7
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Structural and functional radiomics for lung cancer

      review-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Introduction

          Lung cancer ranks second in new cancer cases and first in cancer-related deaths worldwide. Precision medicine is working on altering treatment approaches and improving outcomes in this patient population. Radiological images are a powerful non-invasive tool in the screening and diagnosis of early-stage lung cancer, treatment strategy support, prognosis assessment, and follow-up for advanced-stage lung cancer. Recently, radiological features have evolved from solely semantic to include (handcrafted and deep) radiomic features. Radiomics entails the extraction and analysis of quantitative features from medical images using mathematical and machine learning methods to explore possible ties with biology and clinical outcomes.

          Methods

          Here, we outline the latest applications of both structural and functional radiomics in detection, diagnosis, and prediction of pathology, gene mutation, treatment strategy, follow-up, treatment response evaluation, and prognosis in the field of lung cancer.

          Conclusion

          The major drawbacks of radiomics are the lack of large datasets with high-quality data, standardization of methodology, the black-box nature of deep learning, and reproducibility. The prerequisite for the clinical implementation of radiomics is that these limitations are addressed. Future directions include a safer and more efficient model-training mode, merge multi-modality images, and combined multi-discipline or multi-omics to form “Medomics.”

          Related collections

          Most cited references122

          • Record: found
          • Abstract: found
          • Article: not found

          Cancer statistics, 2020

          Each year, the American Cancer Society estimates the numbers of new cancer cases and deaths that will occur in the United States and compiles the most recent data on population-based cancer occurrence. Incidence data (through 2016) were collected by the Surveillance, Epidemiology, and End Results Program; the National Program of Cancer Registries; and the North American Association of Central Cancer Registries. Mortality data (through 2017) were collected by the National Center for Health Statistics. In 2020, 1,806,590 new cancer cases and 606,520 cancer deaths are projected to occur in the United States. The cancer death rate rose until 1991, then fell continuously through 2017, resulting in an overall decline of 29% that translates into an estimated 2.9 million fewer cancer deaths than would have occurred if peak rates had persisted. This progress is driven by long-term declines in death rates for the 4 leading cancers (lung, colorectal, breast, prostate); however, over the past decade (2008-2017), reductions slowed for female breast and colorectal cancers, and halted for prostate cancer. In contrast, declines accelerated for lung cancer, from 3% annually during 2008 through 2013 to 5% during 2013 through 2017 in men and from 2% to almost 4% in women, spurring the largest ever single-year drop in overall cancer mortality of 2.2% from 2016 to 2017. Yet lung cancer still caused more deaths in 2017 than breast, prostate, colorectal, and brain cancers combined. Recent mortality declines were also dramatic for melanoma of the skin in the wake of US Food and Drug Administration approval of new therapies for metastatic disease, escalating to 7% annually during 2013 through 2017 from 1% during 2006 through 2010 in men and women aged 50 to 64 years and from 2% to 3% in those aged 20 to 49 years; annual declines of 5% to 6% in individuals aged 65 years and older are particularly striking because rates in this age group were increasing prior to 2013. It is also notable that long-term rapid increases in liver cancer mortality have attenuated in women and stabilized in men. In summary, slowing momentum for some cancers amenable to early detection is juxtaposed with notable gains for other common cancers.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Deep learning.

            Deep learning allows computational models that are composed of multiple processing layers to learn representations of data with multiple levels of abstraction. These methods have dramatically improved the state-of-the-art in speech recognition, visual object recognition, object detection and many other domains such as drug discovery and genomics. Deep learning discovers intricate structure in large data sets by using the backpropagation algorithm to indicate how a machine should change its internal parameters that are used to compute the representation in each layer from the representation in the previous layer. Deep convolutional nets have brought about breakthroughs in processing images, video, speech and audio, whereas recurrent nets have shone light on sequential data such as text and speech.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1).

              Assessment of the change in tumour burden is an important feature of the clinical evaluation of cancer therapeutics: both tumour shrinkage (objective response) and disease progression are useful endpoints in clinical trials. Since RECIST was published in 2000, many investigators, cooperative groups, industry and government authorities have adopted these criteria in the assessment of treatment outcomes. However, a number of questions and issues have arisen which have led to the development of a revised RECIST guideline (version 1.1). Evidence for changes, summarised in separate papers in this special issue, has come from assessment of a large data warehouse (>6500 patients), simulation studies and literature reviews. HIGHLIGHTS OF REVISED RECIST 1.1: Major changes include: Number of lesions to be assessed: based on evidence from numerous trial databases merged into a data warehouse for analysis purposes, the number of lesions required to assess tumour burden for response determination has been reduced from a maximum of 10 to a maximum of five total (and from five to two per organ, maximum). Assessment of pathological lymph nodes is now incorporated: nodes with a short axis of 15 mm are considered measurable and assessable as target lesions. The short axis measurement should be included in the sum of lesions in calculation of tumour response. Nodes that shrink to <10mm short axis are considered normal. Confirmation of response is required for trials with response primary endpoint but is no longer required in randomised studies since the control arm serves as appropriate means of interpretation of data. Disease progression is clarified in several aspects: in addition to the previous definition of progression in target disease of 20% increase in sum, a 5mm absolute increase is now required as well to guard against over calling PD when the total sum is very small. Furthermore, there is guidance offered on what constitutes 'unequivocal progression' of non-measurable/non-target disease, a source of confusion in the original RECIST guideline. Finally, a section on detection of new lesions, including the interpretation of FDG-PET scan assessment is included. Imaging guidance: the revised RECIST includes a new imaging appendix with updated recommendations on the optimal anatomical assessment of lesions. A key question considered by the RECIST Working Group in developing RECIST 1.1 was whether it was appropriate to move from anatomic unidimensional assessment of tumour burden to either volumetric anatomical assessment or to functional assessment with PET or MRI. It was concluded that, at present, there is not sufficient standardisation or evidence to abandon anatomical assessment of tumour burden. The only exception to this is in the use of FDG-PET imaging as an adjunct to determination of progression. As is detailed in the final paper in this special issue, the use of these promising newer approaches requires appropriate clinical validation studies.
                Bookmark

                Author and article information

                Contributors
                g.wu@maastrichtuniversity.nl
                Journal
                Eur J Nucl Med Mol Imaging
                Eur J Nucl Med Mol Imaging
                European Journal of Nuclear Medicine and Molecular Imaging
                Springer Berlin Heidelberg (Berlin/Heidelberg )
                1619-7070
                1619-7089
                11 March 2021
                11 March 2021
                2021
                : 48
                : 12
                : 3961-3974
                Affiliations
                [1 ]GRID grid.412966.e, ISNI 0000 0004 0480 1382, The D-Lab, Department of Precision Medicine, GROW–School for Oncology, , Maastricht University Medical Centre+, ; 6229 Maastricht, The Netherlands
                [2 ]GRID grid.33199.31, ISNI 0000 0004 0368 7223, Department of Radiology, Union Hospital, Tongji Medical College, , Huazhong University of Science and Technology, ; Wuhan, China
                [3 ]GRID grid.412839.5, ISNI 0000 0004 1771 3250, Hubei Province Key Laboratory of Molecular Imaging, ; Wuhan, China
                [4 ]GRID grid.411831.e, ISNI 0000 0004 0398 1027, Department of Diagnostic Radiology, Faculty of Applied Medical Sciences, , Jazan University, ; Jazan, Saudi Arabia
                [5 ]GRID grid.412966.e, ISNI 0000 0004 0480 1382, Department of Radiology and Nuclear Medicine, GROW – School for Oncology, , Maastricht University Medical Centre+, ; Maastricht, The Netherlands
                [6 ]GRID grid.4861.b, ISNI 0000 0001 0805 7253, Division of Nuclear Medicine and Oncological Imaging, Department of Medical Physics, , Hospital Center Universitaire De Liege, ; Liege, Belgium
                [7 ]GRID grid.412301.5, ISNI 0000 0000 8653 1507, Department of Nuclear Medicine and Comprehensive Diagnostic Center Aachen (CDCA), , University Hospital RWTH Aachen University, ; Aachen, Germany
                [8 ]GRID grid.284723.8, ISNI 0000 0000 8877 7471, Department of Medical Imaging Center, Nanfang Hospital, , Southern Medical University, ; Guangzhou, China
                Article
                5242
                10.1007/s00259-021-05242-1
                8484174
                33693966
                b34faa80-b055-425c-82e8-e6721e73fcca
                © The Author(s) 2021

                Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

                History
                : 23 November 2020
                : 3 February 2021
                Funding
                Funded by: FundRef http://dx.doi.org/10.13039/501100000781, European Research Council;
                Award ID: ERC-ADG-2015, n° 694812
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100010663, H2020 European Research Council;
                Award ID: ImmunoSABR - n° 733008;PREDICT - ITN - n° 766276;CHAIMELEON - n° 952172
                Award Recipient :
                Categories
                Review Article
                Custom metadata
                © Springer-Verlag GmbH Germany, part of Springer Nature 2021

                Radiology & Imaging
                lung cancer,radiomics,artificial intelligence,medical imaging
                Radiology & Imaging
                lung cancer, radiomics, artificial intelligence, medical imaging

                Comments

                Comment on this article