scispace - formally typeset
Search or ask a question
Journal ArticleDOI

Radiomics: the bridge between medical imaging and personalized medicine

TL;DR: Radiomics, the high-throughput mining of quantitative image features from standard-of-care medical imaging that enables data to be extracted and applied within clinical-decision support systems to improve diagnostic, prognostic, and predictive accuracy, is gaining importance in cancer research as mentioned in this paper.
Abstract: Radiomics, the high-throughput mining of quantitative image features from standard-of-care medical imaging that enables data to be extracted and applied within clinical-decision support systems to improve diagnostic, prognostic, and predictive accuracy, is gaining importance in cancer research. Radiomic analysis exploits sophisticated image analysis tools and the rapid development and validation of medical imaging data that uses image-based signatures for precision diagnosis and treatment, providing a powerful tool in modern medicine. Herein, we describe the process of radiomics, its pitfalls, challenges, opportunities, and its capacity to improve clinical decision making, emphasizing the utility for patients with cancer. Currently, the field of radiomics lacks standardized evaluation of both the scientific integrity and the clinical relevance of the numerous published radiomics investigations resulting from the rapid growth of this area. Rigorous evaluation criteria and reporting guidelines need to be established in order for radiomics to mature as a discipline. Herein, we provide guidance for investigations to meet this urgent need in the field of radiomics.

Summary (1 min read)

Introduction

  • The addition of hydroxylamine to aldehydes and ketones is a well known reaction, indeed the sharp melting points of the highly crystalline oxime derivatives have traditionally been used to characterise the parent carbonyl compounds.
  • A higher stratum of control over the participating centres invokes the employment of remote space filling substituents to minimise the spatial disparity between the reactants.
  • Significantly, reactions which do not otherwise take place are shown to proceed well when steric buttresses are incorporated into the ortho-position/s of a benzene ring (positions analogous to R4 and R5).
  • The article does not cite any examples where bulky substituents, in positions other than ortho (e.g. R1 and R2), are able to act as predictable steric buttresses.

Results and discussion

  • The preparation of the targeted carbonyl substrates 1a–c and 1j–n involves as the key step coupling of the amines 6 with the appropriate acyl halides 7; oxidation of the resulting oamidobenzyl alcohol 8 furnishes the desired aldehydes (Scheme 2).
  • The aldehyde 1j is a tertiary amide analogue of 1a and it may be anticipated that the additional substitution on the olefinic tether (R2 = Me) will influence its reactivity.
  • Increasing the reaction temperature to 80 8C failed to promote further reaction, however heating a solution of the oxime in xylene at reflux (140 8C) leads to quantitative conversion to the tricyclic adduct 5k.

Conclusions

  • The influence of the space filling substituents, R1, R2 and R4 and of the electronic nature of R3 on the course of the reaction of the carbonyl compounds 1 with NH2OH, is quite striking.
  • In those cases where a sterically bulky group (R4≠ H) is positioned ortho to the carbonyl functionality tricyclic isoxazolobenzodiazepinones result, this tricyclic system also arises if the amide nitrogen is tertiary (R2 ≠ H).
  • In no case is an APT reaction observed when the olefinic moiety is substituted with a phenyl group, one explanation considers that this substituent has no significant influence on the electrophilicity of the pendant olefinic centre and therefore no ability to promote the internal cyclisation reaction.
  • That the substituent R2 may have both a steric and electronic role in facilitating tricycle formation cannot be dismissed.

Did you find this useful? Give us your feedback

Content maybe subject to copyright    Report

Radiomics
Citation for published version (APA):
Lambin, P., Leijenaar, R. T. H., Deist, T. M., Peerlings, J., de Jong, E. E. C., van Timmeren, J.,
Sanduleanu, S., Larue, R. T. H. M., Even, A. J. G., Jochems, A., van Wijk, Y., Woodruff, H., van Soest, J.,
Lustberg, T., Roelofs, E., van Elmpt, W., Dekker, A., Mottaghy, F. M., Wildberger, J. E., & Walsh, S.
(2017). Radiomics: the bridge between medical imaging and personalized medicine. Nature Reviews
Clinical Oncology, 14(12), 749-762. https://doi.org/10.1038/nrclinonc.2017.141
Document status and date:
Published: 01/12/2017
DOI:
10.1038/nrclinonc.2017.141
Document Version:
Publisher's PDF, also known as Version of record
Document license:
Taverne
Please check the document version of this publication:
• A submitted manuscript is the version of the article upon submission and before peer-review. There can
be important differences between the submitted version and the official published version of record.
People interested in the research are advised to contact the author for the final version of the publication,
or visit the DOI to the publisher's website.
• The final author version and the galley proof are versions of the publication after peer review.
• The final published version features the final layout of the paper including the volume, issue and page
numbers.
Link to publication
General rights
Copyright and moral rights for the publications made accessible in the public portal are retained by the authors and/or other copyright
owners and it is a condition of accessing publications that users recognise and abide by the legal requirements associated with these
rights.
• Users may download and print one copy of any publication from the public portal for the purpose of private study or research.
• You may not further distribute the material or use it for any profit-making activity or commercial gain
• You may freely distribute the URL identifying the publication in the public portal.
If the publication is distributed under the terms of Article 25fa of the Dutch Copyright Act, indicated by the “Taverne” license above,
please follow below link for the End User Agreement:
www.umlib.nl/taverne-license
Take down policy
If you believe that this document breaches copyright please contact us at:
repository@maastrichtuniversity.nl
providing details and we will investigate your claim.
Download date: 09 Aug. 2022

Imaging is an important technology in medical sci-
ence and is used in clinical practice to aid decision
making
1
. The role of medical imaging, however, is
swiftly evolving from being primarily a diagnos-
tic tool to also include a central role in the context
of personalized precision medicine
2
. In radiomics
3,4
,
digitally encrypted medical images that hold infor-
mation related to tumour pathophysiology are trans-
formed into mineable high-dimensional data
1
. This
information can be harnessed through quantitative
image analyses
5
and leveraged via clinical-decision
support systems (CDSS)
6
to improve medical decision-
making. Radiomics builds upon several decades of
computer-aided diagnosis, prognosis, and thera peutics
research
7,8
. The process used in radiomics involves the
identification of vast arrays of quantitative features
within digital images, storage of such data in federated
databases (that is, a system in which several independ-
ent databases function as a single entity) and the sub-
sequent mining of the data for knowledge extraction
and application
9
. Innumerable quantitative features
can now be extracted using high-throughput comput-
ing from medical images such as CT, MR, and/or PET.
The creation of databases that link immense volumes
of radiomics data (ideally with all other pertinent data)
from millions of patients to form vast, rapid learn-
ing healthcare (RLHC) networks is conceivable, but
presents a considerable data management hurdle
1013
.
Radiomics is not a panacea for clinical decision-
making. Radiomic features (such as intensity, shape,
texture or wavelet) offer information on cancer pheno-
type as well as the tumour microenvironment that is
distinct and complementary to other pertinent data
sources (including clinically obtained, treatment- related
or genomic data)
14
. Radiomics-derived data, when
combined with other pertinent data and correlated
and/or inferred with outcomes data, can produce
accurate robust evidence-basedCDSS.
The potential of radiomics to improve CDSS is
beyond doubt
15
and the field is evolving rapidly. The
principal challenge is the optimal collection and
integration of diverse multimodal data sources in a
quantitative manner that delivers unambiguous clin-
ical predictions that accurately and robustly enable
outcome prediction as a function of the impending
decisions
16
. Many published prediction models that
account for factors related to both disease and treat-
ment are available, but these models lack standardized
1
The D‑Lab: Decision Support
for Precision Medicine,
GROW – School for Oncology
and Developmental Biology,
Maastricht University Medical
Centre, Universiteitssingel 40,
6229 ER, Maastricht,
The Netherlands.
2
Department of Radiology
and Nuclear Medicine,
GROW – School for Oncology
and Developmental Biology,
Maastricht University
Medical Centre, Doctor
Tanslaan 12, 6229 ET,
Maastricht, The Netherlands.
3
Department of Radiation
Oncology (MAASTRO),
GROW – School for Oncology
and Developmental Biology,
Maastricht University
Medical Centre, Doctor
Tanslaan 12, 6229 ET,
Maastricht, The Netherlands.
4
Department of Nuclear
Medicine, University Hospital
RWTH Aachen,
Pauwelsstraße 30,
52074 Aachen, Germany.
*These authors contributed
equally to this work.
Correspondence to P.L.
philippe.lambin@
maastrichtuniversity.nl
doi:10.1038/nrclinonc.2017.141
Published online 4 Oct 2017
Radiomics: the bridge between medical
imaging and personalized medicine
Philippe Lambin
1
, Ralph T.H.Leijenaar
1
*, Timo M.Deist
1
*, Jurgen Peerlings
1,2
,
Evelyn E.C.de Jong
1
, Janita van Timmeren
1
, Sebastian Sanduleanu
1
,
Ruben T.H.M.Larue
1
, Aniek J.G.Even
1
, Arthur Jochems
1
, Yvonka van Wijk
1
,
Henry Woodruff
1
, Johan van Soest
3
, Tim Lustberg
3
, Erik Roelofs
1,3
, Wouter van Elmpt
3
,
Andre Dekker
3
, Felix M.Mottaghy
2,4
, Joachim E.Wildberger
2
and Sean Walsh
1
Abstract
|
Radiomics, the high-throughput mining of quantitative image features from
standard-of-care medical imaging that enables data to be extracted and applied within
clinical-decision support systems to improve diagnostic, prognostic, and predictive accuracy,
is gaining importance in cancer research. Radiomic analysis exploits sophisticated image analysis
tools and the rapid development and validation of medical imaging data that uses image-based
signatures for precision diagnosis and treatment, providing a powerful tool in modern medicine.
Herein, we describe the process of radiomics, its pitfalls, challenges, opportunities, and its
capacity to improve clinical decision making, emphasizing the utility for patients with cancer.
Currently, the field of radiomics lacks standardized evaluation of both the scientific integrity and
the clinical relevance of the numerous published radiomics investigations resulting from the
rapid growth of this area. Rigorous evaluation criteria and reporting guidelines need to be
established in order for radiomics to mature as a discipline. Herein, we provide guidance for
investigations to meet this urgent need in the field of radiomics.
REVIEWS
NATURE REVIEWS
|
CLINICAL ONCOLOGY VOLUME 14
|
DECEMBER 2017
|
749
©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

Exploratory
analysis
Medical
imaging
Data
selection
VOI
Prediction target
Imaging protocols
Test feature stability
Report protocols
Add prediction target
Report algorithms
Store data
Add clinical variables
Archetypal features
Robust segmentation
Feature selection
Internal validation
Radiomics
External validation
Report methodology
Modeling
Feature
extraction
RQS 1
RQS 2
RQS 3
RQS Total 36
Image
protocol
quality
+1 or +2
RQS
checkpoint 1
total: 2
+1+1+1
RQS
checkpoint 2
total: 3
Multiple
segmentation
Phantom
study
Imaging at
multiple
time points
Feature
reduction or
adjustment
for multiple
testing
Multivariable
analysis
Biological
correlates
Cut-off
analysis
Discrimination
statistics
Calibration
statistics
Prospective
study
Validation Comparison
to ‘gold
standard’
Cost-
effectiveness
analysis
Potential
clinical
applications
Open
science
and data
+1 +1 +1 +1 or +2 +1 or +2 +7 +2 +2 +1 -3 or +3 -5 to +5 +1 to +4
RQS
checkpoint 3
total: 31
Nature Reviews | Clinical Oncology
evaluation of their performance, reproducibility,
and/or clinical utility
17
. Consequently, these models
might not be appropriate forCDSS.
In this Review, we describe the process of radiomics
along with latest developments in the field. The pitfalls,
challenges, and opportunities presented by radiomics
to improve CDSS for personalized precision oncology
are highlighted, with an emphasis on the methodo-
logical aspects of radiomics prediction model devel-
opment and validation. We explore the advanced and
innovative information technologies that are essential
for the data management of diverse multimodal data
sources. Finally, we offer a vision of the necessary
steps to ensure continued progression and widespread
acceptance of both radiomics andCDSS.
The workflow of radiomics
Radiomics is defined as the quantitative mapping, that
is, extraction, analysis and modelling of many medical
image features in relation to prediction targets, such as
clinical end points and genomic features. A radiomics
study can be structured in five phases: data selection,
medical imaging, feature extraction, exploratory ana-
lysis, and modelling (FIG.1). To assess the quality of
radiomics studies, we propose the radiomics quality
score (RQS).
Data selection
Radiomic analyses begins with the choice of an imaging
protocol, the volume of interest (VOI) and a prediction
target—the event one wishes to predict. Typically, the
entire primary tumour is analysed and linked to available
data on treatment outcomes, such as survival. Radiomic
analyses can be performed on subregions of the tumour
(habitats), metastatic lesions, as well as in normal tis-
sues. Analysis of these regions might yield radiosensitive
phenotypes, which has implications for treatment plan-
ning strategies. Radiomics analysis, however, is not
restricted to radiotherapy and can be applied to any
image generated in the clinical setting (FIG.2).
The importance of using standardized imaging proto-
cols to eliminate unnecessary confounding variability is
recognized
9,18
; however, nonstandardized imaging proto-
cols are commonplace. Therefore, reproducibility and
Key points
Radiomics is becoming increasingly more important in medical imaging
The explosion of medical imaging data creates an environment ideal for
machine-learning and data-based science
Radiomics-based decision-support systems for precision diagnosis and treatment can
be a powerful tool in modern medicine
Large-scale data sharing is necessary for the validation and full potential that
radiomics represents
Standardized data collection, evaluation criteria, and reporting guidelines are
required for radiomics to mature as a discipline
Figure 1
|
Flowchart depicting the workflow of radiomics and the application of the RQS. The workflow includes the
necessary steps in a radiomic analysis. The RQS both rewards and penalizes the methodology and analyses of a study,
consequently encouraging the best scientific practice. RSQ, radiomics quality score; VOI, volume of interest.
REVIEWS
750
|
DECEMBER 2017
|
VOLUME 14 www.nature.com/nrclinonc
©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

Nature Reviews | Clinical Oncology
Cardiac CT Radiomics MACE Analysis
Phantom studies
An artificial structure that
imitates human tissue
properties is scanned on
multiple machines to
characterize scan output
against a known physical
standard.
comparability of radiomic studies can be achieved only
by extensive disclosure of imaging protocols. We wish
to emphasize this point, and provide examples of how
protocols should be reported in future radiomics studies
(Supplementary informationS1).
Medical imaging
Segmentation. VOIs are segmented manually or (semi-)
automatically
19
. This segmentation determines which vox-
els within an image are analysed, thus, the variability in seg-
mentation can introduce bias in the evaluation of derived
radiomic features
20
. Multiple-segmentation is a method to
limit the extent of this bias. Examples that enable robust
features to be observed
21
include: evaluation by multiple
clinicians, perturb segmentations with noise, combination
of diverse algorithms, or use different stages of the breath-
ing cycle. Key considerations are how the segmentation was
performed, and how sensitive the radiomics analysis is to
different segmentation methods
22
. For example, a semi-
automatic segmentation method can result in different
radiomic features than a manual delineation.
Phantom studies. The determination of inter-scanner and
inter-vendor variability of features is important in radi-
omics
23
. In cases in which radiomic studies rely on data
from multiple scanners, neglecting this variability can
jeopardize the analysis of studies—that is, the proposed
radiomic-based prediction model might not perform ade-
quately on external datasets if new data are acquired on
different scanners. As data from patients scanned on mul-
tiple devices is scarce and subject to uncertainties (such as
organ motion, or different imaging protocols), phantom
studies are a suitable means to gauge these uncertainties
and identify features that rely on the vendor. In essence,
phantom studies provide a risk-mitigation strategy to help
navigate from the current clinical imaging scenario to the
desired optimal imaging scenario.
Imaging at multiple time points. Additional sources
of variability in radiomics features are organ motion or
expansion or shrinkage of the target volume. Radiomics
features that are strongly dependent on these factors can
have limited applicability. To account for these sources of
variability, available test-retest data
24–26
can be exploited
to measure radiomics feature stability. For example, two
datasets of images acquired within a small period of time
from a patientcohort.
Feature extraction
The essence of radiomics is the high-throughput extrac-
tion of quantitative image features to characterize VOIs.
Feature values are dependent upon factors that can
include image pre-processing (for example, filtering, or
intensity discretization) and reconstruction (for exam-
ple, filtered back projection, or iterative reconstruction).
Furthermore, variation exists in feature nomenclature,
mathematical definition, methodology, and software
implementation of the applied feature extraction
algorithms
27–29
. In order to facilitate inter- operability
of radiomic features, differences in nomenclature,
algorithms, software implementations, as well as other
methodological aspects must be elucidated.
Exploratory analysis
Radiomic and non-radiomic features should be com-
bined with the prediction target to create a single
dataset. This approach enables the investigation of
relationships between features. Groups of highly cor-
related radiomics features can be identified via clus-
tering, and these features can be reduced to single
archetypal features per cluster. Radiomic features that
are well-correlated with routine clinical features (such
as tumour stage) do not provide additional information.
Auxiliary feature data collected from multiple segmen-
tations, multiple imaging, and phantom studies, can be
exploited to assess feature robustness. Volatile or robust
features can be identified and subsequently excluded
from model development. For example, a feature that
is robust for the prediction of overall survival for lung
cancer (that is, imaged and segmented in a certain way)
for a given dataset could be volatile for the prediction of
pneumonitis in lung cancer (imaged and segmented in
an alternative way) for a given dataset. Thus, the pro-
cess of feature reduction and/or exclusion should be
described clearly.
Modelling
Radiomic modelling involves three major aspects: fea-
ture selection, modelling methodology, and validation.
Feature selection should be data-driven owing to the
vast in- human range of possible radiomics features; such
analysis should be performed in a robust and transparent
manner. To achieve holistic models, features beyond radi-
omics (such as data from clinical records, data obtained
during treatment or biological and/or genetic) should
also be incorporated. Regarding the choice of modelling
methodology, the identification of optimal machine-
learning methods for radiomic applications is a crucial
step towards stable and clinically relevant CDSS; thus, in
the ideal scenario, multiple machine-learning methods
should be employed
30
and the implementation should be
comprehensively documented. A non-validated model is
Figure 2
|
Radiomics in cardiology. The current gold standard
for quantification of coronary calcifications visible on CT is the
‘Agatston’ method (based upon intensity and volume).
Radiomic features can improve quantification, differentiation
between calcified and non-calcified plaques, and thus the
prediction of Major Adverse Cardiac Events (MACE).
REVIEWS
NATURE REVIEWS
|
CLINICAL ONCOLOGY VOLUME 14
|
DECEMBER 2017
|
751
©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

Calibration-in-the-large
Describes whether the
predictions deviate
systematically (intercept),
whereas the calibration slope
should ideally be equal to 1.
The independence
assumption
The definition in terms of
conditional probabilities is that
the probability of B is not
changed by knowing that A has
occurred. Statistically
independent variables are
always uncorrelated, but the
converse is not necessarily
true.
Feature discretization
The process of converting
continuous features to discrete
binned interval features.
Bootstrapping
Measures the accuracy
(defined in terms of bias,
variance, confidence intervals,
prediction error, etc.) to
characterize the sample
distribution by way of repeated
random sampling methods.
of limited value; validation is an indispensable compo-
nent of a complete radiomic analysis. Models must be
internally validated and, ideally, should be externally
validated.
Feature selection. Depending on the number of filters,
feature categories, and other adjustable parameters, the
possible number of radiomic features that can be extracted
from images is virtually unlimited. The inclusion of
all possible features in a model would inevitably result
in overfitting, which jeopardizes model performance in
patients not previously evaluated. To avoid overfitting,
features that lack robustness against sources of variability
should be eliminated, and archetypal features selected via
dimensionality reduction techniques (such as principal
component analysis or clustering). For example, a feature
that is archetypal for the prediction of overall survival
in patients with lung cancer for a given dataset (imaged
and segmented in a certain way) could be redundant for
the prediction of pneumonitis in lung cancer for a given
dataset (imaged and segmented in an alternative way).
Modelling methodology. The modelling methodology
chosen is often a single technique, selected according to
the preference and experience of those conducting the
study. Different techniques are associated with distinct
inherent limitations, which include the independence
assumption for features in logistic regression, the need for
feature discretization in Bayesian networks, or the network
configuration dependency in deep learning. The choice of
modelling technique has been shown to affect prediction
performance in radiomics
30
. Thus, multiple- modelling
methodology implementations are desirable, but not
essential. The key aspect in the selection of a modelling
methodology is that, when reported, the work must be
entirely reproducible. This goal can be achieved, ideally,
by making the software code available (for example,
via github
31
). (See Supplementary information S1 material
for an overview of machine learning techniques).
Validation. Validation techniques are useful tools
to assess model performance, and thus, internal
and/or preferably external validation must be performed.
Researchers must assess whether the model is predic-
tive for the target patient population or just for a par-
ticular subset of samples analysed. Model performance
is typically measured in terms of discrimination and
calibration. Discrimination can be reported in terms of
the receiver operating characteristic (ROC) curve, or the
area under the ROC curve (AUC). The AUC quantifies
the sensitivity and specificity of the model and repre-
sents the probability that a randomly selected patient
matching an outcome is assigned that outcome by the
prediction model with a larger event-probability than
a randomly chosen patient who does not match the
outcome. Calibration refers to the agreement between
observed outcomes and model predictions, typically
based on grouping of predictions. For example, the
predictions are grouped according to high, medium or
low prob ability. If the mean prediction of tumour recur-
rence in the high-probability group is 25%, the observed
frequency of tumour recurrence in this group should ide-
ally be 25 out of 100 patients. Calibration can be reported
using a calibration plot and calibration-in-the-large/slope.
A measure of overall performance is the Brier score,
the mean squared prediction error. All statistical meth-
ods should be reported for training data and validation
data. Valid models should exhibit statistical consistency
between the training and validation sets. Bootstrapping
techniques can be used to estimate confidence intervals
for the abovementioned statistics and should be reported.
An externally validated model has more credibility than
an internally validated model, because data obtained with
the former approach are considered more independent,
which reinforces the validation. A large body of literature
on validation techniques is available
32–35
.
Reporting open-access scientific data
Validation is the first step towards a model being
accepted in both the scientific and clinical communi-
ties. Independent verification of the results is a necessary
additional step. Reproduction means verification of the
results by independent researchers repeating the ana-
lysis using an identical technique and the same dataset
and/or patient cohort, ensuring that the analysis is con-
ducted without error. Replication means independent
verification of the results by independent researchers
repeating the analysis using the same technique and
different (but appropriately selected) datasets and/or
patient cohorts, aiming for a stronger affirmation of the
findings
36–39
. Radiomic studies involve multiple complex
subprocesses (such as data selection, image acquisition,
feature extraction, or modelling), each one affected by a
wide range of decisions, use of nonstandardized terminol-
ogy, establishment of parameters, and software selection.
Reproducibility and replicability in radiomics are impos-
sible if researchers do not disclose these intricacies. The
amount of necessary information far exceeds the limits
of a traditional manuscript. We propose that future pub-
lications including radiomic results should provide the
following as supplementary material: disclosure of imag-
ing protocols, analysed scans, segmentations of VOIs,
detailed accounts of how features were extracted (includ-
ing the formulae), and of the modelling metho dology
used (ideally, the code). This level of meticulous detail is
required in order to facilitate reproduction and replica-
tion. Furthermore, multiple radiomics software packages
are available and are subject to updates or version-control.
We recognize that the publication of data derived from
patients might not be feasible in all circumstances. As a
minimal means of comparison, and to alleviate this lack of
transparency, we propose that researchers publish numer-
ical values of their investigated features computed on the
digital phantom described in the supplementary material
of this manuscript (available online
40
).
To compare different software implementations for
radiomic feature-extraction algorithms, we present an
example, in which CT-obtained data of the primary
tumour region and the corresponding tumour contours
of four patients with lung cancer serve as ‘real-life’ digital
phantoms (FIG.3). Using the preprocessed image data, we
calculated a set of commonly used features to serve as a
REVIEWS
752
|
DECEMBER 2017
|
VOLUME 14 www.nature.com/nrclinonc
©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

Citations
More filters
Journal ArticleDOI
TL;DR: A set of 169 radiomics features was standardized, which enabled verification and calibration of different radiomics software and could be excellently reproduced.
Abstract: Background Radiomic features may quantify characteristics present in medical imaging. However, the lack of standardized definitions and validated reference values have hampered clinical use. Purpose To standardize a set of 174 radiomic features. Materials and Methods Radiomic features were assessed in three phases. In phase I, 487 features were derived from the basic set of 174 features. Twenty-five research teams with unique radiomics software implementations computed feature values directly from a digital phantom, without any additional image processing. In phase II, 15 teams computed values for 1347 derived features using a CT image of a patient with lung cancer and predefined image processing configurations. In both phases, consensus among the teams on the validity of tentative reference values was measured through the frequency of the modal value and classified as follows: less than three matches, weak; three to five matches, moderate; six to nine matches, strong; 10 or more matches, very strong. In the final phase (phase III), a public data set of multimodality images (CT, fluorine 18 fluorodeoxyglucose PET, and T1-weighted MRI) from 51 patients with soft-tissue sarcoma was used to prospectively assess reproducibility of standardized features. Results Consensus on reference values was initially weak for 232 of 302 features (76.8%) at phase I and 703 of 1075 features (65.4%) at phase II. At the final iteration, weak consensus remained for only two of 487 features (0.4%) at phase I and 19 of 1347 features (1.4%) at phase II. Strong or better consensus was achieved for 463 of 487 features (95.1%) at phase I and 1220 of 1347 features (90.6%) at phase II. Overall, 169 of 174 features were standardized in the first two phases. In the final validation phase (phase III), most of the 169 standardized features could be excellently reproduced (166 with CT; 164 with PET; and 164 with MRI). Conclusion A set of 169 radiomics features was standardized, which enabled verification and calibration of different radiomics software. © RSNA, 2020 Online supplemental material is available for this article. See also the editorial by Kuhl and Truhn in this issue.

1,563 citations


Cites background or methods from "Radiomics: the bridge between medic..."

  • ...We defined a general radiomics image processing scheme based on descriptions in the literature (3,6,17,20)....

    [...]

  • ...noninvasive, readily available in clinical care, and repeatable (3,4)....

    [...]

Journal ArticleDOI
TL;DR: It is found that none of the models identified are of potential clinical use due to methodological flaws and/or underlying biases, which is a major weakness, given the urgency with which validated COVID-19 models are needed.
Abstract: Machine learning methods offer great promise for fast and accurate detection and prognostication of coronavirus disease 2019 (COVID-19) from standard-of-care chest radiographs (CXR) and chest computed tomography (CT) images. Many articles have been published in 2020 describing new machine learning-based models for both of these tasks, but it is unclear which are of potential clinical utility. In this systematic review, we consider all published papers and preprints, for the period from 1 January 2020 to 3 October 2020, which describe new machine learning models for the diagnosis or prognosis of COVID-19 from CXR or CT images. All manuscripts uploaded to bioRxiv, medRxiv and arXiv along with all entries in EMBASE and MEDLINE in this timeframe are considered. Our search identified 2,212 studies, of which 415 were included after initial screening and, after quality screening, 62 studies were included in this systematic review. Our review finds that none of the models identified are of potential clinical use due to methodological flaws and/or underlying biases. This is a major weakness, given the urgency with which validated COVID-19 models are needed. To address this, we give many recommendations which, if followed, will solve these issues and lead to higher-quality model development and well-documented manuscripts. Many machine learning-based approaches have been developed for the prognosis and diagnosis of COVID-19 from medical images and this Analysis identifies over 2,200 relevant published papers and preprints in this area. After initial screening, 62 studies are analysed and the authors find they all have methodological flaws standing in the way of clinical utility. The authors have several recommendations to address these issues.

581 citations

Journal ArticleDOI
TL;DR: The major issues regarding this multi-step process, focussing in particular on challenges of the extraction of radiomic features from data sets provided by computed tomography, positron emission tomographic, and magnetic resonance imaging are summarised.
Abstract: Radiomics is an emerging translational field of research aiming to extract mineable high-dimensional data from clinical images. The radiomic process can be divided into distinct steps with definable inputs and outputs, such as image acquisition and reconstruction, image segmentation, features extraction and qualification, analysis, and model building. Each step needs careful evaluation for the construction of robust and reliable models to be transferred into clinical practice for the purposes of prognosis, non-invasive disease tracking, and evaluation of disease response to treatment. After the definition of texture parameters (shape features; first-, second-, and higher-order features), we briefly discuss the origin of the term radiomics and the methods for selecting the parameters useful for a radiomic approach, including cluster analysis, principal component analysis, random forest, neural network, linear/logistic regression, and other. Reproducibility and clinical value of parameters should be firstly tested with internal cross-validation and then validated on independent external cohorts. This article summarises the major issues regarding this multi-step process, focussing in particular on challenges of the extraction of radiomic features from data sets provided by computed tomography, positron emission tomography, and magnetic resonance imaging.

579 citations


Cites background from "Radiomics: the bridge between medic..."

  • ...Radiomics’ analyses can be performed in tumour regions, metastatic lesions, as well as in normal tissues [2]....

    [...]

Journal ArticleDOI
TL;DR: How the tumour-reprogrammed lung microenvironment can contribute to primary lung tumour progression as well as lung metastasis from extrapulmonary neoplasms by promoting inflammation, angiogenesis, immune modulation and therapeutic responses is discussed.
Abstract: Lung cancer is a major global health problem, as it is the leading cause of cancer-related deaths worldwide. Major advances in the identification of key mutational alterations have led to the development of molecularly targeted therapies, whose efficacy has been limited by emergence of resistance mechanisms. US Food and Drug Administration (FDA)-approved therapies targeting angiogenesis and more recently immune checkpoints have reinvigorated enthusiasm in elucidating the prognostic and pathophysiological roles of the tumour microenvironment in lung cancer. In this Review, we highlight recent advances and emerging concepts for how the tumour-reprogrammed lung microenvironment promotes both primary lung tumours and lung metastasis from extrapulmonary neoplasms by contributing to inflammation, angiogenesis, immune modulation and response to therapies. We also discuss the potential of understanding tumour microenvironmental processes to identify biomarkers of clinical utility and to develop novel targeted therapies against lung cancer.

552 citations

Journal ArticleDOI
TL;DR: An overview of current and next-generation methods for federated, secure and privacy-preserving artificial intelligence with a focus on medical imaging applications, alongside potential attack vectors and future prospects in medical imaging and beyond are presented.
Abstract: The broad application of artificial intelligence techniques in medicine is currently hindered by limited dataset availability for algorithm training and validation, due to the absence of standardized electronic medical records, and strict legal and ethical requirements to protect patient privacy. In medical imaging, harmonized data exchange formats such as Digital Imaging and Communication in Medicine and electronic data storage are the standard, partially addressing the first issue, but the requirements for privacy preservation are equally strict. To prevent patient privacy compromise while promoting scientific research on large datasets that aims to improve patient care, the implementation of technical solutions to simultaneously address the demands for data protection and utilization is mandatory. Here we present an overview of current and next-generation methods for federated, secure and privacy-preserving artificial intelligence with a focus on medical imaging applications, alongside potential attack vectors and future prospects in medical imaging and beyond. Medical imaging data is often subject to privacy and intellectual property restrictions. AI techniques can help out by offering tools like federated learning to bridge the gap between personal data protection and data utilisation for research and clinical routine, but these tools need to be secure.

487 citations

References
More filters
Journal ArticleDOI
TL;DR: A substantial proportion of the worldwide burden of cancer could be prevented through the application of existing cancer control knowledge and by implementing programs for tobacco control, vaccination, and early detection and treatment, as well as public health campaigns promoting physical activity and a healthier dietary intake.
Abstract: The global burden of cancer continues to increase largely because of the aging and growth of the world population alongside an increasing adoption of cancer-causing behaviors, particularly smoking, in economically developing countries. Based on the GLOBOCAN 2008 estimates, about 12.7 million cancer cases and 7.6 million cancer deaths are estimated to have occurred in 2008; of these, 56% of the cases and 64% of the deaths occurred in the economically developing world. Breast cancer is the most frequently diagnosed cancer and the leading cause of cancer death among females, accounting for 23% of the total cancer cases and 14% of the cancer deaths. Lung cancer is the leading cancer site in males, comprising 17% of the total new cancer cases and 23% of the total cancer deaths. Breast cancer is now also the leading cause of cancer death among females in economically developing countries, a shift from the previous decade during which the most common cause of cancer death was cervical cancer. Further, the mortality burden for lung cancer among females in developing countries is as high as the burden for cervical cancer, with each accounting for 11% of the total female cancer deaths. Although overall cancer incidence rates in the developing world are half those seen in the developed world in both sexes, the overall cancer mortality rates are generally similar. Cancer survival tends to be poorer in developing countries, most likely because of a combination of a late stage at diagnosis and limited access to timely and standard treatment. A substantial proportion of the worldwide burden of cancer could be prevented through the application of existing cancer control knowledge and by implementing programs for tobacco control, vaccination (for liver and cervical cancers), and early detection and treatment, as well as public health campaigns promoting physical activity and a healthier dietary intake. Clinicians, public health professionals, and policy makers can play an active role in accelerating the application of such interventions globally.

52,293 citations

Journal ArticleDOI
03 Apr 2015-Science
TL;DR: Treatment efficacy was associated with a higher number of mutations in the tumors, and a tumor-specific T cell response paralleled tumor regression in one patient, suggesting that the genomic landscape of lung cancers shapes response to anti–PD-1 therapy.
Abstract: Immune checkpoint inhibitors, which unleash a patient’s own T cells to kill tumors, are revolutionizing cancer treatment. To unravel the genomic determinants of response to this therapy, we used whole-exome sequencing of non–small cell lung cancers treated with pembrolizumab, an antibody targeting programmed cell death-1 (PD-1). In two independent cohorts, higher nonsynonymous mutation burden in tumors was associated with improved objective response, durable clinical benefit, and progression-free survival. Efficacy also correlated with the molecular smoking signature, higher neoantigen burden, and DNA repair pathway mutations; each factor was also associated with mutation burden. In one responder, neoantigen-specific CD8+ T cell responses paralleled tumor regression, suggesting that anti–PD-1 therapy enhances neoantigen-specific T cell reactivity. Our results suggest that the genomic landscape of lung cancers shapes response to anti–PD-1 therapy.

6,215 citations

Journal ArticleDOI
TL;DR: The recurrence score has been validated as quantifying the likelihood of distant recurrence in tamoxifen-treated patients with node-negative, estrogen-receptor-positive breast cancer and could be used as a continuous function to predict distant recurrent in individual patients.
Abstract: background The likelihood of distant recurrence in patients with breast cancer who have no involved lymph nodes and estrogen-receptor–positive tumors is poorly defined by clinical and histopathological measures. methods We tested whether the results of a reverse-transcriptase–polymerase-chain-reaction (RT-PCR) assay of 21 prospectively selected genes in paraffin-embedded tumor tissue would correlate with the likelihood of distant recurrence in patients with node-negative, tamoxifen-treated breast cancer who were enrolled in the National Surgical Adjuvant Breast and Bowel Project clinical trial B-14. The levels of expression of 16 cancerrelated genes and 5 reference genes were used in a prospectively defined algorithm to calculate a recurrence score and to determine a risk group (low, intermediate, or high) for each patient. results Adequate RT-PCR profiles were obtained in 668 of 675 tumor blocks. The proportions of patients categorized as having a low, intermediate, or high risk by the RT-PCR assay were 51, 22, and 27 percent, respectively. The Kaplan–Meier estimates of the rates of distant recurrence at 10 years in the low-risk, intermediate-risk, and high-risk groups were 6.8 percent (95 percent confidence interval, 4.0 to 9.6), 14.3 percent (95 percent confidence interval, 8.3 to 20.3), and 30.5 percent (95 percent confidence interval, 23.6 to 37.4). The rate in the low-risk group was significantly lower than that in the high-risk group (P<0.001). In a multivariate Cox model, the recurrence score provided significant predictive power that was independent of age and tumor size (P<0.001). The recurrence score was also predictive of overall survival (P<0.001) and could be used as a continuous function to predict distant recurrence in individual patients. conclusions The recurrence score has been validated as quantifying the likelihood of distant recurrence in tamoxifen-treated patients with node-negative, estrogen-receptor–positive breast cancer.

5,685 citations

Journal ArticleDOI
TL;DR: Pembrolizumab had an acceptable side-effect profile and showed antitumor activity in patients with advanced non-small-cell lung cancer and PD-L1 expression in at least 50% of tumor cells correlated with improved efficacy of pembrolIZumab.
Abstract: BackgroundWe assessed the efficacy and safety of programmed cell death 1 (PD-1) inhibition with pembrolizumab in patients with advanced non–small-cell lung cancer enrolled in a phase 1 study. We also sought to define and validate an expression level of the PD-1 ligand 1 (PD-L1) that is associated with the likelihood of clinical benefit. MethodsWe assigned 495 patients receiving pembrolizumab (at a dose of either 2 mg or 10 mg per kilogram of body weight every 3 weeks or 10 mg per kilogram every 2 weeks) to either a training group (182 patients) or a validation group (313 patients). We assessed PD-L1 expression in tumor samples using immunohistochemical analysis, with results reported as the percentage of neoplastic cells with staining for membranous PD-L1 (proportion score). Response was assessed every 9 weeks by central review. ResultsCommon side effects that were attributed to pembrolizumab were fatigue, pruritus, and decreased appetite, with no clear difference according to dose or schedule. Among all ...

4,834 citations

Journal ArticleDOI
TL;DR: This report describes the process of radiomics, its challenges, and its potential power to facilitate better clinical decision making, particularly in the care of patients with cancer.
Abstract: In the past decade, the field of medical image analysis has grown exponentially, with an increased number of pattern recognition tools and an increase in data set sizes. These advances have facilitated the development of processes for high-throughput extraction of quantitative features that result in the conversion of images into mineable data and the subsequent analysis of these data for decision support; this practice is termed radiomics. This is in contrast to the traditional practice of treating medical images as pictures intended solely for visual interpretation. Radiomic data contain first-, second-, and higher-order statistics. These data are combined with other patient data and are mined with sophisticated bioinformatics tools to develop models that may potentially improve diagnostic, prognostic, and predictive accuracy. Because radiomics analyses are intended to be conducted with standard of care images, it is conceivable that conversion of digital images to mineable data will eventually become routine practice. This report describes the process of radiomics, its challenges, and its potential power to facilitate better clinical decision making, particularly in the care of patients with cancer.

4,773 citations

Related Papers (5)
Frequently Asked Questions (19)
Q1. What are the future works mentioned in the paper "Radiomics: the bridge between medical imaging and personalized medicine" ?

Picture archiving and radiomics knowledge systems ( PARKS ) of the future will identify, segment, and extract features from regions of interest. If previous images associated with the same patient are accessible, the earlier identified regions of interest will be automatically identified by the PARKS software. Quantitative image features that are uploaded to a shared database and compared with previous images will be automatically extracted by the PARKS to enhance CDSS for diagnosis, prognosis, and treatment, resulting in improved personalization and precision medicine ( FIG. 7 ). 

Herein, the authors describe the process of radiomics, its pitfalls, challenges, opportunities, and its capacity to improve clinical decision making, emphasizing the utility for patients with cancer. Herein, the authors provide guidance for investigations to meet this urgent need in the field of radiomics. 

Picture archiving and radiomics knowledge systems ( PARKS ) of the future will identify, segment, and extract features from regions of interest. If previous images associated with the same patient are accessible, the earlier identified regions of interest will be automatically identified by the PARKS software. Quantitative image features that are uploaded to a shared database and compared with previous images will be automatically extracted by the PARKS to enhance CDSS for diagnosis, prognosis, and treatment, resulting in improved personalization and precision medicine ( FIG. 7 ). 

In the field of oncology, a promising research area is that of biomarkers — in particular, biomarkers for immunotherapy and imaging biomarkers90,91. 

The effect of SUV discretization in quantitative FDG-PET radiomics: the need for standardized methodology in tumor texture analysis. 

The quantification of the radiosensitivity of human tumours is presently performed on the basis of the ex vivo tumour survival fraction, and the detection of unrepaired DNA double-strand breaks82,83. 

To achieve holistic models, features beyond radiomics (such as data from clinical records, data obtained during treatment or biological and/or genetic) should also be incorporated. 

In addition, universal streamlined solutions through advanced information communication technologies have been central to the realization of this endeavor, readily facilitating synchronized RLHC in each centre without inclusion of sensitive data, which overcomes the classic barriers to data sharing. 

In essence, phantom studies provide a risk-mitigation strategy to help navigate from the current clinical imaging scenario to the desired optimal imaging scenario. 

Radiomic studies should incorporate reproducibility assessments owing to the beneficial ethical, economic and logistical effects they have (such asinforming power calculations and required samples sizes, multicentric trial duration and trial cost). 

Feature selection should be data-driven owing to the vast in- human range of possible radiomics features; such analysis should be performed in a robust and transparent manner. 

A pressing need to embrace knowledge and data-sharing technology106, which transcends institutional and national boundaries107, drives both the research and clinical communities. 

85. Chitnis, M. M. et al. IGF-1R inhibition enhances radiosensitivity and delays double-strand break repair by both non-homologous end-joining and homologous recombination. 

Although the minute technical details of radiomics are tedious, they can greatly influence robustness, generalizability, and confound meta- analyses. 

Examples that enable robust features to be observed21 include: evaluation by multiple clinicians, perturb segmentations with noise, combination of diverse algorithms, or use different stages of the breathing cycle. 

Exploiting this technique, the ontology terms serve as a common reference for the data at each institutional site, permitting a unified process for information retrieval enabled by a semantic gateway to the data. 

these approaches have been undermined by the presence of substantial experimental variability rather than by the existence of interpatient variations in radiosensitivity. 

Groups of highly correlated radiomics features can be identified via clustering, and these features can be reduced to single archetypal features per cluster. 

Such capabilities are on the technological, scientific, and clinical horizons, as most current picture archiving and communication systems have the capability to co-register current images with previous images and perform user-interactive segmentation.