1
|
Fu X, Chen C, Chen Z, Yu J, Wang L. Radiogenomics based survival prediction of small-sample glioblastoma patients by multi-task DFFSP model. BIOMED ENG-BIOMED TE 2024:bmt-2022-0221. [PMID: 39241784 DOI: 10.1515/bmt-2022-0221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/21/2024] [Indexed: 09/09/2024]
Abstract
In this paper, the multi-task dense-feature-fusion survival prediction (DFFSP) model is proposed to predict the three-year survival for glioblastoma (GBM) patients based on radiogenomics data. The contrast-enhanced T1-weighted (T1w) image, T2-weighted (T2w) image and copy number variation (CNV) is used as the input of the three branches of the DFFSP model. This model uses two image extraction modules consisting of residual blocks and one dense feature fusion module to make multi-scale fusion of T1w and T2w image features as backbone. Also, a gene feature extraction module is used to adaptively weight CNV fragments. Besides, a transfer learning module is introduced to solve the small sample problem and an image reconstruction module is adopted to make the model anatomy-aware under a multi-task framework. 256 sample pairs (T1w and corresponding T2w MRI slices) and 187 CNVs of 74 patients were used. The experimental results show that the proposed model can predict the three-year survival of GBM patients with the accuracy of 89.1 %, which is improved by 3.2 and 4.7 % compared with the model without genes and the model using last fusion strategy, respectively. This model could also classify the patients into high-risk and low-risk groups, which will effectively assist doctors in diagnosing GBM patients.
Collapse
Affiliation(s)
- Xue Fu
- Department of Biomedical Engineering, 47854 Nanjing University of Aeronautics and Astronautics , Nanjing, China
| | - Chunxiao Chen
- Department of Biomedical Engineering, 47854 Nanjing University of Aeronautics and Astronautics , Nanjing, China
| | - Zhiying Chen
- Department of Biomedical Engineering, 47854 Nanjing University of Aeronautics and Astronautics , Nanjing, China
| | - Jie Yu
- Department of Biomedical Engineering, 47854 Nanjing University of Aeronautics and Astronautics , Nanjing, China
| | - Liang Wang
- Department of Biomedical Engineering, 47854 Nanjing University of Aeronautics and Astronautics , Nanjing, China
| |
Collapse
|
2
|
Demetriou D, Lockhat Z, Brzozowski L, Saini KS, Dlamini Z, Hull R. The Convergence of Radiology and Genomics: Advancing Breast Cancer Diagnosis with Radiogenomics. Cancers (Basel) 2024; 16:1076. [PMID: 38473432 DOI: 10.3390/cancers16051076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/09/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Despite significant progress in the prevention, screening, diagnosis, prognosis, and therapy of breast cancer (BC), it remains a highly prevalent and life-threatening disease affecting millions worldwide. Molecular subtyping of BC is crucial for predictive and prognostic purposes due to the diverse clinical behaviors observed across various types. The molecular heterogeneity of BC poses uncertainties in its impact on diagnosis, prognosis, and treatment. Numerous studies have highlighted genetic and environmental differences between patients from different geographic regions, emphasizing the need for localized research. International studies have revealed that patients with African heritage are often diagnosed at a more advanced stage and exhibit poorer responses to treatment and lower survival rates. Despite these global findings, there is a dearth of in-depth studies focusing on communities in the African region. Early diagnosis and timely treatment are paramount to improving survival rates. In this context, radiogenomics emerges as a promising field within precision medicine. By associating genetic patterns with image attributes or features, radiogenomics has the potential to significantly improve early detection, prognosis, and diagnosis. It can provide valuable insights into potential treatment options and predict the likelihood of survival, progression, and relapse. Radiogenomics allows for visual features and genetic marker linkage that promises to eliminate the need for biopsy and sequencing. The application of radiogenomics not only contributes to advancing precision oncology and individualized patient treatment but also streamlines clinical workflows. This review aims to delve into the theoretical underpinnings of radiogenomics and explore its practical applications in the diagnosis, management, and treatment of BC and to put radiogenomics on a path towards fully integrated diagnostics.
Collapse
Affiliation(s)
- Demetra Demetriou
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Zarina Lockhat
- Department of Radiology, Faculty of Health Sciences, Steve Biko Academic Hospital, University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Luke Brzozowski
- Translational Research and Core Facilities, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Kamal S Saini
- Fortrea Inc., 8 Moore Drive, Durham, NC 27709, USA
- Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Zodwa Dlamini
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Rodney Hull
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield, Pretoria 0028, South Africa
| |
Collapse
|
3
|
Ghimire P, Kinnersley B, Karami G, Arumugam P, Houlston R, Ashkan K, Modat M, Booth TC. Radiogenomic biomarkers for immunotherapy in glioblastoma: A systematic review of magnetic resonance imaging studies. Neurooncol Adv 2024; 6:vdae055. [PMID: 38680991 PMCID: PMC11046988 DOI: 10.1093/noajnl/vdae055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024] Open
Abstract
Background Immunotherapy is an effective "precision medicine" treatment for several cancers. Imaging signatures of the underlying genome (radiogenomics) in glioblastoma patients may serve as preoperative biomarkers of the tumor-host immune apparatus. Validated biomarkers would have the potential to stratify patients during immunotherapy clinical trials, and if trials are beneficial, facilitate personalized neo-adjuvant treatment. The increased use of whole genome sequencing data, and the advances in bioinformatics and machine learning make such developments plausible. We performed a systematic review to determine the extent of development and validation of immune-related radiogenomic biomarkers for glioblastoma. Methods A systematic review was performed following PRISMA guidelines using the PubMed, Medline, and Embase databases. Qualitative analysis was performed by incorporating the QUADAS 2 tool and CLAIM checklist. PROSPERO registered: CRD42022340968. Extracted data were insufficiently homogenous to perform a meta-analysis. Results Nine studies, all retrospective, were included. Biomarkers extracted from magnetic resonance imaging volumes of interest included apparent diffusion coefficient values, relative cerebral blood volume values, and image-derived features. These biomarkers correlated with genomic markers from tumor cells or immune cells or with patient survival. The majority of studies had a high risk of bias and applicability concerns regarding the index test performed. Conclusions Radiogenomic immune biomarkers have the potential to provide early treatment options to patients with glioblastoma. Targeted immunotherapy, stratified by these biomarkers, has the potential to allow individualized neo-adjuvant precision treatment options in clinical trials. However, there are no prospective studies validating these biomarkers, and interpretation is limited due to study bias with little evidence of generalizability.
Collapse
Affiliation(s)
- Prajwal Ghimire
- Department of Neurosurgery, Kings College Hospital NHS Foundation Trust, London, UK
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, UK
| | - Ben Kinnersley
- Department of Oncology, University College London, London, UK
| | | | | | - Richard Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, Sutton, UK
| | - Keyoumars Ashkan
- Department of Neurosurgery, Kings College Hospital NHS Foundation Trust, London, UK
| | - Marc Modat
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, UK
| | - Thomas C Booth
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, UK
| |
Collapse
|
4
|
Kang W, Qiu X, Luo Y, Luo J, Liu Y, Xi J, Li X, Yang Z. Application of radiomics-based multiomics combinations in the tumor microenvironment and cancer prognosis. J Transl Med 2023; 21:598. [PMID: 37674169 PMCID: PMC10481579 DOI: 10.1186/s12967-023-04437-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/12/2023] [Indexed: 09/08/2023] Open
Abstract
The advent of immunotherapy, a groundbreaking advancement in cancer treatment, has given rise to the prominence of the tumor microenvironment (TME) as a critical area of research. The clinical implications of an improved understanding of the TME are significant and far-reaching. Radiomics has been increasingly utilized in the comprehensive assessment of the TME and cancer prognosis. Similarly, the advancement of pathomics, which is based on pathological images, can offer additional insights into the panoramic view and microscopic information of tumors. The combination of pathomics and radiomics has revolutionized the concept of a "digital biopsy". As genomics and transcriptomics continue to evolve, integrating radiomics with genomic and transcriptomic datasets can offer further insights into tumor and microenvironment heterogeneity and establish correlations with biological significance. Therefore, the synergistic analysis of digital image features (radiomics, pathomics) and genetic phenotypes (genomics) can comprehensively decode and characterize the heterogeneity of the TME as well as predict cancer prognosis. This review presents a comprehensive summary of the research on important radiomics biomarkers for predicting the TME, emphasizing the interplay between radiomics, genomics, transcriptomics, and pathomics, as well as the application of multiomics in decoding the TME and predicting cancer prognosis. Finally, we discuss the challenges and opportunities in multiomics research. In conclusion, this review highlights the crucial role of radiomics and multiomics associations in the assessment of the TME and cancer prognosis. The combined analysis of radiomics, pathomics, genomics, and transcriptomics is a promising research direction with substantial research significance and value for comprehensive TME evaluation and cancer prognosis assessment.
Collapse
Affiliation(s)
- Wendi Kang
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli 17# Chaoyang District, Beijing, 100021, China
| | - Xiang Qiu
- Obstetrics and Gynecology Hospital of, Fudan University, Shanghai, 200011, China
| | - Yingen Luo
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli 17# Chaoyang District, Beijing, 100021, China
| | - Jianwei Luo
- Department of Diagnostic Radiology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 283 Tongzipo Road, Yuelu District, Changsha, 410013, Hunan, China
| | - Yang Liu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Junqing Xi
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli 17# Chaoyang District, Beijing, 100021, China
| | - Xiao Li
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli 17# Chaoyang District, Beijing, 100021, China
| | - Zhengqiang Yang
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli 17# Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
5
|
Liu Z, Duan T, Zhang Y, Weng S, Xu H, Ren Y, Zhang Z, Han X. Radiogenomics: a key component of precision cancer medicine. Br J Cancer 2023; 129:741-753. [PMID: 37414827 PMCID: PMC10449908 DOI: 10.1038/s41416-023-02317-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 05/02/2023] [Accepted: 06/12/2023] [Indexed: 07/08/2023] Open
Abstract
Radiogenomics, focusing on the relationship between genomics and imaging phenotypes, has been widely applied to address tumour heterogeneity and predict immune responsiveness and progression. It is an inevitable consequence of current trends in precision medicine, as radiogenomics costs less than traditional genetic sequencing and provides access to whole-tumour information rather than limited biopsy specimens. By providing voxel-by-voxel genetic information, radiogenomics can allow tailored therapy targeting a complete, heterogeneous tumour or set of tumours. In addition to quantifying lesion characteristics, radiogenomics can also be used to distinguish benign from malignant entities, as well as patient characteristics, to better stratify patients according to disease risk, thereby enabling more precise imaging and screening. Here, we have characterised the radiogenomic application in precision medicine using a multi-omic approach. we outline the main applications of radiogenomics in diagnosis, treatment planning and evaluations in the field of oncology with the aim of developing quantitative and personalised medicine. Finally, we discuss the challenges in the field of radiogenomics and the scope and clinical applicability of these methods.
Collapse
Affiliation(s)
- Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
- Interventional Institute of Zhengzhou University, 450052, Zhengzhou, Henan, China
- Interventional Treatment and Clinical Research Center of Henan Province, 450052, Zhengzhou, Henan, China
| | - Tian Duan
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Yuqing Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Zhenyu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China.
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China.
- Interventional Institute of Zhengzhou University, 450052, Zhengzhou, Henan, China.
- Interventional Treatment and Clinical Research Center of Henan Province, 450052, Zhengzhou, Henan, China.
| |
Collapse
|
6
|
Khalili N, Kazerooni AF, Familiar A, Haldar D, Kraya A, Foster J, Koptyra M, Storm PB, Resnick AC, Nabavizadeh A. Radiomics for characterization of the glioma immune microenvironment. NPJ Precis Oncol 2023; 7:59. [PMID: 37337080 DOI: 10.1038/s41698-023-00413-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 06/02/2023] [Indexed: 06/21/2023] Open
Abstract
Increasing evidence suggests that besides mutational and molecular alterations, the immune component of the tumor microenvironment also substantially impacts tumor behavior and complicates treatment response, particularly to immunotherapies. Although the standard method for characterizing tumor immune profile is through performing integrated genomic analysis on tissue biopsies, the dynamic change in the immune composition of the tumor microenvironment makes this approach not feasible, especially for brain tumors. Radiomics is a rapidly growing field that uses advanced imaging techniques and computational algorithms to extract numerous quantitative features from medical images. Recent advances in machine learning methods are facilitating biological validation of radiomic signatures and allowing them to "mine" for a variety of significant correlates, including genetic, immunologic, and histologic data. Radiomics has the potential to be used as a non-invasive approach to predict the presence and density of immune cells within the microenvironment, as well as to assess the expression of immune-related genes and pathways. This information can be essential for patient stratification, informing treatment decisions and predicting patients' response to immunotherapies. This is particularly important for tumors with difficult surgical access such as gliomas. In this review, we provide an overview of the glioma microenvironment, describe novel approaches for clustering patients based on their tumor immune profile, and discuss the latest progress on utilization of radiomics for immune profiling of glioma based on current literature.
Collapse
Affiliation(s)
- Nastaran Khalili
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Anahita Fathi Kazerooni
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, PA, USA
- AI2D Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ariana Familiar
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Debanjan Haldar
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Institute of Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Adam Kraya
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jessica Foster
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mateusz Koptyra
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Phillip B Storm
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Adam C Resnick
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ali Nabavizadeh
- Center for Data-Driven Discovery in Biomedicine (D3b), Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
7
|
Losurdo P, Gandin I, Belgrano M, Fiorese I, Verardo R, Zanconati F, Cova MA, de Manzini N. microRNAs combined to radiomic features as a predictor of complete clinical response after neoadjuvant radio-chemotherapy for locally advanced rectal cancer: a preliminary study. Surg Endosc 2023; 37:3676-3683. [PMID: 36639577 DOI: 10.1007/s00464-022-09851-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 12/27/2022] [Indexed: 01/15/2023]
Abstract
OBJECTIVE To define a predictive Artificial Intelligence (AI) algorithm based on the integration of a set of biopsy-based microRNAs expression data and radiomic features to understand their potential impact in predicting clinical response (CR) to neoadjuvant radio-chemotherapy (nRCT). The identification of patients who would truly benefit from nRCT for Locally Advanced Rectal Cancer (LARC) could be crucial for an improvement in a tailored therapy. METHODS Forty patients with LARC were retrospectively analyzed. An MRI of the pelvis before and after nRCT was performed. In the diagnostic biopsy, the expression levels of 7 miRNAs were measured and correlated with the tumor response rate (TRG), assessed on the surgical sample. The accuracy of complete CR (cCR) prediction was compared for i) clinical predictors; ii) radiomic features; iii) miRNAs levels; and iv) combination of radiomics and miRNAs. RESULTS Clinical predictors showed the lowest accuracy. The best performing model was based on the integration of radiomic features with miR-145 expression level (AUC-ROC = 0.90). AI algorithm, based on radiomics features and the overexpression of miR-145, showed an association with the TRG class and demonstrated a significant impact on the outcome. CONCLUSION The pre-treatment identification of responders/NON-responders to nRCT could address patients to a personalized strategy, such as total neoadjuvant therapy (TNT) for responders and upfront surgery for non-responders. The combination of radiomic features and miRNAs expression data from images and biopsy obtained through standard of care has the potential to accelerate the discovery of a noninvasive multimodal approach to predict the cCR after nRCT for LARC.
Collapse
Affiliation(s)
- Pasquale Losurdo
- Surgical Clinic Unit, Department of Medical and Surgical Sciences, Hospital of Cattinara, University of Trieste, Strada di Fiume 447, 34149, Trieste, Italy.
| | - Ilaria Gandin
- Biostatistics Unit, Department of Medical and Surgical Sciences, University of Trieste, Strada Di Fiume 447, 34149, Trieste, Italy
| | - Manuel Belgrano
- Radiology Unit, Department of Medical and Surgical Sciences, Hospital of Cattinara, University of Trieste, Strada di Fiume 447, 34149, Trieste, Italy
| | - Ilaria Fiorese
- Radiology Unit, Department of Medical and Surgical Sciences, Hospital of Cattinara, University of Trieste, Strada di Fiume 447, 34149, Trieste, Italy
| | - Roberto Verardo
- LNCIB - Consorzio Interuniversitario per le Biotecnologie c/o BIC Incubatori FVG, Srl - Via Flavia 23/1, 34149, Trieste, Italy
| | - Fabrizio Zanconati
- Pathology Unit, Department of Medical and Surgical Sciences, Hospital of Cattinara, University of Trieste, Strada di Fiume 447, 34149, Trieste, Italy
| | - Maria Assunta Cova
- Radiology Unit, Department of Medical and Surgical Sciences, Hospital of Cattinara, University of Trieste, Strada di Fiume 447, 34149, Trieste, Italy
| | - Nicolò de Manzini
- Surgical Clinic Unit, Department of Medical and Surgical Sciences, Hospital of Cattinara, University of Trieste, Strada di Fiume 447, 34149, Trieste, Italy
| |
Collapse
|
8
|
Radiogenomics: A Valuable Tool for the Clinical Assessment and Research of Ovarian Cancer. J Comput Assist Tomogr 2022; 46:371-378. [DOI: 10.1097/rct.0000000000001279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
9
|
The Role of Radiomics in the Era of Immune Checkpoint Inhibitors: A New Protagonist in the Jungle of Response Criteria. J Clin Med 2022; 11:jcm11061740. [PMID: 35330068 PMCID: PMC8948743 DOI: 10.3390/jcm11061740] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/08/2022] [Accepted: 03/18/2022] [Indexed: 12/15/2022] Open
Abstract
Simple Summary The introduction of immune checkpoint inhibitors has represented a milestone in cancer treatment. Despite PD-L1 expression being the standard biomarker used before the start of therapy, there is still a strict need to identify complementary non-invasive biomarkers in order to better select patients. In this context, radiomics is an emerging approach for examining medical images and clinical data by capturing multiple features hidden from human eye and is potentially able to predict response assessment and survival in the course of immunotherapy. We reviewed the available studies investigating the role of radiomics in cancer patients, focusing on non-small cell lung cancer treated with immune checkpoint inhibitors. Although preliminary research shows encouraging results, different issues need to be solved before radiomics can enter into clinical practice. Abstract Immune checkpoint inhibitors (ICI) have demonstrated encouraging results in terms of durable clinical benefit and survival in several malignancies. Nevertheless, the search to identify an “ideal” biomarker for predicting response to ICI is still far from over. Radiomics is a new translational field of study aiming to extract, by dedicated software, several features from a given medical image, ranging from intensity distribution and spatial heterogeneity to higher-order statistical parameters. Based on these premises, our review aims to summarize the current status of radiomics as a potential predictor of clinical response following immunotherapy treatment. A comprehensive search of PubMed results was conducted. All studies published in English up to and including December 2021 were selected, comprising those that explored computed tomography (CT), magnetic resonance imaging (MRI), and positron emission tomography (PET) for radiomic analyses in the setting of ICI. Several studies have demonstrated the potential applicability of radiomic features in the monitoring of the therapeutic response beyond the traditional morphologic and metabolic criteria, as well as in the prediction of survival or non-invasive assessment of the tumor microenvironment. Nevertheless, important limitations emerge from our review in terms of standardization in feature selection, data sharing, and methods, as well as in external validation. Additionally, there is still need for prospective clinical trials to confirm the potential significant role of radiomics during immunotherapy.
Collapse
|
10
|
Corr F, Grimm D, Saß B, Pojskić M, Bartsch JW, Carl B, Nimsky C, Bopp MHA. Radiogenomic Predictors of Recurrence in Glioblastoma—A Systematic Review. J Pers Med 2022; 12:jpm12030402. [PMID: 35330402 PMCID: PMC8952807 DOI: 10.3390/jpm12030402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 02/23/2022] [Accepted: 03/01/2022] [Indexed: 12/10/2022] Open
Abstract
Glioblastoma, as the most aggressive brain tumor, is associated with a poor prognosis and outcome. To optimize prognosis and clinical therapy decisions, there is an urgent need to stratify patients with increased risk for recurrent tumors and low therapeutic success to optimize individual treatment. Radiogenomics establishes a link between radiological and pathological information. This review provides a state-of-the-art picture illustrating the latest developments in the use of radiogenomic markers regarding prognosis and their potential for monitoring recurrence. Databases PubMed, Google Scholar, and Cochrane Library were searched. Inclusion criteria were defined as diagnosis of glioblastoma with histopathological and radiological follow-up. Out of 321 reviewed articles, 43 articles met these inclusion criteria. Included studies were analyzed for the frequency of radiological and molecular tumor markers whereby radiogenomic associations were analyzed. Six main associations were described: radiogenomic prognosis, MGMT status, IDH, EGFR status, molecular subgroups, and tumor location. Prospective studies analyzing prognostic features of glioblastoma together with radiological features are lacking. By reviewing the progress in the development of radiogenomic markers, we provide insights into the potential efficacy of such an approach for clinical routine use eventually enabling early identification of glioblastoma recurrence and therefore supporting a further personalized monitoring and treatment strategy.
Collapse
Affiliation(s)
- Felix Corr
- Department of Neurosurgery, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany; (D.G.); (B.S.); (M.P.); (J.W.B.); (B.C.); (C.N.); (M.H.A.B.)
- EDU Institute of Higher Education, Villa Bighi, Chaplain’s House, KKR 1320 Kalkara, Malta
- Correspondence:
| | - Dustin Grimm
- Department of Neurosurgery, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany; (D.G.); (B.S.); (M.P.); (J.W.B.); (B.C.); (C.N.); (M.H.A.B.)
- EDU Institute of Higher Education, Villa Bighi, Chaplain’s House, KKR 1320 Kalkara, Malta
| | - Benjamin Saß
- Department of Neurosurgery, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany; (D.G.); (B.S.); (M.P.); (J.W.B.); (B.C.); (C.N.); (M.H.A.B.)
| | - Mirza Pojskić
- Department of Neurosurgery, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany; (D.G.); (B.S.); (M.P.); (J.W.B.); (B.C.); (C.N.); (M.H.A.B.)
| | - Jörg W. Bartsch
- Department of Neurosurgery, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany; (D.G.); (B.S.); (M.P.); (J.W.B.); (B.C.); (C.N.); (M.H.A.B.)
- Center for Mind, Brain and Behavior (CMBB), 35043 Marburg, Germany
| | - Barbara Carl
- Department of Neurosurgery, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany; (D.G.); (B.S.); (M.P.); (J.W.B.); (B.C.); (C.N.); (M.H.A.B.)
- Department of Neurosurgery, Helios Dr. Horst Schmidt Kliniken, Ludwig-Erhard-Strasse 100, 65199 Wiesbaden, Germany
| | - Christopher Nimsky
- Department of Neurosurgery, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany; (D.G.); (B.S.); (M.P.); (J.W.B.); (B.C.); (C.N.); (M.H.A.B.)
- Center for Mind, Brain and Behavior (CMBB), 35043 Marburg, Germany
| | - Miriam H. A. Bopp
- Department of Neurosurgery, University of Marburg, Baldingerstrasse, 35043 Marburg, Germany; (D.G.); (B.S.); (M.P.); (J.W.B.); (B.C.); (C.N.); (M.H.A.B.)
- Center for Mind, Brain and Behavior (CMBB), 35043 Marburg, Germany
| |
Collapse
|
11
|
Hoshino I, Yokota H, Iwatate Y, Mori Y, Kuwayama N, Ishige F, Itami M, Uno T, Nakamura Y, Tatsumi Y, Shimozato O, Nagase H. Prediction of the differences in tumor mutation burden between primary and metastatic lesions by radiogenomics. Cancer Sci 2021; 113:229-239. [PMID: 34689378 PMCID: PMC8748253 DOI: 10.1111/cas.15173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/15/2021] [Accepted: 10/18/2021] [Indexed: 12/13/2022] Open
Abstract
Tumor mutational burden (TMB) is gaining attention as a biomarker for responses to immune checkpoint inhibitors in cancer patients. In this study, we evaluated the status of TMB in primary and liver metastatic lesions in patients with colorectal cancer (CRC). In addition, the status of TMB in primary and liver metastatic lesions was inferred by radiogenomics on the basis of computed tomography (CT) images. The study population included 24 CRC patients with liver metastases. DNA was extracted from primary and liver metastatic lesions obtained from the patients and TMB values were evaluated by next‐generation sequencing. The TMB value was considered high when it equaled to or exceeded 10/100 Mb. Radiogenomic analysis of TMB was performed by machine learning using CT images and the construction of prediction models. In 7 out of 24 patients (29.2%), the TMB status differed between the primary and liver metastatic lesions. Radiogenomic analysis was performed to predict whether TMB status was high or low. The maximum values for the area under the receiver operating characteristic curve were 0.732 and 0.812 for primary CRC and CRC with liver metastasis, respectively. The sensitivity, specificity, and accuracy of the constructed models for TMB status discordance were 0.857, 0.600, and 0.682, respectively. Our results suggested that accurate inference of the TMB status is possible using radiogenomics. Therefore, radiogenomics could facilitate the diagnosis, treatment, and prognosis of patients with CRC in the clinical setting.
Collapse
Affiliation(s)
- Isamu Hoshino
- Division of Gastroenterological Surgery, Chiba Cancer Center, Chiba, Japan
| | - Hajime Yokota
- Department of Diagnostic Radiology and Radiation Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yosuke Iwatate
- Division of Hepato-Biliary-Pancreatic Surgery, Chiba Cancer Center, Chiba, Japan
| | - Yasukuni Mori
- Faculty of Engineering, Graduate School of Engineering, Chiba University, Chiba, Japan
| | - Naoki Kuwayama
- Division of Gastroenterological Surgery, Chiba Cancer Center, Chiba, Japan
| | - Fumitaka Ishige
- Division of Hepato-Biliary-Pancreatic Surgery, Chiba Cancer Center, Chiba, Japan
| | - Makiko Itami
- Division of Clinical Pathology, Chiba Cancer Center, Chiba, Japan
| | - Takashi Uno
- Department of Diagnostic Radiology and Radiation Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yuki Nakamura
- Laboratory of Cancer Genetics, Chiba Cancer Center Research Institute, Chiba Cancer Center, Chiba, Japan
| | - Yasutoshi Tatsumi
- Laboratory of Cancer Genetics, Chiba Cancer Center Research Institute, Chiba Cancer Center, Chiba, Japan
| | - Osamu Shimozato
- Laboratory of Cancer Genetics, Chiba Cancer Center Research Institute, Chiba Cancer Center, Chiba, Japan
| | - Hiroki Nagase
- Laboratory of Cancer Genetics, Chiba Cancer Center Research Institute, Chiba Cancer Center, Chiba, Japan
| |
Collapse
|
12
|
Hill L, Bruns J, Zustiak SP. Hydrogel matrix presence and composition influence drug responses of encapsulated glioblastoma spheroids. Acta Biomater 2021; 132:437-447. [PMID: 34010694 DOI: 10.1016/j.actbio.2021.05.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 04/30/2021] [Accepted: 05/06/2021] [Indexed: 12/26/2022]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive brain tumor with median patient survival of 12-15 months. To facilitate treatment development, bioengineered GBM models that adequately recapitulate the in vivo tumor microenvironment are needed. Matrix-encapsulated multicellular spheroids represent such model because they recapitulate solid tumor characteristics, such as dimensionality, cell-cell, and cell-matrix interactions. Yet, there is no consensus as to which matrix properties are key to improving the predictive capacity of spheroid-based drug screening platforms. We used a hydrogel-encapsulated GBM spheroid model, where matrix properties were independently altered to investigate their effect on GBM spheroid characteristics and drug responsiveness. We focused on hydrogel degradability, tuned via enzymatically degradable crosslinkers, and hydrogel adhesiveness, tuned via integrin ligands. We observed increased cellular infiltration of GBM spheroids and increased resistance to temozolomide in degradable, adhesive hydrogels compared to spheroids in non-degradable, non-adhesive hydrogels or to free-floating spheroids. Further, a higher infiltration index was noted for spheroids in adhesive compared to non-adhesive degradable hydrogels. For spheroids in degradable hydrogels, we determined that infiltrating cells were more susceptible to temozolomide compared to cells in the spheroid core. The temozolomide susceptibility of the infiltrating cells was independent of integrin adhesion. We could not attribute differential drug responses to differential cellular proliferation or to limited drug penetration into the hydrogel matrix. Our results suggest that cell-matrix interactions guide GBM spheroid drug responsiveness and that further elucidation of these interactions could enable the engineering of more predictive drug screening platforms. STATEMENT OF SIGNIFICANCE: Glioblastoma multiforme (GBM) multicellular spheroids hold promise for drug screening and development as they better mimic in vivo cellular responses to therapeutics compared to monolayer cultures. Traditional spheroid models lack an external extracellular matrix (ECM) and fail to mimic the mechanical, physical, and biochemical cues seen in the GBM microenvironment. While embedding spheroids in hydrogel matrices has been shown to better recapitulate the tumor microenvironment, there is still limited understanding as to the key matrix properties that govern spheroid responsiveness to drugs. Here we decoupled and independently altered matrix properties such as degradability, via an enzymatically degradable peptide crosslinker, and cell adhesion, via an adhesive ligand, giving further insight into what matrix properties contribute to GBM chemoresistance.
Collapse
|
13
|
Sun Q, Chen Y, Liang C, Zhao Y, Lv X, Zou Y, Yan K, Zheng H, Liang D, Li ZC. Biologic Pathways Underlying Prognostic Radiomics Phenotypes from Paired MRI and RNA Sequencing in Glioblastoma. Radiology 2021; 301:654-663. [PMID: 34519578 DOI: 10.1148/radiol.2021203281] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Background The biologic meaning of prognostic radiomics phenotypes remains poorly understood, hampered in part by lack of multicenter reproducible evidence. Purpose To uncover the biologic meaning of individual prognostic radiomics phenotypes in glioblastomas using paired MRI and RNA sequencing data and to validate the reproducibility of the identified radiogenomics linkages externally. Materials and Methods This retrospective multicenter study included four data sets gathered between January 2015 and December 2016. From a radiomics analysis set, a 13-feature radiomics signature was built using preoperative MRI for overall survival prediction. Using a radiogenomics training set with both MRI and RNA sequencing, biologic pathways were enriched and correlated with each of the 13 radiomics phenotypes. Radiomics-correlated key genes were identified to derive a prognostic radiomics gene expression (RadGene) score. The reproducibility of identified pathways and genes was validated with an external test set and a public data set (The Cancer Genome Atlas [TCGA]). A log-rank test was performed to assess prognostic significance. Results A total of 435 patients (mean age, 55 years ± 15 [standard deviation]; 263 men) were enrolled. The radiomics signature was associated with overall survival (hazard ratio [HR], 3.68; 95% CI: 2.08, 6.52; P < .001) in the radiomics validation subset. Four types of prognostic radiomics phenotypes were correlated with distinct pathways: immune, proliferative, treatment responsive, and cellular functions (false-discovery rate < 0.10). Thirty radiomics-correlated genes were identified. The prognostic significance of the RadGene score was confirmed in an external test set (HR, 2.02; 95% CI: 1.19, 3.41; P = .01) and a TCGA test set (HR, 1.43; 95% CI: 1.001, 2.04; P = .048). The radiomics-associated pathways and key genes can be replicated in an external test set. Conclusion Individual radiomics phenotypes on MRI scans predictive of overall survival were driven by distinct key pathways involved in immune regulation, tumor proliferation, treatment responses, and cellular functions in glioblastoma, which could be reproduced externally. © RSNA, 2021 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Qiuchang Sun
- From the Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, No. 1068, Xueyuan Ave, Shenzhen 518055, China (Q.S., Y. Zhao, K.Y., H.Z., D.L., Z.C.L.); University of Chinese Academy of Sciences, Beijing, China (Q.S., Z.C.L.); Departments of Neurosurgery/Neuro-oncology (Y.C.) and Medical Imaging (X.L.), Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China; Departments of Neurosurgery (C.L.) and Radiology (Y. Zou), Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Shenzhen Peng Cheng Laboratory, Shenzhen, China (K.Y.); and National Innovation Center for Advanced Medical Devices, Shenzhen, China (H.Z., D.L., Z.C.L.)
| | - Yinsheng Chen
- From the Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, No. 1068, Xueyuan Ave, Shenzhen 518055, China (Q.S., Y. Zhao, K.Y., H.Z., D.L., Z.C.L.); University of Chinese Academy of Sciences, Beijing, China (Q.S., Z.C.L.); Departments of Neurosurgery/Neuro-oncology (Y.C.) and Medical Imaging (X.L.), Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China; Departments of Neurosurgery (C.L.) and Radiology (Y. Zou), Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Shenzhen Peng Cheng Laboratory, Shenzhen, China (K.Y.); and National Innovation Center for Advanced Medical Devices, Shenzhen, China (H.Z., D.L., Z.C.L.)
| | - Chaofeng Liang
- From the Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, No. 1068, Xueyuan Ave, Shenzhen 518055, China (Q.S., Y. Zhao, K.Y., H.Z., D.L., Z.C.L.); University of Chinese Academy of Sciences, Beijing, China (Q.S., Z.C.L.); Departments of Neurosurgery/Neuro-oncology (Y.C.) and Medical Imaging (X.L.), Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China; Departments of Neurosurgery (C.L.) and Radiology (Y. Zou), Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Shenzhen Peng Cheng Laboratory, Shenzhen, China (K.Y.); and National Innovation Center for Advanced Medical Devices, Shenzhen, China (H.Z., D.L., Z.C.L.)
| | - Yuanshen Zhao
- From the Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, No. 1068, Xueyuan Ave, Shenzhen 518055, China (Q.S., Y. Zhao, K.Y., H.Z., D.L., Z.C.L.); University of Chinese Academy of Sciences, Beijing, China (Q.S., Z.C.L.); Departments of Neurosurgery/Neuro-oncology (Y.C.) and Medical Imaging (X.L.), Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China; Departments of Neurosurgery (C.L.) and Radiology (Y. Zou), Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Shenzhen Peng Cheng Laboratory, Shenzhen, China (K.Y.); and National Innovation Center for Advanced Medical Devices, Shenzhen, China (H.Z., D.L., Z.C.L.)
| | - Xiaofei Lv
- From the Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, No. 1068, Xueyuan Ave, Shenzhen 518055, China (Q.S., Y. Zhao, K.Y., H.Z., D.L., Z.C.L.); University of Chinese Academy of Sciences, Beijing, China (Q.S., Z.C.L.); Departments of Neurosurgery/Neuro-oncology (Y.C.) and Medical Imaging (X.L.), Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China; Departments of Neurosurgery (C.L.) and Radiology (Y. Zou), Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Shenzhen Peng Cheng Laboratory, Shenzhen, China (K.Y.); and National Innovation Center for Advanced Medical Devices, Shenzhen, China (H.Z., D.L., Z.C.L.)
| | - Yan Zou
- From the Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, No. 1068, Xueyuan Ave, Shenzhen 518055, China (Q.S., Y. Zhao, K.Y., H.Z., D.L., Z.C.L.); University of Chinese Academy of Sciences, Beijing, China (Q.S., Z.C.L.); Departments of Neurosurgery/Neuro-oncology (Y.C.) and Medical Imaging (X.L.), Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China; Departments of Neurosurgery (C.L.) and Radiology (Y. Zou), Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Shenzhen Peng Cheng Laboratory, Shenzhen, China (K.Y.); and National Innovation Center for Advanced Medical Devices, Shenzhen, China (H.Z., D.L., Z.C.L.)
| | - Kai Yan
- From the Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, No. 1068, Xueyuan Ave, Shenzhen 518055, China (Q.S., Y. Zhao, K.Y., H.Z., D.L., Z.C.L.); University of Chinese Academy of Sciences, Beijing, China (Q.S., Z.C.L.); Departments of Neurosurgery/Neuro-oncology (Y.C.) and Medical Imaging (X.L.), Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China; Departments of Neurosurgery (C.L.) and Radiology (Y. Zou), Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Shenzhen Peng Cheng Laboratory, Shenzhen, China (K.Y.); and National Innovation Center for Advanced Medical Devices, Shenzhen, China (H.Z., D.L., Z.C.L.)
| | - Hairong Zheng
- From the Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, No. 1068, Xueyuan Ave, Shenzhen 518055, China (Q.S., Y. Zhao, K.Y., H.Z., D.L., Z.C.L.); University of Chinese Academy of Sciences, Beijing, China (Q.S., Z.C.L.); Departments of Neurosurgery/Neuro-oncology (Y.C.) and Medical Imaging (X.L.), Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China; Departments of Neurosurgery (C.L.) and Radiology (Y. Zou), Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Shenzhen Peng Cheng Laboratory, Shenzhen, China (K.Y.); and National Innovation Center for Advanced Medical Devices, Shenzhen, China (H.Z., D.L., Z.C.L.)
| | - Dong Liang
- From the Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, No. 1068, Xueyuan Ave, Shenzhen 518055, China (Q.S., Y. Zhao, K.Y., H.Z., D.L., Z.C.L.); University of Chinese Academy of Sciences, Beijing, China (Q.S., Z.C.L.); Departments of Neurosurgery/Neuro-oncology (Y.C.) and Medical Imaging (X.L.), Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China; Departments of Neurosurgery (C.L.) and Radiology (Y. Zou), Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Shenzhen Peng Cheng Laboratory, Shenzhen, China (K.Y.); and National Innovation Center for Advanced Medical Devices, Shenzhen, China (H.Z., D.L., Z.C.L.)
| | - Zhi-Cheng Li
- From the Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, No. 1068, Xueyuan Ave, Shenzhen 518055, China (Q.S., Y. Zhao, K.Y., H.Z., D.L., Z.C.L.); University of Chinese Academy of Sciences, Beijing, China (Q.S., Z.C.L.); Departments of Neurosurgery/Neuro-oncology (Y.C.) and Medical Imaging (X.L.), Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China; Departments of Neurosurgery (C.L.) and Radiology (Y. Zou), Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Shenzhen Peng Cheng Laboratory, Shenzhen, China (K.Y.); and National Innovation Center for Advanced Medical Devices, Shenzhen, China (H.Z., D.L., Z.C.L.)
| |
Collapse
|
14
|
Yin J, Lin C, Jiang M, Tang X, Xie D, Chen J, Ke R. CENPL, ISG20L2, LSM4, MRPL3 are four novel hub genes and may serve as diagnostic and prognostic markers in breast cancer. Sci Rep 2021; 11:15610. [PMID: 34341433 PMCID: PMC8328991 DOI: 10.1038/s41598-021-95068-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 07/14/2021] [Indexed: 12/18/2022] Open
Abstract
As a highly prevalent disease among women worldwide, breast cancer remains in urgent need of further elucidation its molecular mechanisms to improve the patient outcomes. Identifying hub genes involved in the pathogenesis and progression of breast cancer can potentially help to unveil mechanism and also provide novel diagnostic and prognostic markers. In this study, we integrated multiple bioinformatic methods and RNA in situ detection technology to identify and validate hub genes. EZH2 was recognized as a key gene by PPI network analysis. CENPL, ISG20L2, LSM4, MRPL3 were identified as four novel hub genes through the WGCNA analysis and literate search. Among these, many studies on EZH2 in breast cancer have been reported, but no studies are related to the roles of CENPL, ISG20L2, MRPL3 and LSM4 in breast cancer. These four novel hub genes were up-regulated in tumor tissues and associated with cancer progression. The receiver operating characteristic analysis and Kaplan-Meier survival analysis indicated that these four hub genes are promising candidate genes that can serve as diagnostic and prognostic biomarkers for breast cancer. Moreover, these four newly identified hub genes as aberrant molecules in the maintenance of breast cancer development, their exact functional mechanisms deserve further in-depth study.
Collapse
Affiliation(s)
- Jinbao Yin
- School of Medicine, Huaqiao University, Quanzhou, 362021, Fujian, China
- Department of Pathology, Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Chen Lin
- School of Medicine, Huaqiao University, Quanzhou, 362021, Fujian, China
| | - Meng Jiang
- School of Medicine, Huaqiao University, Quanzhou, 362021, Fujian, China
| | - Xinbin Tang
- School of Medicine, Huaqiao University, Quanzhou, 362021, Fujian, China
| | - Danlin Xie
- School of Medicine, Huaqiao University, Quanzhou, 362021, Fujian, China
| | - Jingwen Chen
- School of Medicine, Huaqiao University, Quanzhou, 362021, Fujian, China
| | - Rongqin Ke
- School of Medicine, Huaqiao University, Quanzhou, 362021, Fujian, China.
| |
Collapse
|
15
|
Kinoshita M. [1. Current and Future Prospective of Radiomics in Glioma Imaging]. Nihon Hoshasen Gijutsu Gakkai Zasshi 2021; 77:761-764. [PMID: 34305066 DOI: 10.6009/jjrt.2021_jsrt_77.7.761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
16
|
Glioblastomas within the Subventricular Zone Are Region-Specific Enriched for Mesenchymal Transition Markers: An Intratumoral Gene Expression Analysis. Cancers (Basel) 2021; 13:cancers13153764. [PMID: 34359668 PMCID: PMC8345101 DOI: 10.3390/cancers13153764] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/15/2021] [Accepted: 07/22/2021] [Indexed: 01/31/2023] Open
Abstract
Simple Summary Involvement of the subventricular zone (SVZ) in glioblastoma is associated with poor prognosis and is associated with specific tumor-biological characteristics. In this study, we demonstrate that patient-derived glioblastoma samples from within the SVZ region show increased (epithelial-)mesenchymal transition and angiogenesis/hypoxia signaling as compared to glioblastoma samples from the same patient from outside the SVZ. These results suggest that intratumoral alterations in oncogenic signaling could be mediated by the SVZ microenvironment. Our findings offer rationale for specific targeting of the SVZ in the development of glioblastoma therapy. Abstract Background: Involvement of the subventricular zone (SVZ) in glioblastoma is associated with poor prognosis and is associated with specific tumor-biological characteristics. The SVZ microenvironment can influence gene expression in glioblastoma cells in preclinical models. We aimed to investigate whether the SVZ microenvironment has any influence on intratumoral gene expression patterns in glioblastoma patients. Methods: The publicly available Ivy Glioblastoma database contains clinical, radiological and whole exome sequencing data from multiple regions from resected glioblastomas. SVZ involvement of the various tissue samples was evaluated on MRI scans. In tumors that contacted the SVZ, we performed gene expression analyses and gene set enrichment analyses to compare gene (set) expression in tumor regions within the SVZ to tumor regions outside the SVZ. We also compared these samples to glioblastomas that did not contact the SVZ. Results: Within glioblastomas that contacted the SVZ, tissue samples within the SVZ showed enrichment of gene sets involved in (epithelial-)mesenchymal transition, NF-κB and STAT3 signaling, angiogenesis and hypoxia, compared to the samples outside of the SVZ region from the same tumors (p < 0.05, FDR < 0.25). Comparison of glioblastoma samples within the SVZ region to samples from tumors that did not contact the SVZ yielded similar results. In contrast, we observed no differences when comparing the samples outside of the SVZ from SVZ-contacting glioblastomas with samples from glioblastomas that did not contact the SVZ at all. Conclusion: Glioblastoma samples in the SVZ region are enriched for increased (epithelial-)mesenchymal transition and angiogenesis/hypoxia signaling, possibly mediated by the SVZ microenvironment.
Collapse
|
17
|
Mathios D, Srivastava S, Kim T, Bettegowda C, Lim M. Emerging Technologies for Non-invasive Monitoring of Treatment Response to Immunotherapy for Brain Tumors. Neuromolecular Med 2021; 24:74-87. [PMID: 34297308 DOI: 10.1007/s12017-021-08677-9] [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: 03/09/2021] [Accepted: 07/10/2021] [Indexed: 12/19/2022]
Abstract
Glioblastoma is the most common primary malignant brain tumor and one of the most aggressive tumors across all cancer types with remarkable resistance to any treatment. While immunotherapy has shown a robust clinical benefit in systemic cancers, its benefit is still under investigation in brain cancers. The broader use of immunotherapy in clinical trials for glioblastoma has highlighted the challenges of traditional methods of monitoring progression via imaging. Development of new guidelines, advanced imaging techniques, and immune profiling have emerged to counter premature diagnoses of progressive disease. However, these approaches do not provide a timely diagnosis and are costly and time consuming. Surgery is currently the standard of care for diagnosis of pseudoprogression in cases where MRI is equivocal. However, it is invasive, risky, and disruptive to patient's lives and their oncological treatment. With its increased vascularity, glioblastoma is continually shedding tumor components into the vasculature including tumor cells, genetic material, and extracellular vesicles. These elements can be isolated from routine blood draws and provide a real-time non-invasive indicator of tumor progression. Liquid biopsy therefore presents as an attractive alternative to current methods to guide treatment. While the initial evaluation of liquid biopsy for brain tumors via identification of mutations in the plasma was disappointing, novel technologies and use of alternatives to plasma cell-free DNA analytes provide promise for an effective liquid biopsy approach in brain tumors. This review aims to summarize developments in the use of liquid biopsy to monitor glioblastoma, especially in the context of immunotherapy.
Collapse
Affiliation(s)
- Dimitrios Mathios
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Siddhartha Srivastava
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Timothy Kim
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chetan Bettegowda
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael Lim
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA, USA.
| |
Collapse
|
18
|
Cipollari S, Jamshidi N, Du L, Sung K, Huang D, Margolis DJ, Huang J, Reiter RE, Kuo MD. Tissue clearing techniques for three-dimensional optical imaging of intact human prostate and correlations with multi-parametric MRI. Prostate 2021; 81:521-529. [PMID: 33876838 PMCID: PMC9014804 DOI: 10.1002/pros.24129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 03/26/2021] [Indexed: 11/07/2022]
Abstract
BACKGROUND Tissue clearing technologies have enabled remarkable advancements for in situ characterization of tissues and exploration of the three-dimensional (3D) relationships between cells, however, these studies have predominantly been performed in non-human tissues and correlative assessment with clinical imaging has yet to be explored. We sought to evaluate the feasibility of tissue clearing technologies for 3D imaging of intact human prostate and the mapping of structurally and molecularly preserved pathology data with multi-parametric volumetric MR imaging (mpMRI). METHODS Whole-mount prostates were processed with either hydrogel-based CLARITY or solvent-based iDISCO. The samples were stained with a nuclear dye or fluorescently labeled with antibodies against androgen receptor, alpha-methylacyl coenzyme-A racemase, or p63, and then imaged with 3D confocal microscopy. The apparent diffusion coefficient and Ktrans maps were computed from preoperative mpMRI. RESULTS Quantitative analysis of cleared normal and tumor prostate tissue volumes displayed differences in 3D tissue architecture, marker-specific cell staining, and cell densities that were significantly correlated with mpMRI measurements in this initial, pilot cohort. CONCLUSIONS 3D imaging of human prostate volumes following tissue clearing is a feasible technique for quantitative radiology-pathology correlation analysis with mpMRI and provides an opportunity to explore functional relationships between cellular structures and cross-sectional clinical imaging.
Collapse
Affiliation(s)
- Stefano Cipollari
- Medical Artificial Intelligence Laboratory Program, The University of Hong Kong, Hong Kong SAR
- Department of Radiology, La Sapienza, The University of Rome, Italy
| | - Neema Jamshidi
- Medical Artificial Intelligence Laboratory Program, The University of Hong Kong, Hong Kong SAR
- Department of Radiological Sciences, University of California, Los Angeles, David Geffen School of Medicine, California
| | - Liutao Du
- Department of Radiological Sciences, University of California, Los Angeles, David Geffen School of Medicine, California
| | - Kyunghyun Sung
- Department of Radiological Sciences, University of California, Los Angeles, David Geffen School of Medicine, California
| | - Danshan Huang
- Department of Radiological Sciences, University of California, Los Angeles, David Geffen School of Medicine, California
| | | | - Jiaoti Huang
- Department of Pathology, Duke University School of Medicine, North Carolina
| | - Robert E. Reiter
- Department of Urology, University of California, Los Angeles, David Geffen School of Medicine, California
| | - Michael D. Kuo
- Medical Artificial Intelligence Laboratory Program, The University of Hong Kong, Hong Kong SAR
- Correspondence should be addressed to MDK ()
| |
Collapse
|
19
|
Wang JH, Wahid KA, van Dijk LV, Farahani K, Thompson RF, Fuller CD. Radiomic biomarkers of tumor immune biology and immunotherapy response. Clin Transl Radiat Oncol 2021; 28:97-115. [PMID: 33937530 PMCID: PMC8076712 DOI: 10.1016/j.ctro.2021.03.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 03/20/2021] [Accepted: 03/24/2021] [Indexed: 02/08/2023] Open
Abstract
Immunotherapies are leading to improved outcomes for many cancers, including those with devastating prognoses. As therapies like immune checkpoint inhibitors (ICI) become a mainstay in treatment regimens, many concurrent challenges have arisen - for instance, delineating clinical responders from non-responders. Predicting response has proven to be difficult given a lack of consistent and accurate biomarkers, heterogeneity of the tumor microenvironment (TME), and a poor understanding of resistance mechanisms. For the most part, imaging data have remained an untapped, yet abundant, resource to address these challenges. In recent years, quantitative image analyses have highlighted the utility of medical imaging in predicting tumor phenotypes, prognosis, and therapeutic response. These studies have been fueled by an explosion of resources in high-throughput mining of image features (i.e. radiomics) and artificial intelligence. In this review, we highlight current progress in radiomics to understand tumor immune biology and predict clinical responses to immunotherapies. We also discuss limitations in these studies and future directions for the field, particularly if high-dimensional imaging data are to play a larger role in precision medicine.
Collapse
Affiliation(s)
- Jarey H. Wang
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, United States
| | - Kareem A. Wahid
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Lisanne V. van Dijk
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Keyvan Farahani
- Center for Biomedical Informatics and Information Technology, National Cancer Institute, Bethesda, MD, United States
| | - Reid F. Thompson
- Department of Radiation Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Clifton David Fuller
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
20
|
Taha B, Boley D, Sun J, Chen CC. State of Radiomics in Glioblastoma. Neurosurgery 2021; 89:177-184. [PMID: 33913492 DOI: 10.1093/neuros/nyab124] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 02/13/2021] [Indexed: 12/30/2022] Open
Abstract
Radiomics is an emerging discipline that aims to make intelligent predictions and derive medical insights based on quantitative features extracted from medical images as a means to improve clinical diagnosis or outcome. Pertaining to glioblastoma, radiomics has provided powerful, noninvasive tools for gaining insights into pathogenesis and therapeutic responses. Radiomic studies have yielded meaningful biological understandings of imaging features that are often taken for granted in clinical medicine, including contrast enhancement on glioblastoma magnetic resonance imaging, the distance of a tumor from the subventricular zone, and the extent of mass effect. They have also laid the groundwork for noninvasive detection of mutations and epigenetic events that influence clinical outcomes such as isocitrate dehydrogenase (IDH) and O6-methylguanine-DNA methyltransferase (MGMT). In this article, we review advances in the field of glioblastoma radiomics as they pertain to prediction of IDH mutation status and MGMT promoter methylation status, as well as the development of novel, higher order radiomic parameters.
Collapse
Affiliation(s)
- Birra Taha
- Department of Neurosurgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Daniel Boley
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ju Sun
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Clark C Chen
- Department of Neurosurgery, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
21
|
Shui L, Ren H, Yang X, Li J, Chen Z, Yi C, Zhu H, Shui P. The Era of Radiogenomics in Precision Medicine: An Emerging Approach to Support Diagnosis, Treatment Decisions, and Prognostication in Oncology. Front Oncol 2021; 10:570465. [PMID: 33575207 PMCID: PMC7870863 DOI: 10.3389/fonc.2020.570465] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 12/08/2020] [Indexed: 02/05/2023] Open
Abstract
With the rapid development of new technologies, including artificial intelligence and genome sequencing, radiogenomics has emerged as a state-of-the-art science in the field of individualized medicine. Radiogenomics combines a large volume of quantitative data extracted from medical images with individual genomic phenotypes and constructs a prediction model through deep learning to stratify patients, guide therapeutic strategies, and evaluate clinical outcomes. Recent studies of various types of tumors demonstrate the predictive value of radiogenomics. And some of the issues in the radiogenomic analysis and the solutions from prior works are presented. Although the workflow criteria and international agreed guidelines for statistical methods need to be confirmed, radiogenomics represents a repeatable and cost-effective approach for the detection of continuous changes and is a promising surrogate for invasive interventions. Therefore, radiogenomics could facilitate computer-aided diagnosis, treatment, and prediction of the prognosis in patients with tumors in the routine clinical setting. Here, we summarize the integrated process of radiogenomics and introduce the crucial strategies and statistical algorithms involved in current studies.
Collapse
Affiliation(s)
- Lin Shui
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Haoyu Ren
- Department of General, Visceral and Transplantation Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Xi Yang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jian Li
- Department of Pharmacy, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Ziwei Chen
- Department of Nephrology, Chengdu Integrated TCM and Western Medicine Hospital, Chengdu, China
| | - Cheng Yi
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Zhu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Pixian Shui
- School of Pharmacy, Southwest Medical University, Luzhou, China
| |
Collapse
|
22
|
Liu Z, Wu K, Wu B, Tang X, Yuan H, Pang H, Huang Y, Zhu X, Luo H, Qi Y. Imaging genomics for accurate diagnosis and treatment of tumors: A cutting edge overview. Biomed Pharmacother 2020; 135:111173. [PMID: 33383370 DOI: 10.1016/j.biopha.2020.111173] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 11/30/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023] Open
Abstract
Imaging genomics refers to the establishment of the connection between invasive gene expression features and non-invasive imaging features. Tumor imaging genomics can not only understand the macroscopic phenotype of tumor, but also can deeply analyze the cellular and molecular characteristics of tumor tissue. In recent years, tumor imaging genomics has been a key in the field of medicine. The incidence of cancer in China has increased significantly, which is the main reason of disease death of urban residents. With the rapid development of imaging medicine, depending on imaging genomics, many experts have made remarkable achievements in tumor screening and diagnosis, prognosis evaluation, new treatment targets and understanding of tumor biological mechanism. This review analyzes the relationship between tumor radiology and gene expression, which provides a favorable direction for clinical staging, prognosis evaluation and accurate treatment of tumors.
Collapse
Affiliation(s)
- Zhen Liu
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China
| | - Kefeng Wu
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China
| | - Binhua Wu
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China
| | - Xiaoning Tang
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
| | - Huiqing Yuan
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China
| | - Hao Pang
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China
| | - Yongmei Huang
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China
| | - Xiao Zhu
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China.
| | - Hui Luo
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China.
| | - Yi Qi
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China.
| |
Collapse
|
23
|
Beig N, Bera K, Tiwari P. Introduction to radiomics and radiogenomics in neuro-oncology: implications and challenges. Neurooncol Adv 2020; 2:iv3-iv14. [PMID: 33521636 PMCID: PMC7829475 DOI: 10.1093/noajnl/vdaa148] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Neuro-oncology largely consists of malignancies of the brain and central nervous system including both primary as well as metastatic tumors. Currently, a significant clinical challenge in neuro-oncology is to tailor therapies for patients based on a priori knowledge of their survival outcome or treatment response to conventional or experimental therapies. Radiomics or the quantitative extraction of subvisual data from conventional radiographic imaging has recently emerged as a powerful data-driven approach to offer insights into clinically relevant questions related to diagnosis, prediction, prognosis, as well as assessing treatment response. Furthermore, radiogenomic approaches provide a mechanism to establish statistical correlations of radiomic features with point mutations and next-generation sequencing data to further leverage the potential of routine MRI scans to serve as "virtual biopsy" maps. In this review, we provide an introduction to radiomic and radiogenomic approaches in neuro-oncology, including a brief description of the workflow involving preprocessing, tumor segmentation, and extraction of "hand-crafted" features from the segmented region of interest, as well as identifying radiogenomic associations that could ultimately lead to the development of reliable prognostic and predictive models in neuro-oncology applications. Lastly, we discuss the promise of radiomics and radiogenomic approaches in personalizing treatment decisions in neuro-oncology, as well as the challenges with clinical adoption, which will rely heavily on their demonstrated resilience to nonstandardization in imaging protocols across sites and scanners, as well as in their ability to demonstrate reproducibility across large multi-institutional cohorts.
Collapse
Affiliation(s)
- Niha Beig
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Kaustav Bera
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Pallavi Tiwari
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
24
|
Smedley NF, El-Saden S, Hsu W. Discovering and interpreting transcriptomic drivers of imaging traits using neural networks. Bioinformatics 2020; 36:3537-3548. [PMID: 32101278 DOI: 10.1093/bioinformatics/btaa126] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 01/07/2020] [Accepted: 02/19/2020] [Indexed: 12/20/2022] Open
Abstract
MOTIVATION Cancer heterogeneity is observed at multiple biological levels. To improve our understanding of these differences and their relevance in medicine, approaches to link organ- and tissue-level information from diagnostic images and cellular-level information from genomics are needed. However, these 'radiogenomic' studies often use linear or shallow models, depend on feature selection, or consider one gene at a time to map images to genes. Moreover, no study has systematically attempted to understand the molecular basis of imaging traits based on the interpretation of what the neural network has learned. These studies are thus limited in their ability to understand the transcriptomic drivers of imaging traits, which could provide additional context for determining clinical outcomes. RESULTS We present a neural network-based approach that takes high-dimensional gene expression data as input and performs non-linear mapping to an imaging trait. To interpret the models, we propose gene masking and gene saliency to extract learned relationships from radiogenomic neural networks. In glioblastoma patients, our models outperformed comparable classifiers (>0.10 AUC) and our interpretation methods were validated using a similar model to identify known relationships between genes and molecular subtypes. We found that tumor imaging traits had specific transcription patterns, e.g. edema and genes related to cellular invasion, and 10 radiogenomic traits were significantly predictive of survival. We demonstrate that neural networks can model transcriptomic heterogeneity to reflect differences in imaging and can be used to derive radiogenomic traits with clinical value. AVAILABILITY AND IMPLEMENTATION https://github.com/novasmedley/deepRadiogenomics. CONTACT whsu@mednet.ucla.edu. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Nova F Smedley
- Medical & Imaging Informatics.,Department of Radiological Sciences.,Department of Bioengineering
| | - Suzie El-Saden
- Medical & Imaging Informatics.,Department of Radiological Sciences
| | - William Hsu
- Medical & Imaging Informatics.,Department of Radiological Sciences.,Department of Bioengineering.,Bioinformatics IDP, University of California Los Angeles, Los Angeles, CA 90024, USA
| |
Collapse
|
25
|
Chiang GC, Pisapia DJ, Liechty B, Magge R, Ramakrishna R, Knisely J, Schwartz TH, Fine HA, Kovanlikaya I. The Prognostic Value of MRI Subventricular Zone Involvement and Tumor Genetics in Lower Grade Gliomas. J Neuroimaging 2020; 30:901-909. [PMID: 32721076 DOI: 10.1111/jon.12763] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/20/2020] [Accepted: 07/07/2020] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND AND PURPOSE Glioblastomas (GBMs) that involve the subventricular zone (SVZ) have a poor prognosis, possibly due to recruitment of neural stem cells. The purpose of this study was to evaluate whether SVZ involvement by lower grade gliomas (LGG), WHO grade II and III, similarly predicts poorer outcomes. We further assessed whether tumor genetics and cellularity are associated with SVZ involvement and outcomes. METHODS Forty-five consecutive LGG patients with preoperative imaging and next generation sequencing were included in this study. Regional SVZ involvement and whole tumor apparent diffusion coefficient (ADC) values, as a measure of cellularity, were assessed on magnetic resonance imaging. Progression was determined by RANO criteria. Kaplan-Meier curves and Cox regression analyses were used to determine the hazard ratios (HR) for progression and survival. RESULTS Frontal, parietal, temporal, and overall SVZ involvement and ADC values were not associated with progression or survival (P ≥ .05). However, occipital SVZ involvement, seen in two patients, was associated with a higher risk of tumor progression (HR = 6.6, P = .016) and death (HR = 31.5, P = .015), CDKN2A/B mutations (P = .03), and lower ADC histogram values at the 5th (P = .026) and 10th percentiles (P = .046). Isocitrate dehydrogenase, phosphatase and tensin homolog, epidermal growth factor receptor, and cyclin-dependent kinase 4 mutations were also prognostic (P ≤ .05). CONCLUSIONS Unlike in GBM, overall SVZ involvement was not found to strongly predict poor prognosis in LGGs. However, occipital SVZ involvement, though uncommon, was prognostic and found to be associated with CDKN2A/B mutations and tumor hypercellularity. Further investigation into these molecular mechanisms underlying occipital SVZ involvement in larger cohorts is warranted.
Collapse
Affiliation(s)
- Gloria C Chiang
- Department of Radiology, Division of Neuroradiology, Weill Cornell Medicine, NewYork-Presbyterian Hospital, New York, NY
| | - David J Pisapia
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, NewYork-Presbyterian Hospital, New York, NY
| | - Benjamin Liechty
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, NewYork-Presbyterian Hospital, New York, NY
| | - Rajiv Magge
- Department of Neurology, Weill Cornell Medicine, NewYork-Presbyterian Hospital, New York, NY
| | - Rohan Ramakrishna
- Department of Neurosurgery, Weill Cornell Medicine, NewYork-Presbyterian Hospital, New York, NY
| | - Jonathan Knisely
- Department of Radiation Oncology, Weill Cornell Medicine, NewYork-Presbyterian Hospital, New York, NY
| | - Theodore H Schwartz
- Department of Neurosurgery, Weill Cornell Medicine, NewYork-Presbyterian Hospital, New York, NY
| | - Howard A Fine
- Department of Neurology, Weill Cornell Medicine, NewYork-Presbyterian Hospital, New York, NY
| | - Ilhami Kovanlikaya
- Department of Radiology, Division of Neuroradiology, Weill Cornell Medicine, NewYork-Presbyterian Hospital, New York, NY
| |
Collapse
|
26
|
Zhu Y, Mohamed ASR, Lai SY, Yang S, Kanwar A, Wei L, Kamal M, Sengupta S, Elhalawani H, Skinner H, Mackin DS, Shiao J, Messer J, Wong A, Ding Y, Zhang L, Court L, Ji Y, Fuller CD. Imaging-Genomic Study of Head and Neck Squamous Cell Carcinoma: Associations Between Radiomic Phenotypes and Genomic Mechanisms via Integration of The Cancer Genome Atlas and The Cancer Imaging Archive. JCO Clin Cancer Inform 2020; 3:1-9. [PMID: 30730765 DOI: 10.1200/cci.18.00073] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
PURPOSE Recent data suggest that imaging radiomic features of a tumor could be indicative of important genomic biomarkers. Understanding the relationship between radiomic and genomic features is important for basic cancer research and future patient care. We performed a comprehensive study to discover the imaginggenomic associations in head and neck squamous cell carcinoma (HNSCC) and explore the potential of predicting tumor genomic alternations using radiomic features. METHODS Our retrospective study integrated whole-genome multiomics data from The Cancer Genome Atlas with matched computed tomography imaging data from The Cancer Imaging Archive for the same set of 126 patients with HNSCC. Linear regression and gene set enrichment analysis were used to identify statistically significant associations between radiomic imaging and genomic features. Random forest classifier was used to predict the status of two key HNSCC molecular biomarkers, human papillomavirus and disruptive TP53 mutation, on the basis of radiomic features. RESULTS Widespread and statistically significant associations were discovered between genomic features (including microRNA expression, somatic mutations, and transcriptional activity, copy number variations, and promoter region DNA methylation changes of pathways) and radiomic features characterizing the size, shape, and texture of tumor. Prediction of human papillomavirus and TP53 mutation status using radiomic features achieved areas under the receiver operating characteristic curve of 0.71 and 0.641, respectively. CONCLUSION Our exploratory study suggests that radiomic features are associated with genomic characteristics at multiple molecular layers in HNSCC and provides justification for continued development of radiomics as biomarkers for relevant genomic alterations in HNSCC.
Collapse
Affiliation(s)
- Yitan Zhu
- NorthShore University HealthSystem, Evanston, IL
| | - Abdallah S R Mohamed
- The University of Texas MD Anderson Cancer Center, Houston, TX.,Alexandria University, Alexandria, Egypt
| | - Stephen Y Lai
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Aasheesh Kanwar
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Lin Wei
- NorthShore University HealthSystem, Evanston, IL
| | - Mona Kamal
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | - Heath Skinner
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Dennis S Mackin
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jay Shiao
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jay Messer
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Andrew Wong
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Yao Ding
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Lifei Zhang
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Laurence Court
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Yuan Ji
- NorthShore University HealthSystem, Evanston, IL.,The University of Chicago, Chicago, IL
| | | |
Collapse
|
27
|
Xue W, Zhang J, Tong H, Xie T, Chen X, Zhou B, Wu P, Zhong P, Du X, Guo Y, Yang Y, Liu H, Fang J, Wang S, Wu H, Xu K, Zhang W. Effects of BMPER, CXCL10, and HOXA9 on Neovascularization During Early-Growth Stage of Primary High-Grade Glioma and Their Corresponding MRI Biomarkers. Front Oncol 2020; 10:711. [PMID: 32432046 PMCID: PMC7214627 DOI: 10.3389/fonc.2020.00711] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 04/15/2020] [Indexed: 02/02/2023] Open
Abstract
Neovascularization is required in high-grade glioma (HGG). The objective of this study was to explore neovascularization-related genes and their corresponding MRI biomarkers during the early-growth stage of HGG. Tumor tissues from 30 HGG patients underwent perfusion MRI scanning prior to surgery were used to establish orthotopic xenograft models, pathologically analyze the tumor vasculature and perform transcriptome sequencing. The cases were divided into two groups based on whether the xenograft was successfully established. Microvascular density and BMPER, CXCL10, and HOXA9 expression of surgical specimens in the xenograft-forming group was significantly elevated and the microvascular diameter was significantly reduced, in vitro inhibition of BMPER, CXCL10, or HOXA9 in the glioma stem cell significantly suppressed its tube formation abilities. The in vivo experiment showed that BMPER was highly expressed in the early tumor growth phase (20 days), CXCL10 and HOXA9 expression was elevated with tumor progress, and spatially associated with tumor vasculature. Perfusion weighted MRI (PWI-MRI) derived parameters, rCBV, rCBF, Ktrans, and Vp, were also increased in the xenograft-forming group. In conclusion BMPER, CXCL10, and HOXA9 promote early tumor growth and progression by stimulating neovascularization of primary HGG. The rCBV, rCBF, Ktrans, and Vp can be used as imaging biomarkers to predict the expression statuses of these genes.
Collapse
Affiliation(s)
- Wei Xue
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Junfeng Zhang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Haipeng Tong
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Tian Xie
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Xiao Chen
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Bo Zhou
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Pengfei Wu
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Peng Zhong
- Department of Pathology, Daping Hospital, Army Medical University, Chongqing, China
| | - Xuesong Du
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Yu Guo
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Youyuan Yang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Heng Liu
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
- Department of Radiology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Jingqin Fang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Shunan Wang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Hao Wu
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Kai Xu
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Weiguo Zhang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Clinical Research Centre of Imaging and Nuclear Medicine, Chongqing, China
| |
Collapse
|
28
|
Lo Gullo R, Daimiel I, Morris EA, Pinker K. Combining molecular and imaging metrics in cancer: radiogenomics. Insights Imaging 2020; 11:1. [PMID: 31901171 PMCID: PMC6942081 DOI: 10.1186/s13244-019-0795-6] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 09/25/2019] [Indexed: 02/07/2023] Open
Abstract
Background Radiogenomics is the extension of radiomics through the combination of genetic and radiomic data. Because genetic testing remains expensive, invasive, and time-consuming, and thus unavailable for all patients, radiogenomics may play an important role in providing accurate imaging surrogates which are correlated with genetic expression, thereby serving as a substitute for genetic testing. Main body In this article, we define the meaning of radiogenomics and the difference between radiomics and radiogenomics. We provide an up-to-date review of the radiomics and radiogenomics literature in oncology, focusing on breast, brain, gynecological, liver, kidney, prostate and lung malignancies. We also discuss the current challenges to radiogenomics analysis. Conclusion Radiomics and radiogenomics are promising to increase precision in diagnosis, assessment of prognosis, and prediction of treatment response, providing valuable information for patient care throughout the course of the disease, given that this information is easily obtainable with imaging. Larger prospective studies and standardization will be needed to define relevant imaging biomarkers before they can be implemented into the clinical workflow.
Collapse
Affiliation(s)
- Roberto Lo Gullo
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, 300 E 66th St, New York, NY, 10065, USA.
| | - Isaac Daimiel
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, 300 E 66th St, New York, NY, 10065, USA
| | - Elizabeth A Morris
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, 300 E 66th St, New York, NY, 10065, USA
| | - Katja Pinker
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, 300 E 66th St, New York, NY, 10065, USA.,Department of Biomedical Imaging and Image-guided Therapy, Molecular and Gender Imaging Service, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Wien, Austria
| |
Collapse
|
29
|
Park JE, Kim HS, Park SY, Nam SJ, Chun SM, Jo Y, Kim JH. Prediction of Core Signaling Pathway by Using Diffusion- and Perfusion-based MRI Radiomics and Next-generation Sequencing in Isocitrate Dehydrogenase Wild-type Glioblastoma. Radiology 2019; 294:388-397. [PMID: 31845844 DOI: 10.1148/radiol.2019190913] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background Next-generation sequencing (NGS) enables highly sensitive cancer genomics analysis, but its clinical implications for therapeutic options from imaging-based prediction have been limited. Purpose To predict core signaling pathways in isocitrate dehydrogenase (IDH) wild-type glioblastoma by using diffusion and perfusion MRI radiomics and NGS. Materials and Methods The radiogenomics model was developed by using retrospective patients with glioma who underwent NGS and anatomic, diffusion-, and perfusion-weighted imaging between March 2017 and February 2019. For testing model performance in predicting core signaling pathway, patients with IDH wild-type glioblastoma from a retrospective analysis from a registry (ClinicalTrials.gov NCT02619890) were evaluated. Radiogenomic feature selection was performed by using t tests, least absolute shrinkage and selection operator penalization, and random forest. Combining radiogenomic features, age, and location, the performance of predicting receptor tyrosine kinase (RTK), tumor protein p53 (P53), and retinoblastoma 1 pathways was evaluated by using the area under the receiver operating characteristic curve (AUC). Results There were 120 patients (52 years ± 13 [standard deviation]; 61 women) who were evaluated. Eighty-five patients (51 years ± 13; 43 men) were in the training set and 35 patients with IDH wild-type glioblastoma (56 years ± 12; 19 women) were in the validation set. Radiogenomics model identified 71 features in the RTK, 17 features in P53, and 35 features in the retinoblastoma pathway. The combined model showed better performance than anatomic imaging-based prediction in the RTK (P = .03) and retinoblastoma (P = .03) and perfusion imaging-based prediction in the P53 pathway (P = .04) in the training set. AUC values of the combined model for the prediction of core signaling pathways were 0.88 (95% confidence interval [CI]: 0.74, 1) for RTK, 0.76 (95% CI: 0.59, 0.92) for P53, and 0.81 (95% CI: 0.64, 0.97) for retinoblastoma in the validation set. Conclusion A diffusion- and perfusion-weighted MRI radiomics model can help characterize core signaling pathways and potentially guide targeted therapy for isocitrate dehydrogenase wild-type glioblastoma. © RSNA, 2019 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Ji Eun Park
- From the Department of Radiology and Research Institute of Radiology (J.E.P., H.S.K., Y.J.), Department of Clinical Epidemiology and Biostatistics (S.Y.P.), Department of Pathology (S.J.N., S.M.C.), and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 43 Olympic-ro 88, Songpa-Gu, Seoul 05505, Korea
| | - Ho Sung Kim
- From the Department of Radiology and Research Institute of Radiology (J.E.P., H.S.K., Y.J.), Department of Clinical Epidemiology and Biostatistics (S.Y.P.), Department of Pathology (S.J.N., S.M.C.), and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 43 Olympic-ro 88, Songpa-Gu, Seoul 05505, Korea
| | - Seo Young Park
- From the Department of Radiology and Research Institute of Radiology (J.E.P., H.S.K., Y.J.), Department of Clinical Epidemiology and Biostatistics (S.Y.P.), Department of Pathology (S.J.N., S.M.C.), and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 43 Olympic-ro 88, Songpa-Gu, Seoul 05505, Korea
| | - Soo Jung Nam
- From the Department of Radiology and Research Institute of Radiology (J.E.P., H.S.K., Y.J.), Department of Clinical Epidemiology and Biostatistics (S.Y.P.), Department of Pathology (S.J.N., S.M.C.), and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 43 Olympic-ro 88, Songpa-Gu, Seoul 05505, Korea
| | - Sung-Min Chun
- From the Department of Radiology and Research Institute of Radiology (J.E.P., H.S.K., Y.J.), Department of Clinical Epidemiology and Biostatistics (S.Y.P.), Department of Pathology (S.J.N., S.M.C.), and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 43 Olympic-ro 88, Songpa-Gu, Seoul 05505, Korea
| | - Youngheun Jo
- From the Department of Radiology and Research Institute of Radiology (J.E.P., H.S.K., Y.J.), Department of Clinical Epidemiology and Biostatistics (S.Y.P.), Department of Pathology (S.J.N., S.M.C.), and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 43 Olympic-ro 88, Songpa-Gu, Seoul 05505, Korea
| | - Jeong Hoon Kim
- From the Department of Radiology and Research Institute of Radiology (J.E.P., H.S.K., Y.J.), Department of Clinical Epidemiology and Biostatistics (S.Y.P.), Department of Pathology (S.J.N., S.M.C.), and Department of Neurosurgery (J.H.K.), University of Ulsan College of Medicine, Asan Medical Center, 43 Olympic-ro 88, Songpa-Gu, Seoul 05505, Korea
| |
Collapse
|
30
|
A Comparison of the Efficiency of Using a Deep CNN Approach with Other Common Regression Methods for the Prediction of EGFR Expression in Glioblastoma Patients. J Digit Imaging 2019; 33:391-398. [PMID: 31797142 DOI: 10.1007/s10278-019-00290-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
To estimate epithermal growth factor receptor (EGFR) expression level in glioblastoma (GBM) patients using radiogenomic analysis of magnetic resonance images (MRI). A comparative study using a deep convolutional neural network (CNN)-based regression, deep neural network, least absolute shrinkage and selection operator (LASSO) regression, elastic net regression, and linear regression with no regularization was carried out to estimate EGFR expression of 166 GBM patients. Except for the deep CNN case, overfitting was prevented by using feature selection, and loss values for each method were compared. The loss values in the training phase for deep CNN, deep neural network, Elastic net, LASSO, and the linear regression with no regularization were 2.90, 8.69, 7.13, 14.63, and 21.76, respectively, while in the test phase, the loss values were 5.94, 10.28, 13.61, 17.32, and 24.19 respectively. These results illustrate that the efficiency of the deep CNN approach is better than that of the other methods, including Lasso regression, which is a regression method known for its advantage in high-dimension cases. A comparison between deep CNN, deep neural network, and three other common regression methods was carried out, and the efficiency of the CNN deep learning approach, in comparison with other regression models, was demonstrated.
Collapse
|
31
|
Qian Z, Li Y, Sun Z, Fan X, Xu K, Wang K, Li S, Zhang Z, Jiang T, Liu X, Wang Y. Radiogenomics of lower-grade gliomas: a radiomic signature as a biological surrogate for survival prediction. Aging (Albany NY) 2019; 10:2884-2899. [PMID: 30362964 PMCID: PMC6224242 DOI: 10.18632/aging.101594] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/12/2018] [Indexed: 12/20/2022]
Abstract
Objective: We aimed to identify a radiomic signature to be used as a noninvasive biomarker of prognosis in patients with lower-grade gliomas (LGGs) and to reveal underlying biological processes through comprehensive radiogenomic investigation. Methods: We extracted 55 radiomic features from T2-weighted images of 233 patients with LGGs (training cohort: n = 85; validation cohort: n = 148). Univariate Cox regression and linear risk score formula were applied to generate a radiomic-based signature. Gene ontology analysis of highly expressed genes in the high-risk score group was conducted to establish a radiogenomic map. A nomogram was constructed for individualized survival prediction. Results: The six-feature radiomic signature stratified patients in the training cohort into low- or high-risk groups for overall survival (P = 0.0018). This result was successfully verified in the validation cohort (P = 0.0396). Radiogenomic analysis revealed that the prognostic radiomic signature was associated with hypoxia, angiogenesis, apoptosis, and cell proliferation. The nomogram resulted in high prognostic accuracy (C-index: 0.92, C-index: 0.70) and favorable calibration for individualized survival prediction in the training and validation cohorts. Conclusions: Our results suggest a great potential for the use of radiomic signature as a biological surrogate in providing prognostic information for patients with LGGs.
Collapse
Affiliation(s)
- Zenghui Qian
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Yiming Li
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Zhiyan Sun
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Xing Fan
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Kaibin Xu
- Chinese Academy of Sciences, Institute of Automation, Beijing, China
| | - Kai Wang
- Department of Nuclear Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shaowu Li
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neuroradiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhong Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Tao Jiang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China
| | - Xing Liu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Yinyan Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
32
|
Berendsen S, van Bodegraven E, Seute T, Spliet WGM, Geurts M, Hendrikse J, Schoysman L, Huiszoon WB, Varkila M, Rouss S, Bell EH, Kroonen J, Chakravarti A, Bours V, Snijders TJ, Robe PA. Adverse prognosis of glioblastoma contacting the subventricular zone: Biological correlates. PLoS One 2019; 14:e0222717. [PMID: 31603915 PMCID: PMC6788733 DOI: 10.1371/journal.pone.0222717] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 09/05/2019] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION The subventricular zone (SVZ) in the brain is associated with gliomagenesis and resistance to treatment in glioblastoma. In this study, we investigate the prognostic role and biological characteristics of subventricular zone (SVZ) involvement in glioblastoma. METHODS We analyzed T1-weighted, gadolinium-enhanced MR images of a retrospective cohort of 647 primary glioblastoma patients diagnosed between 2005-2013, and performed a multivariable Cox regression analysis to adjust the prognostic effect of SVZ involvement for clinical patient- and tumor-related factors. Protein expression patterns of a.o. markers of neural stem cellness (CD133 and GFAP-δ) and (epithelial-) mesenchymal transition (NF-κB, C/EBP-β and STAT3) were determined with immunohistochemistry on tissue microarrays containing 220 of the tumors. Molecular classification and mRNA expression-based gene set enrichment analyses, miRNA expression and SNP copy number analyses were performed on fresh frozen tissue obtained from 76 tumors. Confirmatory analyses were performed on glioblastoma TCGA/TCIA data. RESULTS Involvement of the SVZ was a significant adverse prognostic factor in glioblastoma, independent of age, KPS, surgery type and postoperative treatment. Tumor volume and postoperative complications did not explain this prognostic effect. SVZ contact was associated with increased nuclear expression of the (epithelial-) mesenchymal transition markers C/EBP-β and phospho-STAT3. SVZ contact was not associated with molecular subtype, distinct gene expression patterns, or markers of stem cellness. Our main findings were confirmed in a cohort of 229 TCGA/TCIA glioblastomas. CONCLUSION In conclusion, involvement of the SVZ is an independent prognostic factor in glioblastoma, and associates with increased expression of key markers of (epithelial-) mesenchymal transformation, but does not correlate with stem cellness, molecular subtype, or specific (mi)RNA expression patterns.
Collapse
Affiliation(s)
- Sharon Berendsen
- UMC Utrecht Brain Center, Department of Neurology and Neurosurgery, University Medical Center of Utrecht, Utrecht, The Netherlands
| | - Emma van Bodegraven
- UMC Utrecht Brain Center, Department of Translational Neuroscience, University Medical Center of Utrecht, Utrecht, The Netherlands
| | - Tatjana Seute
- UMC Utrecht Brain Center, Department of Neurology and Neurosurgery, University Medical Center of Utrecht, Utrecht, The Netherlands
| | - Wim G. M. Spliet
- Department of Pathology, University Medical Center of Utrecht, Utrecht, The Netherlands
| | - Marjolein Geurts
- UMC Utrecht Brain Center, Department of Neurology and Neurosurgery, University Medical Center of Utrecht, Utrecht, The Netherlands
| | - Jeroen Hendrikse
- Department of Radiology, University Medical Center of Utrecht, Utrecht, The Netherlands
| | - Laurent Schoysman
- Department of Human Genetics, GIGA Research Center, Liège University Hospital, Liège, Belgium
- Department of Radiology, Liège University Hospital, Liège, Belgium
| | - Willemijn B. Huiszoon
- UMC Utrecht Brain Center, Department of Neurology and Neurosurgery, University Medical Center of Utrecht, Utrecht, The Netherlands
| | - Meri Varkila
- UMC Utrecht Brain Center, Department of Neurology and Neurosurgery, University Medical Center of Utrecht, Utrecht, The Netherlands
| | - Soufyan Rouss
- UMC Utrecht Brain Center, Department of Neurology and Neurosurgery, University Medical Center of Utrecht, Utrecht, The Netherlands
| | - Erica H. Bell
- Department of Radiation Oncology, Wexner Medical Center, James Cancer Center, Ohio State University, Columbus, OH, United States of America
| | - Jérôme Kroonen
- UMC Utrecht Brain Center, Department of Neurology and Neurosurgery, University Medical Center of Utrecht, Utrecht, The Netherlands
- Department of Human Genetics, GIGA Research Center, Liège University Hospital, Liège, Belgium
| | - Arnab Chakravarti
- Department of Radiation Oncology, Wexner Medical Center, James Cancer Center, Ohio State University, Columbus, OH, United States of America
| | - Vincent Bours
- Department of Human Genetics, GIGA Research Center, Liège University Hospital, Liège, Belgium
| | - Tom J. Snijders
- UMC Utrecht Brain Center, Department of Neurology and Neurosurgery, University Medical Center of Utrecht, Utrecht, The Netherlands
| | - Pierre A. Robe
- UMC Utrecht Brain Center, Department of Neurology and Neurosurgery, University Medical Center of Utrecht, Utrecht, The Netherlands
- Department of Human Genetics, GIGA Research Center, Liège University Hospital, Liège, Belgium
- Department of Radiation Oncology, Wexner Medical Center, James Cancer Center, Ohio State University, Columbus, OH, United States of America
- * E-mail:
| |
Collapse
|
33
|
Jaju A, Hwang EI, Kool M, Capper D, Chavez L, Brabetz S, Billups C, Li Y, Fouladi M, Packer RJ, Pfister SM, Olson JM, Heier LA. MRI Features of Histologically Diagnosed Supratentorial Primitive Neuroectodermal Tumors and Pineoblastomas in Correlation with Molecular Diagnoses and Outcomes: A Report from the Children's Oncology Group ACNS0332 Trial. AJNR Am J Neuroradiol 2019; 40:1796-1803. [PMID: 31601576 DOI: 10.3174/ajnr.a6253] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 08/21/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND PURPOSE Supratentorial primitive neuroectodermal tumors and pineoblastomas have traditionally been grouped together for treatment purposes. Molecular profiling of these tumors has revealed a number of distinct entities and has led to the term "CNS-primitive neuroectodermal tumors" being removed from the 2016 World Health Organization classification. The purpose of this study was to describe the MR imaging findings of histologically diagnosed primitive neuroectodermal tumors and pineoblastomas and correlate them with molecular diagnoses and outcomes. MATERIALS AND METHODS Histologically diagnosed primitive neuroectodermal tumors and pineoblastomas were enrolled in this Children's Oncology Group Phase III trial, and molecular classification was retrospectively completed using DNA methylation profiling. MR imaging features were systematically studied and correlated with molecular diagnoses and survival. RESULTS Of the 85 patients enrolled, 56 met the inclusion criteria, in whom 28 tumors were in pineal and 28 in nonpineal locations. Methylation profiling revealed a variety of diagnoses, including pineoblastomas (n = 27), high-grade gliomas (n = 17), embryonal tumors (n = 7), atypical teratoid/rhabdoid tumors (n = 3), and ependymomas (n = 2). Thus, 39% overall and 71% of nonpineal tumor diagnoses were discrepant with histopathology. Tumor location, size, margins, and edema were predictors of embryonal-versus-nonembryonal tumors. Larger size and ill-defined margins correlated with poor event-free survival, while metastatic disease by MR imaging did not. CONCLUSIONS In nonpineal locations, only a minority of histologically diagnosed primitive neuroectodermal tumors are embryonal tumors; therefore, high-grade glioma or ependymoma should be high on the radiographic differential. An understanding of molecularly defined tumor entities and their relative frequencies and locations will help the radiologist make more accurate predictions of the tumor types.
Collapse
Affiliation(s)
- A Jaju
- From the Department of Radiology (A.J.), Ann and Robert H Lurie Children's Hospital of Chicago, Chicago, Illinois .,Northwestern University Feinberg School of Medicine (A.J.), Chicago, Illinois
| | - E I Hwang
- Brain Tumor Institute (E.I.H., R.J.P.), Children's National Health System, Washington, DC
| | - M Kool
- Department of Pediatric Neurooncology (M.K., S.B., S.M.P.), German Cancer Research Center, Heidelberg, Baden-Württemberg, Germany
| | - D Capper
- Department of Pediatric Neuropathology (D.C.), University Hospital Heidelberg, Heidelberg, Baden-Württemberg, Germany
| | - L Chavez
- Department of Medicine (L.C.), University of California San Diego, La Jolla, California
| | - S Brabetz
- Department of Pediatric Neurooncology (M.K., S.B., S.M.P.), German Cancer Research Center, Heidelberg, Baden-Württemberg, Germany
| | - C Billups
- Department of Biostatistics (C.B., Y.L.), St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Y Li
- Department of Biostatistics (C.B., Y.L.), St. Jude Children's Research Hospital, Memphis, Tennessee
| | - M Fouladi
- Brain Tumor Center (M.F.), Cincinnati Children's Hospital, Cincinnati, Ohio
| | - R J Packer
- Brain Tumor Institute (E.I.H., R.J.P.), Children's National Health System, Washington, DC
| | - S M Pfister
- Department of Pediatric Neurooncology (M.K., S.B., S.M.P.), German Cancer Research Center, Heidelberg, Baden-Württemberg, Germany
| | - J M Olson
- Fred Hurtchinson Cancer Research Center (J.M.O.), Seattle Children's Hospital, Seattle, Washington
| | - L A Heier
- Department of Radiology (L.A.H.), New York Presbyterian Hospital, New York, New York
| |
Collapse
|
34
|
Glioma through the looking GLASS: molecular evolution of diffuse gliomas and the Glioma Longitudinal Analysis Consortium. Neuro Oncol 2019; 20:873-884. [PMID: 29432615 PMCID: PMC6280138 DOI: 10.1093/neuonc/noy020] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Adult diffuse gliomas are a diverse group of brain neoplasms that inflict a high emotional toll on patients and their families. The Cancer Genome Atlas and similar projects have provided a comprehensive understanding of the somatic alterations and molecular subtypes of glioma at diagnosis. However, gliomas undergo significant cellular and molecular evolution during disease progression. We review the current knowledge on the genomic and epigenetic abnormalities in primary tumors and after disease recurrence, highlight the gaps in the literature, and elaborate on the need for a new multi-institutional effort to bridge these knowledge gaps and how the Glioma Longitudinal Analysis Consortium (GLASS) aims to systemically catalog the longitudinal changes in gliomas. The GLASS initiative will provide essential insights into the evolution of glioma toward a lethal phenotype, with the potential to reveal targetable vulnerabilities and, ultimately, improved outcomes for a patient population in need.
Collapse
|
35
|
Chen X, Fang M, Dong D, Liu L, Xu X, Wei X, Jiang X, Qin L, Liu Z. Development and Validation of a MRI-Based Radiomics Prognostic Classifier in Patients with Primary Glioblastoma Multiforme. Acad Radiol 2019; 26:1292-1300. [PMID: 30660472 DOI: 10.1016/j.acra.2018.12.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 12/06/2018] [Accepted: 12/19/2018] [Indexed: 12/15/2022]
Abstract
RATIONALE AND OBJECTIVES Glioblastoma multiforme (GBM) is the most common and deadly type of primary malignant tumor of the central nervous system. Accurate risk stratification is vital for a more personalized approach in GBM management. The purpose of this study is to develop and validate a MRI-based prognostic quantitative radiomics classifier in patients with newly diagnosed GBM and to evaluate whether the classifier allows stratification with improved accuracy over the clinical and qualitative imaging features risk models. METHODS Clinical and MR imaging data of 127 GBM patients were obtained from the Cancer Genome Atlas and the Cancer Imaging Archive. Regions of interest corresponding to high signal intensity portions of tumor were drawn on postcontrast T1-weighted imaging (post-T1WI) on the 127 patients (allocated in a 2:1 ratio into a training [n = 85] or validation [n = 42] set), then 3824 radiomics features per patient were extracted. The dimension of these radiomics features were reduced using the minimum redundancy maximum relevance algorithm, then Cox proportional hazard regression model was used to build a radiomics classifier for predicting overall survival (OS). The value of the radiomics classifier beyond clinical (gender, age, Karnofsky performance status, radiation therapy, chemotherapy, and type of resection) and VASARI features for OS was assessed with multivariate Cox proportional hazards model. Time-dependent receiver operating characteristic curve analysis was used to assess the predictive accuracy. RESULTS A classifier using four post-T1WI-MRI radiomics features built on the training dataset could successfully separate GBM patients into low- or high-risk group with a significantly different OS in training (HR, 6.307 [95% CI, 3.475-11.446]; p < 0.001) and validation set (HR, 3.646 [95% CI, 1.709-7.779]; p < 0.001). The area under receiver operating characteristic curve of radiomics classifier (training, 0.799; validation, 0.815 for 12-month) was higher compared to that of the clinical risk model (Karnofsky performance status, radiation therapy; training, 0.749; validation, 0.670 for 12-month), and none of the qualitative imaging features was associated with OS. The predictive accuracy was further improved when combined the radiomics classifier with clinical data (training, 0.819; validation: 0.851 for 12-month). CONCLUSION A classifier using radiomics features allows preoperative prediction of survival and risk stratification of patients with GBM, and it shows improved performance compared to that of clinical and qualitative imaging features models.
Collapse
Affiliation(s)
- Xin Chen
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China; Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China; Department of Radiology, Harvard Medical School, Boston 02115, Massachusetts
| | - Mengjie Fang
- Key Laboratory of Molecular Imaging, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Di Dong
- Key Laboratory of Molecular Imaging, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Lingling Liu
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xiangdong Xu
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xinhua Wei
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xinqing Jiang
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Lei Qin
- Department of Imaging, Dana-Farber Cancer Institute, Boston 02115, Massachusetts; Department of Radiology, Harvard Medical School, Boston 02115, Massachusetts.
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China.
| |
Collapse
|
36
|
Broen MPG, Smits M, Wijnenga MMJ, Dubbink HJ, Anten MHME, Schijns OEMG, Beckervordersandforth J, Postma AA, van den Bent MJ. The T2-FLAIR mismatch sign as an imaging marker for non-enhancing IDH-mutant, 1p/19q-intact lower-grade glioma: a validation study. Neuro Oncol 2019; 20:1393-1399. [PMID: 29590424 DOI: 10.1093/neuonc/noy048] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background The purpose of this study was to assess the reproducibility of the previously described T2-fluid attenuated inversion recovery (FLAIR) mismatch sign as a specific imaging marker in non-enhancing isocitrate dehydrogenase (IDH) mutant, 1p/19q non-codeleted lower-grade glioma (LGG), encompassing both diffuse and anaplastic astrocytoma. Methods MR scans (n = 154) from 3 separate databases with genotyped LGG were evaluated by 2 independent reviewers to assess (i) presence/absence of "T2-FLAIR mismatch" sign and (ii) presence/absence of homogeneous signal on T2-weighted images. Interrater agreement with Cohen's kappa (κ) was calculated, as well as diagnostic test performance of the T2-FLAIR mismatch sign to identify IDH-mutant astrocytoma. Results There was substantial interrater agreement for the T2-FLAIR mismatch sign [κ = 0.75 (0.64-0.87)], but only fair agreement for T2 homogeneity [κ = 0.38 (0.25-0.52)]. The T2-FLAIR mismatch sign was present in 38 cases (25%) and had a positive predictive value of 100%, negative predictive value of 68%, a sensitivity of 51%, and a specificity of 100%. Conclusions With a robust interrater agreement, our study confirms that among non-enhancing LGG the T2-FLAIR mismatch sign represents a highly specific imaging marker for IDH-mutant astrocytoma. This non-invasive marker may enable a more informed patient counsel and can aid in the treatment decision processes in a significant proportion of patients presenting with non-enhancing, LGG-like lesions.
Collapse
Affiliation(s)
- Martinus P G Broen
- Department of Neurology, Maastricht University Medical Center, Maastricht, Netherlands.,Department of Neurology, The Brain Tumor Center at Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Marion Smits
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Maarten M J Wijnenga
- Department of Neurology, The Brain Tumor Center at Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Hendrikus J Dubbink
- Department of Pathology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Monique H M E Anten
- Department of Neurology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Olaf E M G Schijns
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, Netherlands
| | - Jan Beckervordersandforth
- Department of Pathology, GROW-school for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Alida A Postma
- Department of Radiology, Maastricht University Medical Center, Maastricht, Netherlands
| | - Martin J van den Bent
- Department of Neurology, The Brain Tumor Center at Erasmus MC Cancer Institute, Rotterdam, Netherlands
| |
Collapse
|
37
|
Fatehi M, Yip S. Commentary: Radiological Characteristics and Natural History of Adult IDH-Wild-Type Astrocytomas With TERT Promoter Mutations. Neurosurgery 2019; 85:E457-E458. [PMID: 30418602 DOI: 10.1093/neuros/nyy529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Accepted: 10/04/2018] [Indexed: 11/13/2022] Open
Affiliation(s)
- Mostafa Fatehi
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Stephen Yip
- Department of Pathology & Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
38
|
Fathi Kazerooni A, Bakas S, Saligheh Rad H, Davatzikos C. Imaging signatures of glioblastoma molecular characteristics: A radiogenomics review. J Magn Reson Imaging 2019; 52:54-69. [PMID: 31456318 DOI: 10.1002/jmri.26907] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 08/09/2019] [Indexed: 02/06/2023] Open
Abstract
Over the past few decades, the advent and development of genomic assessment methods and computational approaches have raised the hopes for identifying therapeutic targets that may aid in the treatment of glioblastoma. However, the targeted therapies have barely been successful in their effort to cure glioblastoma patients, leaving them with a grim prognosis. Glioblastoma exhibits high heterogeneity, both spatially and temporally. The existence of different genetic subpopulations in glioblastoma allows this tumor to adapt itself to environmental forces. Therefore, patients with glioblastoma respond poorly to the prescribed therapies, as treatments are directed towards the whole tumor and not to the specific genetic subregions. Genomic alterations within the tumor develop distinct radiographic phenotypes. In this regard, MRI plays a key role in characterizing molecular signatures of glioblastoma, based on regional variations and phenotypic presentation of the tumor. Radiogenomics has emerged as a (relatively) new field of research to explore the connections between genetic alterations and imaging features. Radiogenomics offers numerous advantages, including noninvasive and global assessment of the tumor and its response to therapies. In this review, we summarize the potential role of radiogenomic techniques to stratify patients according to their specific tumor characteristics with the goal of designing patient-specific therapies. Level of Evidence: 5 Technical Efficacy: Stage 2 J. Magn. Reson. Imaging 2020;52:54-69.
Collapse
Affiliation(s)
- Anahita Fathi Kazerooni
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Spyridon Bakas
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hamidreza Saligheh Rad
- Quantitative MR Imaging and Spectroscopy Group (QMISG), Research Center for Molecular and Cellular Imaging, Tehran University of Medical Sciences, Tehran, Iran
| | - Christos Davatzikos
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
39
|
Korn RL, Rahmanuddin S, Borazanci E. Use of Precision Imaging in the Evaluation of Pancreas Cancer. Cancer Treat Res 2019; 178:209-236. [PMID: 31209847 DOI: 10.1007/978-3-030-16391-4_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Pancreas cancer is an aggressive and fatal disease that will become one of the leading causes of cancer mortality by 2030. An all-out effort is underway to better understand the basic biologic mechanisms of this disease ranging from early development to metastatic disease. In order to change the course of this disease, diagnostic radiology imaging may play a vital role in providing a precise, noninvasive method for early diagnosis and assessment of treatment response. Recent progress in combining medical imaging, advanced image analysis and artificial intelligence, termed radiomics, can offer an innovate approach in detecting the earliest changes of tumor development as well as a rapid method for the detection of response. In this chapter, we introduce the principles of radiomics and demonstrate how it can provide additional information into tumor biology, early detection, and response assessments advancing the goals of precision imaging to deliver the right treatment to the right person at the right time.
Collapse
Affiliation(s)
- Ronald L Korn
- Virginia G Piper Cancer Center at HonorHealth, Scottsdale, AZ, USA. .,Translational Genomics Research Institute, An Affiliate of City of Hope, Phoenix, AZ, USA. .,Imaging Endpoints Core Lab, Scottsdale, AZ, USA.
| | | | - Erkut Borazanci
- Virginia G Piper Cancer Center at HonorHealth, Scottsdale, AZ, USA.,Translational Genomics Research Institute, An Affiliate of City of Hope, Phoenix, AZ, USA
| |
Collapse
|
40
|
Liu Z, Liu H, Liu Z, Zhang J. Oligodendroglial tumours: subventricular zone involvement and seizure history are associated with CIC mutation status. BMC Neurol 2019; 19:134. [PMID: 31215432 PMCID: PMC6582578 DOI: 10.1186/s12883-019-1362-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 06/06/2019] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND CIC-mutant oligodendroglial tumours linked to better prognosis. We aim to investigate associations between CIC gene mutation status, MR characteristics and clinical features. METHODS Imaging and genomic data from the Cancer Genome Atlas and the Cancer Imaging Archive (TCGA/TCIA) for 59 patients with oligodendroglial tumours were used. Differences between CIC mutation and CIC wild-type were tested using Chi-square test and binary logistic regression analysis. RESULTS In univariate analysis, the clinical variables and MR features, which consisted 3 selected features (subventricular zone[SVZ] involvement, volume and seizure history) were associated with CIC mutation status (all p < 0.05). A multivariate logistic regression analysis identified that seizure history (no vs. yes odd ratio [OR]: 28.960, 95 confidence interval [CI]:2.625-319.49, p = 0.006) and SVZ involvement (SVZ- vs. SVZ+ OR: 77.092, p = 0.003; 95% CI: 4.578-1298.334) were associated with a higher incidence of CIC mutation status. The nomogram showed good discrimination, with a C-index of 0.906 (95% CI: 0.812-1.000) and was well calibrated. SVZ- group has increased (SVZ- vs. SVZ+, hazard ratio [HR]: 4.500, p = 0.04; 95% CI: 1.069-18.945) overall survival. CONCLUSIONS Absence of seizure history and SVZ involvement (-) was associated with a higher incidence of CIC mutation.
Collapse
Affiliation(s)
- Zhenyin Liu
- Department of medical imaging, Guangzhou women and children's medical center, Guangzhou medical university, Jinsui road 9 #, Guangzhou City, 510623, People's Republic of China
| | - Hongsheng Liu
- Department of medical imaging, Guangzhou women and children's medical center, Guangzhou medical university, Jinsui road 9 #, Guangzhou City, 510623, People's Republic of China
| | - Zhenqing Liu
- Department of medical imaging, Guangzhou women and children's medical center, Guangzhou medical university, Jinsui road 9 #, Guangzhou City, 510623, People's Republic of China
| | - Jing Zhang
- Department of medical imaging, Guangzhou women and children's medical center, Guangzhou medical university, Jinsui road 9 #, Guangzhou City, 510623, People's Republic of China.
| |
Collapse
|
41
|
MR imaging phenotype correlates with extent of genome-wide copy number abundance in IDH mutant gliomas. Neuroradiology 2019; 61:1023-1031. [PMID: 31134296 DOI: 10.1007/s00234-019-02219-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 04/29/2019] [Indexed: 12/15/2022]
Abstract
PURPOSE There is variability in survival within IDH mutant gliomas determined by chromosomal events. Copy number variation (CNV) abundance associated with survival in low-grade and IDH mutant astrocytoma has been reported. Our purpose was to correlate the extent of genome-wide CNV abundance in IDH mutant astrocytomas with MRI features. METHODS Presurgical MRI and CNV plots derived from Illumina 850k EPIC DNA methylation arrays of 18 cases of WHO grade II-IV IDH mutant astrocytomas were reviewed. IDH mutant astrocytomas were divided into CNV stable group (CNV-S) with ≤ 3 chromosomal gains or losses and lack of focal gene amplifications and CNV unstable group (CNV-U) with > 3 large chromosomal gains/losses and/or focal amplifications. The associations between MR features, relative cerebral blood volume (rCBV), CNV abundance, and time to progression were assessed. Tumor rCBV estimates were obtained using DSC T2* perfusion analysis. RESULTS There were nine (50%) CNV-S and nine (50%) CNV-U IDH mutant astrocytomas. CNV-U tumors showed larger mean tumor size (P = 0.004) and maximum diameter on FLAIR (P = 0.004) and also demonstrated significantly higher median rCBV than CNV-S tumors (2.62 vs 0.78, P = 0.019). CNV-U tumors tended to have shorter time to progression although without statistical significance (P = 0.393). CONCLUSIONS Larger size/diameter and higher rCBVs were seen associated CNV-U astrocytomas, suggesting a correlation of aggressive imaging phenotype with unstable and aggressive genotype in IDH mutant astrocytomas.
Collapse
|
42
|
Bowen L, Xiaojing L. Radiogenomics of Clear Cell Renal Cell Carcinoma: Associations Between mRNA-Based Subtyping and CT Imaging Features. Acad Radiol 2019; 26:e32-e37. [PMID: 30064916 DOI: 10.1016/j.acra.2018.05.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 05/15/2018] [Accepted: 05/16/2018] [Indexed: 12/11/2022]
Abstract
PURPOSE To investigate associations between clear-cell renal cell carcinoma mRNA-based subtyping and CT features. MATERIALS AND METHODS The CT data from 177 patients generated with The Cancer Imaging Archive were reviewed. The correlation was analyzed using chi-square test and univariate regression analysis. RESULTS Identified were 124 (53.2%) m1, 67 (28.8%) m2, 17 (7.3%) m3, and 14 (8.7%) m4 subtypes. m1-subtype rates were significantly higher in well-defined margin lesions (p = 0.041). m3-subtype rates were significantly higher in ill-defined margin lesions (p = 0.012), in collecting system invasion lesions (p = 0.028) and collecting system invasion lesions (p = 0.026).On univariate logistic regression analysis, tumor margin (well-defined margin vs ill-defined margin, OR: 2.104; p = 0.041; 95% CI: 1.024-4.322) was associated with m1-subtype. Tumor margin (well-defined margin vs ill-defined margin, OR: 2.104; p = 0.012; 95% CI: 0.212-0.834) and collecting system invasion (yes vs no, OR: 0.421; p = 0.028; 95% CI: 0.212-0.834) and renal vein invasion (yes vs no, OR: 2.164; p = 0.026; 95% CI: 1.090-4.294) were associated with m3-subtype. There was no significant difference between mRNA-based subtyping (m2 vs other; m4 vs other) and the CT features. CONCLUSIONS This preliminary radiogenomics analysis of clear-cell renal cell carcinoma revealed associations between CT features and mRNA-based subtyping which warrant further investigation and validation.
Collapse
|
43
|
Kim MM, Speers C, Li P, Schipper M, Junck L, Leung D, Orringer D, Heth J, Umemura Y, Spratt DE, Wahl DR, Cao Y, Lawrence TS, Tsien CI. Dose-intensified chemoradiation is associated with altered patterns of failure and favorable survival in patients with newly diagnosed glioblastoma. J Neurooncol 2019; 143:313-319. [PMID: 30977058 DOI: 10.1007/s11060-019-03166-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 04/08/2019] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND PURPOSE We evaluated whether dose-intensified chemoradiation alters patterns of failure and is associated with favorable survival in the temozolomide era. MATERIALS AND METHODS Between 2003 and 2015, 82 patients with newly diagnosed glioblastoma were treated with 66-81 Gy in 30 fractions using conventional magnetic resonance imaging. Progression-free (PFS) and overall survival (OS) were calculated using Kaplan-Meier methods. Factors associated with improved PFS, OS, and time to progression were assessed using multivariate Cox model and linear regression. RESULTS Median follow-up was 23 months (95% CI 4-124 months). Sixty-one percent of patients underwent subtotal resection or biopsy, and 38% (10/26) of patients with available data had MGMT promoter methylation. Median PFS was 8.4 months (95% CI 7.3-11.0) and OS was 18.7 months (95% CI 13.1-25.3). Only 30 patients (44%) experienced central recurrence, 6 (9%) in-field, 16 (23.5%) marginal and 16 (23.5%) distant. On multivariate analysis, younger age (HR 0.95, 95% CI 0.93-0.97, p = 0.0001), higher performance status (HR 0.39, 95% CI 0.16-0.95, p = 0.04), gross total resection (GTR) versus biopsy (HR 0.37, 95% CI 0.16-0.85, p = 0.02) and MGMT methylation (HR 0.25, 95% CI 0.09-0.71, p = 0.009) were associated with improved OS. Only distant versus central recurrence (p = 0.03) and GTR (p = 0.02) were associated with longer time to progression. Late grade 3 neurologic toxicity was rare (6%) in patients experiencing long-term survival. CONCLUSION Dose-escalated chemoRT resulted in lower rates of central recurrence and prolonged time to progression compared to historical controls, although a significant number of central recurrences were still observed. Advanced imaging and correlative molecular studies may enable targeted treatment advances that reduce rates of in- and out-of-field progression.
Collapse
Affiliation(s)
- Michelle M Kim
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA.
| | - Corey Speers
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Pin Li
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Matthew Schipper
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Larry Junck
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Denise Leung
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Daniel Orringer
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Jason Heth
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Yoshie Umemura
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Daniel E Spratt
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Daniel R Wahl
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Yue Cao
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Theodore S Lawrence
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Christina I Tsien
- Department of Radiation Oncology, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
44
|
Prasanna P, Mitra J, Beig N, Nayate A, Patel J, Ghose S, Thawani R, Partovi S, Madabhushi A, Tiwari P. Mass Effect Deformation Heterogeneity (MEDH) on Gadolinium-contrast T1-weighted MRI is associated with decreased survival in patients with right cerebral hemisphere Glioblastoma: A feasibility study. Sci Rep 2019; 9:1145. [PMID: 30718547 PMCID: PMC6362117 DOI: 10.1038/s41598-018-37615-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 12/04/2018] [Indexed: 12/04/2022] Open
Abstract
Subtle tissue deformations caused by mass-effect in Glioblastoma (GBM) are often not visually evident, and may cause neurological deficits, impacting survival. Radiomic features provide sub-visual quantitative measures to uncover disease characteristics. We present a new radiomic feature to capture mass effect-induced deformations in the brain on Gadolinium-contrast (Gd-C) T1w-MRI, and their impact on survival. Our rationale is that larger variations in deformation within functionally eloquent areas of the contralateral hemisphere are likely related to decreased survival. Displacements in the cortical and subcortical structures were measured by aligning the Gd-C T1w-MRI to a healthy atlas. The variance of deformation magnitudes was measured and defined as Mass Effect Deformation Heterogeneity (MEDH) within the brain structures. MEDH values were then correlated with overall-survival of 89 subjects on the discovery cohort, with tumors on the right (n = 41) and left (n = 48) cerebral hemispheres, and evaluated on a hold-out cohort (n = 49 subjects). On both cohorts, decreased survival time was found to be associated with increased MEDH in areas of language comprehension, social cognition, visual perception, emotion, somato-sensory, cognitive and motor-control functions, particularly in the memory areas in the left-hemisphere. Our results suggest that higher MEDH in functionally eloquent areas of the left-hemisphere due to GBM in the right-hemisphere may be associated with poor-survival.
Collapse
Affiliation(s)
- Prateek Prasanna
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, USA
| | - Jhimli Mitra
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, USA
- General Electric Global Research, New York, USA
| | - Niha Beig
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, USA
| | - Ameya Nayate
- Department of Radiology, University Hospitals Cleveland Medical Center, Cleveland, USA
| | - Jay Patel
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, USA
| | - Soumya Ghose
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, USA
| | - Rajat Thawani
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, USA
| | - Sasan Partovi
- Department of Radiology, University Hospitals Cleveland Medical Center, Cleveland, USA
| | - Anant Madabhushi
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, USA
| | - Pallavi Tiwari
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, USA.
| |
Collapse
|
45
|
Texture analysis on conventional MRI images accurately predicts early malignant transformation of low-grade gliomas. Eur Radiol 2019; 29:2751-2759. [PMID: 30617484 DOI: 10.1007/s00330-018-5921-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/31/2018] [Accepted: 11/27/2018] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Texture analysis performed on MRI images can provide additional quantitative information that is invisible to human assessment. This study aimed to evaluate the feasibility of texture analysis on preoperative conventional MRI images in predicting early malignant transformation from low- to high-grade glioma and compare its utility to histogram analysis alone. METHODS A total of 68 patients with low-grade glioma (LGG) were included in this study, 15 of which showed malignant transformation. Patients were randomly divided into training (60%) and testing (40%) sets. Texture analyses were performed to obtain the most discriminant factor (MDF) values for both training and testing data. Receiver operating characteristic (ROC) curve analyses were performed on MDF values and 9 histogram parameters in the training data to obtain cutoff values for determining the correct rates of discrimination between two groups in the testing data. RESULTS The ROC analyses on MDF values resulted in an area under the curve (AUC) of 0.90 (sensitivity 85%, specificity 84%) for T2w FLAIR, 0.92 (86%, 94%) for ADC, 0.96 (97%, 84%) for T1w, and 0.82 (78%, 75%) for T1w + Gd and correctly discriminated between the two groups in 93%, 100%, 93%, and 92% of cases in testing data, respectively. In the astrocytoma subgroup, AUCs were 0.92 (88%, 83%) for T2w FLAIR and 0.90 (92%, 74%) for T1w + Gd and correctly discriminated two groups in 100% and 92% of cases. The MDF outperformed all 9 of the histogram parameters. CONCLUSION Texture analysis on conventional preoperative MRI images can accurately predict early malignant transformation of LGGs, which may guide therapeutic planning. KEY POINTS • Texture analysis performed on MRI images can provide additional quantitative information that is invisible to human assessment. • Texture analysis based on conventional preoperative MR images can accurately predict early malignant transformation from low- to high-grade glioma. • Texture analysis is a clinically feasible technique that may provide an alternative and effective way of determining the likelihood of early malignant transformation and help guide therapeutic decisions.
Collapse
|
46
|
Napel S, Mu W, Jardim‐Perassi BV, Aerts HJWL, Gillies RJ. Quantitative imaging of cancer in the postgenomic era: Radio(geno)mics, deep learning, and habitats. Cancer 2018; 124:4633-4649. [PMID: 30383900 PMCID: PMC6482447 DOI: 10.1002/cncr.31630] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 07/11/2018] [Accepted: 07/17/2018] [Indexed: 11/07/2022]
Abstract
Although cancer often is referred to as "a disease of the genes," it is indisputable that the (epi)genetic properties of individual cancer cells are highly variable, even within the same tumor. Hence, preexisting resistant clones will emerge and proliferate after therapeutic selection that targets sensitive clones. Herein, the authors propose that quantitative image analytics, known as "radiomics," can be used to quantify and characterize this heterogeneity. Virtually every patient with cancer is imaged radiologically. Radiomics is predicated on the beliefs that these images reflect underlying pathophysiologies, and that they can be converted into mineable data for improved diagnosis, prognosis, prediction, and therapy monitoring. In the last decade, the radiomics of cancer has grown from a few laboratories to a worldwide enterprise. During this growth, radiomics has established a convention, wherein a large set of annotated image features (1-2000 features) are extracted from segmented regions of interest and used to build classifier models to separate individual patients into their appropriate class (eg, indolent vs aggressive disease). An extension of this conventional radiomics is the application of "deep learning," wherein convolutional neural networks can be used to detect the most informative regions and features without human intervention. A further extension of radiomics involves automatically segmenting informative subregions ("habitats") within tumors, which can be linked to underlying tumor pathophysiology. The goal of the radiomics enterprise is to provide informed decision support for the practice of precision oncology.
Collapse
Affiliation(s)
- Sandy Napel
- Department of RadiologyStanford UniversityStanfordCalifornia
| | - Wei Mu
- Department of Cancer PhysiologyH. Lee Moffitt Cancer CenterTampaFlorida
| | | | - Hugo J. W. L. Aerts
- Dana‐Farber Cancer Institute, Department of Radiology, Brigham and Women’s HospitalHarvard Medical SchoolBostonMassachusetts
| | - Robert J. Gillies
- Department of Cancer PhysiologyH. Lee Moffitt Cancer CenterTampaFlorida
| |
Collapse
|
47
|
Iv M, Zhou M, Shpanskaya K, Perreault S, Wang Z, Tranvinh E, Lanzman B, Vajapeyam S, Vitanza NA, Fisher PG, Cho YJ, Laughlin S, Ramaswamy V, Taylor MD, Cheshier SH, Grant GA, Young Poussaint T, Gevaert O, Yeom KW. MR Imaging-Based Radiomic Signatures of Distinct Molecular Subgroups of Medulloblastoma. AJNR Am J Neuroradiol 2018; 40:154-161. [PMID: 30523141 DOI: 10.3174/ajnr.a5899] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/06/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND PURPOSE Distinct molecular subgroups of pediatric medulloblastoma confer important differences in prognosis and therapy. Currently, tissue sampling is the only method to obtain information for classification. Our goal was to develop and validate radiomic and machine learning approaches for predicting molecular subgroups of pediatric medulloblastoma. MATERIALS AND METHODS In this multi-institutional retrospective study, we evaluated MR imaging datasets of 109 pediatric patients with medulloblastoma from 3 children's hospitals from January 2001 to January 2014. A computational framework was developed to extract MR imaging-based radiomic features from tumor segmentations, and we tested 2 predictive models: a double 10-fold cross-validation using a combined dataset consisting of all 3 patient cohorts and a 3-dataset cross-validation, in which training was performed on 2 cohorts and testing was performed on the third independent cohort. We used the Wilcoxon rank sum test for feature selection with assessment of area under the receiver operating characteristic curve to evaluate model performance. RESULTS Of 590 MR imaging-derived radiomic features, including intensity-based histograms, tumor edge-sharpness, Gabor features, and local area integral invariant features, extracted from imaging-derived tumor segmentations, tumor edge-sharpness was most useful for predicting sonic hedgehog and group 4 tumors. Receiver operating characteristic analysis revealed superior performance of the double 10-fold cross-validation model for predicting sonic hedgehog, group 3, and group 4 tumors when using combined T1- and T2-weighted images (area under the curve = 0.79, 0.70, and 0.83, respectively). With the independent 3-dataset cross-validation strategy, select radiomic features were predictive of sonic hedgehog (area under the curve = 0.70-0.73) and group 4 (area under the curve = 0.76-0.80) medulloblastoma. CONCLUSIONS This study provides proof-of-concept results for the application of radiomic and machine learning approaches to a multi-institutional dataset for the prediction of medulloblastoma subgroups.
Collapse
Affiliation(s)
- M Iv
- From the Department of Radiology (M.I., M.Z., K.S., E.T., B.L., K.W.Y.)
| | - M Zhou
- From the Department of Radiology (M.I., M.Z., K.S., E.T., B.L., K.W.Y.).,Stanford Center for Biomedical Informatics (M.Z., O.G., Z.W.)
| | - K Shpanskaya
- From the Department of Radiology (M.I., M.Z., K.S., E.T., B.L., K.W.Y.)
| | - S Perreault
- Department of Pediatrics (S.P.), Pediatric Neurology, Centre Hospitalier Universitaire Sainte Justine, University of Montréal, Montreal, Quebec, Canada
| | - Z Wang
- Stanford Center for Biomedical Informatics (M.Z., O.G., Z.W.)
| | - E Tranvinh
- From the Department of Radiology (M.I., M.Z., K.S., E.T., B.L., K.W.Y.)
| | - B Lanzman
- From the Department of Radiology (M.I., M.Z., K.S., E.T., B.L., K.W.Y.)
| | - S Vajapeyam
- Department of Radiology (S.V., T.Y.P.), Boston Children's Hospital, Harvard University, Boston, Massachusetts
| | - N A Vitanza
- Department Pediatrics Hematology-Oncology (N.A.V.), Seattle Children's Hospital, University of Washington, Seattle, Washington
| | - P G Fisher
- Department of Pediatrics (P.G.F.), Pediatric Neurology
| | - Y J Cho
- Department of Pediatrics (Y.J.C.), Pediatric Neurology, Oregon Health & Science University, Portland, Oregon
| | - S Laughlin
- Departments of Radiology, Neuro-Oncology, and Neurosurgery (S.L., V.R., M.D.T.), Hospital for Sick Children, Toronto, Ontario, Canada
| | - V Ramaswamy
- Departments of Radiology, Neuro-Oncology, and Neurosurgery (S.L., V.R., M.D.T.), Hospital for Sick Children, Toronto, Ontario, Canada
| | - M D Taylor
- Departments of Radiology, Neuro-Oncology, and Neurosurgery (S.L., V.R., M.D.T.), Hospital for Sick Children, Toronto, Ontario, Canada
| | - S H Cheshier
- Department of Neurosurgery (S.H.C.), Pediatric Neurosurgery, University of Utah, Salt Lake City, Utah
| | - G A Grant
- Department of Neurosurgery (G.A.G.), Pediatric Neurosurgery, Lucile Packard Children's Hospital, Stanford University, Palo Alto, California
| | - T Young Poussaint
- Department of Radiology (S.V., T.Y.P.), Boston Children's Hospital, Harvard University, Boston, Massachusetts
| | - O Gevaert
- Stanford Center for Biomedical Informatics (M.Z., O.G., Z.W.)
| | - K W Yeom
- From the Department of Radiology (M.I., M.Z., K.S., E.T., B.L., K.W.Y.) .,Department of Radiology (K.W.Y.), Artificial Intelligence in Medicine and Imaging, Stanford University, Stanford, California
| |
Collapse
|
48
|
Khan IN, Ullah N, Hussein D, Saini KS. Current and emerging biomarkers in tumors of the central nervous system: Possible diagnostic, prognostic and therapeutic applications. Semin Cancer Biol 2018; 52:85-102. [PMID: 28774835 DOI: 10.1016/j.semcancer.2017.07.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 07/25/2017] [Indexed: 12/15/2022]
Affiliation(s)
- Ishaq N Khan
- PK-Neurooncology Research Group, Institute of Basic Medical Sciences, Khyber Medical University, Peshawar 25100, Pakistan; Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Najeeb Ullah
- Department of Anatomy, Institute of Basic Medical Sciences, Khyber Medical University, Peshawar 25100, Pakistan.
| | - Deema Hussein
- Neurooncology Translational Group, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Kulvinder S Saini
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Department of Biotechnology, Eternal University, Baru Sahib, Himachal Pradesh 173101, India.
| |
Collapse
|
49
|
Treiber JM, Steed TC, Brandel MG, Patel KS, Dale AM, Carter BS, Chen CC. Molecular physiology of contrast enhancement in glioblastomas: An analysis of The Cancer Imaging Archive (TCIA). J Clin Neurosci 2018; 55:86-92. [PMID: 29934058 DOI: 10.1016/j.jocn.2018.06.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 03/06/2018] [Accepted: 06/04/2018] [Indexed: 12/23/2022]
Abstract
The physiologic processes underlying MRI contrast enhancement in glioblastoma patients remain poorly understood. MRIs of 148 glioblastoma subjects from The Cancer Imaging Archive were segmented using Iterative Probabilistic Voxel Labeling (IPVL). Three aspects of contrast enhancement (CE) were parametrized: the mean intensity of all CE voxels (CEi), the intensity heterogeneity in CE (CEh), and volumetric ratio of CE to necrosis (CEr). Associations between these parameters and patterns of gene expression were analyzed using DAVID functional enrichment analysis. Glioma CpG island methylator phenotype (G-CIMP) glioblastomas were poorly enhancing. Otherwise, no differences in CE parameters were found between proneural, neural, mesenchymal, and classical glioblastomas. High CEi was associated with expression of genes that mediate inflammatory responses. High CEh was associated with increased expression of genes that regulate remodeling of extracellular matrix (ECM) and endothelial permeability. High CEr was associated with increased expression of genes that mediate cellular response to stressful metabolic states, including hypoxia and starvation. Our results indicate that CE in glioblastoma is associated with distinct biological processes involved in inflammatory response and tissue hypoxia. Integrative analysis of these CE parameters may yield meaningful information pertaining to the biologic state of glioblastomas and guide future therapeutic paradigms.
Collapse
Affiliation(s)
- Jeffrey M Treiber
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA.
| | - Tyler C Steed
- Department of Neurosurgery, Emory University, Atlanta, GA, USA.
| | - Michael G Brandel
- Department of Neurosurgery, University of California, San Diego, La Jolla, CA, USA.
| | - Kunal S Patel
- Department of Neurosurgery, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA.
| | - Anders M Dale
- Multimodal Imaging Laboratory, University of California San Diego, La Jolla, CA, USA; Department of Radiology, University of California San Diego, La Jolla, CA, USA.
| | - Bob S Carter
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA.
| | - Clark C Chen
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
50
|
Arita H, Kinoshita M, Kawaguchi A, Takahashi M, Narita Y, Terakawa Y, Tsuyuguchi N, Okita Y, Nonaka M, Moriuchi S, Takagaki M, Fujimoto Y, Fukai J, Izumoto S, Ishibashi K, Nakajima Y, Shofuda T, Kanematsu D, Yoshioka E, Kodama Y, Mano M, Mori K, Ichimura K, Kanemura Y. Lesion location implemented magnetic resonance imaging radiomics for predicting IDH and TERT promoter mutations in grade II/III gliomas. Sci Rep 2018; 8:11773. [PMID: 30082856 PMCID: PMC6078954 DOI: 10.1038/s41598-018-30273-4] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 07/27/2018] [Indexed: 11/30/2022] Open
Abstract
Molecular biological characterization of tumors has become a pivotal procedure for glioma patient care. The aim of this study is to build conventional MRI-based radiomics model to predict genetic alterations within grade II/III gliomas attempting to implement lesion location information in the model to improve diagnostic accuracy. One-hundred and ninety-nine grade II/III gliomas patients were enrolled. Three molecular subtypes were identified: IDH1/2-mutant, IDH1/2-mutant with TERT promoter mutation, and IDH-wild type. A total of 109 radiomics features from 169 MRI datasets and location information from 199 datasets were extracted. Prediction modeling for genetic alteration was trained via LASSO regression for 111 datasets and validated by the remaining 58 datasets. IDH mutation was detected with an accuracy of 0.82 for the training set and 0.83 for the validation set without lesion location information. Diagnostic accuracy improved to 0.85 for the training set and 0.87 for the validation set when lesion location information was implemented. Diagnostic accuracy for predicting 3 molecular subtypes of grade II/III gliomas was 0.74 for the training set and 0.56 for the validation set with lesion location information implemented. Conventional MRI-based radiomics is one of the most promising strategies that may lead to a non-invasive diagnostic technique for molecular characterization of grade II/III gliomas.
Collapse
Affiliation(s)
- Hideyuki Arita
- Department of Neurosurgery, Osaka International Cancer Institute, Osaka, 541-8567, Japan
- Kansai Molecular Diagnosis Network for CNS Tumors, Osaka, 540-0006, Japan
- Department of Neurosurgery, Osaka University Graduate School of Medicine, Suita, 565-0871, Japan
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, 104-0045, Japan
| | - Manabu Kinoshita
- Department of Neurosurgery, Osaka International Cancer Institute, Osaka, 541-8567, Japan.
- Kansai Molecular Diagnosis Network for CNS Tumors, Osaka, 540-0006, Japan.
| | - Atsushi Kawaguchi
- Center for Comprehensive Community Medicine, Center for Comprehensive Community Medicine, Faculty of Medicine, Saga University, Saga, 849-8501, Japan
| | - Masamichi Takahashi
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Yoshitaka Narita
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Yuzo Terakawa
- Kansai Molecular Diagnosis Network for CNS Tumors, Osaka, 540-0006, Japan
- Department of Neurosurgery, Osaka City University Graduate School of Medicine, Osaka, 545-0051, Japan
| | - Naohiro Tsuyuguchi
- Kansai Molecular Diagnosis Network for CNS Tumors, Osaka, 540-0006, Japan
- Department of Neurosurgery, Osaka City University Graduate School of Medicine, Osaka, 545-0051, Japan
- Department of Neurosurgery, Kindai University Faculty of Medicine, Sayama, 589-8511, Japan
| | - Yoshiko Okita
- Kansai Molecular Diagnosis Network for CNS Tumors, Osaka, 540-0006, Japan
- Department of Neurosurgery, National Hospital Organization Osaka National Hospital, Osaka, 540-0006, Japan
| | - Masahiro Nonaka
- Kansai Molecular Diagnosis Network for CNS Tumors, Osaka, 540-0006, Japan
- Department of Neurosurgery, National Hospital Organization Osaka National Hospital, Osaka, 540-0006, Japan
- Department of Neurosurgery, Kansai Medical University, Hirakata, 573-1191, Japan
| | - Shusuke Moriuchi
- Kansai Molecular Diagnosis Network for CNS Tumors, Osaka, 540-0006, Japan
- Department of Neurosurgery, National Hospital Organization Osaka National Hospital, Osaka, 540-0006, Japan
- Department of Neurosurgery, Rinku General Medical Center, Izumisano, 598-8577, Japan
| | - Masatoshi Takagaki
- Kansai Molecular Diagnosis Network for CNS Tumors, Osaka, 540-0006, Japan
- Department of Neurosurgery, Kawachi General Hospital, Higashi-Osaka, 578-0954, Japan
| | - Yasunori Fujimoto
- Kansai Molecular Diagnosis Network for CNS Tumors, Osaka, 540-0006, Japan
- Department of Neurosurgery, Osaka University Graduate School of Medicine, Suita, 565-0871, Japan
| | - Junya Fukai
- Kansai Molecular Diagnosis Network for CNS Tumors, Osaka, 540-0006, Japan
- Department of Neurosurgery, Wakayama Medical University, Wakayama, 641-8509, Japan
| | - Shuichi Izumoto
- Kansai Molecular Diagnosis Network for CNS Tumors, Osaka, 540-0006, Japan
- Department of Neurosurgery, Kindai University Faculty of Medicine, Sayama, 589-8511, Japan
| | - Kenichi Ishibashi
- Kansai Molecular Diagnosis Network for CNS Tumors, Osaka, 540-0006, Japan
- Department of Neurosurgery, Osaka City General Hospital, Osaka, 534-0021, Japan
| | - Yoshikazu Nakajima
- Kansai Molecular Diagnosis Network for CNS Tumors, Osaka, 540-0006, Japan
- Department of Neurosurgery, Sakai City Medical Center, Sakai, 593-8304, Japan
| | - Tomoko Shofuda
- Kansai Molecular Diagnosis Network for CNS Tumors, Osaka, 540-0006, Japan
- Division of Stem Cell Research, Institute for Clinical Research, Osaka National Hospital, National Hospital Organization, Osaka, 540-0006, Japan
| | - Daisuke Kanematsu
- Kansai Molecular Diagnosis Network for CNS Tumors, Osaka, 540-0006, Japan
- Division of Regenerative Medicine, Institute for Clinical Research, Osaka National Hospital, National Hospital Organization, Osaka, 540-0006, Japan
| | - Ema Yoshioka
- Kansai Molecular Diagnosis Network for CNS Tumors, Osaka, 540-0006, Japan
- Division of Regenerative Medicine, Institute for Clinical Research, Osaka National Hospital, National Hospital Organization, Osaka, 540-0006, Japan
| | - Yoshinori Kodama
- Kansai Molecular Diagnosis Network for CNS Tumors, Osaka, 540-0006, Japan
- Department of Pathology and Applied Neurobiology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
- Department of Central Laboratory and Surgical Pathology, Osaka National Hospital, National Hospital Organization, Osaka, 540-0006, Japan
| | - Masayuki Mano
- Kansai Molecular Diagnosis Network for CNS Tumors, Osaka, 540-0006, Japan
- Department of Central Laboratory and Surgical Pathology, Osaka National Hospital, National Hospital Organization, Osaka, 540-0006, Japan
| | - Kanji Mori
- Kansai Molecular Diagnosis Network for CNS Tumors, Osaka, 540-0006, Japan
- Department of Neurosurgery, Kansai Rosai Hospital, Amagasaki, 660-8511, Japan
| | - Koichi Ichimura
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, 104-0045, Japan
| | - Yonehiro Kanemura
- Kansai Molecular Diagnosis Network for CNS Tumors, Osaka, 540-0006, Japan
- Division of Regenerative Medicine, Institute for Clinical Research, Osaka National Hospital, National Hospital Organization, Osaka, 540-0006, Japan
| |
Collapse
|