51
|
Patel M, Zhan J, Natarajan K, Flintham R, Davies N, Sanghera P, Grist J, Duddalwar V, Peet A, Sawlani V. Machine learning-based radiomic evaluation of treatment response prediction in glioblastoma. Clin Radiol 2021; 76:628.e17-628.e27. [PMID: 33941364 DOI: 10.1016/j.crad.2021.03.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 03/29/2021] [Indexed: 11/16/2022]
Abstract
AIM To investigate machine learning based models combining clinical, radiomic, and molecular information to distinguish between early true progression (tPD) and pseudoprogression (psPD) in patients with glioblastoma. MATERIALS AND METHODS A retrospective analysis was undertaken of 76 patients (46 tPD, 30 psPD) with early enhancing disease following chemoradiotherapy for glioblastoma. Outcome was determined on follow-up until 6 months post-chemoradiotherapy. Models comprised clinical characteristics, O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation status, and 307 quantitative imaging features extracted from enhancing disease and perilesional oedema masks on early post-chemoradiotherapy contrast-enhanced T1-weighted imaging, T2-weighted imaging (T2WI), and apparent diffusion coefficient (ADC) maps. Feature selection was performed within bootstrapped cross-validated recursive feature elimination with a random forest algorithm. Naive Bayes five-fold cross-validation was used to validate the final model. RESULTS Top selected features included age, MGMT promoter methylation status, two shape-based features from the enhancing disease mask, three radiomic features from the enhancing disease mask on ADC, and one radiomic feature from the perilesional oedema mask on T2WI. The final model had an area under the receiver operating characteristics curve (AUC) of 0.80, sensitivity 78.2%, specificity 66.7%, and accuracy of 73.7%. CONCLUSION Incorporating a machine learning-based approach using quantitative radiomic features from standard-of-care magnetic resonance imaging (MRI), in combination with clinical characteristics and MGMT promoter methylation status has a complementary effect and improves model performance for early prediction of glioblastoma treatment response.
Collapse
Affiliation(s)
- M Patel
- University of Birmingham, Birmingham, UK; Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - J Zhan
- Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK; The Affiliated Hospital of Qingdao University, Qingdao Shi, Shandong Sheng, China
| | - K Natarajan
- Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - R Flintham
- Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - N Davies
- Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - P Sanghera
- University of Birmingham, Birmingham, UK; Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - J Grist
- University of Birmingham, Birmingham, UK
| | - V Duddalwar
- Departments of Radiology, Urology and Biomedical Engineering, University of Southern California, USA
| | - A Peet
- University of Birmingham, Birmingham, UK; Birmingham Children's Hospital, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - V Sawlani
- University of Birmingham, Birmingham, UK; Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.
| |
Collapse
|
52
|
Cheung HMC, Rubin D. Challenges and opportunities for artificial intelligence in oncological imaging. Clin Radiol 2021; 76:728-736. [PMID: 33902889 DOI: 10.1016/j.crad.2021.03.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/15/2021] [Indexed: 02/08/2023]
Abstract
Imaging plays a key role in oncology, including the diagnosis and detection of cancer, determining clinical management, assessing treatment response, and complications of treatment or disease. The current use of clinical oncology is predominantly qualitative in nature with some relatively crude size-based measurements of tumours for assessment of disease progression or treatment response; however, it is increasingly understood that there may be significantly more information about oncological disease that can be obtained from imaging that is not currently utilized. Artificial intelligence (AI) has the potential to harness quantitative techniques to improve oncological imaging. These may include improving the efficiency or accuracy of traditional roles of imaging such as diagnosis or detection. These may also include new roles for imaging such as risk-stratifying patients for different types of therapy or determining biological tumour subtypes. This review article outlines several major areas in oncological imaging where there may be opportunities for AI technology. These include (1) screening and detection of cancer, (2) diagnosis and risk stratification, (3) tumour segmentation, (4) precision oncology, and (5) predicting prognosis and assessing treatment response. This review will also address some of the potential barriers to AI research in oncological imaging.
Collapse
Affiliation(s)
- H M C Cheung
- Department of Medical Imaging, Sunnybrook Health Sciences Centre, University of Toronto, Canada
| | - D Rubin
- Department of Radiology, Stanford University, CA, USA.
| |
Collapse
|
53
|
Hajjo R, Sabbah DA, Bardaweel SK, Tropsha A. Identification of Tumor-Specific MRI Biomarkers Using Machine Learning (ML). Diagnostics (Basel) 2021; 11:742. [PMID: 33919342 PMCID: PMC8143297 DOI: 10.3390/diagnostics11050742] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 02/06/2023] Open
Abstract
The identification of reliable and non-invasive oncology biomarkers remains a main priority in healthcare. There are only a few biomarkers that have been approved as diagnostic for cancer. The most frequently used cancer biomarkers are derived from either biological materials or imaging data. Most cancer biomarkers suffer from a lack of high specificity. However, the latest advancements in machine learning (ML) and artificial intelligence (AI) have enabled the identification of highly predictive, disease-specific biomarkers. Such biomarkers can be used to diagnose cancer patients, to predict cancer prognosis, or even to predict treatment efficacy. Herein, we provide a summary of the current status of developing and applying Magnetic resonance imaging (MRI) biomarkers in cancer care. We focus on all aspects of MRI biomarkers, starting from MRI data collection, preprocessing and machine learning methods, and ending with summarizing the types of existing biomarkers and their clinical applications in different cancer types.
Collapse
Affiliation(s)
- Rima Hajjo
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, P.O. Box 130, Amman 11733, Jordan;
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, The University of North Carlina at Chapel Hill, Chapel Hill, NC 27599, USA;
- National Center for Epidemics and Communicable Disease Control, Amman 11118, Jordan
| | - Dima A. Sabbah
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, P.O. Box 130, Amman 11733, Jordan;
| | - Sanaa K. Bardaweel
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Jordan, Amman 11942, Jordan;
| | - Alexander Tropsha
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, The University of North Carlina at Chapel Hill, Chapel Hill, NC 27599, USA;
| |
Collapse
|
54
|
Chuntova P, Chow F, Watchmaker PB, Galvez M, Heimberger AB, Newell EW, Diaz A, DePinho RA, Li MO, Wherry EJ, Mitchell D, Terabe M, Wainwright DA, Berzofsky JA, Herold-Mende C, Heath JR, Lim M, Margolin KA, Chiocca EA, Kasahara N, Ellingson BM, Brown CE, Chen Y, Fecci PE, Reardon DA, Dunn GP, Liau LM, Costello JF, Wick W, Cloughesy T, Timmer WC, Wen PY, Prins RM, Platten M, Okada H. Unique challenges for glioblastoma immunotherapy-discussions across neuro-oncology and non-neuro-oncology experts in cancer immunology. Meeting Report from the 2019 SNO Immuno-Oncology Think Tank. Neuro Oncol 2021; 23:356-375. [PMID: 33367885 PMCID: PMC7992879 DOI: 10.1093/neuonc/noaa277] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cancer immunotherapy has made remarkable advances with over 50 separate Food and Drug Administration (FDA) approvals as first- or second-line indications since 2015. These include immune checkpoint blocking antibodies, chimeric antigen receptor-transduced T cells, and bispecific T-cell-engaging antibodies. While multiple cancer types now benefit from these immunotherapies, notable exceptions thus far include brain tumors, such as glioblastoma. As such, it seems critical to gain a better understanding of unique mechanistic challenges underlying the resistance of malignant gliomas to immunotherapy, as well as to acquire insights into the development of future strategies. An Immuno-Oncology Think Tank Meeting was held during the 2019 Annual Society for Neuro-Oncology Scientific Conference. Discussants in the fields of neuro-oncology, neurosurgery, neuro-imaging, medical oncology, and cancer immunology participated in the meeting. Sessions focused on topics such as the tumor microenvironment, myeloid cells, T-cell dysfunction, cellular engineering, and translational aspects that are critical and unique challenges inherent with primary brain tumors. In this review, we summarize the discussions and the key messages from the meeting, which may potentially serve as a basis for advancing the field of immune neuro-oncology in a collaborative manner.
Collapse
Affiliation(s)
- Pavlina Chuntova
- Department of Neurological Surgery, UCSF, San Francisco, California
| | - Frances Chow
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | | | - Mildred Galvez
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, Los Angeles, California
| | - Amy B Heimberger
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Evan W Newell
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Aaron Diaz
- Department of Neurological Surgery, UCSF, San Francisco, California
| | - Ronald A DePinho
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ming O Li
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - E John Wherry
- Department of Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Duane Mitchell
- Department of Neurosurgery, University of Florida College of Medicine, Gainesville, Florida
| | - Masaki Terabe
- Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Derek A Wainwright
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Jay A Berzofsky
- Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | | | | | - Michael Lim
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kim A Margolin
- Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, California
| | - E Antonio Chiocca
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts
| | | | - Benjamin M Ellingson
- Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Christine E Brown
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, California
| | - Yvonne Chen
- Department of Microbiology, Immunology & Molecular Genetics, UCLA, Los Angeles, California
| | - Peter E Fecci
- Department of Neurosurgery, Duke University School of Medicine, Durham, North Carolina
| | - David A Reardon
- Department of Medicine/Medical Oncology, Harvard Medical School, Boston, Massachusetts
| | - Gavin P Dunn
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Linda M Liau
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, California
| | | | - Wolfgang Wick
- Department of Neurology, University Hospital Heidelberg, Heidelberg, Germany
| | - Timothy Cloughesy
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - William C Timmer
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, Maryland
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Robert M Prins
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, Los Angeles, California.,Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - Michael Platten
- Department of Neurology, Medical Faculty Mannheim, MCTN, University of Heidelberg, Mannheim, Germany.,DKTK CCU Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hideho Okada
- Department of Neurological Surgery, UCSF, San Francisco, California.,Parker Institute for Cancer Immunotherapy, San Francisco, California
| |
Collapse
|
55
|
Rogachev AD, Alemasov NA, Ivanisenko VA, Ivanisenko NV, Gaisler EV, Oleshko OS, Cheresiz SV, Mishinov SV, Stupak VV, Pokrovsky AG. Correlation of Metabolic Profiles of Plasma and Cerebrospinal Fluid of High-Grade Glioma Patients. Metabolites 2021; 11:metabo11030133. [PMID: 33669010 PMCID: PMC7996604 DOI: 10.3390/metabo11030133] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/09/2021] [Accepted: 02/22/2021] [Indexed: 01/17/2023] Open
Abstract
This work compares the metabolic profiles of plasma and the cerebrospinal fluid (CSF) of the patients with high-grade (III and IV) gliomas and the conditionally healthy controls using the wide-range targeted screening of low molecular metabolites by HPLC-MS/MS. The obtained data were analyzed using robust linear regression with Huber’s M-estimates, and a number of metabolites with correlated content in plasma and CSF was identified. The statistical analysis shows a significant correlation of metabolite content in plasma and CSF samples for the majority of metabolites. Several metabolites were shown to have high correlation in the control samples, but not in the glioma patients. This can be due to the specific metabolic processes in the glioma patients or to the damaged integrity of blood-brain barrier. The results of our study may be useful for the understanding of molecular mechanisms underlying the development of gliomas, as well as for the search of potential biomarkers for the minimally invasive diagnostic procedures of gliomas.
Collapse
Affiliation(s)
- Artem D. Rogachev
- V. Zelman Institute for Medicine and Psychology, Novosibirsk State University, Pirogov str., 2, 630090 Novosibirsk, Russia; (E.V.G.); (O.S.O.); (S.V.C.); (A.G.P.)
- N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, acad. Lavrentiev ave., 9, 630090 Novosibirsk, Russia
- Correspondence: ; Tel.: +7-(383)-330-97-47
| | - Nikolay A. Alemasov
- Institute of Cytology and Genetics of Siberian Branch of Russian Academy of Sciences, acad. Lavrentiev ave., 10, 630090 Novosibirsk, Russia; (N.A.A.); (V.A.I.); (N.V.I.)
| | - Vladimir A. Ivanisenko
- Institute of Cytology and Genetics of Siberian Branch of Russian Academy of Sciences, acad. Lavrentiev ave., 10, 630090 Novosibirsk, Russia; (N.A.A.); (V.A.I.); (N.V.I.)
| | - Nikita V. Ivanisenko
- Institute of Cytology and Genetics of Siberian Branch of Russian Academy of Sciences, acad. Lavrentiev ave., 10, 630090 Novosibirsk, Russia; (N.A.A.); (V.A.I.); (N.V.I.)
| | - Evgeniy V. Gaisler
- V. Zelman Institute for Medicine and Psychology, Novosibirsk State University, Pirogov str., 2, 630090 Novosibirsk, Russia; (E.V.G.); (O.S.O.); (S.V.C.); (A.G.P.)
| | - Olga S. Oleshko
- V. Zelman Institute for Medicine and Psychology, Novosibirsk State University, Pirogov str., 2, 630090 Novosibirsk, Russia; (E.V.G.); (O.S.O.); (S.V.C.); (A.G.P.)
| | - Sergey V. Cheresiz
- V. Zelman Institute for Medicine and Psychology, Novosibirsk State University, Pirogov str., 2, 630090 Novosibirsk, Russia; (E.V.G.); (O.S.O.); (S.V.C.); (A.G.P.)
| | - Sergey V. Mishinov
- FSBI “Novosibirsk Research Institute of Traumatology and Orthopedics Named after Ya. L. Tsiviyan”, Frunze str., 17, 630091 Novosibirsk, Russia; (S.V.M.); (V.V.S.)
| | - Vyacheslav V. Stupak
- FSBI “Novosibirsk Research Institute of Traumatology and Orthopedics Named after Ya. L. Tsiviyan”, Frunze str., 17, 630091 Novosibirsk, Russia; (S.V.M.); (V.V.S.)
| | - Andrey G. Pokrovsky
- V. Zelman Institute for Medicine and Psychology, Novosibirsk State University, Pirogov str., 2, 630090 Novosibirsk, Russia; (E.V.G.); (O.S.O.); (S.V.C.); (A.G.P.)
| |
Collapse
|
56
|
Vitale A, Villa R, Ugga L, Romeo V, Stanzione A, Cuocolo R. Artificial intelligence applied to neuroimaging data in Parkinsonian syndromes: Actuality and expectations. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2021; 18:1753-1773. [PMID: 33757209 DOI: 10.3934/mbe.2021091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Idiopathic Parkinson's Disease (iPD) is a common motor neurodegenerative disorder. It affects more frequently the elderly population, causing a significant emotional burden both for the patient and caregivers, due to the disease-related onset of motor and cognitive disabilities. iPD's clinical hallmark is the onset of cardinal motor symptoms such as bradykinesia, rest tremor, rigidity, and postural instability. However, these symptoms appear when the neurodegenerative process is already in an advanced stage. Furthermore, the greatest challenge is to distinguish iPD from other similar neurodegenerative disorders, "atypical parkinsonisms", such as Multisystem Atrophy, Progressive Supranuclear Palsy and Cortical Basal Degeneration, since they share many phenotypic manifestations, especially in the early stages. The diagnosis of these neurodegenerative motor disorders is essentially clinical. Consequently, the diagnostic accuracy mainly depends on the professional knowledge and experience of the physician. Recent advances in artificial intelligence have made it possible to analyze the large amount of clinical and instrumental information in the medical field. The application machine learning algorithms to the analysis of neuroimaging data appear to be a promising tool for identifying microstructural alterations related to the pathological process in order to explain the onset of symptoms and the spread of the neurodegenerative process. In this context, the search for quantitative biomarkers capable of identifying parkinsonian patients in the prodromal phases of the disease, of correctly distinguishing them from atypical parkinsonisms and of predicting clinical evolution and response to therapy represent the main goal of most current clinical research studies. Our aim was to review the recent literature and describe the current knowledge about the contribution given by machine learning applications to research and clinical management of parkinsonian syndromes.
Collapse
Affiliation(s)
- Annalisa Vitale
- Department of Advanced Biomedical Sciences, University of Naples "Federico Ⅱ", Via S. Pansini 5, 80131-Naples, Italy
| | - Rossella Villa
- Department of Advanced Biomedical Sciences, University of Naples "Federico Ⅱ", Via S. Pansini 5, 80131-Naples, Italy
| | - Lorenzo Ugga
- Department of Advanced Biomedical Sciences, University of Naples "Federico Ⅱ", Via S. Pansini 5, 80131-Naples, Italy
| | - Valeria Romeo
- Department of Advanced Biomedical Sciences, University of Naples "Federico Ⅱ", Via S. Pansini 5, 80131-Naples, Italy
| | - Arnaldo Stanzione
- Department of Advanced Biomedical Sciences, University of Naples "Federico Ⅱ", Via S. Pansini 5, 80131-Naples, Italy
| | - Renato Cuocolo
- Department of Clinical Medicine and Surgery, University of Naples "Federico Ⅱ", Via S. Pansini 5, 80131-Naples, Italy
| |
Collapse
|
57
|
Sheng C, Chen Z, Lei J, Zhu J, Song S. Development and Multi-Data Set Verification of an RNA Binding Protein Signature for Prognosis Prediction in Glioma. Front Med (Lausanne) 2021; 8:637803. [PMID: 33634155 PMCID: PMC7900154 DOI: 10.3389/fmed.2021.637803] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/11/2021] [Indexed: 12/13/2022] Open
Abstract
Objective: Increasing evidence emphasizes the clinical implications of RNA binding proteins (RBPs) in cancers. This study aimed to develop a RBP signature for predicting prognosis in glioma. Methods: Two glioma datasets as training (n = 693) and validation (n = 325) sets were retrieved from the CGGA database. In the training set, univariate Cox regression analysis was conducted to screen prognosis-related RBPs based on differentially expressed RBPs between WHO grade II and IV. A ten-RBP signature was then established. The predictive efficacy was evaluated by ROCs. The applicability was verified in the validation set. The pathways involving the risk scores were analyzed by ssGSEA. scRNA-seq was utilized for evaluating their expression in different glioma cell types. Moreover, their expression was externally validated between glioma and control samples. Results: Based on 39 prognosis-related RBPs, a ten RBP signature was constructed. High risk score distinctly indicated a poorer prognosis than low risk score. AUCs were separately 0.838 and 0.822 in the training and validation sets, suggesting its well performance for prognosis prediction. Following adjustment of other clinicopathological characteristics, the signature was an independent risk factor. Various cancer-related pathways were significantly activated in samples with high risk score. The scRNA-seq identified that risk RBPs were mainly expressed in glioma malignant cells. Their high expression was also found in glioma than control samples. Conclusion: This study developed a novel RBP signature for robustly predicting prognosis of glioma following multi-data set verification. These RBPs may affect the progression of glioma.
Collapse
Affiliation(s)
- Chunpeng Sheng
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhihua Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianwei Lei
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianming Zhu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shuxin Song
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
58
|
Booth TC, Thompson G, Bulbeck H, Boele F, Buckley C, Cardoso J, Dos Santos Canas L, Jenkinson D, Ashkan K, Kreindler J, Huskens N, Luis A, McBain C, Mills SJ, Modat M, Morley N, Murphy C, Ourselin S, Pennington M, Powell J, Summers D, Waldman AD, Watts C, Williams M, Grant R, Jenkinson MD. A Position Statement on the Utility of Interval Imaging in Standard of Care Brain Tumour Management: Defining the Evidence Gap and Opportunities for Future Research. Front Oncol 2021; 11:620070. [PMID: 33634034 PMCID: PMC7900557 DOI: 10.3389/fonc.2021.620070] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 01/06/2021] [Indexed: 12/19/2022] Open
Abstract
OBJECTIV E To summarise current evidence for the utility of interval imaging in monitoring disease in adult brain tumours, and to develop a position for future evidence gathering while incorporating the application of data science and health economics. METHODS Experts in 'interval imaging' (imaging at pre-planned time-points to assess tumour status); data science; health economics, trial management of adult brain tumours, and patient representatives convened in London, UK. The current evidence on the use of interval imaging for monitoring brain tumours was reviewed. To improve the evidence that interval imaging has a role in disease management, we discussed specific themes of data science, health economics, statistical considerations, patient and carer perspectives, and multi-centre study design. Suggestions for future studies aimed at filling knowledge gaps were discussed. RESULTS Meningioma and glioma were identified as priorities for interval imaging utility analysis. The "monitoring biomarkers" most commonly used in adult brain tumour patients were standard structural MRI features. Interval imaging was commonly scheduled to provide reported imaging prior to planned, regular clinic visits. There is limited evidence relating interval imaging in the absence of clinical deterioration to management change that alters morbidity, mortality, quality of life, or resource use. Progression-free survival is confounded as an outcome measure when using structural MRI in glioma. Uncertainty from imaging causes distress for some patients and their caregivers, while for others it provides an important indicator of disease activity. Any study design that changes imaging regimens should consider the potential for influencing current or planned therapeutic trials, ensure that opportunity costs are measured, and capture indirect benefits and added value. CONCLUSION Evidence for the value, and therefore utility, of regular interval imaging is currently lacking. Ongoing collaborative efforts will improve trial design and generate the evidence to optimise monitoring imaging biomarkers in standard of care brain tumour management.
Collapse
Affiliation(s)
- Thomas C. Booth
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, United Kingdom
- Department of Neuroradiology, King’s College Hospital NHS Foundation Trust, London, United Kingdom
| | - Gerard Thompson
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Florien Boele
- Leeds Institute of Medical Research at St James’s, St James’s University Hospital, Leeds, United Kingdom
- Faculty of Medicine and Health, Leeds Institute of Health Sciences, University of Leeds, Leeds, United Kingdom
| | | | - Jorge Cardoso
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, United Kingdom
| | - Liane Dos Santos Canas
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, United Kingdom
| | | | - Keyoumars Ashkan
- Department of Neurosurgery, King’s College Hospital NHS Foundation Trust, London, United Kingdom
| | | | - Nicky Huskens
- The Tessa Jowell Brain Cancer Mission, London, United Kingdom
| | - Aysha Luis
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, United Kingdom
- Lysholm Department of Neuroradiology, National Hospital for Neurology and Neurosurgery, London, United Kingdom
| | - Catherine McBain
- Department of Oncology, Christie Hospital NHS Foundation Trust, Manchester, United Kingdom
| | - Samantha J. Mills
- Department of Neuroradiology, The Walton Centre NHS Foundation Trust, Liverpool, United Kingdom
| | - Marc Modat
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, United Kingdom
| | - Nick Morley
- Department of Radiology, Wales Research and Diagnostic PET Imaging Centre, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Caroline Murphy
- King’s College Trials Unit, King’s College London, London, United Kingdom
| | - Sebastian Ourselin
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London, United Kingdom
| | - Mark Pennington
- King’s Health Economics, King’s College London, London, United Kingdom
| | - James Powell
- Department of Oncology, Velindre Cancer Centre, Cardiff, United Kingdom
| | - David Summers
- Department of Neuroradiology, Western General Hospital, Edinburgh, United Kingdom
| | - Adam D. Waldman
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Colin Watts
- Birmingham Brain Cancer Program, University of Birmingham, Birmingham, United Kingdom
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Matthew Williams
- Department of Neuro-oncology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Robin Grant
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Michael D. Jenkinson
- Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
- Department of Neurosurgery, The Walton Centre NHS Foundation Trust, Liverpool, United Kingdom
| |
Collapse
|
59
|
GLIMPSE: a glioblastoma prognostication model using ensemble learning-a surveillance, epidemiology, and end results study. Health Inf Sci Syst 2021; 9:5. [PMID: 33489102 DOI: 10.1007/s13755-020-00134-4] [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: 07/22/2020] [Accepted: 11/04/2020] [Indexed: 12/21/2022] Open
Abstract
Purpose Glioblastoma is one of the most common and aggressive brain tumors in the world with a poor prognosis. A glioblastoma prognostication model has the potential to improve the cancer's standard of care. No other paper has looked at using ensemble learning with a population database to predict multiple binary glioblastoma survival outcomes. Methods We utilized ensemble learning to design, build, and test a prognostication system for glioblastoma for short-, intermediate- and long-term survival, based on various clinical features. We used the population database SEER which covers 17 different registries. The most important prognostic features were identified and used as a clinical feature set. The statistical feature set was determined using Random Forests. The accuracy, sensitivity, specificity, area under the receiver operating characteristic (AUROC), positive predictive value (PPV), and negative predictive value (NPV) were reported. Results Statistically-determined feature sets had the best performance. All the top models for short, intermediate, and long-term survival were random forests. With regards to short-term survival, top model had metrics AUROC = 0.937, accuracy = 86%, specificity = 88%, sensitivity = 85%, NPV = 85%, and PPV = 87%. For long-term survival, the top model had AUROC = 0.893, accuracy = 81%, specificity = 79%, sensitivity = 83%, NPV = 82%, and PPV = 79%. The top intermediate-term survival prediction had AUROC ≥ 0.780 and the other metrics were at least 70%. Conclusions Our ensemble models were high-performing and achieved AUROCs as high as 0.94, highlighting the importance of balancing, using ensemble techniques and statistical feature selection. Our models can potentially be used by clinicians after external validation.
Collapse
|
60
|
Favorable role of IDH1/2 mutations aided with MGMT promoter gene methylation in the outcome of patients with malignant glioma. Future Sci OA 2020; 7:FSO663. [PMID: 33552543 PMCID: PMC7849969 DOI: 10.2144/fsoa-2020-0057] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Aim The implications of molecular biomarkers IDH1/2 mutations and MGMT gene promoter methylation were evaluated for prognostic outcome of glioma patients. Materials & methods Glioma cases were analyzed for IDH1/2 mutations and MGMT promoter methylation by DNA sequencing and methylation-specific PCR, respectively. Results Mutations found in IDH1/2 genes totaled 63.4% (N = 40) wherein IDH1 mutations were significantly associated with oligidendrioglioma (p = 0.005) and astrocytoma (p = 0.0002). IDH1 mutants presented more, 60.5% in MGMT promoter-methylated cases (p = 0.03). IDH1 mutant cases had better survival for glioblastoma and oligodendrioglioma (log-rank p = 0.01). Multivariate analysis confirmed better survival in MGMT methylation carriers (hazard ratio [HR]: 0.59; p = 0.031). Combination of both biomarkers showed better prognosis on temozolomide (p < 0.05). Conclusion IDH1/2 mutations proved independent prognostic factors in glioma and associated with MGMT methylation for better survival.
Collapse
|
61
|
Machine Learning Model to Predict Pseudoprogression Versus Progression in Glioblastoma Using MRI: A Multi-Institutional Study (KROG 18-07). Cancers (Basel) 2020; 12:cancers12092706. [PMID: 32967367 PMCID: PMC7564954 DOI: 10.3390/cancers12092706] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 01/28/2023] Open
Abstract
Simple Summary Even after the introduction of a standard regimen consisting of concurrent chemoradiotherapy and adjuvant temozolomide, patients with glioblastoma multiforme mostly experience disease progression. Clinicians often encounter a situation where they need to distinguish progressive disease from pseudoprogression after treatment. We tried to investigate the feasibility of machine learning algorithm to distinguish pseudoprogression from progressive disease. In multi-institutional dataset, the developed machine learning model showed an acceptable performance. This algorithm involving MRI data and clinical features could help making decision during patients’ disease course. For the practical use, we calibrated the machine learning model to offer the probability of pseudoprogression to clinicians, then we constructed the web-based user interface to access the model. Abstract Some patients with glioblastoma show a worsening presentation in imaging after concurrent chemoradiation, even when they receive gross total resection. Previously, we showed the feasibility of a machine learning model to predict pseudoprogression (PsPD) versus progressive disease (PD) in glioblastoma patients. The previous model was based on the dataset from two institutions (termed as the Seoul National University Hospital (SNUH) dataset, N = 78). To test this model in a larger dataset, we collected cases from multiple institutions that raised the problem of PsPD vs. PD diagnosis in clinics (Korean Radiation Oncology Group (KROG) dataset, N = 104). The dataset was composed of brain MR images and clinical information. We tested the previous model in the KROG dataset; however, that model showed limited performance. After hyperparameter optimization, we developed a deep learning model based on the whole dataset (N = 182). The 10-fold cross validation revealed that the micro-average area under the precision-recall curve (AUPRC) was 0.86. The calibration model was constructed to estimate the interpretable probability directly from the model output. After calibration, the final model offers clinical probability in a web-user interface.
Collapse
|
62
|
Diffusion- and Perfusion-Weighted Magnetic Resonance Imaging Methods in Nonenhancing Gliomas. World Neurosurg 2020; 141:123-130. [DOI: 10.1016/j.wneu.2020.05.278] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 05/29/2020] [Accepted: 05/30/2020] [Indexed: 12/21/2022]
|
63
|
McGrath H, Li P, Dorent R, Bradford R, Saeed S, Bisdas S, Ourselin S, Shapey J, Vercauteren T. Manual segmentation versus semi-automated segmentation for quantifying vestibular schwannoma volume on MRI. Int J Comput Assist Radiol Surg 2020; 15:1445-1455. [PMID: 32676869 PMCID: PMC7419453 DOI: 10.1007/s11548-020-02222-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 06/20/2020] [Indexed: 12/21/2022]
Abstract
Purpose Management of vestibular schwannoma (VS) is based on tumour size as observed on T1 MRI scans with contrast agent injection. The current clinical practice is to measure the diameter of the tumour in its largest dimension. It has been shown that volumetric measurement is more accurate and more reliable as a measure of VS size. The reference approach to achieve such volumetry is to manually segment the tumour, which is a time intensive task. We suggest that semi-automated segmentation may be a clinically applicable solution to this problem and that it could replace linear measurements as the clinical standard. Methods Using high-quality software available for academic purposes, we ran a comparative study of manual versus semi-automated segmentation of VS on MRI with 5 clinicians and scientists. We gathered both quantitative and qualitative data to compare the two approaches; including segmentation time, segmentation effort and segmentation accuracy. Results We found that the selected semi-automated segmentation approach is significantly faster (167 s vs 479 s, \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$p<0.001$$\end{document}p<0.001), less temporally and physically demanding and has approximately equal performance when compared with manual segmentation, with some improvements in accuracy. There were some limitations, including algorithmic unpredictability and error, which produced more frustration and increased mental effort in comparison with manual segmentation. Conclusion We suggest that semi-automated segmentation could be applied clinically for volumetric measurement of VS on MRI. In future, the generic software could be refined for use specifically for VS segmentation, thereby improving accuracy. Electronic supplementary material The online version of this article (10.1007/s11548-020-02222-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hari McGrath
- GKT School of Medical Education, King's College London, London, UK.
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK.
| | - Peichao Li
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Reuben Dorent
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Robert Bradford
- Queen Square Radiosurgery Centre (Gamma Knife), National Hospital for Neurology and Neurosurgery, London, UK
- Department of Neurosurgery, National Hospital for Neurology and Neurosurgery, London, UK
| | - Shakeel Saeed
- Department of Neurosurgery, National Hospital for Neurology and Neurosurgery, London, UK
- The Ear Institute, UCL, London, UK
- The Royal National Throat Nose and Ear Hospital, London, UK
| | - Sotirios Bisdas
- Neuroradiology Department, National Hospital for Neurology and Neurosurgery, London, UK
| | - Sebastien Ourselin
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Jonathan Shapey
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
- Department of Neurosurgery, National Hospital for Neurology and Neurosurgery, London, UK
- Wellcome/EPSRC Centre for Interventional and Surgical Sciences, UCL, London, UK
| | - Tom Vercauteren
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| |
Collapse
|