indexed: 02/05/2023] Open
Abstract
Well-trained machine learning (ML) and artificial intelligence (AI) systems can provide clinicians with therapeutic assistance, potentially increasing efficiency and improving efficacy. ML has demonstrated high accuracy in oncology-related diagnostic imaging, including screening mammography interpretation, colon polyp detection, glioma classification, and grading. By utilizing ML techniques, the manual steps of detecting and segmenting lesions are greatly reduced. ML-based tumor imaging analysis is independent of the experience level of evaluating physicians, and the results are expected to be more standardized and accurate. One of the biggest challenges is its generalizability worldwide. The current detection and screening methods for colon polyps and breast cancer have a vast amount of data, so they are ideal areas for studying the global standardization of artificial intelligence. Central nervous system cancers are rare and have poor prognoses based on current management standards. ML offers the prospect of unraveling undiscovered features from routinely acquired neuroimaging for improving treatment planning, prognostication, monitoring, and response assessment of CNS tumors such as gliomas. By studying AI in such rare cancer types, standard management methods may be improved by augmenting personalized/precision medicine. This review aims to provide clinicians and medical researchers with a basic understanding of how ML works and its role in oncology, especially in breast cancer, colorectal cancer, and primary and metastatic brain cancer. Understanding AI basics, current achievements, and future challenges are crucial in advancing the use of AI in oncology.
Collapse
Affiliation(s)
- Nikitha Vobugari
- Department of Internal Medicine, Medstar Washington Hospital Center, Washington, DC 20010, USA; (N.V.); (K.G.)
| | - Vikranth Raja
- Department of Medicine, P.S.G Institute of Medical Sciences and Research, Coimbatore 641004, Tamil Nadu, India;
| | - Udhav Sethi
- School of Computer Science, University of Waterloo, Waterloo, ON N2L 3G1, Canada;
| | - Kejal Gandhi
- Department of Internal Medicine, Medstar Washington Hospital Center, Washington, DC 20010, USA; (N.V.); (K.G.)
| | - Kishore Raja
- Department of Pediatric Cardiology, University of Minnesota, Minneapolis, MN 55454, USA;
| | - Salim R. Surani
- Department of Pulmonary and Critical Care, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
28
|
Tian Y, Komolafe TE, Chen T, Zhou B, Yang X. Prediction of TACE Treatment Response in a Preoperative MRI via Analysis of Integrating Deep Learning and Radiomics Features. J Med Biol Eng 2022. [DOI: 10.1007/s40846-022-00692-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
29
|
Li G, Li L, Li Y, Qian Z, Wu F, He Y, Jiang H, Li R, Wang D, Zhai Y, Wang Z, Jiang T, Zhang J, Zhang W. An MRI radiomics approach to predict survival and tumour-infiltrating macrophages in gliomas. Brain 2022; 145:1151-1161. [PMID: 35136934 PMCID: PMC9050568 DOI: 10.1093/brain/awab340] [Citation(s) in RCA: 137] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/15/2021] [Accepted: 08/18/2021] [Indexed: 01/08/2023] Open
Abstract
Preoperative MRI is one of the most important clinical results for the diagnosis and treatment of glioma patients. The objective of this study was to construct a stable and validatable preoperative T2-weighted MRI-based radiomics model for predicting the survival of gliomas. A total of 652 glioma patients across three independent cohorts were covered in this study including their preoperative T2-weighted MRI images, RNA-seq and clinical data. Radiomic features (1731) were extracted from preoperative T2-weighted MRI images of 167 gliomas (discovery cohort) collected from Beijing Tiantan Hospital and then used to develop a radiomics prediction model through a machine learning-based method. The performance of the radiomics prediction model was validated in two independent cohorts including 261 gliomas from the The Cancer Genomae Atlas database (external validation cohort) and 224 gliomas collected in the prospective study from Beijing Tiantan Hospital (prospective validation cohort). RNA-seq data of gliomas from discovery and external validation cohorts were applied to establish the relationship between biological function and the key radiomics features, which were further validated by single-cell sequencing and immunohistochemical staining. The 14 radiomic features-based prediction model was constructed from preoperative T2-weighted MRI images in the discovery cohort, and showed highly robust predictive power for overall survival of gliomas in external and prospective validation cohorts. The radiomic features in the prediction model were associated with immune response, especially tumour macrophage infiltration. The preoperative T2-weighted MRI radiomics prediction model can stably predict the survival of glioma patients and assist in preoperatively assessing the extent of macrophage infiltration in glioma tumours.
Collapse
Affiliation(s)
- Guanzhang Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Lin Li
- Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Yiming Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Zenghui Qian
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Fan Wu
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China
| | - Yufei He
- Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Haoyu Jiang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Renpeng Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Di Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - You Zhai
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China
| | - Zhiliang Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Tao Jiang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China.,Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing 100070, China.,China National Clinical Research Center for Neurological Diseases, Beijing 100070, China.,Chinese Glioma Genome Atlas Network and Asian Glioma Genome Atlas Network, Beijing, China
| | - Jing Zhang
- Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Wei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China.,Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China.,China National Clinical Research Center for Neurological Diseases, Beijing 100070, China.,Chinese Glioma Genome Atlas Network and Asian Glioma Genome Atlas Network, Beijing, China
| |
Collapse
|
30
|
The application of radiomics in predicting gene mutations in cancer. Eur Radiol 2022; 32:4014-4024. [DOI: 10.1007/s00330-021-08520-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 12/11/2021] [Accepted: 12/14/2021] [Indexed: 12/24/2022]
|
31
|
Cui W, Wang Y, Ren J, Hubbard CS, Fu X, Fang S, Wang D, Zhang H, Li Y, Li L, Jiang T, Liu H. Personalized
fMRI
delineates functional regions preserved within brain tumors. Ann Neurol 2022; 91:353-366. [PMID: 35023218 PMCID: PMC9107064 DOI: 10.1002/ana.26303] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 01/09/2022] [Accepted: 01/10/2022] [Indexed: 11/09/2022]
Abstract
Objective Methods Results Interpretation
Collapse
Affiliation(s)
- Weigang Cui
- Department of Automation Science and Electrical Engineering Beihang University Beijing China
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology Massachusetts General Hospital, Harvard Medical School Charlestown MA USA
- Department of Neuroscience Medical University of South Carolina Charleston SC USA
- School of Engineering Medicine, Beihang University Beijing China
| | - Yinyan Wang
- Department of Neurosurgery Beijing Tiantan Hospital, Capital Medical University Beijing China
- Beijing Neurosurgical Institute, Capital Medical University Beijing China
| | - Jianxun Ren
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology Massachusetts General Hospital, Harvard Medical School Charlestown MA USA
- National Engineering Laboratory for Neuromodulation School of Aerospace Engineering, Tsinghua University Beijing China
| | - Catherine S. Hubbard
- Department of Neuroscience Medical University of South Carolina Charleston SC USA
| | - Xiaoxuan Fu
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology Massachusetts General Hospital, Harvard Medical School Charlestown MA USA
- Department of Neuroscience Medical University of South Carolina Charleston SC USA
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment Hebei University of Technology Tianjin China
| | - Shengyu Fang
- Beijing Neurosurgical Institute, Capital Medical University Beijing China
| | - Danhong Wang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology Massachusetts General Hospital, Harvard Medical School Charlestown MA USA
| | - Hao Zhang
- Department of Neurological Rehabilitation Beijing Bo'ai Hospital, China Rehabilitation Research Center Beijing China
| | - Yang Li
- Department of Automation Science and Electrical Engineering Beihang University Beijing China
| | - Luming Li
- National Engineering Laboratory for Neuromodulation School of Aerospace Engineering, Tsinghua University Beijing China
- Precision Medicine & Healthcare Research Center, Tsinghua‐Berkeley Shenzhen Institute, Tsinghua University Shenzhen Guangdong China
- IDG/McGovern Institute for Brain Research at Tsinghua University Beijing China
| | - Tao Jiang
- Department of Neurosurgery Beijing Tiantan Hospital, Capital Medical University Beijing China
- Beijing Neurosurgical Institute, Capital Medical University Beijing China
| | - Hesheng Liu
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology Massachusetts General Hospital, Harvard Medical School Charlestown MA USA
- Department of Neuroscience Medical University of South Carolina Charleston SC USA
| |
Collapse
|
32
|
Nowakowski A, Lahijanian Z, Panet-Raymond V, Siegel PM, Petrecca K, Maleki F, Dankner M. Radiomics as an emerging tool in the management of brain metastases. Neurooncol Adv 2022; 4:vdac141. [PMID: 36284932 PMCID: PMC9583687 DOI: 10.1093/noajnl/vdac141] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Brain metastases (BM) are associated with significant morbidity and mortality in patients with advanced cancer. Despite significant advances in surgical, radiation, and systemic therapy in recent years, the median overall survival of patients with BM is less than 1 year. The acquisition of medical images, such as computed tomography (CT) and magnetic resonance imaging (MRI), is critical for the diagnosis and stratification of patients to appropriate treatments. Radiomic analyses have the potential to improve the standard of care for patients with BM by applying artificial intelligence (AI) with already acquired medical images to predict clinical outcomes and direct the personalized care of BM patients. Herein, we outline the existing literature applying radiomics for the clinical management of BM. This includes predicting patient response to radiotherapy and identifying radiation necrosis, performing virtual biopsies to predict tumor mutation status, and determining the cancer of origin in brain tumors identified via imaging. With further development, radiomics has the potential to aid in BM patient stratification while circumventing the need for invasive tissue sampling, particularly for patients not eligible for surgical resection.
Collapse
Affiliation(s)
- Alexander Nowakowski
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Québec, Canada
| | - Zubin Lahijanian
- McGill University Health Centre, Department of Diagnostic Radiology, McGill University, Montreal, Québec, Canada
| | - Valerie Panet-Raymond
- McGill University Health Centre, Department of Diagnostic Radiology, McGill University, Montreal, Québec, Canada
| | - Peter M Siegel
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Québec, Canada
| | - Kevin Petrecca
- Montreal Neurological Institute-Hospital, McGill University, Montreal, Québec, Canada
| | - Farhad Maleki
- Department of Computer Science, University of Calgary, Calgary, Alberta, Canada
| | - Matthew Dankner
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Québec, Canada
| |
Collapse
|
33
|
Li L, Su Q, Yang H. Preoperative prediction of microvascular invasion in hepatocellular carcinoma: a radiomic nomogram based on MRI. Clin Radiol 2021; 77:e269-e279. [PMID: 34980458 DOI: 10.1016/j.crad.2021.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 12/08/2021] [Indexed: 11/18/2022]
Abstract
AIM To develop a reliable model to predict microvascular invasion (MVI) in patients with hepatocellular carcinoma (HCC) by combining a large number of clinical and imaging examinations, especially the radiomic features of magnetic resonance imaging (MRI). MATERIALS AND METHODS Three hundred and one consecutive patients from two centres were enrolled. Least absolute shrinkage and selection operator (LASSO) regression was used to shrink the feature size, and logistic regression was used to construct a predictive radiomic signature. The ability of the nomogram to discriminate MVI in patients with HCC was evaluated using area under the curve (AUC) of receiver operating characteristics (ROC), accuracy, and calibration curves. RESULTS The radiomic signature showed a significant association with MVI (p<0.001 for all data sets). Other useful predictors of MVI included non-smooth tumour margin, internal arteries, and the alpha-fetoprotein (AFP) level. The nomogram demonstrated a strong prognostic capability in the training set and both validation sets, providing AUCs of 0.914 (95% confidence interval [CI] 0.853-0.956), 0.872 (95% CI: 0.757-0.946), and 0.881 (95% CI: 0.806-0.934), respectively. CONCLUSIONS The preoperative radiomic nomogram, incorporating clinical risk factors and a radiomic signature, could predict MVI in patients with HCC. The MRI-based radiomic-clinical model predicted the MVI of HCC effectively and was more efficient compared with the radiomic model or clinical model alone.
Collapse
Affiliation(s)
- L Li
- Department of Hepatobiliary Surgery, The People's Hospital of Qijiang, Chongqing, China
| | - Q Su
- Department of Hepatopancreatobiliary Surgery, The Affiliated Calmette Hospital of Kunming Medical University, The First People's Hospital of Kunming, Calmette Hospital Kunming, Yunnan Province, China.
| | - H Yang
- Department of Hepatobiliary Surgery, The People's Hospital of Qijiang, Chongqing, China.
| |
Collapse
|
34
|
Wang Z, Tang X, Wu J, Zhang Z, He K, Wu D, Chen S, Xiao X. Radiomics features based on T2-weighted fluid-attenuated inversion recovery MRI predict the expression levels of CD44 and CD133 in lower-grade gliomas. Future Oncol 2021; 18:807-819. [PMID: 34783576 DOI: 10.2217/fon-2021-1173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Objective: To verify the association between CD44 and CD133 expression levels and the prognosis of patients with lower-grade gliomas (LGGs) and constructing radiomic models to predict those two genes' expression levels before surgery. Materials & methods: Genomic data of patients with LGG and the corresponding T2-weighted fluid-attenuated inversion recovery images were downloaded from the Cancer Genome Atlas and the Cancer Imaging Archive, which were utilized for prognosis analysis, radiomic feature extraction and model construction, respectively. Results & conclusion: CD44 and CD133 expression levels in LGG can significantly affect the prognosis of patients with LGG. Based on the T2-weighted fluid-attenuated inversion recovery images, the radiomic features can effectively predict the expression levels of CD44 and CD133 before surgery.
Collapse
Affiliation(s)
- Zhenhua Wang
- Department of Radiology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Xiaoping Tang
- Department of Radiology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Ji Wu
- Department of Radiology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Zhaotao Zhang
- Department of Radiology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Keng He
- Department of Radiology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Di Wu
- Department of Radiology, First Affiliated Hospital of GanNan Medical College, GanZhou, China
| | - ShiQi Chen
- Department of Radiology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Xinlan Xiao
- Department of Radiology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| |
Collapse
|
35
|
Gore S, Chougule T, Jagtap J, Saini J, Ingalhalikar M. A Review of Radiomics and Deep Predictive Modeling in Glioma Characterization. Acad Radiol 2021; 28:1599-1621. [PMID: 32660755 DOI: 10.1016/j.acra.2020.06.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/11/2020] [Accepted: 06/11/2020] [Indexed: 12/22/2022]
Abstract
Recent developments in glioma categorization based on biological genotypes and application of computational machine learning or deep learning based predictive models using multi-modal MRI biomarkers to assess these genotypes provides potential assurance for optimal and personalized treatment plans and efficacy. Artificial intelligence based quantified assessment of glioma using MRI derived hand-crafted or auto-extracted features have become crucial as genomic alterations can be associated with MRI based phenotypes. This survey integrates all the recent work carried out in state-of-the-art radiomics, and Artificial Intelligence based learning solutions related to molecular diagnosis, prognosis, and treatment monitoring with the aim to create a structured resource on radiogenomic analysis of glioma. Challenges such as inter-scanner variability, requirement of benchmark datasets, prospective validations for clinical applicability are discussed with further scope for designing optimal solutions for glioma stratification with immediate recommendations for further diagnostic decisions and personalized treatment plans for glioma patients.
Collapse
|
36
|
Abdel Razek AAK, Alksas A, Shehata M, AbdelKhalek A, Abdel Baky K, El-Baz A, Helmy E. Clinical applications of artificial intelligence and radiomics in neuro-oncology imaging. Insights Imaging 2021; 12:152. [PMID: 34676470 PMCID: PMC8531173 DOI: 10.1186/s13244-021-01102-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 09/26/2021] [Indexed: 12/15/2022] Open
Abstract
This article is a comprehensive review of the basic background, technique, and clinical applications of artificial intelligence (AI) and radiomics in the field of neuro-oncology. A variety of AI and radiomics utilized conventional and advanced techniques to differentiate brain tumors from non-neoplastic lesions such as inflammatory and demyelinating brain lesions. It is used in the diagnosis of gliomas and discrimination of gliomas from lymphomas and metastasis. Also, semiautomated and automated tumor segmentation has been developed for radiotherapy planning and follow-up. It has a role in the grading, prediction of treatment response, and prognosis of gliomas. Radiogenomics allowed the connection of the imaging phenotype of the tumor to its molecular environment. In addition, AI is applied for the assessment of extra-axial brain tumors and pediatric tumors with high performance in tumor detection, classification, and stratification of patient's prognoses.
Collapse
Affiliation(s)
| | - Ahmed Alksas
- Biomaging Lab, Department of Bioengineering, University of Louisville, Louisville, KY, 40292, USA
| | - Mohamed Shehata
- Biomaging Lab, Department of Bioengineering, University of Louisville, Louisville, KY, 40292, USA
| | - Amr AbdelKhalek
- Internship at Mansoura University Hospital, Mansoura Faculty of Medicine, Mansoura, Egypt
| | - Khaled Abdel Baky
- Department of Diagnostic Radiology, Faculty of Medicine, Port Said University, Port Said, Egypt
| | - Ayman El-Baz
- Biomaging Lab, Department of Bioengineering, University of Louisville, Louisville, KY, 40292, USA
| | - Eman Helmy
- Department of Diagnostic Radiology, Faculty of Medicine, Mansoura University, Elgomheryia Street, Mansoura, 3512, Egypt.
| |
Collapse
|
37
|
Tian Y, Komolafe TE, Zheng J, Zhou G, Chen T, Zhou B, Yang X. Assessing PD-L1 Expression Level via Preoperative MRI in HCC Based on Integrating Deep Learning and Radiomics Features. Diagnostics (Basel) 2021; 11:1875. [PMID: 34679573 PMCID: PMC8534850 DOI: 10.3390/diagnostics11101875] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/02/2021] [Accepted: 10/04/2021] [Indexed: 12/30/2022] Open
Abstract
To assess if quantitative integrated deep learning and radiomics features can predict the PD-L1 expression level in preoperative MRI of hepatocellular carcinoma (HCC) patients. The data in this study consist of 103 hepatocellular carcinoma patients who received immunotherapy in a single center. These patients were divided into a high PD-L1 expression group (30 patients) and a low PD-L1 expression group (73 patients). Both radiomics and deep learning features were extracted from their MRI sequence of T2-WI, which were merged into an integrative feature space for machine learning for the prediction of PD-L1 expression. The five-fold cross-validation was adopted to validate the performance of the model, while the AUC was used to assess the predictive ability of the model. Based on the five-fold cross-validation, the integrated model achieved the best prediction performance, with an AUC score of 0.897 ± 0.084, followed by the deep learning-based model with an AUC of 0.852 ± 0.043 then the radiomics-based model with AUC of 0.794 ± 0.035. The feature set integrating radiomics and deep learning features is more effective in predicting PD-L1 expression level than only one feature type. The integrated model can achieve fast and accurate prediction of PD-L1 expression status in preoperative MRI of HCC patients.
Collapse
Affiliation(s)
- Yuchi Tian
- Academy of Engineering and Technology, Fudan University, Shanghai 200433, China;
| | | | - Jian Zheng
- Department of Medical Imaging, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China;
| | - Guofeng Zhou
- Department of Radiology, Zhongshan Hospital, Shanghai 200032, China;
| | - Tao Chen
- School of Information Science and Technology, Fudan University, Shanghai 200433, China;
| | - Bo Zhou
- Department of Interventional Radiology, Zhongshan Hospital, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Xiaodong Yang
- Academy of Engineering and Technology, Fudan University, Shanghai 200433, China;
- Department of Medical Imaging, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China;
| |
Collapse
|
38
|
Wang ZH, Xiao XL, Zhang ZT, He K, Hu F. A Radiomics Model for Predicting Early Recurrence in Grade II Gliomas Based on Preoperative Multiparametric Magnetic Resonance Imaging. Front Oncol 2021; 11:684996. [PMID: 34540662 PMCID: PMC8443788 DOI: 10.3389/fonc.2021.684996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 08/12/2021] [Indexed: 12/23/2022] Open
Abstract
Objective This study aimed to develop a radiomics model to predict early recurrence (<1 year) in grade II glioma after the first resection. Methods The pathological, clinical, and magnetic resonance imaging (MRI) data of patients diagnosed with grade II glioma who underwent surgery and had a recurrence between 2017 and 2020 in our hospital were retrospectively analyzed. After a rigorous selection, 64 patients were eligible and enrolled in the study. Twenty-two cases had a pathologically confirmed recurrent glioma. The cases were randomly assigned using a ratio of 7:3 to either the training set or validation set. T1-weighted image (T1WI), T2-weighted image (T2WI), and contrast-enhanced T1-weighted image (T1CE) were acquired. The minimum-redundancy-maximum-relevancy (mRMR) method alone or in combination with univariate logistic analysis were used to identify the most optimal predictive feature from the three image sequences. Multivariate logistic regression analysis was then used to develop a predictive model using the screened features. The performance of each model in both training and validation datasets was assessed using a receiver operating characteristic (ROC) curve, calibration curve, and decision curve analysis (DCA). Results A total of 396 radiomics features were initially extracted from each image sequence. After running the mRMR and univariate logistic analysis, nine predictive features were identified and used to build the multiparametric radiomics model. The model had a higher AUC when compared with the univariate models in both training and validation data sets with an AUC of 0.966 (95% confidence interval: 0.949–0.99) and 0.930 (95% confidence interval: 0.905–0.973), respectively. The calibration curves indicated a good agreement between the predictable and the actual probability of developing recurrence. The DCA demonstrated that the predictive value of the model improved when combining the three MRI sequences. Conclusion Our multiparametric radiomics model could be used as an efficient and accurate tool for predicting the recurrence of grade II glioma.
Collapse
Affiliation(s)
- Zhen-Hua Wang
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xin-Lan Xiao
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhao-Tao Zhang
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Keng He
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Feng Hu
- Department of Radiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
39
|
Han Y, Zhang L, Niu S, Chen S, Yang B, Chen H, Zheng F, Zang Y, Zhang H, Xin Y, Chen X. Differentiation Between Glioblastoma Multiforme and Metastasis From the Lungs and Other Sites Using Combined Clinical/Routine MRI Radiomics. Front Cell Dev Biol 2021; 9:710461. [PMID: 34513840 PMCID: PMC8427511 DOI: 10.3389/fcell.2021.710461] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 08/09/2021] [Indexed: 01/17/2023] Open
Abstract
Background Differentiation between cerebral glioblastoma multiforme (GBM) and solitary brain metastasis (MET) is important. The existing radiomic differentiation method ignores the clinical and routine magnetic resonance imaging (MRI) features. Purpose To differentiate between GBM and MET and between METs from the lungs (MET-lung) and other sites (MET-other) through clinical and routine MRI, and radiomics analyses. Methods and Materials A total of 350 patients were collected from two institutions, including 182 patients with GBM and 168 patients with MET, which were all proven by pathology. The ROI of the tumor was obtained on axial postcontrast MRI which was performed before operation. Seven radiomic feature selection methods and four classification algorithms constituted 28 classifiers in two classification strategies, with the best classifier serving as the final radiomics model. The clinical and combination models were constructed using the nomograms developed. The performance of the nomograms was evaluated in terms of calibration, discrimination, and clinical usefulness. Student’s t-test or the chi-square test was used to assess the differences in the clinical and radiological characteristics between the training and internal validation cohorts. Receiver operating characteristic curve analysis was performed to assess the performance of developed models with the area under the curve (AUC). Results The classifier fisher_decision tree (fisher_DT) showed the best performance (AUC: 0.696, 95% CI:0.608-0.783) for distinguishing between GBM and MET in internal validation cohorts; the classifier reliefF_random forest (reliefF_RF) showed the best performance (AUC: 0.759, 95% CI: 0.613-0.904) for distinguishing between MET-lung and MET-other in internal validation cohorts. The combination models incorporating the radiomics signature and clinical-radiological characteristics were superior to the clinical-radiological models in the two classification strategies (AUC: 0.764 for differentiation between GBM in internal validation cohorts and MET and 0.759 or differentiation between MET-lung and MET-other in internal validation cohorts). The nomograms showed satisfactory performance and calibration and were considered clinically useful, as revealed in the decision curve analysis. Data Conclusion The combination of radiomic and non-radiomic features is helpful for the differentiation among GBM, MET-lung, and MET-other.
Collapse
Affiliation(s)
- Yuqi Han
- School of Life Sciences and Technology, Xidian University, Xi'an, China.,Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Lingling Zhang
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shuzi Niu
- Institute of Software, Chinese Academy of Sciences, Beijing, China
| | - Shuguang Chen
- School of Mathematical Sciences, Nankai University, Tianjin, China
| | - Bo Yang
- Department of Computing, The Hong Kong Polytechnic University, Hong Kong, China
| | - Hongyan Chen
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Fei Zheng
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yuying Zang
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hongbo Zhang
- Department of Neurosurgery, Huizhou Third People's Hospital, Guangzhou Medical University, Huizhou, China
| | - Yu Xin
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xuzhu Chen
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
40
|
Wei RL, Wei XT. Advanced Diagnosis of Glioma by Using Emerging Magnetic Resonance Sequences. Front Oncol 2021; 11:694498. [PMID: 34422648 PMCID: PMC8374052 DOI: 10.3389/fonc.2021.694498] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 07/19/2021] [Indexed: 12/15/2022] Open
Abstract
Glioma, the most common primary brain tumor in adults, can be difficult to discern radiologically from other brain lesions, which affects surgical planning and follow-up treatment. Recent advances in MRI demonstrate that preoperative diagnosis of glioma has stepped into molecular and algorithm-assisted levels. Specifically, the histology-based glioma classification is composed of multiple different molecular subtypes with distinct behavior, prognosis, and response to therapy, and now each aspect can be assessed by corresponding emerging MR sequences like amide proton transfer-weighted MRI, inflow-based vascular-space-occupancy MRI, and radiomics algorithm. As a result of this novel progress, the clinical practice of glioma has been updated. Accurate diagnosis of glioma at the molecular level can be achieved ahead of the operation to formulate a thorough plan including surgery radical level, shortened length of stay, flexible follow-up plan, timely therapy response feedback, and eventually benefit patients individually.
Collapse
Affiliation(s)
- Ruo-Lun Wei
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin-Ting Wei
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
41
|
Fournier L, Costaridou L, Bidaut L, Michoux N, Lecouvet FE, de Geus-Oei LF, Boellaard R, Oprea-Lager DE, Obuchowski NA, Caroli A, Kunz WG, Oei EH, O'Connor JPB, Mayerhoefer ME, Franca M, Alberich-Bayarri A, Deroose CM, Loewe C, Manniesing R, Caramella C, Lopci E, Lassau N, Persson A, Achten R, Rosendahl K, Clement O, Kotter E, Golay X, Smits M, Dewey M, Sullivan DC, van der Lugt A, deSouza NM, European Society Of Radiology. Incorporating radiomics into clinical trials: expert consensus endorsed by the European Society of Radiology on considerations for data-driven compared to biologically driven quantitative biomarkers. Eur Radiol 2021; 31:6001-6012. [PMID: 33492473 PMCID: PMC8270834 DOI: 10.1007/s00330-020-07598-8] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/16/2020] [Accepted: 12/03/2020] [Indexed: 02/07/2023]
Abstract
Existing quantitative imaging biomarkers (QIBs) are associated with known biological tissue characteristics and follow a well-understood path of technical, biological and clinical validation before incorporation into clinical trials. In radiomics, novel data-driven processes extract numerous visually imperceptible statistical features from the imaging data with no a priori assumptions on their correlation with biological processes. The selection of relevant features (radiomic signature) and incorporation into clinical trials therefore requires additional considerations to ensure meaningful imaging endpoints. Also, the number of radiomic features tested means that power calculations would result in sample sizes impossible to achieve within clinical trials. This article examines how the process of standardising and validating data-driven imaging biomarkers differs from those based on biological associations. Radiomic signatures are best developed initially on datasets that represent diversity of acquisition protocols as well as diversity of disease and of normal findings, rather than within clinical trials with standardised and optimised protocols as this would risk the selection of radiomic features being linked to the imaging process rather than the pathology. Normalisation through discretisation and feature harmonisation are essential pre-processing steps. Biological correlation may be performed after the technical and clinical validity of a radiomic signature is established, but is not mandatory. Feature selection may be part of discovery within a radiomics-specific trial or represent exploratory endpoints within an established trial; a previously validated radiomic signature may even be used as a primary/secondary endpoint, particularly if associations are demonstrated with specific biological processes and pathways being targeted within clinical trials. KEY POINTS: • Data-driven processes like radiomics risk false discoveries due to high-dimensionality of the dataset compared to sample size, making adequate diversity of the data, cross-validation and external validation essential to mitigate the risks of spurious associations and overfitting. • Use of radiomic signatures within clinical trials requires multistep standardisation of image acquisition, image analysis and data mining processes. • Biological correlation may be established after clinical validation but is not mandatory.
Collapse
Affiliation(s)
- Laure Fournier
- PARCC, INSERM, Radiology Department, AP-HP, Hopital europeen Georges Pompidou, Université de Paris, F-75015, Paris, France
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
| | - Lena Costaridou
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
- School of Medicine, University of Patras, University Campus, Rio, 26 500, Patras, Greece
| | - Luc Bidaut
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- College of Science, University of Lincoln, Lincoln, LN6 7TS, UK
| | - Nicolas Michoux
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiology, Institut de Recherche Expérimentale et Clinique (IREC), Cliniques Universitaires Saint Luc, Université Catholique de Louvain (UCLouvain), B-1200, Brussels, Belgium
| | - Frederic E Lecouvet
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiology, Institut de Recherche Expérimentale et Clinique (IREC), Cliniques Universitaires Saint Luc, Université Catholique de Louvain (UCLouvain), B-1200, Brussels, Belgium
| | - Lioe-Fee de Geus-Oei
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Biomedical Photonic Imaging Group, University of Twente, Enschede, The Netherlands
| | - Ronald Boellaard
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
- Department of Radiology & Nuclear Medicine, Cancer Centre Amsterdam, Amsterdam University Medical Centers (VU University), Amsterdam, The Netherlands
- Quantitative Imaging Biomarkers Alliance, Radiological Society of North America, Oak Brook, IL, USA
| | - Daniela E Oprea-Lager
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiology & Nuclear Medicine, Cancer Centre Amsterdam, Amsterdam University Medical Centers (VU University), Amsterdam, The Netherlands
| | - Nancy A Obuchowski
- Quantitative Imaging Biomarkers Alliance, Radiological Society of North America, Oak Brook, IL, USA
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Anna Caroli
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
- Department of Biomedical Engineering, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Wolfgang G Kunz
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Edwin H Oei
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
- Department of Radiology & Nuclear Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - James P B O'Connor
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Marius E Mayerhoefer
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Manuela Franca
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
- Department of Radiology, Centro Hospitalar Universitário do Porto, Instituto de Ciências Biomédicas de Abel Salazar, University of Porto, Porto, Portugal
| | - Angel Alberich-Bayarri
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
- Quantitative Imaging Biomarkers in Medicine (QUIBIM), Valencia, Spain
| | - Christophe M Deroose
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Christian Loewe
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
- Division of Cardiovascular and Interventional Radiology, Dept. for Bioimaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Rashindra Manniesing
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Caroline Caramella
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Radiology Department, Hôpital Marie Lannelongue, Institut d'Oncologie Thoracique, Université Paris-Saclay, Le Plessis-Robinson, France
| | - Egesta Lopci
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Nuclear Medicine, Humanitas Clinical and Research Hospital - IRCCS, Rozzano, MI, Italy
| | - Nathalie Lassau
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Quantitative Imaging Biomarkers Alliance, Radiological Society of North America, Oak Brook, IL, USA
- Imaging Department, Gustave Roussy Cancer Campus Grand, Paris, UMR 1281, INSERM, CNRS, CEA, Universite Paris-Saclay, Saint-Aubin, France
| | - Anders Persson
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
- Department of Radiology, and Department of Health, Medicine and Caring Sciences, Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Rik Achten
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
- Department of Radiology and Medical Imaging, Ghent University Hospital, Gent, Belgium
| | - Karen Rosendahl
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
- Department of Radiology, University Hospital of North Norway, Tromsø, Norway
| | - Olivier Clement
- PARCC, INSERM, Radiology Department, AP-HP, Hopital europeen Georges Pompidou, Université de Paris, F-75015, Paris, France
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
| | - Elmar Kotter
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
- Department of Radiology, University Medical Center Freiburg, Freiburg, Germany
| | - Xavier Golay
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
- Quantitative Imaging Biomarkers Alliance, Radiological Society of North America, Oak Brook, IL, USA
- Queen Square Institute of Neurology, University College London, London, UK
| | - Marion Smits
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiology & Nuclear Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Marc Dewey
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
- Department of Radiology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Daniel C Sullivan
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
- Quantitative Imaging Biomarkers Alliance, Radiological Society of North America, Oak Brook, IL, USA
- Dept. of Radiology, Duke University, 311 Research Dr, Durham, NC, 27710, USA
| | - Aad van der Lugt
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
- Department of Radiology & Nuclear Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Nandita M deSouza
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria.
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium.
- Quantitative Imaging Biomarkers Alliance, Radiological Society of North America, Oak Brook, IL, USA.
- Division of Radiotherapy and Imaging, The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, London, UK.
| | | |
Collapse
|
42
|
Jiang Z, Dong Y, Yang L, Lv Y, Dong S, Yuan S, Li D, Liu L. CT-Based Hand-crafted Radiomic Signatures Can Predict PD-L1 Expression Levels in Non-small Cell Lung Cancer: a Two-Center Study. J Digit Imaging 2021; 34:1073-1085. [PMID: 34327623 DOI: 10.1007/s10278-021-00484-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 05/27/2021] [Accepted: 06/21/2021] [Indexed: 12/01/2022] Open
Abstract
Here, we used pre-treatment CT images to develop and evaluate a radiomic signature that can predict the expression of programmed death ligand 1 (PD-L1) in non-small cell lung cancer (NSCLC). We then verified its predictive performance by cross-referencing its results with clinical characteristics. This two-center retrospective analysis included 125 patients with histologically confirmed NSCLC. A total of 1287 hand-crafted radiomic features were observed from manually determined tumor regions. Valuable features were then selected with a ridge regression-based recursive feature elimination approach. Machine learning-based prediction models were then built from this and compared each other. The final radiomic signature was built using logistic regression in the primary cohort, and then tested in a validation cohort. Finally, we compared the efficacy of the radiomic signature to the clinical model and the radiomic-clinical nomogram. Among the 125 patients, 89 were classified as having PD-L1 positive expression. However, there was no significant difference in PD-L1 expression levels determined by clinical characteristics (P = 0.109-0.955). Upon selecting 9 radiomic features, we found that the logistic regression-based prediction model performed the best (AUC = 0.96, P < 0.001). In the external cohort, our radiomic signature showed an AUC of 0.85, which outperformed both the clinical model (AUC = 0.38, P < 0.001) and the radiomics-nomogram model (AUC = 0.61, P < 0.001). Our CT-based hand-crafted radiomic signature model can effectively predict PD-L1 expression levels, providing a noninvasive means of better understanding PD-L1 expression in patients with NSCLC.
Collapse
Affiliation(s)
- Zekun Jiang
- Shandong Key Laboratory of Medical Physics and Image Processing, Shandong Institute of Industrial Technology for Health Sciences and Precision Medicine, School of Physics and Electronics, Shandong Normal University, Jinan, 250358, Shandong, China
| | - Yinjun Dong
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China.,Liaocheng People's Hospital, Liaocheng, 252002, Shandong, China.,Shandong University, Jinan, 250117, Shandong, China
| | - Linke Yang
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Yunhong Lv
- Department of Mathematics and Information Technology, Xingtai University, Xingtai, 054001, Hebei, China.,Department of Mathematics and Statistics, University of Windsor, Windsor, ON, N9B 3P4, Canada
| | - Shuai Dong
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Shuanghu Yuan
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China.
| | - Dengwang Li
- Shandong Key Laboratory of Medical Physics and Image Processing, Shandong Institute of Industrial Technology for Health Sciences and Precision Medicine, School of Physics and Electronics, Shandong Normal University, Jinan, 250358, Shandong, China.
| | - Liheng Liu
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
43
|
Ren M, Yang H, Lai Q, Shi D, Liu G, Shuang X, Su J, Xie L, Dong Y, Jiang X. MRI-based radiomics analysis for predicting the EGFR mutation based on thoracic spinal metastases in lung adenocarcinoma patients. Med Phys 2021; 48:5142-5151. [PMID: 34318502 DOI: 10.1002/mp.15137] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 07/08/2021] [Accepted: 07/21/2021] [Indexed: 11/11/2022] Open
Abstract
PURPOSE This study aims to develop and evaluate multi-parametric MRI-based radiomics for preoperative identification of epidermal growth factor receptor (EGFR) mutation, which is important in treatment planning for patients with thoracic spinal metastases from primary lung adenocarcinoma. METHODS A total of 110 patients were enrolled between January 2016 and March 2019 as a primary cohort. A time-independent validation cohort was conducted containing 52 patients consecutively enrolled from July 2019 to April 2021. The patients were pathologically diagnosed with thoracic spinal metastases from primary lung adenocarcinoma; all underwent T1-weighted (T1W), T2-weighted (T2W), and T2-weighted fat-suppressed (T2FS) MRI scans of the thoracic spinal. Handcrafted and deep learning-based features were extracted and selected from each MRI modality, and used to build the radiomics signature. Various machine learning classifiers were developed and compared. A clinical-radiomics nomogram integrating the combined rad signature and the most important clinical factor was constructed with receiver operating characteristic (ROC), calibration, and decision curves analysis (DCA) to evaluate the prediction performance. RESULTS The combined radiomics signature derived from the joint of three modalities can effectively classify EGFR mutation and EGFR wild-type patients, with an area under the ROC curve (AUC) of 0.886 (95% confidence interval [CI]: 0.826-0.947, SEN =0.935, SPE =0.688) in the training group and 0.803 (95% CI: 0.682-0.924, SEN = 0.700, SPE = 0.818) in the time-independent validation group. The nomogram incorporating the combined radiomics signature and smoking status achieved the best prediction performance in the training (AUC = 0.888, 95% CI: 0.849-0.958, SEN = 0.839, SPE = 0.792) and time-independent validation (AUC = 0.821, 95% CI: 0.692-0.929, SEN = 0.667, SPE = 0.909) cohorts. The DCA confirmed potential clinical usefulness of our nomogram. CONCLUSION Our study demonstrated the potential of multi-parametric MRI-based radiomics on preoperatively predicting the EGFR mutation. The proposed nomogram model can be considered as a new biomarker to guide the selection of individual treatment strategies for patients with thoracic spinal metastases from primary lung adenocarcinoma.
Collapse
Affiliation(s)
- Meihong Ren
- Department of Biomedical Engineering, School of Fundamental Sciences, China Medical University, Shenyang, P.R. China
| | - Huazhe Yang
- Department of Biophysics, School of Fundamental Sciences, China Medical University, Shenyang, P.R. China
| | - Qingyuan Lai
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, P.R. China
| | - Dabao Shi
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, P.R. China
| | - Guanyu Liu
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, P.R. China
| | - Xue Shuang
- Department of Biomedical Engineering, School of Fundamental Sciences, China Medical University, Shenyang, P.R. China
| | - Juan Su
- Department of Biomedical Engineering, School of Fundamental Sciences, China Medical University, Shenyang, P.R. China
| | - Liping Xie
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, P.R. China
| | - Yue Dong
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, P.R. China
| | - Xiran Jiang
- Department of Biomedical Engineering, School of Fundamental Sciences, China Medical University, Shenyang, P.R. China
| |
Collapse
|
44
|
Safai A, Shinde S, Jadhav M, Chougule T, Indoria A, Kumar M, Santosh V, Jabeen S, Beniwal M, Konar S, Saini J, Ingalhalikar M. Developing a Radiomics Signature for Supratentorial Extra-Ventricular Ependymoma Using Multimodal MR Imaging. Front Neurol 2021; 12:648092. [PMID: 34367044 PMCID: PMC8339322 DOI: 10.3389/fneur.2021.648092] [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: 12/31/2020] [Accepted: 06/14/2021] [Indexed: 11/25/2022] Open
Abstract
Rationale and Objectives: To build a machine learning-based diagnostic model that can accurately distinguish adult supratentorial extraventricular ependymoma (STEE) from similarly appearing high-grade gliomas (HGG) using quantitative radiomic signatures from a multi-parametric MRI framework. Materials and Methods: We computed radiomic features on the preprocessed and segmented tumor masks from a pre-operative multimodal MRI dataset [contrast-enhanced T1 (T1ce), T2, fluid-attenuated inversion recovery (FLAIR), apparent diffusion coefficient (ADC)] from STEE (n = 15), HGG-Grade IV (HGG-G4) (n = 24), and HGG-Grade III (HGG-G3) (n = 36) patients, followed by an optimum two-stage feature selection and multiclass classification. Performance of multiple classifiers were evaluated on both unimodal and multimodal feature sets and most discriminative radiomic features involved in classification of STEE from HGG subtypes were obtained. Results: Multimodal features demonstrated higher classification performance over unimodal feature set in discriminating STEE and HGG subtypes with an accuracy of 68% on test data and above 80% on cross validation, along with an overall above 90% specificity. Among unimodal feature sets, those extracted from FLAIR demonstrated high classification performance in delineating all three tumor groups. Texture-based radiomic features particularly from FLAIR were most important in discriminating STEE from HGG-G4, whereas first-order features from T2 and ADC consistently ranked higher in differentiating multiple tumor groups. Conclusions: This study illustrates the utility of radiomics-based multimodal MRI framework in accurately discriminating similarly appearing adult STEE from HGG subtypes. Radiomic features from multiple MRI modalities could capture intricate and complementary information for a robust and highly accurate multiclass tumor classification.
Collapse
Affiliation(s)
- Apoorva Safai
- Symbiosis Center for Medical Image Analysis, Symbiosis Institute of Technology, Symbiosis International University, Pune, India
| | - Sumeet Shinde
- Symbiosis Center for Medical Image Analysis, Symbiosis Institute of Technology, Symbiosis International University, Pune, India
| | - Manali Jadhav
- Symbiosis Center for Medical Image Analysis, Symbiosis Institute of Technology, Symbiosis International University, Pune, India
| | - Tanay Chougule
- Symbiosis Center for Medical Image Analysis, Symbiosis Institute of Technology, Symbiosis International University, Pune, India
| | - Abhilasha Indoria
- Department of Neuroimaging & Interventional Radiology, National Institute of Mental Health & Neurosciences, Bangalore, India
| | - Manoj Kumar
- Department of Neuroimaging & Interventional Radiology, National Institute of Mental Health & Neurosciences, Bangalore, India
| | - Vani Santosh
- Department of Neuropathology, National Institute of Mental Health & Neurosciences, Bangalore, India
| | - Shumyla Jabeen
- Department of Neuroimaging & Interventional Radiology, National Institute of Mental Health & Neurosciences, Bangalore, India
| | - Manish Beniwal
- Department of Neurosurgery, National Institute of Mental Health & Neurosciences, Bangalore, India
| | - Subhash Konar
- Department of Neurosurgery, National Institute of Mental Health & Neurosciences, Bangalore, India
| | - Jitender Saini
- Department of Neuroimaging & Interventional Radiology, National Institute of Mental Health & Neurosciences, Bangalore, India
| | - Madhura Ingalhalikar
- Symbiosis Center for Medical Image Analysis, Symbiosis Institute of Technology, Symbiosis International University, Pune, India
| |
Collapse
|
45
|
Jafari SH, Rabiei N, Taghizadieh M, Mirazimi SMA, Kowsari H, Farzin MA, Razaghi Bahabadi Z, Rezaei S, Mohammadi AH, Alirezaei Z, Dashti F, Nejati M. Joint application of biochemical markers and imaging techniques in the accurate and early detection of glioblastoma. Pathol Res Pract 2021; 224:153528. [PMID: 34171601 DOI: 10.1016/j.prp.2021.153528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 06/09/2021] [Accepted: 06/14/2021] [Indexed: 11/28/2022]
Abstract
Glioblastoma is a primary brain tumor with the most metastatic effect in adults. Despite the wide range of multidimensional treatments, tumor heterogeneity is one of the main causes of tumor spread and gives great complexity to diagnostic and therapeutic methods. Therefore, featuring noble noninvasive prognostic methods that are focused on glioblastoma heterogeneity is perceived as an urgent need. Imaging neuro-oncological biomarkers including MGMT (O6-methylguanine-DNA methyltransferase) promoter methylation status, tumor grade along with other tumor characteristics and demographic features (e.g., age) are commonly referred to during diagnostic, therapeutic and prognostic processes. Therefore, the use of new noninvasive prognostic methods focused on glioblastoma heterogeneity is considered an urgent need. Some neuronal biomarkers, including the promoter methylation status of the promoter MGMT, the characteristics and grade of the tumor, along with the patient's demographics (such as age and sex) are involved in diagnosis, treatment, and prognosis. Among the wide array of imaging techniques, magnetic resonance imaging combined with the more physiologically detailed technique of H-magnetic resonance spectroscopy can be useful in diagnosing neurological cancer patients. In addition, intracranial tumor qualitative analysis and sometimes tumor biopsies help in accurate diagnosis. This review summarizes the evidence for biochemical biomarkers being a reliable biomarker in the early detection and disease management in GBM. Moreover, we highlight the correlation between Imaging techniques and biochemical biomarkers and ask whether they can be combined.
Collapse
Affiliation(s)
- Seyed Hamed Jafari
- Medical Imaging Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nikta Rabiei
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Taghizadieh
- Department of Pathology, School of Medicine, Center for Women's Health Research Zahra, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sayad Mohammad Ali Mirazimi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamed Kowsari
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Amin Farzin
- Department of Laboratory Medicine, School of Allied Medical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Zahra Razaghi Bahabadi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Samaneh Rezaei
- Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Hossein Mohammadi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Zahra Alirezaei
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Paramedical School, Bushehr University of Medical Sciences, Bushehr, Iran.
| | - Fatemeh Dashti
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran.
| | - Majid Nejati
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
46
|
La Greca Saint-Esteven A, Vuong D, Tschanz F, van Timmeren JE, Dal Bello R, Waller V, Pruschy M, Guckenberger M, Tanadini-Lang S. Systematic Review on the Association of Radiomics with Tumor Biological Endpoints. Cancers (Basel) 2021; 13:cancers13123015. [PMID: 34208595 PMCID: PMC8234501 DOI: 10.3390/cancers13123015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 12/23/2022] Open
Abstract
Radiomics supposes an alternative non-invasive tumor characterization tool, which has experienced increased interest with the advent of more powerful computers and more sophisticated machine learning algorithms. Nonetheless, the incorporation of radiomics in cancer clinical-decision support systems still necessitates a thorough analysis of its relationship with tumor biology. Herein, we present a systematic review focusing on the clinical evidence of radiomics as a surrogate method for tumor molecular profile characterization. An extensive literature review was conducted in PubMed, including papers on radiomics and a selected set of clinically relevant and commonly used tumor molecular markers. We summarized our findings based on different cancer entities, additionally evaluating the effect of different modalities for the prediction of biomarkers at each tumor site. Results suggest the existence of an association between the studied biomarkers and radiomics from different modalities and different tumor sites, even though a larger number of multi-center studies are required to further validate the reported outcomes.
Collapse
Affiliation(s)
- Agustina La Greca Saint-Esteven
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland; (D.V.); (J.E.v.T.); (R.D.B.); (M.G.); (S.T.-L.)
- Correspondence:
| | - Diem Vuong
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland; (D.V.); (J.E.v.T.); (R.D.B.); (M.G.); (S.T.-L.)
| | - Fabienne Tschanz
- Laboratory of Applied Radiobiology, Department of Radiation Oncology, University of Zurich, 8091 Zurich, Switzerland; (F.T.); (V.W.); (M.P.)
| | - Janita E. van Timmeren
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland; (D.V.); (J.E.v.T.); (R.D.B.); (M.G.); (S.T.-L.)
| | - Riccardo Dal Bello
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland; (D.V.); (J.E.v.T.); (R.D.B.); (M.G.); (S.T.-L.)
| | - Verena Waller
- Laboratory of Applied Radiobiology, Department of Radiation Oncology, University of Zurich, 8091 Zurich, Switzerland; (F.T.); (V.W.); (M.P.)
| | - Martin Pruschy
- Laboratory of Applied Radiobiology, Department of Radiation Oncology, University of Zurich, 8091 Zurich, Switzerland; (F.T.); (V.W.); (M.P.)
| | - Matthias Guckenberger
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland; (D.V.); (J.E.v.T.); (R.D.B.); (M.G.); (S.T.-L.)
| | - Stephanie Tanadini-Lang
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland; (D.V.); (J.E.v.T.); (R.D.B.); (M.G.); (S.T.-L.)
| |
Collapse
|
47
|
Hope A, Verduin M, Dilling TJ, Choudhury A, Fijten R, Wee L, Aerts HJWL, El Naqa I, Mitchell R, Vooijs M, Dekker A, de Ruysscher D, Traverso A. Artificial Intelligence Applications to Improve the Treatment of Locally Advanced Non-Small Cell Lung Cancers. Cancers (Basel) 2021; 13:2382. [PMID: 34069307 PMCID: PMC8156328 DOI: 10.3390/cancers13102382] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/21/2021] [Accepted: 05/03/2021] [Indexed: 11/16/2022] Open
Abstract
Locally advanced non-small cell lung cancer patients represent around one third of newly diagnosed lung cancer patients. There remains a large unmet need to find treatment strategies that can improve the survival of these patients while minimizing therapeutical side effects. Increasing the availability of patients' data (imaging, electronic health records, patients' reported outcomes, and genomics) will enable the application of AI algorithms to improve therapy selections. In this review, we discuss how artificial intelligence (AI) can be integral to improving clinical decision support systems. To realize this, a roadmap for AI must be defined. We define six milestones involving a broad spectrum of stakeholders, from physicians to patients, that we feel are necessary for an optimal transition of AI into the clinic.
Collapse
Affiliation(s)
- Andrew Hope
- Department of Radiation Oncology, University of Toronto, Toronto, ON 5MT 1P5, Canada;
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON 5MT 1P5, Canada
| | - Maikel Verduin
- Department of Radiation Oncology (Maastro) GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre+, 6229 ET Maastricht, The Netherlands; (M.V.); (A.C.); (R.F.); (L.W.); (M.V.); (A.D.); (D.d.R.)
| | - Thomas J Dilling
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA;
| | - Ananya Choudhury
- Department of Radiation Oncology (Maastro) GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre+, 6229 ET Maastricht, The Netherlands; (M.V.); (A.C.); (R.F.); (L.W.); (M.V.); (A.D.); (D.d.R.)
| | - Rianne Fijten
- Department of Radiation Oncology (Maastro) GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre+, 6229 ET Maastricht, The Netherlands; (M.V.); (A.C.); (R.F.); (L.W.); (M.V.); (A.D.); (D.d.R.)
| | - Leonard Wee
- Department of Radiation Oncology (Maastro) GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre+, 6229 ET Maastricht, The Netherlands; (M.V.); (A.C.); (R.F.); (L.W.); (M.V.); (A.D.); (D.d.R.)
| | - Hugo JWL Aerts
- Artificial Intelligence in Medicine (AIM) Program, Mass General Brigham, Harvard Medical School, Boston, MA 02115, USA;
- Department of Radiation Oncology, Brigham and Women’s Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
- Radiology and Nuclear Medicine, CARIM & GROW, Maastricht University, 6228 ET Maastricht, The Netherlands
| | - Issam El Naqa
- Department of Machine Learning, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; (I.E.N.); (R.M.)
| | - Ross Mitchell
- Department of Machine Learning, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; (I.E.N.); (R.M.)
| | - Marc Vooijs
- Department of Radiation Oncology (Maastro) GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre+, 6229 ET Maastricht, The Netherlands; (M.V.); (A.C.); (R.F.); (L.W.); (M.V.); (A.D.); (D.d.R.)
| | - Andre Dekker
- Department of Radiation Oncology (Maastro) GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre+, 6229 ET Maastricht, The Netherlands; (M.V.); (A.C.); (R.F.); (L.W.); (M.V.); (A.D.); (D.d.R.)
| | - Dirk de Ruysscher
- Department of Radiation Oncology (Maastro) GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre+, 6229 ET Maastricht, The Netherlands; (M.V.); (A.C.); (R.F.); (L.W.); (M.V.); (A.D.); (D.d.R.)
| | - Alberto Traverso
- Department of Radiation Oncology (Maastro) GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre+, 6229 ET Maastricht, The Netherlands; (M.V.); (A.C.); (R.F.); (L.W.); (M.V.); (A.D.); (D.d.R.)
| |
Collapse
|
48
|
Buchlak QD, Esmaili N, Leveque JC, Bennett C, Farrokhi F, Piccardi M. Machine learning applications to neuroimaging for glioma detection and classification: An artificial intelligence augmented systematic review. J Clin Neurosci 2021; 89:177-198. [PMID: 34119265 DOI: 10.1016/j.jocn.2021.04.043] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 04/30/2021] [Indexed: 12/13/2022]
Abstract
Glioma is the most common primary intraparenchymal tumor of the brain and the 5-year survival rate of high-grade glioma is poor. Magnetic resonance imaging (MRI) is essential for detecting, characterizing and monitoring brain tumors but definitive diagnosis still relies on surgical pathology. Machine learning has been applied to the analysis of MRI data in glioma research and has the potential to change clinical practice and improve patient outcomes. This systematic review synthesizes and analyzes the current state of machine learning applications to glioma MRI data and explores the use of machine learning for systematic review automation. Various datapoints were extracted from the 153 studies that met inclusion criteria and analyzed. Natural language processing (NLP) analysis involved keyword extraction, topic modeling and document classification. Machine learning has been applied to tumor grading and diagnosis, tumor segmentation, non-invasive genomic biomarker identification, detection of progression and patient survival prediction. Model performance was generally strong (AUC = 0.87 ± 0.09; sensitivity = 0.87 ± 0.10; specificity = 0.0.86 ± 0.10; precision = 0.88 ± 0.11). Convolutional neural network, support vector machine and random forest algorithms were top performers. Deep learning document classifiers yielded acceptable performance (mean 5-fold cross-validation AUC = 0.71). Machine learning tools and data resources were synthesized and summarized to facilitate future research. Machine learning has been widely applied to the processing of MRI data in glioma research and has demonstrated substantial utility. NLP and transfer learning resources enabled the successful development of a replicable method for automating the systematic review article screening process, which has potential for shortening the time from discovery to clinical application in medicine.
Collapse
Affiliation(s)
- Quinlan D Buchlak
- School of Medicine, The University of Notre Dame Australia, Sydney, NSW, Australia.
| | - Nazanin Esmaili
- School of Medicine, The University of Notre Dame Australia, Sydney, NSW, Australia; Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW, Australia
| | | | - Christine Bennett
- School of Medicine, The University of Notre Dame Australia, Sydney, NSW, Australia
| | - Farrokh Farrokhi
- Neuroscience Institute, Virginia Mason Medical Center, Seattle, WA, USA
| | - Massimo Piccardi
- Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW, Australia
| |
Collapse
|
49
|
Park YW, Park JE, Ahn SS, Kim EH, Kang SG, Chang JH, Kim SH, Choi SH, Kim HS, Lee SK. Magnetic Resonance Imaging Parameters for Noninvasive Prediction of Epidermal Growth Factor Receptor Amplification in Isocitrate Dehydrogenase-Wild-Type Lower-Grade Gliomas: A Multicenter Study. Neurosurgery 2021; 89:257-265. [PMID: 33913501 DOI: 10.1093/neuros/nyab136] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/21/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The epidermal growth factor receptor (EGFR) amplification status of isocitrate dehydrogenase-wild-type (IDHwt) lower-grade gliomas (LGGs; grade II/III) is one of the key markers for diagnosing molecular glioblastoma. However, the association between EGFR status and imaging parameters is unclear. OBJECTIVE To identify noninvasive imaging parameters from diffusion-weighted and dynamic susceptibility contrast imaging for predicting the EGFR amplification status of IDHwt LGGs. METHODS A total of 86 IDHwt LGG patients with known EGFR amplification status (62 nonamplified and 24 amplified) from 3 tertiary institutions were included. Qualitative and quantitative imaging features, including histogram parameters from apparent diffusion coefficient (ADC), normalized cerebral blood volume (nCBV), and normalized cerebral blood flow (nCBF), were assessed. Univariable and multivariable logistic regression models were constructed. RESULTS On multivariable analysis, multifocal/multicentric distribution (odds ratio [OR] = 11.77, P = .006), mean ADC (OR = 0.01, P = .044), 5th percentile of ADC (OR = 0.01, P = .046), and 95th percentile of nCBF (OR = 1.24, P = .031) were independent predictors of EGFR amplification. The diagnostic performance of the model with qualitative imaging parameters increased significantly when quantitative imaging parameters were added, with areas under the curves of 0.81 and 0.93, respectively (P = .004). CONCLUSION The presence of multifocal/multicentric distribution patterns, lower mean ADC, lower 5th percentile of ADC, and higher 95th percentile of nCBF may be useful imaging biomarkers for EGFR amplification in IDHwt LGGs. Moreover, quantitative imaging biomarkers may add value to qualitative imaging parameters.
Collapse
Affiliation(s)
- Yae Won Park
- Department of Radiology and Research Institute of Radiological Science and Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Ji Eun Park
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Seoul, Korea
| | - Sung Soo Ahn
- Department of Radiology and Research Institute of Radiological Science and Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Eui Hyun Kim
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Seok-Gu Kang
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Jong Hee Chang
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Se Hoon Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Seung Hong Choi
- Department of Radiology, Seoul National University Hospital, Seoul, South Korea
| | - Ho Sung Kim
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Seoul, Korea
| | - Seung-Koo Lee
- Department of Radiology and Research Institute of Radiological Science and Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
50
|
Tu SJ, Chen WY, Wu CT. Uncertainty measurement of radiomics features against inherent quantum noise in computed tomography imaging. Eur Radiol 2021; 31:7865-7875. [PMID: 33852047 DOI: 10.1007/s00330-021-07943-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/18/2021] [Accepted: 03/25/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Quantum noise is a random process in X-ray-based imaging systems. We addressed and measured the uncertainty of radiomics features against this quantum noise in computed tomography (CT) images. METHODS A clinical multi-detector CT scanner, two homogeneous phantom sets, and four heterogeneous samples were used. A solid tumor tissue removed from a male BALB/c mouse was included. We the placed phantom sets on the CT scanning table and repeated 20 acquisitions with identical imaging settings. Regions of interest were delineated for feature extraction. Statistical quantities-average, standard deviation, and percentage uncertainty-were calculated from these 20 repeated scans. Percentage uncertainty was used to measure and quantify feature stability against quantum noise. Twelve radiomics features were measured. Random noise was added to study the robustness of machine learning classifiers against feature uncertainty. RESULTS We found the ranges of percentage uncertainties from homogeneous soft tissue phantoms, homogeneous bone phantoms, and solid tumor tissue to be 0.01-2138%, 0.02-15%, and 0.18-16%, respectively. Overall, it was found that the CT features ShortRunHighGrayLevelEmpha (SRHGE) (0.01-0.18%), ShortRunLowGrayLevelEmpha (SRLGE) (0.01-0.41%), LowGrayLevelRunEmpha (LGRE) (0.01-0.39%), and LongRunLowGrayLevelEmpha (LRLGE) (0.02-0.66%) were the most stable features against the inherent quantum noise. The most unstable features were cluster shade (1-2138%) and max probability (1-16%). The impact of random noise to the prediction accuracy by different machine learning classifiers was found to be between 0 and 12%. CONCLUSIONS Twelve features were used for uncertainty measurements. The upper and lower bounds of percentage uncertainties were determined. The quantum noise effect on machine learning classifiers is model dependent. KEY POINTS • Quantum noise is a random process and is intrinsic to X-ray-based imaging systems. This inherent quantum noise creates unpredictable fluctuations in the gray-level intensities of image pixels. Extra cautions and further validations are strongly recommended when unstable radiomics features are selected by a predictive model for disease classification or treatment outcome prognosis. • We addressed and used the statistical quantity of percentage uncertainty to measure the uncertainty of radiomics features against the inherent quantum noise in computed tomography (CT) images. • A clinical multi-detector CT scanner, two homogeneous phantom sets, and four heterogeneous samples were used in the stability measurement. A solid tumor tissue removed from a male BALB/c mouse was included in the heterogeneous sample.
Collapse
Affiliation(s)
- Shu-Ju Tu
- Department of Medical Imaging and Radiological Sciences, College of Medicine, Chang Gung University, 259 Wen-Hua First Road, Kwei-Shan, Tao-Yuan, 333, Taiwan. .,Department of Medical Imaging and Intervention, Linkou Chang Gung Memorial Hospital, Tao-Yuan, Taiwan.
| | - Wei-Yuan Chen
- Department of Medical Imaging and Radiological Sciences, College of Medicine, Chang Gung University, 259 Wen-Hua First Road, Kwei-Shan, Tao-Yuan, 333, Taiwan.,Department of Medical Imaging and Intervention, Linkou Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
| | - Chen-Te Wu
- Department of Medical Imaging and Radiological Sciences, College of Medicine, Chang Gung University, 259 Wen-Hua First Road, Kwei-Shan, Tao-Yuan, 333, Taiwan.,Department of Medical Imaging and Intervention, Linkou Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
| |
Collapse
|