Skip to main content
Erschienen in: European Radiology 8/2021

Open Access 25.01.2021 | Imaging Informatics and Artificial Intelligence

Incorporating radiomics into clinical trials: expert consensus endorsed by the European Society of Radiology on considerations for data-driven compared to biologically driven quantitative biomarkers

verfasst von: Laure Fournier, Lena Costaridou, Luc Bidaut, Nicolas Michoux, Frederic E. Lecouvet, Lioe-Fee de Geus-Oei, Ronald Boellaard, Daniela E. Oprea-Lager, Nancy A Obuchowski, Anna Caroli, Wolfgang G. Kunz, Edwin H. Oei, James P. B. O’Connor, Marius E. Mayerhoefer, Manuela Franca, Angel Alberich-Bayarri, Christophe M. Deroose, Christian Loewe, Rashindra Manniesing, Caroline Caramella, Egesta Lopci, Nathalie Lassau, Anders Persson, Rik Achten, Karen Rosendahl, Olivier Clement, Elmar Kotter, Xavier Golay, Marion Smits, Marc Dewey, Daniel C. Sullivan, Aad van der Lugt, Nandita M. deSouza, European Society of Radiology

Erschienen in: European Radiology | Ausgabe 8/2021

Abstract

Existing quantitative imaging biomarkers (QIBs) are associated with known biological tissue characteristics and follow a well-understood path of technical, biological and clinical validation before incorporation into clinical trials. In radiomics, novel data-driven processes extract numerous visually imperceptible statistical features from the imaging data with no a priori assumptions on their correlation with biological processes. The selection of relevant features (radiomic signature) and incorporation into clinical trials therefore requires additional considerations to ensure meaningful imaging endpoints. Also, the number of radiomic features tested means that power calculations would result in sample sizes impossible to achieve within clinical trials. This article examines how the process of standardising and validating data-driven imaging biomarkers differs from those based on biological associations. Radiomic signatures are best developed initially on datasets that represent diversity of acquisition protocols as well as diversity of disease and of normal findings, rather than within clinical trials with standardised and optimised protocols as this would risk the selection of radiomic features being linked to the imaging process rather than the pathology. Normalisation through discretisation and feature harmonisation are essential pre-processing steps. Biological correlation may be performed after the technical and clinical validity of a radiomic signature is established, but is not mandatory. Feature selection may be part of discovery within a radiomics-specific trial or represent exploratory endpoints within an established trial; a previously validated radiomic signature may even be used as a primary/secondary endpoint, particularly if associations are demonstrated with specific biological processes and pathways being targeted within clinical trials.

Key Points

• Data-driven processes like radiomics risk false discoveries due to high-dimensionality of the dataset compared to sample size, making adequate diversity of the data, cross-validation and external validation essential to mitigate the risks of spurious associations and overfitting.
• Use of radiomic signatures within clinical trials requires multistep standardisation of image acquisition, image analysis and data mining processes.
• Biological correlation may be established after clinical validation but is not mandatory.
Hinweise
The original online version of this article was revised: Firstly, “endorsed by the European Society of Radiology” was missing in the article title. Secondly, the institutional author “European Society of Radiology” was missing in the author line, including the related affiliation 34. Thirdly, the following sentence was missing in the Acknowledgements: This paper was endorsed by the ESR Executive Council in December 2020.
A correction to this article is available online at https://​doi.​org/​10.​1007/​s00330-021-07721-3.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ADC
Apparent diffusion coefficient
CE
Conformite Europeenne
CNN
Convolutional neural networks
CT
Computerised tomography
DL
Deep learning
DR
Deep radiomics
EGFR
Epidermal growth factor receptor
FDG
Fluorodeoxyglucose
IBSI
Image biomarker standardisation initiative
MeSH
Medical Subject Headings
MRI
Magnetic resonance imaging
PET
Positron emission tomography
QA
Quality assurance
QC
Quality control
QIBs
Quantitative imaging biomarkers
SPECT
Single photon emission computed tomography
SUV
Standardised uptake value
VOI
Volume of interest

Introduction

Quantitative imaging biomarkers (QIBs) are associated with tissue characteristics that are altered by disease and its treatment. Necrosis decreases tissue cellularity and increases water content manifesting as an increase in T2 [1], a reduction in glucose uptake [2] and an increase in elasticity [3]. Perfusion imaging detects and characterises hypervascular lesions such as cancers, or monitors the effect of anti-angiogenic drugs [4, 5]. Implementation of QIBs into clinical trials follows a well-defined path from discovery, through a process of technical and biological validation, to implementation and clinical validation. A roadmap defining the process was published as a consensus statement from multiple stakeholders [6]. Despite this, QIBs have been slow to be adopted as trial endpoints because of the relative complexity of imaging protocols and variability of the quantified output under differing conditions (e.g. hardware, software, protocol and observer variability) [7].
Recently, a new approach to derive imaging biomarkers has been advocated through the concept of radiomics [8, 9]. This data-driven framework ‘discovers’ quantitative information within images by extracting high-dimensional data (‘features’) beyond that visually perceptible, using computational statistics (often based on machine learning algorithms) to predict or establish association with a meaningful clinical endpoint [10, 11]. Technical and clinical performance of the ‘radiomic signature’ (specific combination of mathematically derived features) determines its appropriateness. If considered necessary, a link to a biological process is explored a posteriori [12]. Radiomic signatures have been associated with outcome or response [13], and may be used together with clinical, histological and genomic metrics as part of a nomogram of features [14]. The exponential rise in publications involving data-driven biomarkers has not been accompanied by a mechanism-based understanding of their nature but focuses on their ability to classify disease and patient outcome (Fig. 1). Radiomics has been used for detecting cancer [15], cancer staging [16], performing classifications [17], assessing response to chemotherapy [18], radiation therapies [1922], immunotherapy [2326] and predicting/prognosing survival [27].
A major disadvantage of a non-mechanistic data-driven approach is that random chance associations may occur. Most studies look at the associations between a large number of features extracted from discretised images and prognosis/response/outcome in an inadequate number of samples. For biomarker profiles that rely on statistical rather than biological associations, generalisation and scalability to multicentre trials requires more than a simple standardisation process. Also, their validation pathway needs to incorporate measures that may differ substantially from traditionally accepted methods. This article prepared by imaging experts from the European Society of Radiology EIBALL (European Imaging Biomarker ALLiance) and the EORTC (European Organisation for Research and Treatment of Cancer) Imaging Group with representatives from QIBA (Quantitative Imaging Biomarkers Alliance) examines how the process of standardising and validating data-driven imaging biomarkers differs from those based on biological associations, and what measures need to be considered when implementing them into clinical trials and, eventually, into clinical routine. Structured discussions were conducted via teleconferencing and written communications.

Standardising the radiomics process for clinical trials

Radiomics analyses rely on image acquisition, image analysis and computational statistics [28], so standardisation of these domains is mandatory prior to their validation (Table 1). As radiomics analyses have been applied to CT [2931], MRI [3236], nuclear medicine using FDG-PET [3742] and other tracers [43, 44], and ultrasound [45], image acquisition standardisation needs to consider modality, scanner and scan protocol. Standardisation of image analysis needs to consider software (consistency of technical implementation) and subjectivity (human interaction). Standardisation of computational statistics needs to consider adequacy, performance and requirements for validation of algorithms and models (Fig. 2).
Table 1
Comparison of standardisation steps for biologically driven and data-driven biomarkers (QA, quality assurance; QC, quality Control; VOI, volume of interest)
Steps
Biologically driven quantitative biomarkers
Data-driven quantitative biomarkers
Image acquisition
• Standardised protocols (single and multicentre)
• QA/QC process across instruments, sites
• Stability of measurement monitored with phantom studies; may be strengthened by human subject test-retest
• Non-standardised protocols in discovery phase followed by standardised protocols within trials
• QA/QC process across instruments, sites
• Stability of measurement requires human subject test-retest
VOI delineation
• Can be manual or semi-automated
• Can be machine-learnt
• Deep learning available but infrequently used
• Can be manual or semi-automated
• Can be machine-learnt
• Can be derived from fully convolutional neural networks
Data analysis
• Commercial or academic software applicable to datasets regardless of their source
• Algorithms used are specific to image datasets and may require adaptation and standardisation for individual situations or new datasets*
Biomarker extraction
• Follows standard formula that describes the biological feature (e.g. tissue density, perfusion, diffusion, standardised uptake of radiotracers related to a biological process/receptor status)
• Algorithm-based mathematical feature extraction not directly linked to a biological process, followed by selection of feature combination that best separate disease from no disease, good from poor outcome (e.g. shape features such as diameter, sphericity; histogram-derived features such as median, skewness, entropy; texture features such as contrast, homogeneity, Haralick variance)
Biomarker interpretation
• Directly linked to biological process
• Indirect associations with biological process assumed
Image acquisition and normalisation
An element of diversity of acquisition protocols or machines is advantageous at the discovery phase of data-driven biomarkers so that the identified radiomic signatures used in clinical trials are robust enough across a range of platforms [46]. Datasets utilised for radiomic signature development must be representative of the disease and capture the variability and severity for which they will be used. Within a clinical trials framework, as with previously published recommendations and guidelines [6, 4749], an optimised tightly controlled standardised imaging protocol ensures image quality (low level of noise, artifact-free, spatial resolution) and stability over time, with known intra- and inter-site reproducibility that does not exceed the expected level of change associated with the trial intervention [50]. Phantom studies are limited for quality control of high-dimensionality information [51] because a suitable phantom would need to exhibit high-dimensionality in a realistic setting and cover the requirements of each type of feature.
Basic methods of image normalisation include pixel size resampling by filtering [52] and/or resampling (rescaling) values with respect to global or local mean and standard deviation of reference image/tissue, or by adjusting the histograms [53]. Normalisation methods affect reproducibility of image features [54, 55]. For second-order statistics features, reduction of matrix dimension post-normalisation is needed. This is achieved by discretisation (quantisation, grey-level resampling, histogram re-binning) and reduces noise from clustered intensity values. Choice of the absolute (fixed bin size) or the relative (fixed bin number) method significantly affects the values of texture features and requires optimisation depending on the clinical task at hand [5658]. Shape features (area, centroid, perimeter, roundness, Feret’s diameter) are less sensitive to differences in intensity values. Both types of features remain dependent on the spatial resolution of the image. Numerical harmonisation of features as an alternative to standardisation of image acquisition and pre-processing is based on transformation of variable feature distributions to a common batch-effect free reference space, to deal with varying imaging conditions [59, 60]
The Image Biomarker Standardization Initiative (IBSI) [61] offers a common reference of definitions and benchmarking of radiomic features and provides recommendations for comprehensive reporting of image acquisition parameters and pre-processing methods.
Image analysis—segmentation
As with biologically driven biomarkers, manual region of interest delineation introduces inter- and intra-observer variability because of variation in border perception. Observer training and working to protocol assists in this regard. Semi-automated segmentation methods, e.g. region-growing or level set active contour models [62] and deep learning methods [63], are more reproducible [64], but they are dependent on their training set, which may introduce other errors. Quantitative verification metrics [65], such as Dice coefficient, and Hausdorff distance metrics, help determine segmentation reproducibility. Images that require alignment for different time series data, parametric maps and modalities should evaluate deviations in locations (distance) of pairs of homologous landmark points, especially important for non-rigid image registration [66, 67].
Image analysis—feature extraction
‘Hand-crafted’ radiomics extracts predefined human-engineered features from the volume-of-interest (VOI) [17]. These include shape characteristics, intensity histogram metrics and texture parameters (local binary patterns, grey-level co-occurrence, run-length, zone-length and neighbourhood different matrices, auto-regressive model, Markov random fields, Riesz wavelets, S-transform, fractals) which require specific assumptions in their computation, so that software implementations on different platforms (even if all are IBSI compliant) and between different versions of the same software can lead to different results [68]. Recommendations on calculating and reporting radiomic features have been proposed, and both mathematical equations and pre-processing applied should be reported. The information and framework provided through IBSI [61] should also be followed as much as possible to ensure the quality and relevance of the post-processing (denoising, resampling, enhancement, spatial alignment correction, segmentation and feature extraction). Other descriptive (radiologist-scored), functional (SUV, ADC, Ktrans) or clinical parameters may be added to the radiomic signature if pertinent.
Computational statistics—feature selection
Several tools are described [6972]. To identify relevant, non-redundant and stable features with which to build models, three categories of technique are employed. Filter methods (ANOVA, correlation, RELIEF [73]) rely on a criterion function, have low computational cost and are less prone to overfitting, by separating selection from model building; however, they are more unstable to different datasets. Wrapper methods (forward selection, backward elimination, stepwise selection) incorporate a specific machine learning algorithm to eliminate features but have increased computational cost and high probability of overfitting, since model training uses feature combinations that include common features. Embedded methods (LASSO, RIDGE regression) embed features successively and penalise the coefficients of a model that contribute to overfitting at each iteration. They represent a trade-off between filter and wrapper methods.
Computational statistics—classifier/model
After dimension reduction, selected features are investigated for their association with clinical outcome using tools such as univariable or multivariable logistic regression, decision tree, random forest, support vector machine, neural networks, all described extensively in previous publications [6568] and used for QIBs and radiomic analyses [24]. Classifiers are differentiated depending on the nature of the clinical outcome, i.e. discrete (mainly binary) or continuous [74, 75]. No tool has proved universally superior and most require a compromise between complexity of tuning versus interpretability of results.
Computational statistics—deep radiomics (DR)
A recent evolution has been the integration of radiomics with deep learning (DL) [7678]. ‘Discovery Radiomics’ automatically extracts deep features relevant to a given query (e.g. diagnosis, prognosis) from the data, and the resulting trained model can be applied to complete datasets, avoiding the error-prone segmentation step. As DL can include multiple data types, relevant information in electronic patient records can be exploited.

Validating the radiomics output

Technical validation
Following identification of a radiomics signature associated with disease/outcome, two fully independent datasets are needed, one for training and cross-validation (internal validation), and at least one other to test the final model and confirm generalisability and performance (external validation). Both training and testing datasets should be of sufficient uniform quality (data balancing) and representative for the patient population for which the radiomics model is intended. An adequate sample (size and diversity) is essential for the training and validation datasets, with respect to the number and type of features (‘signature’) considered. Testing the model with a dataset containing a different prevalence of cases and/or a high degree of imbalance may result in overoptimistic conclusions. Feature selection avoids over-parameterised models, reduces dimensionality of the feature space (data dimension reduction) and ensures that only a small and stable subset of original features relevant to the task are retained. A strategy to cross-validate the structure of the model requires careful considerations regarding sample size, accuracy estimation and the choice of the validation method (hold-out, k-fold cross-validation, bootstrap). Grid searches pose the danger of overfitting, leading to overoptimistic model performance that is not reproduced on other datasets or in clinical practice. Finally, repeatability and reproducibility of the signature in a multicentre context (affected by imaging apparatus, acquisition protocols and analysis methods) is a crucial step in technical validation [7981]. As with QIBs, radiomics models should be tested with cross-institutional clinical training and testing datasets to guarantee generalisability to representative patient populations.
Biological validation
Biological correlation with liquid/tissue biopsies may be performed after the technical and clinical validity of a radiomic signature is established but is not mandatory. A radiomic signature that is related to survival outcomes may potentially reflect a tissue phenotype associated with a specific biology. Biological validation reduces the likelihood that radiomic features are selected by statistical chance or may be attributed to the nature of the data sample used for model development. It also offers the opportunity to reduce the number of selected features.
Clinical validation
The process by which the clinical utility of a single quantitative feature, or multiple features embedded in a statistical model is demonstrated, allowing improvement of health outcomes (improved diagnosis or therapeutic management of a disease or individual patient) is being addressed slowly for radiomics. Following initial ‘discovery’, new and independent datasets are required to replicate the performance of the identified model and validate it clinically. Performance metrics, e.g. sensitivity and specificity, should be evaluated ideally in prospective trials, or prospectively in the clinic using routinely obtained clinical data (real-life conditions) in order to avoid bias. Table 2 lists some exemplar studies and their clinical use. Broadly speaking, standard recommendations for clinical validation and clinical utility assessment of any QIB should be followed and applied.
Table 2
Exemplar radiomics signature studies and their clinical use
Radiomic analysis
Radiomic feature (process)
Modality
Tissue types investigated
Decision-making role
Second-order statistics
Textural (Haralick, Gabor)
CT [2931]
MRI [2426]
PET/CT [3742]
Lung, breast, brain, liver, prostate, head and neck, lymph node, cervix
• Prognostic
• Predictive
• Response
• Survival
• EGFR expression
• p53 mutation status
Higher-order statistics
Filter grids extract repetitive or non-repetitive patterns
Wavelets
CT [8287]
MRI [8890]
PET/CT [91, 92]
Lung, oesophagus, brain, pancreas, breast, head and neck
• Diagnostic
• Prognostic
• Predictive
• Response
• Survival
• Surgical resection margins
Laplacian transforms (bandpass filters)
CT [93, 94]
MRI [9597]
PET/CT [92]
Brain, lung, rectum, cervix, kidney
Prognostic
Response
Minkowski functions (patterns of voxels with intensity above threshold)
Fractal dimensions (patterns imposed on image and number of grid elements containing voxels of a specified value is computed)
Delta radiomics
Change in radiomic features
PET/CT [98, 99]
Lung
Response
Dynamic radiomic studies
Pharmacokinetic radiomic features
PET/CT [100]
Lung
Response, data highly correlated to data from static studies

Biological correlates of radiomic features

Images provide an averaged macroscopic view (with large partial volume effects, both in space and time) of the geometry and/or function of the tissue. Radiomic features are statistical descriptors characterising the macroscopic visual aspect of images and only indirectly relate to the microscopic histological characteristics of the imaged tissue. Such features are then used as a statistical/phenomenological description of the outcome, and not embedded into an actual biological/physical model of this outcome that would unambiguously establish causality between features and outcome.
Radiomic information on visually imperceptible phenotypic characteristics such as intensity, shape, size and texture distinguish benign and malignant tumours, likely reflecting different cellular morphology [101]. In cervix cancer, radiomic features of low-volume tumours with radiomic profiles similar to high-volume tumours had a worse prognosis implying a more aggressive phenotype at an earlier stage [36]. In a lung cancer study, texture entropy and cluster features, as well as voxel intensity variance features, were associated with the immune system, the p53 pathway, pathways involved in cell cycle regulation [102] and for predicting EGFR mutation status [103]. Nevertheless, why specific features are associated with specific pathways remains unexplored and the relationship between radiomic signature and cell morphology, density, distribution pattern, alignment and organelle composition need further elucidation.
Although it is possible to extract mathematically hundreds or thousands of radiomic features from digital images, most studies to date suggest that less than 20 are indicative of unfavourable biology, and these largely relate to shape and textural uniformity. 2D shape features indicate more rapidly progressive disease with reduced overall survival in glioblastoma multiforme [104]. Shape and textural features from CT scans of lung cancer have been shown to predict unfavourable biology (nodal and distant metastases respectively) [105]. In prostate cancer, Gabor textural features (defining spatial frequency patterns within the image) were predictive of Gleason grade on MRI. As gland lumen shape features relate to Gleason grade, discriminability of Gabor features is a likely consequence of variations in gland shape and morphology at the tissue level [106]. In future, prospective selection of a handful of relevant features should become possible to interrogate specific biological processes and pathways being manipulated within clinical trials so that it may be possible for the clinical question to drive the choice of biomarker usage and analysis. However, understanding the biological basis for a biomarker to facilitate its acceptance into clinical practice is not the primary objective of a data-driven process such as radiomics. It may well be that reliable modelling of the outcome with a relatively high and clinically acceptable performance means that biological validation would not be a primary concern [107].

Limitations of data-driven processes

When defining training datasets for radiomic feature extraction and selection in clinical trials, case-control data may be considered but may underrepresent the disease. Enrichment of training datasets with normal and abnormal cases of varying disease severity is mandatory to achieve appropriate balance. Bias in the training datasets limits generalisability. For example, a radiomic signature developed on lung nodules detected on chest x-rays in a population with a high prevalence of tuberculosis and few cancers will overdiagnose tuberculosis in a population with a high prevalence of cancer. Image acquisition bias (cases recognised as disease acquired with a specific protocol or device) where selected features are linked to image acquisition rather than to image content may fail to predict disease when applied to an independent population. Manual VOI segmentation and use of locally developed methodology risks discovery of features that are not generalisable and may be influenced by hardware or software-related factors rather than the disease itself. Diverse but balanced image acquisition conditions in the training dataset should counteract these effects. Though balance and diversity are necessary at the discovery stage, it is crucial to evaluate performance only on populations representative of the natural prevalence.
The radiomic process, which tests combinations of hundreds and thousands of parameters, risks false discovery. Traditional statistical corrections for multiple tests would lead to p values impossible to reach. Strategies to reduce spurious correlations and overfitting include artificially increasing the number of samples by data augmentation (datasets flipped, rotated and deformed to simulate new patients). Cross-validation or bootstrapping are alternative strategies, but an independent dataset to confirm the findings is always required.

Implementation of radiomics in clinical trials

Although the discovery phase requires image acquisition diversity, standardised protocols, pre- and post-processing methods, tools and algorithms for feature extraction are needed for incorporating into clinical trials and facilitated by centralised data analyses and publicly available analysis software (Table 3). To incorporate radiomics in clinical trials, three potential scenarios can be considered. Firstly, where radiomic signature discovery is the objective, a trial should follow the steps described and illustrated (Fig. 2). Secondly, a radiomic ‘exploratory end-point’ may form an ancillary study within an established trial. Here, a two-phase process would involve an initial phase utilising more than two-thirds of the final cohort data (training cohort) to identify the most promising feature(s) and a subsequent phase using the remaining patients (independent cohort) to evaluate the performance of the identified radiomic signature. Thirdly, where a previously validated radiomic signature is used, this could be incorporated into a clinical trial as a primary or secondary endpoint. In this last case, the pathway of a data-driven biomarker does not differ from a QIB.
Table 3
Recommended process for inclusion of data-driven biomarkers into clinical trials
Step
Recommended process for clinical trial inclusion
Image acquisition
Standardised protocol agreed with site with vendor-specific amendments (incl. software version control) to achieve reproducibility of other QIBs within accepted published standards
Image acquisition—normalisation
Raw data saved. Image normalisation predefined
Image analysis—segmentation
If manual or semi-automated, done by centralised/core laboratory by > 1 observer to establish reproducibility. If automated, can be done with CE-marked software with established limits of agreement at local sites
Image analysis—feature extraction
Use of validated features with established error margins, adapted for individual situations. Discard redundant features. Test reproducibility, repeatability within trial setting
Computational statistics—feature and model selection
Based on performance by association with trial endpoint (e.g. response/survival)
Validation
Adequate sample size, test data on samples with similar characteristics, cross-validation strategies, avoid over-fitted models
Biomarker interpretation
Association with positive diagnosis, prognosis or outcome

Summary and future perspective

Data-driven imaging biomarkers provide information beyond that perceived by human readers. Their benefits may be exploited if specific standardisation and validation pathways are defined and the different/additional hurdles compared to more traditional QIBs are addressed. Effects of different types of processing on subsequent extracted feature variability and predictive model performance is an open area of research [13]. Availability of public access patient cohorts with well-documented image datasets is expected to facilitate consensus regarding pre- and post-processing methods and determine utility of radiomics within clinical trials.
While radiomics may eventually encompass all quantitative image-derived information into a common framework, current implementations mostly relate to intensity, shape and textural features within a VOI. In the future, quantitative (or even qualitative) functional information, e.g. derived from PET, SPECT, pharmacokinetic modelling and other parametric imaging modalities, may form part of the radiomic signature, and require a smaller or biologically more meaningful set of parameters. Deep radiomics may also be deployed in trials, and recent studies have already demonstrated the potential of such approaches [108111].
Regardless of definitive biological correlation, once adopted and properly deployed, data-driven biomarkers may be combined with clinical data and other biomarkers (biochemical, genetic, epigenetic, transcription factors, proteins). Such expanded use of radiomics should eventually improve disease characterisation, prognostic stratification and response prediction in clinical trials, ultimately advancing precision medicine.

Acknowledgements

This paper was endorsed by the ESR Executive Council in December 2020.

Compliance with ethical standards

Guarantor

Nandita M deSouza.

Conflict of interest

LF - Speaker fees from Sanofi, Novartis, Jannssen, General Electric.
Congress sponsorship from Guerbet. Industrial grant on radiomics from Invectys, Novartis. Co-investigator in grant with Philips, Ariana Pharma, Evolucare.
CC — personal fees from Pfizer, BMS, MSD, Roche and advisory role for Astra Zeneca.
CMD - Consulting or advisory roles with Ipsen, Novartis, Terumo, and Advanced Accelerator Applications; participation in speakers’ bureaus with Terumo and Advanced Accelerator Applications; and travel, accommodations, or expenses with General Electric and Terumo.
XG: CEO of Gold Standard Phantoms, a company designing calibration devices for quantitative MRI.
All other authors- none.

Statistics and biometry

No complex statistical methods were necessary for this paper.
Not applicable in this perspectives paper.

Ethical approval

Not applicable in this special report.

Methodology

• Special report
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/​.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Radiologie

Kombi-Abonnement

Mit e.Med Radiologie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Radiologie, den Premium-Inhalten der radiologischen Fachzeitschriften, inklusive einer gedruckten Radiologie-Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Santamaria G, Velasco M, Bargallo X, Caparros X, Farrus B, Luis Fernandez P (2010) Radiologic and pathologic findings in breast tumors with high signal intensity on T2-weighted MR images. Radiographics 30:533–548PubMedCrossRef Santamaria G, Velasco M, Bargallo X, Caparros X, Farrus B, Luis Fernandez P (2010) Radiologic and pathologic findings in breast tumors with high signal intensity on T2-weighted MR images. Radiographics 30:533–548PubMedCrossRef
2.
Zurück zum Zitat Parghane RV, Basu S (2020) PET/computed tomography in treatment response assessment in cancer: an overview with emphasis on the evolving role in response evaluation to immunotherapy and radiation therapy. PET Clin 15:101–123PubMedCrossRef Parghane RV, Basu S (2020) PET/computed tomography in treatment response assessment in cancer: an overview with emphasis on the evolving role in response evaluation to immunotherapy and radiation therapy. PET Clin 15:101–123PubMedCrossRef
3.
Zurück zum Zitat Lee SH, Moon WK, Cho N et al (2014) Shear-wave elastographic features of breast cancers: comparison with mechanical elasticity and histopathologic characteristics. Invest Radiol 49:147–155PubMedCrossRef Lee SH, Moon WK, Cho N et al (2014) Shear-wave elastographic features of breast cancers: comparison with mechanical elasticity and histopathologic characteristics. Invest Radiol 49:147–155PubMedCrossRef
4.
Zurück zum Zitat de Bazelaire C, Calmon R, Chapellier M, Pluvinage A, Frija J, de Kerviler E (2010) CT and MRI imaging in tumoral angiogenesis. Bull Cancer 97:79–90PubMed de Bazelaire C, Calmon R, Chapellier M, Pluvinage A, Frija J, de Kerviler E (2010) CT and MRI imaging in tumoral angiogenesis. Bull Cancer 97:79–90PubMed
5.
Zurück zum Zitat Ammari S, Thiam R, Cuenod CA et al (2014) Radiological evaluation of response to treatment: application to metastatic renal cancers receiving anti-angiogenic treatment. Diagn Interv Imaging 95:527–539PubMedCrossRef Ammari S, Thiam R, Cuenod CA et al (2014) Radiological evaluation of response to treatment: application to metastatic renal cancers receiving anti-angiogenic treatment. Diagn Interv Imaging 95:527–539PubMedCrossRef
6.
Zurück zum Zitat O’Connor JP, Aboagye EO, Adams JE et al (2017) Imaging biomarker roadmap for cancer studies. Nat Rev Clin Oncol 14:169–186PubMedCrossRef O’Connor JP, Aboagye EO, Adams JE et al (2017) Imaging biomarker roadmap for cancer studies. Nat Rev Clin Oncol 14:169–186PubMedCrossRef
7.
Zurück zum Zitat deSouza NM, Achten E, Alberich-Bayarri A et al (2019) Validated imaging biomarkers as decision-making tools in clinical trials and routine practice: current status and recommendations from the EIBALL* subcommittee of the European Society of Radiology (ESR). Insights Imaging 10:87PubMedPubMedCentralCrossRef deSouza NM, Achten E, Alberich-Bayarri A et al (2019) Validated imaging biomarkers as decision-making tools in clinical trials and routine practice: current status and recommendations from the EIBALL* subcommittee of the European Society of Radiology (ESR). Insights Imaging 10:87PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Gillies RJ, Kinahan PE, Hricak H (2016) Radiomics: images are more than pictures, they are data. Radiology 278:563–577PubMedCrossRef Gillies RJ, Kinahan PE, Hricak H (2016) Radiomics: images are more than pictures, they are data. Radiology 278:563–577PubMedCrossRef
10.
Zurück zum Zitat Lambin P, Leijenaar RTH, Deist TM et al (2017) Radiomics: the bridge between medical imaging and personalized medicine. Nat Rev Clin Oncol 14:749–762PubMedCrossRef Lambin P, Leijenaar RTH, Deist TM et al (2017) Radiomics: the bridge between medical imaging and personalized medicine. Nat Rev Clin Oncol 14:749–762PubMedCrossRef
11.
Zurück zum Zitat van Timmeren JE, Cester D, Tanadini-Lang S, Alkadhi H, Baessler B (2020) Radiomics in medical imaging-“how-to” guide and critical reflection. Insights Imaging 11:91PubMedPubMedCentralCrossRef van Timmeren JE, Cester D, Tanadini-Lang S, Alkadhi H, Baessler B (2020) Radiomics in medical imaging-“how-to” guide and critical reflection. Insights Imaging 11:91PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Sanduleanu S, Woodruff HC, de Jong EEC et al (2018) Tracking tumor biology with radiomics: a systematic review utilizing a radiomics quality score. Radiother Oncol 127:349–360PubMedCrossRef Sanduleanu S, Woodruff HC, de Jong EEC et al (2018) Tracking tumor biology with radiomics: a systematic review utilizing a radiomics quality score. Radiother Oncol 127:349–360PubMedCrossRef
13.
Zurück zum Zitat Nie K, Al-Hallaq H, Li XA et al (2019) NCTN assessment on current applications of radiomics in oncology. Int J Radiat Oncol Biol Phys 104:302–315PubMedPubMedCentralCrossRef Nie K, Al-Hallaq H, Li XA et al (2019) NCTN assessment on current applications of radiomics in oncology. Int J Radiat Oncol Biol Phys 104:302–315PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Wang T, Gao T, Yang J et al (2019) Preoperative prediction of pelvic lymph nodes metastasis in early-stage cervical cancer using radiomics nomogram developed based on T2-weighted MRI and diffusion-weighted imaging. Eur J Radiol 114:128–135PubMedCrossRef Wang T, Gao T, Yang J et al (2019) Preoperative prediction of pelvic lymph nodes metastasis in early-stage cervical cancer using radiomics nomogram developed based on T2-weighted MRI and diffusion-weighted imaging. Eur J Radiol 114:128–135PubMedCrossRef
15.
Zurück zum Zitat Cameron A, Khalvati F, Haider MA, Wong A (2016) MAPS: a quantitative radiomics approach for prostate cancer detection. IEEE Trans Biomed Eng 63:1145–1156PubMedCrossRef Cameron A, Khalvati F, Haider MA, Wong A (2016) MAPS: a quantitative radiomics approach for prostate cancer detection. IEEE Trans Biomed Eng 63:1145–1156PubMedCrossRef
16.
Zurück zum Zitat Ma X, Shen F, Jia Y, Xia Y, Li Q, Lu J (2019) MRI-based radiomics of rectal cancer: preoperative assessment of the pathological features. BMC Med Imaging 19:86PubMedPubMedCentralCrossRef Ma X, Shen F, Jia Y, Xia Y, Li Q, Lu J (2019) MRI-based radiomics of rectal cancer: preoperative assessment of the pathological features. BMC Med Imaging 19:86PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Yun J, Park JE, Lee H, Ham S, Kim N, Kim HS (2019) Radiomic features and multilayer perceptron network classifier: a robust MRI classification strategy for distinguishing glioblastoma from primary central nervous system lymphoma. Sci Rep 9:5746PubMedPubMedCentralCrossRef Yun J, Park JE, Lee H, Ham S, Kim N, Kim HS (2019) Radiomic features and multilayer perceptron network classifier: a robust MRI classification strategy for distinguishing glioblastoma from primary central nervous system lymphoma. Sci Rep 9:5746PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Shi L, He Y, Yuan Z et al (2018) Radiomics for response and outcome assessment for non-small cell lung cancer. Technol Cancer Res Treat 17:1533033818782788PubMedPubMedCentralCrossRef Shi L, He Y, Yuan Z et al (2018) Radiomics for response and outcome assessment for non-small cell lung cancer. Technol Cancer Res Treat 17:1533033818782788PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Peeken JC, Bernhofer M, Wiestler B et al (2018) Radiomics in radiooncology - challenging the medical physicist. Phys Med 48:27–36PubMedCrossRef Peeken JC, Bernhofer M, Wiestler B et al (2018) Radiomics in radiooncology - challenging the medical physicist. Phys Med 48:27–36PubMedCrossRef
20.
Zurück zum Zitat Reuze S, Schernberg A, Orlhac F et al (2018) Radiomics in nuclear medicine applied to radiation therapy: methods, pitfalls, and challenges. Int J Radiat Oncol Biol Phys 102:1117–1142PubMedCrossRef Reuze S, Schernberg A, Orlhac F et al (2018) Radiomics in nuclear medicine applied to radiation therapy: methods, pitfalls, and challenges. Int J Radiat Oncol Biol Phys 102:1117–1142PubMedCrossRef
21.
Zurück zum Zitat Elhalawani H, Lin TA, Volpe S et al (2018) Machine learning applications in head and neck radiation oncology: lessons from open-source radiomics challenges. Front Oncol 8:294PubMedPubMedCentralCrossRef Elhalawani H, Lin TA, Volpe S et al (2018) Machine learning applications in head and neck radiation oncology: lessons from open-source radiomics challenges. Front Oncol 8:294PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat El Naqa I, Ten Haken RK (2018) Can radiomics personalise immunotherapy? Lancet Oncol 19:1138–1139PubMedCrossRef El Naqa I, Ten Haken RK (2018) Can radiomics personalise immunotherapy? Lancet Oncol 19:1138–1139PubMedCrossRef
24.
Zurück zum Zitat Sun R, Limkin EJ, Vakalopoulou M et al (2018) A radiomics approach to assess tumour-infiltrating CD8 cells and response to anti-PD-1 or anti-PD-L1 immunotherapy: an imaging biomarker, retrospective multicohort study. Lancet Oncol 19:1180–1191PubMedCrossRef Sun R, Limkin EJ, Vakalopoulou M et al (2018) A radiomics approach to assess tumour-infiltrating CD8 cells and response to anti-PD-1 or anti-PD-L1 immunotherapy: an imaging biomarker, retrospective multicohort study. Lancet Oncol 19:1180–1191PubMedCrossRef
25.
Zurück zum Zitat Trebeschi S, Drago SG, Birkbak NJ et al (2019) Predicting response to cancer immunotherapy using noninvasive radiomic biomarkers. Ann Oncol 30:998–1004PubMedPubMedCentralCrossRef Trebeschi S, Drago SG, Birkbak NJ et al (2019) Predicting response to cancer immunotherapy using noninvasive radiomic biomarkers. Ann Oncol 30:998–1004PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Capobianco E, Dominietto M (2020) From medical imaging to radiomics: role of data science for advancing precision health. J Pers Med 10 Capobianco E, Dominietto M (2020) From medical imaging to radiomics: role of data science for advancing precision health. J Pers Med 10
29.
Zurück zum Zitat Bogowicz M, Riesterer O, Ikenberg K et al (2017) Computed tomography radiomics predicts HPV status and local tumor control after definitive radiochemotherapy in head and neck squamous cell carcinoma. Int J Radiat Oncol Biol Phys 99:921–928PubMedCrossRef Bogowicz M, Riesterer O, Ikenberg K et al (2017) Computed tomography radiomics predicts HPV status and local tumor control after definitive radiochemotherapy in head and neck squamous cell carcinoma. Int J Radiat Oncol Biol Phys 99:921–928PubMedCrossRef
30.
Zurück zum Zitat Zhong Y, Yuan M, Zhang T, Zhang YD, Li H, Yu TF (2018) Radiomics approach to prediction of occult mediastinal lymph node metastasis of lung adenocarcinoma. AJR Am J Roentgenol 211:109–113PubMedCrossRef Zhong Y, Yuan M, Zhang T, Zhang YD, Li H, Yu TF (2018) Radiomics approach to prediction of occult mediastinal lymph node metastasis of lung adenocarcinoma. AJR Am J Roentgenol 211:109–113PubMedCrossRef
31.
Zurück zum Zitat Dou TH, Coroller TP, van Griethuysen JJM, Mak RH, Aerts H (2018) Peritumoral radiomics features predict distant metastasis in locally advanced NSCLC. PLoS One 13:e0206108PubMedPubMedCentralCrossRef Dou TH, Coroller TP, van Griethuysen JJM, Mak RH, Aerts H (2018) Peritumoral radiomics features predict distant metastasis in locally advanced NSCLC. PLoS One 13:e0206108PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Kim JY, Park JE, Jo Y et al (2019) Incorporating diffusion- and perfusion-weighted MRI into a radiomics model improves diagnostic performance for pseudoprogression in glioblastoma patients. Neuro Oncol 21:404–414PubMedCrossRef Kim JY, Park JE, Jo Y et al (2019) Incorporating diffusion- and perfusion-weighted MRI into a radiomics model improves diagnostic performance for pseudoprogression in glioblastoma patients. Neuro Oncol 21:404–414PubMedCrossRef
33.
Zurück zum Zitat Suh HB, Choi YS, Bae S et al (2018) Primary central nervous system lymphoma and atypical glioblastoma: differentiation using radiomics approach. Eur Radiol 28:3832–3839PubMedCrossRef Suh HB, Choi YS, Bae S et al (2018) Primary central nervous system lymphoma and atypical glioblastoma: differentiation using radiomics approach. Eur Radiol 28:3832–3839PubMedCrossRef
34.
Zurück zum Zitat Li Y, Liu X, Xu K et al (2018) MRI features can predict EGFR expression in lower grade gliomas: a voxel-based radiomic analysis. Eur Radiol 28:356–362PubMedCrossRef Li Y, Liu X, Xu K et al (2018) MRI features can predict EGFR expression in lower grade gliomas: a voxel-based radiomic analysis. Eur Radiol 28:356–362PubMedCrossRef
35.
Zurück zum Zitat Li Y, Qian Z, Xu K et al (2018) MRI features predict p53 status in lower-grade gliomas via a machine-learning approach. Neuroimage Clin 17:306–311PubMedCrossRef Li Y, Qian Z, Xu K et al (2018) MRI features predict p53 status in lower-grade gliomas via a machine-learning approach. Neuroimage Clin 17:306–311PubMedCrossRef
36.
Zurück zum Zitat Wormald BW, Doran SJ, Ind TE, D’Arcy J, Petts J, deSouza NM (2020) Radiomic features of cervical cancer on T2-and diffusion-weighted MRI: prognostic value in low-volume tumors suitable for trachelectomy. Gynecol Oncol 156:107–114PubMedPubMedCentralCrossRef Wormald BW, Doran SJ, Ind TE, D’Arcy J, Petts J, deSouza NM (2020) Radiomic features of cervical cancer on T2-and diffusion-weighted MRI: prognostic value in low-volume tumors suitable for trachelectomy. Gynecol Oncol 156:107–114PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Cook GJ, Yip C, Siddique M et al (2013) Are pretreatment 18F-FDG PET tumor textural features in non-small cell lung cancer associated with response and survival after chemoradiotherapy? J Nucl Med 54:19–26PubMedCrossRef Cook GJ, Yip C, Siddique M et al (2013) Are pretreatment 18F-FDG PET tumor textural features in non-small cell lung cancer associated with response and survival after chemoradiotherapy? J Nucl Med 54:19–26PubMedCrossRef
38.
Zurück zum Zitat Tixier F, Hatt M, Valla C et al (2014) Visual versus quantitative assessment of intratumor 18F-FDG PET uptake heterogeneity: prognostic value in non-small cell lung cancer. J Nucl Med 55:1235–1241PubMedCrossRef Tixier F, Hatt M, Valla C et al (2014) Visual versus quantitative assessment of intratumor 18F-FDG PET uptake heterogeneity: prognostic value in non-small cell lung cancer. J Nucl Med 55:1235–1241PubMedCrossRef
39.
Zurück zum Zitat Cook GJ, O’Brien ME, Siddique M et al (2015) Non-small cell lung cancer treated with erlotinib: heterogeneity of (18)F-FDG uptake at PET-association with treatment response and prognosis. Radiology 276:883–893PubMedCrossRef Cook GJ, O’Brien ME, Siddique M et al (2015) Non-small cell lung cancer treated with erlotinib: heterogeneity of (18)F-FDG uptake at PET-association with treatment response and prognosis. Radiology 276:883–893PubMedCrossRef
40.
Zurück zum Zitat Parmar C, Leijenaar RT, Grossmann P et al (2015) Radiomic feature clusters and prognostic signatures specific for Lung and Head & Neck cancer. Sci Rep 5:11044PubMedPubMedCentralCrossRef Parmar C, Leijenaar RT, Grossmann P et al (2015) Radiomic feature clusters and prognostic signatures specific for Lung and Head & Neck cancer. Sci Rep 5:11044PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Li S, Ding C, Zhang H, Song J, Wu L (2019) Radiomics for the prediction of EGFR mutation subtypes in non-small cell lung cancer. Med Phys 46:4545–4552PubMedCrossRef Li S, Ding C, Zhang H, Song J, Wu L (2019) Radiomics for the prediction of EGFR mutation subtypes in non-small cell lung cancer. Med Phys 46:4545–4552PubMedCrossRef
42.
Zurück zum Zitat Mattonen SA, Davidzon GA, Benson J et al (2019) Bone marrow and tumor radiomics at (18)F-FDG PET/CT: impact on outcome prediction in non-small cell lung cancer. Radiology 293:451–459PubMedCrossRef Mattonen SA, Davidzon GA, Benson J et al (2019) Bone marrow and tumor radiomics at (18)F-FDG PET/CT: impact on outcome prediction in non-small cell lung cancer. Radiology 293:451–459PubMedCrossRef
43.
Zurück zum Zitat Antunes J, Viswanath S, Rusu M et al (2016) Radiomics analysis on FLT-PET/MRI for characterization of early treatment response in renal cell carcinoma: a proof-of-concept study. Transl Oncol 9:155–162PubMedPubMedCentralCrossRef Antunes J, Viswanath S, Rusu M et al (2016) Radiomics analysis on FLT-PET/MRI for characterization of early treatment response in renal cell carcinoma: a proof-of-concept study. Transl Oncol 9:155–162PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Zamboglou C, Carles M, Fechter T et al (2019) Radiomic features from PSMA PET for non-invasive intraprostatic tumor discrimination and characterization in patients with intermediate- and high-risk prostate cancer - a comparison study with histology reference. Theranostics 9:2595–2605PubMedPubMedCentralCrossRef Zamboglou C, Carles M, Fechter T et al (2019) Radiomic features from PSMA PET for non-invasive intraprostatic tumor discrimination and characterization in patients with intermediate- and high-risk prostate cancer - a comparison study with histology reference. Theranostics 9:2595–2605PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Zheng X, Yao Z, Huang Y et al (2020) Deep learning radiomics can predict axillary lymph node status in early-stage breast cancer. Nat Commun 11:1236PubMedPubMedCentralCrossRef Zheng X, Yao Z, Huang Y et al (2020) Deep learning radiomics can predict axillary lymph node status in early-stage breast cancer. Nat Commun 11:1236PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Caramella C, Allorant A, Orlhac F et al (2018) Can we trust the calculation of texture indices of CT images? A phantom study. Med Phys 45:1529–1536PubMedCrossRef Caramella C, Allorant A, Orlhac F et al (2018) Can we trust the calculation of texture indices of CT images? A phantom study. Med Phys 45:1529–1536PubMedCrossRef
47.
Zurück zum Zitat Raunig DL, McShane LM, Pennello G et al (2015) Quantitative imaging biomarkers: a review of statistical methods for technical performance assessment. Stat Methods Med Res 24:27–67PubMedCrossRef Raunig DL, McShane LM, Pennello G et al (2015) Quantitative imaging biomarkers: a review of statistical methods for technical performance assessment. Stat Methods Med Res 24:27–67PubMedCrossRef
48.
Zurück zum Zitat Shaikh F, Franc B, Allen E et al (2018) Translational radiomics: defining the strategy pipeline and considerations for application-part 2: from clinical implementation to enterprise. J Am Coll Radiol 15:543–549PubMedPubMedCentralCrossRef Shaikh F, Franc B, Allen E et al (2018) Translational radiomics: defining the strategy pipeline and considerations for application-part 2: from clinical implementation to enterprise. J Am Coll Radiol 15:543–549PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Shaikh F, Franc B, Allen E et al (2018) Translational radiomics: defining the strategy pipeline and considerations for application-part 1: from methodology to clinical implementation. J Am Coll Radiol 15:538–542PubMedPubMedCentralCrossRef Shaikh F, Franc B, Allen E et al (2018) Translational radiomics: defining the strategy pipeline and considerations for application-part 1: from methodology to clinical implementation. J Am Coll Radiol 15:538–542PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Pfaehler E, van Sluis J, Merema BBJ et al (2020) Experimental multicenter and multivendor evaluation of the performance of PET radiomic features using 3-dimensionally printed phantom inserts. J Nucl Med 61:469–476PubMedPubMedCentralCrossRef Pfaehler E, van Sluis J, Merema BBJ et al (2020) Experimental multicenter and multivendor evaluation of the performance of PET radiomic features using 3-dimensionally printed phantom inserts. J Nucl Med 61:469–476PubMedPubMedCentralCrossRef
52.
53.
Zurück zum Zitat Nyul LG, Udupa JK, Zhang X (2000) New variants of a method of MRI scale standardization. IEEE Trans Med Imaging 19:143–150PubMedCrossRef Nyul LG, Udupa JK, Zhang X (2000) New variants of a method of MRI scale standardization. IEEE Trans Med Imaging 19:143–150PubMedCrossRef
54.
Zurück zum Zitat Isaksson LJ, Raimondi S, Botta F et al (2020) Effects of MRI image normalization techniques in prostate cancer radiomics. Phys Med 71:7–13PubMedCrossRef Isaksson LJ, Raimondi S, Botta F et al (2020) Effects of MRI image normalization techniques in prostate cancer radiomics. Phys Med 71:7–13PubMedCrossRef
55.
Zurück zum Zitat Scalco E, Belfatto A, Mastropietro A et al (2020) T2w-MRI signal normalization affects radiomics features reproducibility. Med Phys 47:1680–1691PubMedCrossRef Scalco E, Belfatto A, Mastropietro A et al (2020) T2w-MRI signal normalization affects radiomics features reproducibility. Med Phys 47:1680–1691PubMedCrossRef
56.
Zurück zum Zitat Leijenaar RT, Nalbantov G, Carvalho S et al (2015) The effect of SUV discretization in quantitative FDG-PET radiomics: the need for standardized methodology in tumor texture analysis. Sci Rep 5:11075PubMedPubMedCentralCrossRef Leijenaar RT, Nalbantov G, Carvalho S et al (2015) The effect of SUV discretization in quantitative FDG-PET radiomics: the need for standardized methodology in tumor texture analysis. Sci Rep 5:11075PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Lee SH, Cho HH, Lee HY, Park H (2019) Clinical impact of variability on CT radiomics and suggestions for suitable feature selection: a focus on lung cancer. Cancer Imaging 19:54PubMedPubMedCentralCrossRef Lee SH, Cho HH, Lee HY, Park H (2019) Clinical impact of variability on CT radiomics and suggestions for suitable feature selection: a focus on lung cancer. Cancer Imaging 19:54PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Duron L, Balvay D, Vande Perre S et al (2019) Gray-level discretization impacts reproducible MRI radiomics texture features. PLoS One 14:e0213459PubMedPubMedCentralCrossRef Duron L, Balvay D, Vande Perre S et al (2019) Gray-level discretization impacts reproducible MRI radiomics texture features. PLoS One 14:e0213459PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Orlhac F, Frouin F, Nioche C, Ayache N, Buvat I (2019) Validation of a method to compensate multicenter effects affecting CT radiomics. Radiology 291:53–59PubMedCrossRef Orlhac F, Frouin F, Nioche C, Ayache N, Buvat I (2019) Validation of a method to compensate multicenter effects affecting CT radiomics. Radiology 291:53–59PubMedCrossRef
61.
Zurück zum Zitat Zwanenburg A, Vallieres M, Abdalah MA et al (2020) The image biomarker standardization initiative: standardized quantitative radiomics for high-throughput image-based phenotyping. Radiology 295:328–338PubMedCrossRef Zwanenburg A, Vallieres M, Abdalah MA et al (2020) The image biomarker standardization initiative: standardized quantitative radiomics for high-throughput image-based phenotyping. Radiology 295:328–338PubMedCrossRef
62.
Zurück zum Zitat Owens CA, Peterson CB, Tang C et al (2018) Lung tumor segmentation methods: impact on the uncertainty of radiomics features for non-small cell lung cancer. PLoS One 13:e0205003PubMedPubMedCentralCrossRef Owens CA, Peterson CB, Tang C et al (2018) Lung tumor segmentation methods: impact on the uncertainty of radiomics features for non-small cell lung cancer. PLoS One 13:e0205003PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Caballo M, Pangallo DR, Mann RM, Sechopoulos I (2020) Deep learning-based segmentation of breast masses in dedicated breast CT imaging: radiomic feature stability between radiologists and artificial intelligence. Comput Biol Med 118:103629PubMedCrossRef Caballo M, Pangallo DR, Mann RM, Sechopoulos I (2020) Deep learning-based segmentation of breast masses in dedicated breast CT imaging: radiomic feature stability between radiologists and artificial intelligence. Comput Biol Med 118:103629PubMedCrossRef
64.
Zurück zum Zitat Hatt M, Lee JA, Schmidtlein CR et al (2017) Classification and evaluation strategies of auto-segmentation approaches for PET: report of AAPM task group No. 211. Med Phys 44:e1–e42PubMedCrossRef Hatt M, Lee JA, Schmidtlein CR et al (2017) Classification and evaluation strategies of auto-segmentation approaches for PET: report of AAPM task group No. 211. Med Phys 44:e1–e42PubMedCrossRef
65.
Zurück zum Zitat Waninger JJ, Green MD, Cheze Le Rest C, Rosen B, El Naqa I (2019) Integrating radiomics into clinical trial design. Q J Nucl Med Mol Imaging 63:339–346PubMedCrossRef Waninger JJ, Green MD, Cheze Le Rest C, Rosen B, El Naqa I (2019) Integrating radiomics into clinical trial design. Q J Nucl Med Mol Imaging 63:339–346PubMedCrossRef
66.
Zurück zum Zitat Ciardo D, Jereczek-Fossa BA, Petralia G et al (2017) Multimodal image registration for the identification of dominant intraprostatic lesion in high-precision radiotherapy treatments. Br J Radiol 90:20170021PubMedPubMedCentralCrossRef Ciardo D, Jereczek-Fossa BA, Petralia G et al (2017) Multimodal image registration for the identification of dominant intraprostatic lesion in high-precision radiotherapy treatments. Br J Radiol 90:20170021PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Ou Y, Weinstein SP, Conant EF et al (2015) Deformable registration for quantifying longitudinal tumor changes during neoadjuvant chemotherapy. Magn Reson Med 73:2343–2356PubMedCrossRef Ou Y, Weinstein SP, Conant EF et al (2015) Deformable registration for quantifying longitudinal tumor changes during neoadjuvant chemotherapy. Magn Reson Med 73:2343–2356PubMedCrossRef
68.
Zurück zum Zitat Fornacon-Wood I, Mistry H, Ackermann CJ et al (2020) Reliability and prognostic value of radiomic features are highly dependent on choice of feature extraction platform. Eur Radiol 30:6241–6250PubMedPubMedCentralCrossRef Fornacon-Wood I, Mistry H, Ackermann CJ et al (2020) Reliability and prognostic value of radiomic features are highly dependent on choice of feature extraction platform. Eur Radiol 30:6241–6250PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat Dhall DKR, Juneja M (2020) Machine learning: a review of the algorithms and its applications. In: Singh PKASY, Kolekar M, Tanwar S (eds) Proceedings of ICRIC 2019 Lecture Notes in Electrical Engineering. Springer, Cham Dhall DKR, Juneja M (2020) Machine learning: a review of the algorithms and its applications. In: Singh PKASY, Kolekar M, Tanwar S (eds) Proceedings of ICRIC 2019 Lecture Notes in Electrical Engineering. Springer, Cham
70.
Zurück zum Zitat Ozgur C, Kleckner M, Li Y (2015) Selection of statistical software for solving big data problems: a guide for businesses, students, and universities. Sage Open 5:1–12CrossRef Ozgur C, Kleckner M, Li Y (2015) Selection of statistical software for solving big data problems: a guide for businesses, students, and universities. Sage Open 5:1–12CrossRef
71.
Zurück zum Zitat Pillai R, Oza P, Sharma P (2020) Review of machine learning techniques in health care. In: Singh P, Kar A, Singh Y, Kolekar M, Tanwar S (eds) Proceedings of ICRIC 2019 Lecture Notes in Electrical Engineering. Springer, Cham Pillai R, Oza P, Sharma P (2020) Review of machine learning techniques in health care. In: Singh P, Kar A, Singh Y, Kolekar M, Tanwar S (eds) Proceedings of ICRIC 2019 Lecture Notes in Electrical Engineering. Springer, Cham
72.
Zurück zum Zitat Tanwani AK, Alfridi J, Shafiq Z, Farooq M (2009) Guidelines to select machine learning scheme for classification of biomedical datasets. In: Pizzuti C, Ritchie MD, Giacobini M (eds) Evolutionary Computation, Machine Learning and Data Mining in Bioinformatics EvoBIO 2009 Lecture Notes in Computer Science. Springer, Berlin, Heidelberg, pp 128–139 Tanwani AK, Alfridi J, Shafiq Z, Farooq M (2009) Guidelines to select machine learning scheme for classification of biomedical datasets. In: Pizzuti C, Ritchie MD, Giacobini M (eds) Evolutionary Computation, Machine Learning and Data Mining in Bioinformatics EvoBIO 2009 Lecture Notes in Computer Science. Springer, Berlin, Heidelberg, pp 128–139
73.
Zurück zum Zitat Chen T, Ning Z, Xu L et al (2019) Radiomics nomogram for predicting the malignant potential of gastrointestinal stromal tumours preoperatively. Eur Radiol 29:1074–1082PubMedCrossRef Chen T, Ning Z, Xu L et al (2019) Radiomics nomogram for predicting the malignant potential of gastrointestinal stromal tumours preoperatively. Eur Radiol 29:1074–1082PubMedCrossRef
74.
75.
Zurück zum Zitat Leger S, Zwanenburg A, Pilz K et al (2017) A comparative study of machine learning methods for time-to-event survival data for radiomics risk modelling. Sci Rep 7:13206PubMedPubMedCentralCrossRef Leger S, Zwanenburg A, Pilz K et al (2017) A comparative study of machine learning methods for time-to-event survival data for radiomics risk modelling. Sci Rep 7:13206PubMedPubMedCentralCrossRef
76.
Zurück zum Zitat Afshar P, Mohammadi A, Plataniotis KN, Oikonomou A, Benali H (2019) From handcrafted to deep-learning-based cancer radiomics challenges and opportunities. Ieee Signal Processing Magazine 36:132–160CrossRef Afshar P, Mohammadi A, Plataniotis KN, Oikonomou A, Benali H (2019) From handcrafted to deep-learning-based cancer radiomics challenges and opportunities. Ieee Signal Processing Magazine 36:132–160CrossRef
77.
Zurück zum Zitat Vial A, Stirling D, Field M et al (2018) The role of deep learning and radiomic feature extraction in cancer-specific predictive modelling: a review. Transl Cancer Res 7:803–816 Vial A, Stirling D, Field M et al (2018) The role of deep learning and radiomic feature extraction in cancer-specific predictive modelling: a review. Transl Cancer Res 7:803–816
78.
Zurück zum Zitat Avanzo M, Wei L, Stancanello J et al (2020) Machine and deep learning methods for radiomics. Med Phys 47:e185–e202PubMedCrossRef Avanzo M, Wei L, Stancanello J et al (2020) Machine and deep learning methods for radiomics. Med Phys 47:e185–e202PubMedCrossRef
79.
Zurück zum Zitat Peerlings J, Woodruff HC, Winfield JM et al (2019) Stability of radiomics features in apparent diffusion coefficient maps from a multi-centre test-retest trial. Sci Rep 9:4800PubMedPubMedCentralCrossRef Peerlings J, Woodruff HC, Winfield JM et al (2019) Stability of radiomics features in apparent diffusion coefficient maps from a multi-centre test-retest trial. Sci Rep 9:4800PubMedPubMedCentralCrossRef
80.
Zurück zum Zitat Traverso A, Wee L, Dekker A, Gillies R (2018) Repeatability and reproducibility of radiomic features: a systematic review. Int J Radiat Oncol Biol Phys 102:1143–1158PubMedPubMedCentralCrossRef Traverso A, Wee L, Dekker A, Gillies R (2018) Repeatability and reproducibility of radiomic features: a systematic review. Int J Radiat Oncol Biol Phys 102:1143–1158PubMedPubMedCentralCrossRef
81.
Zurück zum Zitat AlBadawy EA, Saha A, Mazurowski MA (2018) Deep learning for segmentation of brain tumors: impact of cross-institutional training and testing. Med Phys 45:1150–1158PubMedCrossRef AlBadawy EA, Saha A, Mazurowski MA (2018) Deep learning for segmentation of brain tumors: impact of cross-institutional training and testing. Med Phys 45:1150–1158PubMedCrossRef
82.
Zurück zum Zitat Larue R, Klaassen R, Jochems A et al (2018) Pre-treatment CT radiomics to predict 3-year overall survival following chemoradiotherapy of esophageal cancer. Acta Oncol 57:1475–1481PubMedCrossRef Larue R, Klaassen R, Jochems A et al (2018) Pre-treatment CT radiomics to predict 3-year overall survival following chemoradiotherapy of esophageal cancer. Acta Oncol 57:1475–1481PubMedCrossRef
83.
Zurück zum Zitat Soufi M, Arimura H, Nagami N (2018) Identification of optimal mother wavelets in survival prediction of lung cancer patients using wavelet decomposition-based radiomic features. Med Phys 45:5116–5128PubMedCrossRef Soufi M, Arimura H, Nagami N (2018) Identification of optimal mother wavelets in survival prediction of lung cancer patients using wavelet decomposition-based radiomic features. Med Phys 45:5116–5128PubMedCrossRef
84.
Zurück zum Zitat Xu X, Huang L, Chen J et al (2019) Application of radiomics signature captured from pretreatment thoracic CT to predict brain metastases in stage III/IV ALK-positive non-small cell lung cancer patients. J Thorac Dis 11:4516–4528PubMedPubMedCentralCrossRef Xu X, Huang L, Chen J et al (2019) Application of radiomics signature captured from pretreatment thoracic CT to predict brain metastases in stage III/IV ALK-positive non-small cell lung cancer patients. J Thorac Dis 11:4516–4528PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Li H, Xie Y, Wang X, Chen F, Sun J, Jiang X (2019) Radiomics features on non-contrast computed tomography predict early enlargement of spontaneous intracerebral hemorrhage. Clin Neurol Neurosurg 185:105491PubMedCrossRef Li H, Xie Y, Wang X, Chen F, Sun J, Jiang X (2019) Radiomics features on non-contrast computed tomography predict early enlargement of spontaneous intracerebral hemorrhage. Clin Neurol Neurosurg 185:105491PubMedCrossRef
86.
Zurück zum Zitat Huynh E, Coroller TP, Narayan V et al (2016) CT-based radiomic analysis of stereotactic body radiation therapy patients with lung cancer. Radiother Oncol 120:258–266PubMedCrossRef Huynh E, Coroller TP, Narayan V et al (2016) CT-based radiomic analysis of stereotactic body radiation therapy patients with lung cancer. Radiother Oncol 120:258–266PubMedCrossRef
87.
Zurück zum Zitat Hui B, Qiu JJ, Liu JH, Ke NW (2020) Identification of pancreaticoduodenectomy resection for pancreatic head adenocarcinoma: a preliminary study of radiomics. Comput Math Methods Med 2020:2761627PubMedPubMedCentralCrossRef Hui B, Qiu JJ, Liu JH, Ke NW (2020) Identification of pancreaticoduodenectomy resection for pancreatic head adenocarcinoma: a preliminary study of radiomics. Comput Math Methods Med 2020:2761627PubMedPubMedCentralCrossRef
88.
Zurück zum Zitat Leithner D, Horvat JV, Marino MA et al (2019) Radiomic signatures with contrast-enhanced magnetic resonance imaging for the assessment of breast cancer receptor status and molecular subtypes: initial results. Breast Cancer Res 21:106PubMedPubMedCentralCrossRef Leithner D, Horvat JV, Marino MA et al (2019) Radiomic signatures with contrast-enhanced magnetic resonance imaging for the assessment of breast cancer receptor status and molecular subtypes: initial results. Breast Cancer Res 21:106PubMedPubMedCentralCrossRef
89.
Zurück zum Zitat Zhang Y, Yan P, Liang F, Ma C, Liang S, Jiang C (2019) Predictors of epilepsy presentation in unruptured brain arteriovenous malformations: a quantitative evaluation of location and radiomics features on T2-weighted imaging. World Neurosurg 125:e1008–e1015PubMedCrossRef Zhang Y, Yan P, Liang F, Ma C, Liang S, Jiang C (2019) Predictors of epilepsy presentation in unruptured brain arteriovenous malformations: a quantitative evaluation of location and radiomics features on T2-weighted imaging. World Neurosurg 125:e1008–e1015PubMedCrossRef
90.
Zurück zum Zitat Zhou J, Lu J, Gao C et al (2020) Predicting the response to neoadjuvant chemotherapy for breast cancer: wavelet transforming radiomics in MRI. BMC Cancer 20:100PubMedPubMedCentralCrossRef Zhou J, Lu J, Gao C et al (2020) Predicting the response to neoadjuvant chemotherapy for breast cancer: wavelet transforming radiomics in MRI. BMC Cancer 20:100PubMedPubMedCentralCrossRef
92.
95.
Zurück zum Zitat Bhatia A, Birger M, Veeraraghavan H et al (2019) MRI radiomic features are associated with survival in melanoma brain metastases treated with immune checkpoint inhibitors. Neuro Oncol 21:1578–1586PubMedPubMedCentralCrossRef Bhatia A, Birger M, Veeraraghavan H et al (2019) MRI radiomic features are associated with survival in melanoma brain metastases treated with immune checkpoint inhibitors. Neuro Oncol 21:1578–1586PubMedPubMedCentralCrossRef
96.
Zurück zum Zitat Shayesteh SP, Alikhassi A, Fard Esfahani A et al (2019) Neo-adjuvant chemoradiotherapy response prediction using MRI based ensemble learning method in rectal cancer patients. Phys Med 62:111–119PubMedCrossRef Shayesteh SP, Alikhassi A, Fard Esfahani A et al (2019) Neo-adjuvant chemoradiotherapy response prediction using MRI based ensemble learning method in rectal cancer patients. Phys Med 62:111–119PubMedCrossRef
97.
Zurück zum Zitat Fiset S, Welch ML, Weiss J et al (2019) Repeatability and reproducibility of MRI-based radiomic features in cervical cancer. Radiother Oncol 135:107–114PubMedCrossRef Fiset S, Welch ML, Weiss J et al (2019) Repeatability and reproducibility of MRI-based radiomic features in cervical cancer. Radiother Oncol 135:107–114PubMedCrossRef
98.
Zurück zum Zitat Fave X, Zhang L, Yang J et al (2017) Delta-radiomics features for the prediction of patient outcomes in non-small cell lung cancer. Sci Rep 7:588PubMedPubMedCentralCrossRef Fave X, Zhang L, Yang J et al (2017) Delta-radiomics features for the prediction of patient outcomes in non-small cell lung cancer. Sci Rep 7:588PubMedPubMedCentralCrossRef
99.
Zurück zum Zitat Dong X, Sun X, Sun L et al (2016) Early change in metabolic tumor heterogeneity during chemoradiotherapy and its prognostic value for patients with locally advanced non-small cell lung cancer. PLoS One 11:e0157836PubMedPubMedCentralCrossRef Dong X, Sun X, Sun L et al (2016) Early change in metabolic tumor heterogeneity during chemoradiotherapy and its prognostic value for patients with locally advanced non-small cell lung cancer. PLoS One 11:e0157836PubMedPubMedCentralCrossRef
100.
Zurück zum Zitat Tixier F, Vriens D, Cheze-Le Rest C et al (2016) Comparison of tumor uptake heterogeneity characterization between static and parametric 18F-FDG PET images in non-small cell lung cancer. J Nucl Med 57:1033–1039PubMedCrossRef Tixier F, Vriens D, Cheze-Le Rest C et al (2016) Comparison of tumor uptake heterogeneity characterization between static and parametric 18F-FDG PET images in non-small cell lung cancer. J Nucl Med 57:1033–1039PubMedCrossRef
101.
Zurück zum Zitat Yin P, Mao N, Zhao C, Wu J, Chen L, Hong N (2019) A triple-classification radiomics model for the differentiation of primary chordoma, giant cell tumor, and metastatic tumor of sacrum based on T2-weighted and contrast-enhanced T1-weighted MRI. J Magn Reson Imaging 49:752–759PubMedCrossRef Yin P, Mao N, Zhao C, Wu J, Chen L, Hong N (2019) A triple-classification radiomics model for the differentiation of primary chordoma, giant cell tumor, and metastatic tumor of sacrum based on T2-weighted and contrast-enhanced T1-weighted MRI. J Magn Reson Imaging 49:752–759PubMedCrossRef
102.
Zurück zum Zitat Grossmann P, Stringfield O, El-Hachem N et al (2017) Defining the biological basis of radiomic phenotypes in lung cancer. Elife 6 Grossmann P, Stringfield O, El-Hachem N et al (2017) Defining the biological basis of radiomic phenotypes in lung cancer. Elife 6
103.
Zurück zum Zitat Tu W, Sun G, Fan L et al (2019) Radiomics signature: a potential and incremental predictor for EGFR mutation status in NSCLC patients, comparison with CT morphology. Lung Cancer 132:28–35PubMedCrossRef Tu W, Sun G, Fan L et al (2019) Radiomics signature: a potential and incremental predictor for EGFR mutation status in NSCLC patients, comparison with CT morphology. Lung Cancer 132:28–35PubMedCrossRef
104.
Zurück zum Zitat Sanghani P, Ang BT, King NKK, Ren H (2018) Overall survival prediction in glioblastoma multiforme patients from volumetric, shape and texture features using machine learning. Surg Oncol 27:709–714PubMedCrossRef Sanghani P, Ang BT, King NKK, Ren H (2018) Overall survival prediction in glioblastoma multiforme patients from volumetric, shape and texture features using machine learning. Surg Oncol 27:709–714PubMedCrossRef
105.
Zurück zum Zitat Ferreira Junior JR, Koenigkam-Santos M, Cipriano FEG, Fabro AT, Azevedo-Marques PM (2018) Radiomics-based features for pattern recognition of lung cancer histopathology and metastases. Comput Methods Programs Biomed 159:23–30PubMedCrossRef Ferreira Junior JR, Koenigkam-Santos M, Cipriano FEG, Fabro AT, Azevedo-Marques PM (2018) Radiomics-based features for pattern recognition of lung cancer histopathology and metastases. Comput Methods Programs Biomed 159:23–30PubMedCrossRef
106.
Zurück zum Zitat Penzias G, Singanamalli A, Elliott R et al (2018) Identifying the morphologic basis for radiomic features in distinguishing different Gleason grades of prostate cancer on MRI: preliminary findings. PLoS One 13:e0200730PubMedPubMedCentralCrossRef Penzias G, Singanamalli A, Elliott R et al (2018) Identifying the morphologic basis for radiomic features in distinguishing different Gleason grades of prostate cancer on MRI: preliminary findings. PLoS One 13:e0200730PubMedPubMedCentralCrossRef
108.
Zurück zum Zitat Oakden-Rayner L, Carneiro G, Bessen T, Nascimento JC, Bradley AP, Palmer LJ (2017) Precision radiology: predicting longevity using feature engineering and deep learning methods in a radiomics framework. Sci Rep 7:1648PubMedPubMedCentralCrossRef Oakden-Rayner L, Carneiro G, Bessen T, Nascimento JC, Bradley AP, Palmer LJ (2017) Precision radiology: predicting longevity using feature engineering and deep learning methods in a radiomics framework. Sci Rep 7:1648PubMedPubMedCentralCrossRef
109.
Zurück zum Zitat Bibault JE, Giraud P, Housset M et al (2018) Deep learning and radiomics predict complete response after neo-adjuvant chemoradiation for locally advanced rectal cancer. Sci Rep 8:12611PubMedPubMedCentralCrossRef Bibault JE, Giraud P, Housset M et al (2018) Deep learning and radiomics predict complete response after neo-adjuvant chemoradiation for locally advanced rectal cancer. Sci Rep 8:12611PubMedPubMedCentralCrossRef
110.
Zurück zum Zitat Ning Z, Luo J, Li Y et al (2019) Pattern classification for gastrointestinal stromal tumors by integration of radiomics and deep convolutional features. IEEE J Biomed Health Inform 23:1181–1191PubMedCrossRef Ning Z, Luo J, Li Y et al (2019) Pattern classification for gastrointestinal stromal tumors by integration of radiomics and deep convolutional features. IEEE J Biomed Health Inform 23:1181–1191PubMedCrossRef
111.
Zurück zum Zitat Shboul ZA, Alam M, Vidyaratne L, Pei L, Elbakary MI, Iftekharuddin KM (2019) Feature-guided deep radiomics for glioblastoma patient survival prediction. Front Neurosci 13:966PubMedPubMedCentralCrossRef Shboul ZA, Alam M, Vidyaratne L, Pei L, Elbakary MI, Iftekharuddin KM (2019) Feature-guided deep radiomics for glioblastoma patient survival prediction. Front Neurosci 13:966PubMedPubMedCentralCrossRef
Metadaten
Titel
Incorporating radiomics into clinical trials: expert consensus endorsed by the European Society of Radiology on considerations for data-driven compared to biologically driven quantitative biomarkers
verfasst von
Laure Fournier
Lena Costaridou
Luc Bidaut
Nicolas Michoux
Frederic E. Lecouvet
Lioe-Fee de Geus-Oei
Ronald Boellaard
Daniela E. Oprea-Lager
Nancy A Obuchowski
Anna Caroli
Wolfgang G. Kunz
Edwin H. Oei
James P. B. O’Connor
Marius E. Mayerhoefer
Manuela Franca
Angel Alberich-Bayarri
Christophe M. Deroose
Christian Loewe
Rashindra Manniesing
Caroline Caramella
Egesta Lopci
Nathalie Lassau
Anders Persson
Rik Achten
Karen Rosendahl
Olivier Clement
Elmar Kotter
Xavier Golay
Marion Smits
Marc Dewey
Daniel C. Sullivan
Aad van der Lugt
Nandita M. deSouza
European Society of Radiology
Publikationsdatum
25.01.2021
Verlag
Springer Berlin Heidelberg
Erschienen in
European Radiology / Ausgabe 8/2021
Print ISSN: 0938-7994
Elektronische ISSN: 1432-1084
DOI
https://doi.org/10.1007/s00330-020-07598-8

Weitere Artikel der Ausgabe 8/2021

European Radiology 8/2021 Zur Ausgabe

Akuter Schwindel: Wann lohnt sich eine MRT?

28.04.2024 Schwindel Nachrichten

Akuter Schwindel stellt oft eine diagnostische Herausforderung dar. Wie nützlich dabei eine MRT ist, hat eine Studie aus Finnland untersucht. Immerhin einer von sechs Patienten wurde mit akutem ischämischem Schlaganfall diagnostiziert.

Screening-Mammografie offenbart erhöhtes Herz-Kreislauf-Risiko

26.04.2024 Mammografie Nachrichten

Routinemäßige Mammografien helfen, Brustkrebs frühzeitig zu erkennen. Anhand der Röntgenuntersuchung lassen sich aber auch kardiovaskuläre Risikopatientinnen identifizieren. Als zuverlässiger Anhaltspunkt gilt die Verkalkung der Brustarterien.

S3-Leitlinie zu Pankreaskrebs aktualisiert

23.04.2024 Pankreaskarzinom Nachrichten

Die Empfehlungen zur Therapie des Pankreaskarzinoms wurden um zwei Off-Label-Anwendungen erweitert. Und auch im Bereich der Früherkennung gibt es Aktualisierungen.

Fünf Dinge, die im Kindernotfall besser zu unterlassen sind

18.04.2024 Pädiatrische Notfallmedizin Nachrichten

Im Choosing-Wisely-Programm, das für die deutsche Initiative „Klug entscheiden“ Pate gestanden hat, sind erstmals Empfehlungen zum Umgang mit Notfällen von Kindern erschienen. Fünf Dinge gilt es demnach zu vermeiden.

Update Radiologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.