You are on page 1of 17

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/351238113

Combining liquid biopsy and radiomics for personalized treatment of lung


cancer patients. State of the art and new perspectives

Article  in  Pharmacological Research · April 2021


DOI: 10.1016/j.phrs.2021.105643

CITATIONS READS

2 62

12 authors, including:

Federico Cucchiara Iacopo Petrini


Università di Pisa Università di Pisa
42 PUBLICATIONS   132 CITATIONS    119 PUBLICATIONS   2,119 CITATIONS   

SEE PROFILE SEE PROFILE

Chiara Romei Stefania Crucitta


Azienda Ospedaliero-Universitaria Pisana Università di Pisa
45 PUBLICATIONS   173 CITATIONS    74 PUBLICATIONS   792 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Pharmacogenetics & breast cancer View project

CftDNA and CRC View project

All content following this page was uploaded by Federico Cucchiara on 14 May 2021.

The user has requested enhancement of the downloaded file.


Pharmacological Research 169 (2021) 105643

Contents lists available at ScienceDirect

Pharmacological Research
journal homepage: www.elsevier.com/locate/yphrs

Review

Combining liquid biopsy and radiomics for personalized treatment of lung


cancer patients. State of the art and new perspectives
Federico Cucchiara a, Iacopo Petrini b, Chiara Romei c, Stefania Crucitta a, Maurizio Lucchesi b,
Simona Valleggi b, Cristina Scavone d, Annalisa Capuano d, Annalisa De Liperi c, Antonio Chella b,
Romano Danesi a, *, Marzia Del Re a
a
Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
b
Unit of Pneumology, Department of Translational Research and New Technologies in Medicine, University Hospital of Pisa, Pisa, Italy
c
Unit II of Radio-diagnostics, Department of Diagnostic and Imaging, University Hospital of Pisa, Pisa, Italy
d
Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy

A R T I C L E I N F O A B S T R A C T

Keywords: Lung cancer has become a paradigm for precision medicine in oncology, and liquid biopsy (LB) together with
Lung cancer radiomics may have a great potential in this scenario. They are both minimally invasive, easy to perform, and can
Liquid biopsy (LB) be repeated during patient’s follow-up. Also, increasing evidence suggest that LB and radiomics may provide an
Radiomics
efficient way to screen and diagnose tumors at an early stage, including the monitoring of any change in the
Artificial Intelligence (AI)
Precision medicine
tumor molecular profile. This could allow treatment optimization, improvement of patients’ quality of life, and
healthcare-related costs reduction. Latest reports on lung cancer patients suggest a combination of these two
strategies, along with cutting-edge data analysis, to decode valuable information regarding tumor type,
aggressiveness, progression, and response to treatment. The approach seems more compatible with clinical
practice than the current standard, and provides new diagnostic companions being able to suggest the best
treatment strategy compared to conventional methods. To implement radiomics and liquid biopsy directly into
clinical practice, an artificial intelligence (AI)-based system could help to link patients’ clinical data together
with tumor molecular profiles and imaging characteristics. AI could also solve problems and limitations related
to LB and radiomics methodologies. Further work is needed, including new health policies and the access to large
amounts of high-quality and well-organized data, allowing a complementary and synergistic combination of LB
and imaging, to provide an attractive choice e in the personalized treatment of lung cancer.

2–5%), v-RAF murine sarcoma viral homolog B1 (BRAF; 1–3%) and


1. Introduction mitogen-activated protein (MAP) kinase/extracellular signal-regulated
kinase (ERK) 1 (MEK1; 1%) genes, mesenchymal-epithelial transition
Lung cancer is the second most common neoplasm in both males and factor (c-MET) gene amplification (3.5%), and rearrangements in the
females [1] and the leading cause of tumor death in western countries anaplastic lymphoma kinase (ALK; 3%) or ROS proto-oncogene 1
[1,2] accounting for 18.4% of mortality, with 1.8 million deaths receptor tyrosine kinase (ROS1; 1%) genes [4,5]. Moreover,
worldwide every year [1,2]. Since 2015, tumor molecular profile is part remarkable differences have been reported in the frequency of the
of the histopathological and immunohistochemical characterization at genetic landscape of smoking-related versus non-smoking-related can­
disease diagnosis [3]. Several oncogenic drivers have been identified in cers [6,7], highlighting also a different spectrum of mutated genes, such
non-small cell lung cancer (NSCLC), including the epidermal growth as c-MET, RAS, tumor protein 53 (TP53), BRAF, AT-rich interaction
factor receptor (EGFR; 10–30% of cases), fibroblast growth factor re­ domain 1 A (ARID1A) and mismatch repair genes mutations in smokers
ceptor 1 (FGFR1; 20%), Kirsten-rat sarcoma (KRAS; 15–30%), versus EGFR mutations, ALK and ROS1 fusions in never-smokers [7].
phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit alpha The identification of such mutations led to the development of the
(PIK3CA; 2–5%), human epidermal growth factor receptor 2 (HER2; paradigm of targeted anticancer therapies and has raised the clinical

* Correspondence to: Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, 56126,
Pisa, Italy.
E-mail address: romano.danesi@unipi.it (R. Danesi).

https://doi.org/10.1016/j.phrs.2021.105643
Received 11 March 2021; Received in revised form 22 April 2021; Accepted 22 April 2021
Available online 30 April 2021
1043-6618/© 2021 Elsevier Ltd. All rights reserved.
F. Cucchiara et al. Pharmacological Research 169 (2021) 105643

Nomenclature MS-PCR methylation-specific PCR


NBC naive bayesian classifier
ALK anaplastic lymphoma kinase NGS next generation sequencing
ARID1A AT-rich interaction domain 1A NSCLC non-small cell lung cancer
ARMS-PCR amplification-refractory mutation system PCR ORR overall response rate HU, Hounsfield Unit
AUROC area under the receiver operator characteristic curve OS overall survival
BFAST blood first assay screening trial PCR polymerase chain reaction
BRAF v-RAF murine sarcoma viral homolog B1 PD-1 programmed cell death-protein-1
CD cluster of differentiation PD-L1 programmed death-ligand 1
cfDNA cell free DNA PET positron emission tomography
cfRNA cell free RNA PFS progression-free survival
cMET mesenchymal-epithelial transition factor Pik platelet isolation kit
CT computed tomography PIK3CA phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic
CTAG1B Cancer/Testis Antigen 1B subunit alpha
CTCs circulating tumor cells PLAP placental alkaline phosphatase
CTLA-4 cytotoxic T-lymphocytes associated with protein 4 PPV positive predictive values
ddPCR droplet digital PCR PTGER4 prostaglandin E receptor 4
dsDNA double-stranded DNA qPCR quantitative PCR
DT decision tree RET rearranged during transfection
EGFR epidermal growth factor receptor RF random forest
EMA European Medicines Agency ROS1 ROS proto-oncogene 1 receptor tyrosine kinase
FA fractional abundance SCLC small cell lung cancer
FDA Food and Drug Administration SEC Size exclusion chromatography
FDG fluorodeoxyglucose SFTPD surfactant protein D
FGFR1 fibroblast growth factor receptor 1 SHOX2 short stature homeobox 2
FISH Fluorescence in situ hybridization SNV single-nucleotide variants
HER2 human epidermal growth factor receptor 2 SUV standardized uptake value
IHC immunohistochemistry SVM support vector machine
IFS immuno fluorescent staining TCGA the cancer genome atlas
KNN K-nearest neighbor TEPs tumor educated platelet
KRAS Kirsten-rat sarcoma TKI tyrosine kinase inhibitor
LB liquid biopsy TMB tumor mutational burden
MEK mitogen-activated protein (MAP) kinase/extracellular TP53 tumor protein 53
signal-regulated kinase (ERK) 1 TSPAN8 tetraspanin 8
miR miRNA TTF time-to-treatment failure
miRNA micro-RNA VOI volume of interest
MLR multinomial logistic regression WES whole exome sequencing
mRNA messenger RNA

need to develop sensitive technologies for the search of prognostic and combination of LB and imaging quantitative features; (3) the study re­
predictive biomarkers. ported data from abstracts on such methodologies presented at inter­
national meetings but not yet published in extenso on scientific journals.
2. Sources and selection criteria Moreover, additional articles, such as reviews and editorials which dealt
with the general concepts of the review, were also included.
This narrative review looked at published data from PubMed, Sco­
pus, and Web of Science. The databases were investigated for relevant 3. Genomics through liquid biopsy. The reason why
research articles published between January 2010 and September 2020, At a molecular level, tumor progression arises from the selection of
using the key search terms “cancer”, “lung cancer”, “NSCLC”, “liquid independent genetic mutations in distinct tumor-cell sub-populations
biopsy”, “radiomic”, “radiogenomic”, “biomarkers”, “artificial intelli­ over time, and across different disease sites [8]. These sub-clones have
gence”, “machine learning”, “deep learning”, “prognostic”, “predictive”, different capacities to grow, invade, and metastasize, but also determine
“targeted therapy”, “TKI”, “immunotherapy”. These searches were primary and acquired resistance to treatment [9,10]. Interestingly, Hu
combined using appropriate Boolean operators to yield 2292 abstracts, Zheng and colleagues [11] compared the whole exosome sequencing
which were hand searched. The full text was obtained for articles (WES) data of 457 paired primary tumors and metastatic samples from
considered to be eligible by each author. Research articles published in 136 patients with breast, colorectal or lung cancer. Untreated and
English that reported the preclinical and clinical application of both treated metastases were included, showing that treated metastases often
liquid biopsy (LB) and imaging quantitative features were included in harbored new driver mutations - not detected in the primitive lesion -
this review. No restrictions were adopted for the geographical origin of whereas untreated metastasis did not, suggesting a clonal evolution
the articles: data from Asia, Africa, Europe, North and South America, promotion by medication. The ratio of non-synonymous versus synon­
and Oceania were allowed. Research studies were included in the review ymous single-nucleotide variants (SNVs) is another way to infer the
if they met the following criteria: (1) the study looked at the diagnostic, selective pressures on protein-coding sequences [12]. A ratio greater
prognostic and/or predictive value of LB in lung cancer; (2) the study than 1 implies positive or Darwinian selection (driving change), less
looked at the diagnostic, prognostic and/or predictive value of imaging than 1 implies purifying or stabilizing selection (acting against change),
quantitative features in lung cancer; (3) the study examined the clinical and a ratio of exactly 1 indicates neutral (i.e., no) selection. Hu Zheng

2
F. Cucchiara et al. Pharmacological Research 169 (2021) 105643

et al. also investigated this ratio among putative driver genes in me­ microenvironmental interactions, and therapeutic vulnerabilities. NGS
tastases, pointing out a minor selective pressure relative to colorectal is a high-throughput sequencing method that can simultaneously
and breast cancer development, but not lung cancer [11]. These data interrogate variable areas of the genome, and a growing body of evi­
suggest that treatment has a pivotal role in determining longitudinal dence is pointing to its usefulness in clinical practice by detecting so­
molecular trajectories of NSCLC. Furthermore, since treatment also in­ matic mutations, including single-nucleotide variations (SNVs), copy
fluences the clonality of metastases [11], it would be expected that number variations (CNVs), insertions/deletions or gene fusions [29].
polyclonal seeding of distant metastases might be more common among Different NGS-based methods have been developed and subsequently
untreated patients than treated ones. Hence, as the prevalence of poly­ validated for mutation detection on cfDNA in lung cancer [30]. Efficient
clonal seeding was variable across the primary lesion and the metastatic use of cfDNA NGS-based methods could be exploited to highlight
sites, and as it depends on treatment, the concept of a single-site biopsy distinct effects of individual oncogenic alleles (e.g., activating EGFR
to monitor disease dynamics is practically unfeasible, because it is mutations in NSCLC [31]) and patterns of co-mutations yielding diver­
invasive and may result in an underestimation of tumor heterogeneity. gent clinical behaviors and variable sensitivity to anticancer therapies
To overcome certain scientific and practical issues, the interest in the within the same driver mutation-defined subgroups [32]. From an
development of minimally invasive LBs has grown. In today’s era of evolutionary standpoint, co-occurrence of oncogenic mutations implies
precision oncology, LBs could be implemented both at pre-treatment functional cooperation, whereas mutual exclusivity indicates redun­
baseline as predictive marker, and longitudinally, to monitor response dancy or antagonism [32,33]. Exploration of concomitant mutations
and anticipate treatment resistance [13]. LBs often involve the extrac­ network can expand knowledge of essential biological interactions
tion of circulating tumor DNA (ctDNA) [14–17], a fraction of befalling over tumor growth, and could be related with radiomic fea­
plasma-derived cell free DNA (cfDNA) to evaluate somatic mutations, tures to envisage tumor resistance to treatment resulting from the
circulating tumor cells (CTCs) [15–17], tumor microvesiscles [16–18], accumulation of subclones.
tumor educated platelet (TEPs) [16,17,19], cell free RNA (cfRNA) [16,
17,20] and circulating micro-RNA (miRNA) [16,17,20] (Table 1 and 4. Radiomic through imaging. A promising contribution
Fig. 1). Medical imaging play a major role in decision-making processes for
Besides, the recent use of immunotherapy has raised new clinical cancer patients, even if with a semi-quantitative (semantic [34,35])
needs. Assessment of tumor-derived programmed death-ligand 1 (PD- meaning. Usually, an experienced radiologist/oncologist assign a score
L1) expression in tumor tissues is currently used to predict drug to certain parameters, stemming from expertise-based observation.
response [21,22]. Nonetheless, a broadly and functional Nonetheless, out of the information contained in digital medical pic­
pro-inflammatory environment is required for treatment to be effec­ tures, sight can only extract about 10%.
tive, and evaluation of tumor immunogenicity is also mandatory [23]. A In 2012, Lambin et al. [36] and Kumar et al. [37] formally defined
possible indirect expression of tumor immunogenicity is represented by “radiomics” as a new automated and reproducible analysis to extract
the tumor mutational burden (TMB) [24]. The underlying hypothesis is large amounts of quantitative features from medical images using mul­
that the more non-synonymous mutations are present in the tumor, the tiple data-characterization algorithms and powerful computers. Among
greater the chance that neoantigens will arise, triggering an immune radiomic features, shape and texture are mainly investigated in lung
response [24]. TMB can be assessed by quantifying the number of cancer images. Shape features describe the geometry volume of interest
non-synonymous mutations in the whole exome or in a genetic panel (VOI) [38], while texture ones aim at quantifying its variability in the
defined by next generation sequencing (NGS) [25]. Published data are grey-scale levels [39,40] (Table 2).
growing [26,27], confirming that TMB analysis in ctDNA and molecular Ganeshan et al. in 2012 [41] and Mattonen et al. in 2015 [42,43]
cargo of exosomes derived from the peripheral tumor microenviron­ showed the potential of changes in computed tomography (CT) texture
ment, potentially adding new LB candidates to a growing list of blood analysis as predictive markers of local recurrence and poorer survival. A
tests to manage cancer patients. retrospective analysis [44] on 1,160 radiomics features obtained from
Additionally, with advances in artificial intelligence (AI) software the largest measurable lesion from 188 patients with NSCLC enrolled in
and hardware, especially machine learning algorithms [28], the next 3 multicenter clinical trials [45–47], showed the power of a radiomic
challenges include the possibility to use NGS to quantify and qualify signature in foreseeing sensitivity to treatment. The signature resulting
somatic mutations respect to tumor pathogenesis, biology, from the selection and combination of the four most distinguishable

Table 1
Comparison of the analysis capabilities of ctDNA, CTCs, exosomes, TEPs and cfRNA.
ctDNA CTCs exosomes TEPs cfRNA

Current technologies ARMS-PCR MS-PCR Flow cytometry IHC; IFS; SEC; chemical precipitation PIK qPCR; qPCR
qPCR; ddPCR NGS FISH qPCR; ddPCR NGS Immunocapture qPCR; ddPCR NGS ddPCR NGS ddPCR NGS

Applications Physical properties ✓ ✓


Biological features ✓ ✓
Metabolomic analyses ✓ ✓
Methylation patterns ✓
Epigenetic alterations ✓
Gene expression ✓ ✓ ✓ ✓
Somatic mutations ✓ ✓ ✓
CNA ✓
Gene fusion ✓ ✓ ✓ ✓
Splicing variants ✓ ✓ ✓
miRNA analyses ✓ ✓
DNA/RNA and protein-based ✓ ✓ ✓
molecular profiling

Abbreviations: ctDNA, circulating tumor DNA; CTCs, circulating tumor cells; TEPs, tumor educated platelets; cfRNA, cell free RNA; miRNA, micro RNA; PCR, poly­
merase chain reaction; ARMS-PCR, amplification-refractory mutation system PCR; MS-PCR, methylation-specific PCR; ddPCR, droplet digital PCR; qPCR, quantitative
PCR; NGS, next generation sequencing; IHC, immunocytochemistry; IFS, immuno fluorescent staining; FISH, Fluorescence in situ hybridization; SEC, Size exclusion
chromatography; Pik, platelet isolation kit.

3
F. Cucchiara et al. Pharmacological Research 169 (2021) 105643

Fig. 1. Liquid biopsy to assess tumor heterogeneity. Schematic of clones dynamicity model (on the left), where treatment exerts pressure for selection of resistant
subclones in lung cancer. Metastases can originate from the major (driver) clones of the primitive lesion, leading to homogeneity between paired primary tumor and
metastases, or they can originate from minor clones in the primary tumor, leading to spatial heterogeneity among lesions. Seeding of polyclonal disseminated cells, as
well as their treatment-affected plasticity over tumor growth and disease progression, contributes to the spatial and temporal heterogeneity that characterizes the
neoplasm. Liquid biopsy (on the right) can cover such heterogeneity and overcome practical and scientific issues of canonical tumor sampling. CTCs, ctDNA, cfRNA
(miRNA), tumor exosomes and tumor educated platelet can be isolated from many biological fluids (mainly blood), both for a qualitative and quantitative analysis
that allows early cancer diagnosis and monitoring of disease progression.

Table 2
Radiomic features.
Size Shape Textures

Effective Diameter of the imaginary Roundness It is a dimensionless measure. The value Contrast Contrast is a measure of the local grey-level intensity
diameter ellipsoid intersecting the tumor range is 0 < roundness ≤ 1, where 1 (HU) variation
(mm) surface indicates a perfect circle. Roundness looks
for tumor spherical disproportion
Maximum The greatest Euclidean distance Smoothness Regularity of the edges of the geometric Correlation Correlation shows the linear dependency of gray-
linear size between two vertices of the figure which is built on the neoplastic mass level values to their respective voxels in a grey-level
(mm) polygon mesh built on the co-occurrence matrix. The value range is
tumor surface. 0 < correlation ≤ 1, where 1 indicates a perfect
correlation
Surface area Tumor lesion surface area Regularity Geometry associated with the tumor lesion. Entropy Entropy specifies the randomness in neighborhood
(mm2) The greater the regularity, the simpler the voxel-related grey-level intensity value differences.
geometric shape rrelated to the neoplastic
mass
Volume Tumor lesion volume Compactness Density of the neoplastic mass Homogeneity Uniformity of the image array. A greater
(mm3) homogeneity implies a smaller range of discrete grey-
level intensity values.
Surface-to- Relationship between tumor Elongation Elongation shows the relationship between Variance Variance is the mean of the squared distances of each
volume lesion surface area and volume the two largest principal components in the grey-level intensity value from the grey-level mean
ratio ROI shape value.
It is a measure of the spread of the distribution about
the mean.

Abbreviations: mm, millimeters; VOI, volume of interest; HU, Hounsfield Unit

features, suggested a supporting role in risk stratification and manage­ radio-genomics [65] (Fig. 2).
ment of patients.
Radiomic analysis can also reflect pathophysiological processes un­ 5. Combining liquid biopsy and radiomics for diagnostic and
derlying tumor development, and growing evidence suggest its use to prognostic purposes
decode and monitor tumor genotype [48,49,58–64,50–57]. As an
example, several papers emphasize the association between NSCLC 5.1. The diagnostic and prognostic values of liquid biopsy
radiomic quantitative features and EGFR mutations [48,49,58,63,
50–57], ALK rearrangement status [51,59–61], or other significant al­ Currently, LB is mainly used as an auxiliary tool for routine exami­
terations such as KRAS mutations, ROS1 and rearranged during trans­ nation in lung cancer [66]. Although it seems to have a limited impact
fection (RET) gene fusions [51,59,63]. This additional imaging-derived on diagnosis and staging, recent studies proved that LB might confer an
information, allowed to obtain new correlations between cellular gene advantage on the initial molecular investigation [13]. Yang and col­
expression and tissue-scale imaging, introducing the notion of laborators [67] - investigating the prognostic role of CTCs in 107

4
F. Cucchiara et al. Pharmacological Research 169 (2021) 105643

Fig. 2. Outline of radiogenomic pipeline. After image acquisition, areas of interest are manually or automatically segmented through computational approaches.
Shape, texture and several other radiomic features are then extracted thanks to the use of specialized user-defined algorithms or through machine learning techniques
(deep learning). Subsequently, the imaging traits could be integrated with the gene expression map and other clinical data, to select a radiogenomic model using
either statistical or machine learning methodologies. Models can be purely correlative or prognostic/predictive. The model is used to gain clinical data, including
prognostic and predictive information, for a given set of imaging features and genomic information. Incorporation of outcomes data into the radiogenomic model
leads to further refinement and model validation.

patients with EGFR-mutated advanced NSCLC receiving first-line ther­ hypomethylated [75]. For instance, specific enrichment of methylated
apy with either erlotinib or gefitinib - found that subjects with low fragments of cfDNA, such as methylated SHOX2 and PTGER4 [76],
baseline CTCs count (< 5 CTCs) showed longer median progression-free proved to distinguish early-stage lung cancer from healthy control, with
survival (PFS) and time-to-treatment failure (TTF) than the others (11.1 91–98% area under the receiver operator characteristic curve (AUROC)
vs. 6.8 months, p = 0.009 and 12.4 vs. 8.0 months, p = 0.0107; [76,77]. Conversely, cfRNA methylation was demonstrated to mediate
respectively). Conversely, although a significant difference was miRNA expression and cancer cell migration [78], however further
observed between the two groups also on day 28 for PFS (11.6 vs. 6.3 studies are needed to validate its use for early diagnosis. miRNAs are the
months, p < 0.0001), no difference emerged for TTF. most suitable cfRNA molecules in fluid samples and various researchers
In line with this, other papers showed that dynamic ctDNA mea­ proposed to look at them for diagnosis. The detection of different miRNA
surements also have prognostic significance in EGFR-mutated lung levels [79] and patterns [80] can be used not only to identify patients
cancer treated with tyrosine kinase inhibitors (TKIs) [68–72], regardless with stage I-IIIA lung cancer but also for histological classification,
of tumor histological type or patient characteristics like age and smoking whether it is a benign or malignant nodule [81], squamous cell carci­
habits. On this topic, Cargnin and colleagues [73] performed a system­ noma or adenocarcinoma [79], NSCLC or small cell lung cancer (SCLC)
atic review and meta-analysis. Interestingly, they observed a harmful [20].
impact of high baseline ctDNA levels on lung cancer patients’ survival Tumor-derived proteins and metabolites have also been described as
(p < 0.001, out of 1723 patients enrolled in 16 studies), while no as­ LB markers in lung carcinoma diagnosis [82,83], and can be used to
sociation with PFS was reported (p = 0.29, in 640 patients from five distinguish benign and malignant nodules within a CT screening [84].
studies). By contrast, mutations in ctDNA appeared less useful to detect
and diagnose lung cancer at an early stage [9], showing only 50% 5.1.1. Exosomes
sensitivity in early detection (possibly due to the lower detectable DNA, RNA, and proteins are also carried by microvesicles, including
amounts of ctDNA released by early-stage lung neoplasms [74]). exosomes. Exosomes are 30–150 nm vesicles of endosomes that can be
Besides, epigenetic biomarkers - including cfDNA/RNA methylation - released into the extracellular space as messengers by different cell types
gained increasing attention as a non-invasive material for lung cancer [85–88], including cancer cells [89]. Rab-GTPases, including Rab27a
diagnosis. It has been reported that in the early stage, tumor-suppressor and Rab27b, are the key regulators of exosomes secretion and Rab27 is
genes are usually hypermethylated, and proto-oncogenes closely related to tumor onset and development [90], which indicates

5
F. Cucchiara et al. Pharmacological Research 169 (2021) 105643

Table 3
Advantages, drawbacks, and potential synergies of liquid biopsy and radiomics approaches.
Liquid biopsy Radiomics

Advantages Isolation of different biomarkers from many biological fluids and sources: Multimodality
• CTCs are tumoral cells derived from solid tumors circulating in peripheral Conversion of clinical images into high-dimensional, mineable data (beyond
blood which have relevance for initial investigation, metastatic process, and the human sight) via high-throughput extraction of quantitative features
disease progression thanks machine-learning algorithm.
• cfDNA/RNA are small fragments of nucleic acid with short half-life, which are Local and global approach
easily and quickly extracted from biological fluids (e.g. blood) and analysed, Cost-effectiveness
both qualitatively and quantitatively
• exosomes are membrane-bound EVs that are produced in the endosomal
compartment of both healthy and cancer cells. Tumor EVs can be isolated. They
are high concentrated in blood (≥ 109 EVs/ml), carrying abundant content and
they are stable (i.e. maintain the integrity of contents)
Assessment of tumor heterogeneity and dynamics Assessment of tumor heterogeneity and dynamics
Minimally invasive techniques (minimal pain/risk) for early cancer diagnosis and Minimally invasive techniques (minimal pain/risk) for early cancer diagnosis
monitoring of disease progression and monitoring of disease progression
Easily repeated. Possibility to perform continuous follow-up examination and Easily repeated. Possibility to perform continuous follow-up examination and
real-time monitoring for drug-response and resistance (e.g. CTCs allow for real-time monitoring for drug-response and resistance
immuno-labelling based approaches and for molecular characterization of both
cellular and subcellular level; ctDNA are sensitive for detection of disease burden,
may predicting acquired drug resistance)
Drawbacks Still too few laboratory application. Molecular protocols need to be standardized Radiation
(different isolation technologies and different detection rates)
Difficulty in detection: Heterogeneity of analysed cohorts. Dearth of standardization for image
• Low-input amount and isolation of rare CTCs with limited capture techniques acquisition. Dearth of standardization for computational approaches and
• Challenging discrimination of ctDNA from normal cfDNA features selection
• Laborious isolation and purification of exosomes
Handling of small quantities and easily degradable material after harvesting, thus Biased discovery rates and type I error inflation
requiring extremely sensitive and specific analytic methods
Microenvironmental changes may influence the release of the amount of Need of validation on large cohort of data, collected from multiple centres,
biological materials ideally prospectively and analysed using modern methods capable of
Poor information on adequate controls addressing their heterogeneity
False positive and false negative results: risk of not correctly evaluating the Black-box issue (non-interpretable advanced machine-learning algorithms
efficacy of a pharmacological treatment work like black boxes, hindering clinical translation)
It is still unclear if liquid biopsy provides a representative sampling of all genetic Indirect and not-detailed quantification of underlying biological processes
clones or there is a propensity for specific subpopulation within the intra-tumor
heterogeneity landscape
Potential Correlating biopsies and radiomics at early cancer diagnosis and over monitoring of disease progression
synergy Create common patient-oriented liquid biopsies and radiomic databases (medical imaging and blood withdrawn are part of standard of care)
Avoid unnecessary radiation by using liquid biopsy
Use radiomics to refine liquid biopsy results and provide a full-field analyses of patient’s lesions in virtually real-time response

Abbreviations: CTCs, circulating tumor cells; cfDNA/RNA, cell free DNA/RNA; EVs, extracellular vesicles; ctDNA, circulating tumor DNA; ml, millilitres.

the role of exosomes secretion in tumor biology. Tumor-derived exo­ 5.2. The contribution of radiomics
somes are carrier of important information of the tumor cell and can be
found in many body fluids such as blood, urine, pleural effusions, asci­ Combining imaging-derived quantitative features could strengthen
tes, and bronchoalveolar fluid [91]. Given their minimally invasive the aforementioned diagnostic and prognostic values of LB. Radiologic
isolation, exosomes can be considered as potential circulating bio­ examination, like CT scans, is known as a useful tool to screen and detect
markers for tumor diagnosis and prognosis [91]. As an example, as early lung cancer at an early stage [97]. However, the application is limited by
as in 2009 it was shown that exosome-derived miRNA isolated from the false-positive rate [97] and radiation exposure [98]. A combination
NSCLC patients mirrored the miRNA pattern expressed in NSCLC tissue of minimally invasive circulating biomarkers and screening scans could
(i.e. overexpression of miR-17–3p, miR-21, miR-106a, miR-146, be more valuable and reliable than the independent use of the two
miR-155, miR-191, miR192, miR-203, miR-205, miR-210, miR-212, strategies. As an example, a panel of three miRNAs selected from The
and miR-214), suggesting their use as surrogate material for NSCLC Cancer Genome Atlas (TCGA) database demonstrated a sensitivity of
[92]. Along the same lines, other notable studies [93,94] also demon­ 81.2% to discriminate lung cancer from healthy controls, while
strated the diagnostic accuracy of a multi-marker model including the combining two miRNAs from the same panel with measuring pulmonary
overexpression of several exosome-derived proteins (e.g. CD151, nodules size improved diagnostic sensitivity to 89.9% [99]. Moreover,
TSPAN8, CD171, SFTPD, CD82, PLAP, CTAG1B, HER2, flotillin-1 and 102 patients with untreated stage I-IIIA NSCLC who underwent radical
mucin-16 [94]) in diagnose lung cancer. Conversely, although Mon­ resection were enrolled at Virgen de las Nieves University Hospital.
termini and colleagues [95] successfully detected EGFR-mutations in CTCs were detected before and one month after surgery, and compared
plasma-derived exosome double-stranded DNA (dsDNA) from xeno­ with the maximum standardized uptake value (SUVmax) measured on
grafts mice of human lung cancer (A549) treated with TKIs, there is still preoperative fluorodeoxyglucose (FDG)-positron emission tomography
no clinical evidence [96]. (PET)/CT scans [100]. In this single-center prospective study,
Bayarri-Lara and colleagues interestingly found that SUVmax is an

6
F. Cucchiara et al. Pharmacological Research 169 (2021) 105643

independent predictor for the presence of CTCs after surgery [100] and fractional abundance (FA) is also an additional tool in interpreting a
that CTCs were also blended with a reduction in PFS on multivariate liquid biopsy test, by informing the clinician whether T790M is the
analysis, regardless of disease staging [100]. In this context, other recent dominant mechanism of resistance or a subclonal phenomenon within
studies also showed that prediction models based on radiomic imaging heterogeneous biology (i.e. T790M FA <10%). The detection of EGFR
features can further help distinguishing between benign, malignant and driver mutation at high FA could confirm that ctDNA is shed by the
inflammatory lung nodules [101–106], between different histological tumor and would make a negative T790M result more likely than a true
subtypes [107–112], and between primary and metastatic lesions [113]. negative, although its presence as a minor subclone is still possible.
The latter reports suggest that the combination of LB and radiomics has Overall, Oxnard and colleagues also found ~30% of false negative
the potential to serve as an alarm signal in patients with a high proba­ T790M due to a lack of sensitivity/DNA shedding [126]. A negative
bility of lung cancer onset, thus providing an efficient way to screen and result should always be validated by tissue sampling, which will enable
find tumor at an early stage. This could potentially eliminate or signif­ the detection of other mechanisms of resistance, not covered by
icantly reduce the risk of radiation exposure by performing multiple currently approved cfDNA assays (e.g. histotype transformation [128,
follow-up radiologic investigations for stable lung injury and reduce the 129], MET [130] and HER2 [131] amplification, or PIK3CA mutations
complications related to the high false-positive rates observed in [132]).
low-dose radiologic screening (Table 3). In the future, the values of the Besides T790M mutation, C797S also hinders third-generation EGFR-
combination of liquid and radiomic biopsy in the diagnosis of lung TKI inhibition by preventing irreversible binding, between the small
cancer, particularly during the screening of early lung cancer in the molecule and the ATP-binding pocket [133]. Thress and colleagues
high-risk population, require well-designed prospective studies confirmed its emergence as a specific mechanism of acquired resistance
involving large cohorts. to osimertinib by screening cfDNA from fifteen EGFR T790M mutant
patients during treatment [134].
6. Combine liquid biopsy and radiomics for predictive purposes Furthermore, in addition to EGFR, recent studies have also evaluated
the possibility of using LB to detect both ALK rearrangement and point
6.1. Target therapy mutations, prior and during ALK-inhibitors treatment [135–139].
Among them, positive results came from a single-arm cohort of the
6.1.1. The predictive value of liquid biopsy Phase II/III blood first assay screening trial (BFAST) [139], evaluating
LB can predict treatment response, especially for patients with blood-based NGS to detect specific fusions in 2188 patients with
oncogene-addicted tumors undertaking targeted-therapies. About 15% advanced NSCLC. Blood-based detection of ALK fusions produced high
of western and 40% of Asian patients with NSCLC harbor EGFR sensitive overall response rate (ORR) and clinical benefits in patients receiving
mutations (e.g. ex19del and L858R) [114], to which first/second alectinib, validating the clinical utility of blood-based NGS as an addi­
[115–117] and third-generation [118] EGFR-TKIs are effective. Also, the tional companion diagnostic tool to inform clinical decision making in
T790M mutation inevitably occur in 50–60% of patients with acquired ALK-positive NSCLC.
resistance to first- or second-generation TKIs [119], but it is sensitive to Papadimitrakopoulou’s group also proved cfDNA to be a clinically
the third-generation TKI osimertinib [120,121]. viable source for other guideline-recommended biomarkers, including
Nowadays, exon 20 insertions remain an area of unmet need and ALK [140]. By looking at 282 patients with newly diagnosed metastatic
should be considered. Moreover, limited available clinical data suggest NSCLC, it was found a 100% positive predictive value for cfDNA versus
that other rare variants, including E709X, L747P/S, exon 18 mutations, tissue, with a concordance greater than 98.2%. Utilizing cfDNA in
and certain rare insertion/deletions of the EGFR exon 19 are sensitive to addition to tissue increased detection by 48%, including those with
afatinib or osimertinib [122]. In 2015, the European Medicines Agency negative, not assessed, or insufficient tissue results. Moreover, cfDNA
(EMA) approved the Qiagen Therascreen EGFR RGQ PCR kit for the median turnaround time was significantly shorter than tissue (9 vs 15
discovering and monitoring of both ex19del and T790M mutations in days; p < 0.0001).
ctDNA [17]. Likewise, one year later the Food and Drug Administration The aforementioned results support a new paradigm for detecting
(FDA) approved the use of the Roche cobas® EGFR Mutation Test v2 on mutations at progression, with plasma genotyping proposed as a first-
plasma samples [17]. In the WJOG8114LTR [123] prospective line screening test - since its lower turnaround time - and tumor bi­
multi-institutional study, the clinical significance of detecting ctDNA opsy requested in the case of negative results.
was evaluated in 57 EGFR mutant lung adenocarcinoma patients treated
with afatinib. At baseline, 62.5% of patients were also positive for EGFR 6.1.2. The contribution of radiomics
mutations in plasma, and the rate of detection was higher among pa­ Since genomic analysis is becoming conventional, research interest
tients with a systemic disease. Interestingly, among positive patients, in investigating whether radiomics can predict the genetic status of the
those with undetectable circulating EGFR mutations within four weeks neoplasm is also rising. Radiogenomic research on lung cancer started in
after treatment showed a significantly longer PFS than those who did not 2012 with Gevaert et al. [141], by correlating PET/CT features and
achieve negative conversion (13.6 vs. 5.1 months, p < 0.0001). aggregated gene expression patterns, with encouraging preliminary re­
More notably, LB constitutes a very appealing option at progression, sults. Two years later, Halpenny and colleagues [142] started a series of
to track mechanisms of resistance [124–126]. This setting is different publications linking specific imaging parameters to selected mutations,
from the initial diagnosis because tumor biology is more heterogeneous such as EGFR [51,53,150–152,63,143–149], ALK [51,152] and KRAS
and subclonal [127]. In 2016, Sundaresan and collaborators [125] [51,63,145,147]. So far, many research teams have shown that radiomic
analyzed the T790M both in CTCs and ctDNA, comparing it with data analysis can lead to the molecular classification of the tumor, although
from simultaneously collected tumor biopsies in 40 patients with the selected features and approaches were not consistent with each
EGFR-mutant lung cancers progressing on TKIs. Although CTC- and other. As an example, EGFR activating mutation was independently
ctDNA-based genotyping failed in about 20% of cases, the two assays associated to selected features such as, contrast [143], Laws-Energy
together enabled genotyping in all patients with available blood sam­ [144], median Hounsfield Unit (HU) [145], SUVmax [146], pleural
ples, and identified the T790M mutation in 14 (35%) patients in whom retraction [51], small size [51,147], speculation [147], irregular nod­
the concurrent biopsy was negative or indeterminate. Even Oxnard et al. ules [148], poorly defined margins [148], ground glass [53,148],
[126] showed that ~30% of patients who test negative for the presence emphysema [149], locoregional infiltration [149], normalized inverse
of T790M in tissue are positive in plasma and benefit from osimertinib difference moment [150], as well as combined radiomic profiles [63,
treatment, establishing that these apparent false positives were not 151]; ALK rearrangement was linked to pleural effusion [51] and
sequencing errors, but the result of tumor heterogeneity [126]. T790M lobulated margin [152]; while KRAS was related to round shape [51],

7
F. Cucchiara et al. Pharmacological Research 169 (2021) 105643

nodules in non-tumor lobes [51], multiple small nodules [147], as well to identify CTCs by fluorescent immunohistochemistry (IHC) [175].
as general radiomic profiles [63]. Nonetheless, CTCs data in lung cancer are very preliminary and
Machine learning approaches could overcome the problem and hurdles concerning their isolation have also emerged [176–178]. The
compute the most distinguishable radiomic features - whatever they are hypothesis that PD-L1 expression on CTCs could be used as a predictive
- through n-fold bootstrapped training sets and then incorporating them marker of response to ICIs has unfortunately not been confirmed so far.
into a model for mutation status prediction, fitted on the specific studied Furthermore, it remains to be clarified whether PD-L1 is expressed on all
cohort. Different studies used different approaches. Most frequently, top CTCs or in some subpopulations; whether prognosis and predictive
ranking features were incorporated in a signature via multinomial lo­ response to immunotherapy is correlated with CTC-derived PD-L1
gistic regression (MLR; often used in small data sets due to their re­ expression at baseline or during follow-up of treated patients; and
sistivity to overfitting) [50–52,54,57,58,144], although some authors whether the expression of PD-L1 in CTCs is related to synchronously
preferred other models, such as naive bayesian classifier (NBC) [58], matched tissue biopsies. Only Ilie et al. [168] were able to show good
K-nearest neighbor (KNN) [58], random forest (RF) [55,56,58,63], concordance between tissue and CTCs, but the rate of positive samples
support vector machine (SVM) [58] and decision tree (DT) [53,58]. All was low and correlation with response to PD1/PD-L1 inhibitors could
works assessed model performance by measuring the AUROC in the not be investigated.
validation cohort, evidencing good capability with acceptable repre­ Apart, recent work by Lee and Rho’s groups reports that lung cancer
sentativeness for predicting mutation status. cells release PD-L1 positive extracellular vesicles into the circulation,
Yet, a dearth of standardization for image acquisition and the het­ mainly exosomes [179]. First, they found that the exosomal expression
erogeneity of the small cohorts analyzed, still means a lack of repro­ did not differ among cultures of three distinct NSCLC cell lines, although
ducibility and generalizability in methodology [153]. Furthermore, the amount of PD-L1 in the exosomes was proportional to the quantity of
radiomic indices should be considered as indirect and non-detailed PD-L1 expression on the cells from which the exosomes were derived.
quantification of underlying biological processes. Hence, they next tested that PD-L1-containing exosomes can affect
To identify truly reliable radiogenomic features or signatures, large tumor growth in vitro and in vivo by systemically suppressing the im­
cohorts of heterogeneous patients should be collected from multiple mune system. Finally, the clinical relevance of the findings was
centers, ideally prospectively, and analyzed using modern methods confirmed by observing a correlation between the proportion of
capable of addressing such data heterogeneity [154]. To strengthen the PD-L1-positive exosomes and the PD-L1 expression level in the related
trustworthiness of the results, radiomics-based genotype predictions cancer tissue upon 24 patients. Consistent with these results, Danesi
could also be interestingly compared with information from LB [155], study-group investigated the association between PD-L1 mRNA in
over time. As previously mentioned, a combination of these two mini­ plasma-derived exosomes and response to nivolumab and pem­
mally invasive strategies, together with cutting-edge data analysis brolizumab in patients with NSCLC (8) or melanoma (18) [180]. Exo­
strategies, could be more valuable and reliable than their independent somal PD-L1 mRNA was obtained from blood at baseline and after 2
use, and may help decode tumor valuable information regarding type, months of treatment, and measured by digital droplet PCR. A significant
aggressiveness, progression, and response to treatment [144,154]. A reduction in PD-L1 mRNA copies/ml was found at baseline vs 2 months
study from the University of Oklahoma reported that while radiomics of treatment for patients with complete and partial responses, but not for
(AUROC = 0.78) and genomics (AUROC = 0.78) models were capable of patients with stable disease. Vice versa, a significant increase in PD-L1
predicting survival, accuracy significantly improved (AUROC = 0.84) levels was observed in patients who progressed.
when both data were combined [156]. This approach seems more This finding suggests that PD-L1 positive extracellular vesicles are
compatible with clinic practice than current standard, providing new also able to counteract the immune pressure at the effector stage and
instrumental and more objective diagnostic support, being able to sug­ offer a novel marker for immunotherapy beside CTCs [181].
gest a change in treatment strategy earlier than conventional methods
(Table 3). 6.2.1.1. Tumor mutational burden. TMB has recently been evaluated
prospectively, alone or in association with PD-L1 IHC, for the stratifi­
6.2. Immunotherapy cation of late stage NSCLC patients who benefit from PD-1 blockade [23,
182]. In addition to exosomal PD-L1, WES for determination of TMB in
6.2.1. The predictive value of liquid biopsy LB from patients with advanced NSCLC suggests that blood-based TMB
Given the LB’s promising results obtained in targeted therapies, the could also be used as a clinically actionable biomarker for predicting
growing effort in use it to predict response to immunotherapy is anti-PD-L1 therapy, especially if tumor biopsy is not accessible or has
unsurprising. been resampled [183]. However, the first results on the concordance
Immunotherapy is able to switch off the inhibitory effect of tumor on between tissue and cfDNA are contrasting [184]. One of the major issues
the immune system [157]. Among them, immune checkpoint inhibitors in the TMB evaluation is the lack of standardization. For instance, the
(ICIs; i.e. ipilimumab [158,159], pembrolizumab [160–162], nivolumab TMB calculation, intended as the type of somatic mutations (e.g. pas­
[158,159], atezolizumab [163,164] and durvalumab [165,166]) pre­ senger or driver mutations), varied among the studies. And this also
vent the binding of PD-L1 and cluster of differentiation 86 (CD86), to because different sequencing panels covered different genomic regions.
their respective receptors programmed cell death-protein-1 (PD-1) and Also, no clear binary cut-point is used to stratify patients with low/high
cytotoxic T-lymphocytes associated with protein 4 (CTLA-4), on cyto­ TMB [185]. Furthermore, others challenges persist and, for instance,
toxic T-cell, which would otherwise contribute to the host immune patients with both high TMB and PD-L1 tumor cells positivity, immu­
system evasion by promoting self-tolerance and suppressing inflamma­ notherapy might still fail, revealing both the complexity and our
tory activity [167]. incomplete understanding of the immunopathology of cancer [186].
Assessment of PD-L1 expression in NSCLC tissues is currently used to Additionally, a critical review by Passaro et al. [187] confirmed ECOG
predict drug response in the clinics [21,22]; however, since CTCs arise PS, bone metastasis and liver metastases to negatively affect the efficacy
from more than one tumor site, they should provide a better overall of ICIs, while the combination of chemo- and immuno-therapy seems to
representation of PD-L1 expression in the metastatic setting [168]. be more effective in women and in patients with a history of smoking.
Studies on CTC PD-L1 expression have accumulated for several solid However, although useful in clinical practice for risk–benefit ratio
tumors including lung [168–174], establishing their feasibility. There stratification, the authors conclude that a single prognostic factor is not
are many methods for detecting CTCs. An attractive one would be to the best way to understand tumor clinical behavior and
acquire CTCs by a negative enrichment method that used biological-related microenvironment. Thus, given the immune response
magnetic-activated cell sorting, and then fix the derived dried specimen

8
F. Cucchiara et al. Pharmacological Research 169 (2021) 105643

heterogeneity and both tumor and patient diversity, the use of a single relatively rare and fragile compared to the number of hematological
predictor is impracticable and new approaches using additional and molecules found in the blood sample, and ctDNA analysis could be
more robust biomarkers should be associated with ICIs. contaminated by genomic DNA as well [205]. Therefore, there are dif­
ficulties in detection [205]. Besides, it is unclear whether the LB pro­
6.2.2. The contribution of radiomics vides a representative sampling of all genetic clones or there is a
As proposed for target therapy, Tang et al. [188] presented a NSCLC propensity for a specific subpopulation within the intra-tumor hetero­
radiomic signature using unsupervised classification approach. Four geneity landscape [206]. Concerning this, an NGS effort could be
distinct clusters were grouped by combining tissue-based CD3 count and attempted to explore the co-occurrence patterns of subclone-derived
PD-L1 low/high expressions from an early-stage NSCLCs cohort. Texture mutations during tumor progression and related to divergent clinical
features and intensity measures were then correlated to these four behaviors and treatment resistance. Depending on the size of the
clusters. Therefore, corresponding survival models were carried out, analyzed panel, the advantages of ctDNA-based NGS methods lie in the
indicating the highest overall survival (OS) in the radiomic cluster that simultaneous detection of dozens of targetable genetic abnormalities,
was strongly correlated with high CD3 and low PD-L1 expressions. including single nucleotide variations (SNV), copy number variations
Radiomic features pertaining to vessel tortuosity extracted from (CNV), insertions/deletions, or fusion gene, reflecting the overall het­
baseline pretherapy CT scans have also been recently identified as a erogeneity of the intrapatient tumor, while space-limited tissue NGS
potential independent predictor of response to nivolumab in locally could lose new potential therapeutic targets [207]. This is particularly
advanced NSCLC [189], and changes in CT radiomic features concerning relevant considering the expected increase in enrollment of umbrella
intra- and peri-tumor areas were associated with lymphocyte distribu­ trials or expanded access programs requiring molecular assignment.
tion [190–192], leveraging on predicting OS and response to immuno­ Conversely, subsequent tissue NGS could identify substantial differences
therapy [190,193]. Similar findings were obtained by including most between neoplastic masses separated by time (temporal) or anatomic
predictive radiomic features and clinical covariates in DT models to location (spatial) in the same patient. A paper from Kurzrock’s Center
stratify patients into survival risk-groups [194]. By doing so, a texture for Personalized Cancer Therapy showed high concordance between
radiomic feature (the Gray-Level Co-Occurrence Matrix inverse difference) genomic results of ctDNA and tissue NGS, but demonstrated that there
was found to be not only significantly related with OS in four inde­ could be significant differences if the acquisition of of LB from tissue
pendent NSCLC cohorts (HR = 2.74; 95% CI 1.04 – 7.24), but also biopsy exceedes 6 months [208]. Therefore, a complementary NGS
positively associated with the hypoxia-related carbonic anhydrase, analysis of tissue and ctDNA could capture intrapatient tumor hetero­
tumor acidosis, and treatment resistance. Dr. Aerts and his group carried geneity, being repeatable during treatment. CtDNA-based NGS methods
out an AI-based characterization of NSCLC primary and metastatic le­ also allow tracking variant allelic frequency (VAF), which is the relative
sions on the pre-treatment contrast-enhanced CT imaging data to proportion of mutant forms compared to wild-type ones at a given locus.
develop and validate a noninvasive machine learning biomarker capable VAF monitoring during patient’s treatment is under investigation and
of distinguishing between immunotherapy responding and non­ offers a promising opportunity for long-term disease management as
responding (AUROC = 0.83, p < 0.001) [195]. It was also found highly well [209]. However, given the considerable dilution of somatic muta­
significant associations with pathways involved in mitosis, indicating a tions from plasma, the innate error rates of NGS calls are a primary
relationship between increased proliferative potential and preferential concern for ctDNA analysis and may affect data interpretation. To
response to immunotherapy [195]. address this, many labs are adopting sophisticated foolproof computa­
To conclude, it is also interesting to describe an early report on 228 tional approaches to greatly improve sequencing specificity, reduce the
NSCLC patients treated with single agent or double agent immuno­ chance of false-positive calls and thus increase sensitivity [210,211].
therapy identified a clinical-radiomic model of both TTF Overall, the improvement of analytical methods is necessary to in­
(AUROC = 0.804% and 73.41% accuracy) and hyper-progressive dis­ crease LB sensitivity [212].
ease (AUROC = 0.865% and 82.28% accuracy). The models have Additionally, the viability of platforms in one’s own region are other
potentially important practical implications in identifying highly issues for the use of ctDNA-based NGS methods, and each clinical setting
vulnerable NSCLC patients treated with immunotherapy that experience will need to evaluate and balance between assay availability, cost, and
rapid disease progression and poor survival outcomes [196]. sensitivity.
Radiomics could ameliorate NGS cost-effectiveness by adding new
7. Conclusion high-throughput imaging features reflecting underlying gene expression
patterns and revealing heterogeneous tumor metabolism and anatomy
LB and radiomics have promising and attractive characteristics, they [213]. This extraction is typically preparatory to a data mining process
are quantifiable and can be repeated to evaluate tumor progression. [11] to increase precision in diagnosis, assessment of prognosis, pre­
Furthermore, in comparison to tissue molecular profiling, limited by the diction of treatment response, and provide valuable information for
invasiveness of the procedure, medical imaging and withdrawal of blood patient care throughout the disease [214].
samples are more commonly part of the standard of care [197] and may More should also be learned about the functioning of radiomics to
provide a full-field analysis of the lesion, in virtually real-time response. understand the underlying clinical and/or molecular connotation [215]
Nevertheless, the proposed literature must be carefully considered. of imaging features (i.e. the black-box problem).
Publications naturally tend to overestimate the positive results over the We posit that the embedding of LB-related genomic testing and
negative ones [198,199], however, the lack of reproducibility is known radiomic features through AI could overcome problems and limitations
and is mainly due to the absence of standardization in all the workflow related to methodologies and permit research findings implementation
phases [200,201]. Additionally, many studies suffer from severe vali­ directly into clinical practice. The purpose is twofold: provide a cost-
dation limitations, including improper statistical analysis (e.g. lack of effective treatment personalization, by using an approach where clini­
p-value adjustment for multiple tests) and/or absence of an independent cians and patients promptly share the best minimally invasive available
dataset to confirm results. This can easily lead to biased discovery rates evidence when faced with the task of making decision.
and type I error inflation [202]. The global precision medicine (diagnostics/therapeutics) market
For instance, due to the risk of false-negative results, LB is not yet size is projected to reach $ 85.5 billion by 2025 [216], and progress and
considered a standard alternative to the tissue biopsy but is rather used promises of multiple new treatment choices for lung cancer come with
as a complementary test for patients undergoing TKI if tissue-specimens exorbitant prices, too. Quickly diagnosis of the tumor, detecting any
are lacking [203] (and there is still no indication to perform it on pa­ change in tumor molecular profile, responsiveness to treatment and
tients selected for immunotherapy [204]). CTCs and ctDNA are tumor recurrence, would focus treatment options for patient well-being,

9
F. Cucchiara et al. Pharmacological Research 169 (2021) 105643

increase the success rate, improve patient QoL and reduce Competing interests
healthcare-related costs. As reported by Sholl and colleagues, the mo­
lecular genetics of tumors has yet to take hold in clinical oncology The authors declare no conflict of interest.
training, and many oncologists are not comfortable interpreting mo­
lecular tumor findings [212]. Therefore, to achieve the goal and reduce Acknowledgments
the risk of interpretation they will have to consult a multidisciplinary
group of experts trained in molecular pathology [212], combine a new None.
narrative, mechanistic and mathematical thinking in their training [217,
218] and consider a new bio-psycho-social model of patient-centered References
disease [212,217,218].
Although these skills are not even currently accredited in medical [1] R.L. Siegel, K.D. Miller, A. Jemal, Cancer statistics, 2019, CA Cancer J. Clin. 69
(2019) 7–34, https://doi.org/10.3322/caac.21551.
education programs, a survey conducted by the American Medical As­ [2] F. Bray, J. Ferlay, I. Soerjomataram, R.L. Siegel, L.A. Torre, A. Jemal, Global
sociation (AMA) in 2016 shows that 85% of physicians perceived ben­ cancer statistics 2018: GLOBOCAN estimates of incidence and mortality
efits from new digital tools [219]. Yet, the implementation of AI in worldwide for 36 cancers in 185 countries, CA Cancer J. Clin. 68 (2018)
394–424, https://doi.org/10.3322/caac.21492.
medical education is challenging because the tuition will beef up the [3] W.D. Travis, E. Brambilla, A.P. Burke, A. Marx, A.G. Nicholson, Introduction to
existing curriculum rather than replace existing coursework, and The 2015 World Health Organization classification of tumors of the lung, pleura,
another practical problem is that many senior physicians have little to thymus, and heart, J. Thorac. Oncol. Publ. Int. Assoc. Study Lung Cancer 10
(2015) 1240–1242, https://doi.org/10.1097/JTO.0000000000000663.
no experience with AI. [4] U. Testa, G. Castelli, E. Pelosi, Lung Cancers: Molecular Characterization, Clonal
Finally, there are also important legal and regulatory aspects to Heterogeneity and Evolution, and Cancer Stem Cells, in: Cancers (Basel), 10,
address [220] concerning the use of AI in the healthcare system, such as 2018, https://doi.org/10.3390/cancers10080248.
[5] M. Shea, D.B. Costa, D. Rangachari, Management of advanced non-small cell lung
the data privacy and ownership [221]. The key to success will require
cancers with known mutations or rearrangements: latest evidence and treatment
policies that properly regulate large reserves of linked information approaches. Ther. Adv. Respir. Dis. 10 (2016) 113–129, https://doi.org/
(around seven gigabytes of health data for single patient [222]), 10.1177/1753465815617871.
ensuring accessibility and security. Ideally, data should be easily avail­ [6] S. Couraud, P.-J. Souquet, C. Paris, P. Dô, H. Doubre, E. Pichon, A. Dixmier,
I. Monnet, B. Etienne-Mastroianni, M. Vincent, J. Trédaniel, M. Perrichon,
able for those who need, while security should be efficient to prevent P. Foucher, B. Coudert, D. Moro-Sibilot, E. Dansin, S. Labonne, P. Missy, F. Morin,
inappropriate access. Strong user authentication methods should be in H. Blanché, G. Zalcman, BioCAST/IFCT-1002: epidemiological and molecular
place to consider the high-security constraints, and traceability should features of lung cancer in never-smokers. Eur. Respir. J. 45 (2015) 1403–1414,
https://doi.org/10.1183/09031936.00097214.
also be important at all levels of data processing. Linking and coordi­ [7] R. Govindan, L. Ding, M. Griffith, J. Subramanian, N.D. Dees, K.L. Kanchi, C.
nating medical records are also steps not to be overlooked and often A. Maher, R. Fulton, L. Fulton, J. Wallis, K. Chen, J. Walker, S. McDonald,
require a third party to govern these procedures. R. Bose, D. Ornitz, D. Xiong, M. You, D.J. Dooling, M. Watson, E.R. Mardis, R.
K. Wilson, Genomic landscape of non-small cell lung cancer in smokers and
These challenges, while not trivial, also present significant new op­ never-smokers, Cell 150 (2012) 1121–1134, https://doi.org/10.1016/j.
portunities for the development of novel theoretical approaches and cell.2012.08.024.
practical applications in data-driven medical decisions. If new health [8] I. Dagogo-Jack, A.T. Shaw, Tumour heterogeneity and resistance to cancer
therapies, Nat. Rev. Clin. Oncol. 15 (2018) 81–94, https://doi.org/10.1038/
policies take hold, and new studies will access to large amounts of well- nrclinonc.2017.166.
organized high-quality data, the complementary and possibly synergis­ [9] M. Jamal-Hanjani, G.A. Wilson, N. McGranahan, N.J. Birkbak, T.B.K. Watkins,
tic combination of LB and imaging could provide an attractive choice to S. Veeriah, S. Shafi, D.H. Johnson, R. Mitter, R. Rosenthal, M. Salm, S. Horswell,
M. Escudero, N. Matthews, A. Rowan, T. Chambers, D.A. Moore, S. Turajlic,
the traditional molecular pathology profile in the personalized treat­
H. Xu, S.-M. Lee, M.D. Forster, T. Ahmad, C.T. Hiley, C. Abbosh, M. Falzon,
ment of NSCLC. E. Borg, T. Marafioti, D. Lawrence, M. Hayward, S. Kolvekar, N. Panagiotopoulos,
S.M. Janes, R. Thakrar, A. Ahmed, F. Blackhall, Y. Summers, R. Shah, L. Joseph,
Ethics approval and consent to participate A.M. Quinn, P.A. Crosbie, B. Naidu, G. Middleton, G. Langman, S. Trotter,
M. Nicolson, H. Remmen, K. Kerr, M. Chetty, L. Gomersall, D.A. Fennell,
A. Nakas, S. Rathinam, G. Anand, S. Khan, P. Russell, V. Ezhil, B. Ismail, M. Irvin-
Not applicable. Sellers, V. Prakash, J.F. Lester, M. Kornaszewska, R. Attanoos, H. Adams,
H. Davies, S. Dentro, P. Taniere, B. O’Sullivan, H.L. Lowe, J.A. Hartley, N. Iles,
H. Bell, Y. Ngai, J.A. Shaw, J. Herrero, Z. Szallasi, R.F. Schwarz, A. Stewart, S.
Consent for publication A. Quezada, J. Le Quesne, P. Van Loo, C. Dive, A. Hackshaw, C. Swanton,
Tracking the evolution of non-small-cell lung cancer, N. Engl. J. Med 376 (2017)
Not applicable. 2109–2121, https://doi.org/10.1056/NEJMoa1616288.
[10] C.M. Lovly, A.K.S. Salama, R. Salgia, Tumor heterogeneity and therapeutic
resistance. Am. Soc. Clin. Oncol. Educ. Book. Am. Soc. Clin. Oncol. Annu. Meet.
Availability of data and materials 35 (2016) e585–e593, https://doi.org/10.1200/EDBK_158808.
[11] Z. Hu, Z. Li, Z. Ma, C. Curtis, Multi-cancer analysis of clonality and the timing of
systemic spread in paired primary tumors and metastases, Nat. Genet. 52 (2020)
Not applicable. 701–708, https://doi.org/10.1038/s41588-020-0628-z.
[12] K. Hanada, S.-H. Shiu, W.-H. Li, The nonsynonymous/synonymous substitution
Funding rate ratio versus the radical/conservative replacement rate ratio in the evolution
of mammalian genes, Mol. Biol. Evol. 24 (2007) 2235–2241, https://doi.org/
10.1093/molbev/msm152.
This research was funded to Romano Danesi under grant n. [13] C. Rolfo, P.C. Mack, G.V. Scagliotti, P. Baas, F. Barlesi, T.G. Bivona, R.S. Herbst, T.
2017NR7W5K (PRIN 2017) from MIUR, Italy. S. Mok, N. Peled, R. Pirker, L.E. Raez, M. Reck, J.W. Riess, L.V. Sequist, F.
A. Shepherd, L.M. Sholl, D.S.W. Tan, H.A. Wakelee, I.I. Wistuba, M.W. Wynes, D.
P. Carbone, F.R. Hirsch, D.R. Gandara, Liquid biopsy for advanced non-small cell
Authors’ contributions lung cancer (NSCLC): a statement paper from the IASLC, J. Thorac. Oncol. 13
(2018) 1248–1268, https://doi.org/10.1016/j.jtho.2018.05.030.
Federico Cucchiara, Marzia Del Re, Romano Danesi: Conceptuali­ [14] M. Elazezy, S.A. Joosse, Techniques of using circulating tumor DNA as a liquid
biopsy component in cancer management. Comput. Struct. Biotechnol. J. 16
zation. All authors: Investigation, Formal analysis. All authors: Data (2018) 370–378, https://doi.org/10.1016/j.csbj.2018.10.002.
curation. Federico Cucchiara: Writing - original draft preparation. [15] M.H.D. Neumann, S. Bender, T. Krahn, T. Schlange, ctDNA and CTCs in liquid
Marzia Del Re: Writing - review & editing. Romano Danesi: Visualiza­ biopsy − current status and where we need to progress, Comput. Struct.
Biotechnol. J. 16 (2018) 190–195, https://doi.org/10.1016/j.csbj.2018.05.002.
tion. Romano Danesi: Supervision. All authors have read and agreed to [16] G. De Rubis, S. Rajeev Krishnan, M. Bebawy, Liquid biopsies in cancer diagnosis,
the published version of the manuscript. monitoring, and prognosis, Trends Pharmacol. Sci. 40 (2019) 172–186, https://
doi.org/10.1016/j.tips.2019.01.006.

10
F. Cucchiara et al. Pharmacological Research 169 (2021) 105643

[17] G. Siravegna, S. Marsoni, S. Siena, A. Bardelli, Integrating liquid biopsies into the challenges, Magn. Reson. Imaging 30 (2012) 1234–1248, https://doi.org/
management of cancer, Nat. Rev. Clin. Oncol. 14 (2017) 531–548, https://doi. 10.1016/j.mri.2012.06.010.
org/10.1038/nrclinonc.2017.14. [38] E.J. Limkin, S. Reuzé, A. Carré, R. Sun, A. Schernberg, A. Alexis, E. Deutsch,
[18] S. Cui, Z. Cheng, W. Qin, L. Jiang, Exosomes as a liquid biopsy for lung cancer, C. Ferté, C. Robert, The complexity of tumor shape, spiculatedness, correlates
Lung Cancer 116 (2018) 46–54, https://doi.org/10.1016/j.lungcan.2017.12.012. with tumor radiomic shape features, Sci. Rep. 9 (2019) 4329, https://doi.org/
[19] L. Liu, F. Lin, X. Ma, Z. Chen, J. Yu, Tumor-educated platelet as liquid biopsy in 10.1038/s41598-019-40437-5.
lung cancer patients, Crit. Rev. Oncol. Hematol. 146 (2020), 102863, https://doi. [39] R.M. Haralick, K. Shanmugam, I. Dinstein, Textural features for image
org/10.1016/j.critrevonc.2020.102863. classification, IEEE Trans. Syst. Man. Cybern. SMC-3 (1973) 610–621, https://
[20] S. Lu, H. Kong, Y. Hou, D. Ge, W. Huang, J. Ou, D. Yang, L. Zhang, G. Wu, Y. Song, doi.org/10.1109/TSMC.1973.4309314.
X. Zhang, C. Zhai, Q. Wang, H. Zhu, Y. Wu, C. Bai, Two plasma microRNA panels [40] F. Han, H. Wang, G. Zhang, H. Han, B. Song, L. Li, W. Moore, H. Lu, H. Zhao,
for diagnosis and subtype discrimination of lung cancer, Lung Cancer 123 (2018) Z. Liang, Texture feature analysis for computer-aided diagnosis on pulmonary
44–51, https://doi.org/10.1016/j.lungcan.2018.06.027. nodules, J. Digit. Imaging 28 (2015) 99–115, https://doi.org/10.1007/s10278-
[21] E. Jimenez Aguilar, H. Rizvi, S. Kravets, C.A. Lydon, A.E. Adeni, S. Subegdjo, M. 014-9718-8.
D. Hellmann, M.M. Awad, Comparison of outcomes with PD-L1 tumor proportion [41] B. Ganeshan, E. Panayiotou, K. Burnand, S. Dizdarevic, K. Miles, Tumour
score (TPS) of 50-74% vs 75-100% in patients with non-small cell lung cancer heterogeneity in non-small cell lung carcinoma assessed by CT texture analysis: a
(NSCLC) treated with first-line PD-1 inhibitors, JCO 36 (2018) 9037, https://doi. potential marker of survival, Eur. Radiol. 22 (2012) 796–802, https://doi.org/
org/10.1200/JCO.2018.36.15_suppl.9037. 10.1007/s00330-011-2319-8.
[22] K. Ancevski Hunter, M.A. Socinski, L.C. Villaruz, PD-L1 testing in guiding patient [42] S.A. Mattonen, D.A. Palma, C. Johnson, A.V. Louie, M. Landis, G. Rodrigues,
selection for PD-1/PD-L1 inhibitor therapy in lung cancer, Mol. Diagn. Ther. 22 I. Chan, R. Etemad-Rezai, T.P.C. Yeung, S. Senan, A.D. Ward, Detection of local
(2018) 1–10, https://doi.org/10.1007/s40291-017-0308-6. cancer recurrence after stereotactic ablative radiation therapy for lung cancer:
[23] N.A. Rizvi, M.D. Hellmann, A. Snyder, P. Kvistborg, V. Makarov, J.J. Havel, physician performance versus radiomic assessment, Int. J. Radiat. Oncol. Biol.
W. Lee, J. Yuan, P. Wong, T.S. Ho, M.L. Miller, N. Rekhtman, A.L. Moreira, Phys. 94 (2016) 1121–1128, https://doi.org/10.1016/j.ijrobp.2015.12.369.
F. Ibrahim, C. Bruggeman, B. Gasmi, R. Zappasodi, Y. Maeda, C. Sander, E. [43] S.A. Mattonen, S. Tetar, D.A. Palma, A.V. Louie, S. Senan, A.D. Ward, Imaging
B. Garon, T. Merghoub, J.D. Wolchok, T.N. Schumacher, T.A. Chan, Cancer texture analysis for automated prediction of lung cancer recurrence after
immunology. Mutational landscape determines sensitivity to PD-1 blockade in stereotactic radiotherapy, J. Med Imaging (Bellingham) 2 (2015), 041010,
non-small cell lung cancer, Science 348 (2015) 124–128, https://doi.org/ https://doi.org/10.1117/1.JMI.2.4.041010.
10.1126/science.aaa1348. [44] L. Dercle, M. Fronheiser, L. Lu, S. Du, W. Hayes, D.K. Leung, A. Roy, J. Wilkerson,
[24] F. Galuppini, C.A. Dal Pozzo, J. Deckert, F. Loupakis, M. Fassan, R. Baffa, Tumor P. Guo, A.T. Fojo, L.H. Schwartz, B. Zhao, Identification of non–small cell lung
mutation burden: from comprehensive mutational screening to the clinic, Cancer cancer sensitive to systemic cancer therapies using radiomics, Clin. Cancer Res.
Cell Int. 19 (2019) 209, https://doi.org/10.1186/s12935-019-0929-4. 26 (2020) 2151–2162, https://doi.org/10.1158/1078-0432.CCR-19-2942.
[25] H. Rizvi, F. Sanchez-Vega, K. La, W. Chatila, P. Jonsson, D. Halpenny, [45] Study of BMS-936558 (Nivolumab) Compared to Docetaxel in Previously Treated
A. Plodkowski, N. Long, J.L. Sauter, N. Rekhtman, T. Hollmann, K.A. Schalper, J. Advanced or Metastatic Squamous Cell Non-small Cell Lung Cancer (NSCLC)
F. Gainor, R. Shen, A. Ni, K.C. Arbour, T. Merghoub, J. Wolchok, A. Snyder, J. (CheckMate 017) - Full Text View - ClinicalTrials.gov, n.d. 〈https://www.
E. Chaft, M.G. Kris, C.M. Rudin, N.D. Socci, M.F. Berger, B.S. Taylor, A. Zehir, D. clinicaltrials.gov/ct2/show/NCT01642004〉 (accessed 18 July 2020).
B. Solit, M.E. Arcila, M. Ladanyi, G.J. Riely, N. Schultz, M.D. Hellmann, Molecular [46] Phase II Trial to Correlate Radiographic Response Induced By Gefitinib With
determinants of response to anti-programmed cell death (PD)-1 and anti- Mutations in the Protein-Tyrosine Kinase Domain of the EGF Receptor Gene - Full
programmed death-ligand 1 (PD-L1) blockade in patients with non-small-cell Text View - ClinicalTrials.gov, n.d. 〈https://www.clinicaltrials.gov/ct2/show/
lung cancer profiled with targeted next-generation sequencing, J. Clin. Oncol. J. NCT00588445〉 (accessed 18 July 2020).
Am. Soc. Clin. Oncol. 36 (2018) 633–641, https://doi.org/10.1200/ [47] Study of Nivolumab (BMS-936558) in Patients With Advanced or Metastatic
JCO.2017.75.3384. Squamous Cell Nonsmall-cell Lung Cancer Who Have Received At Least 2 Prior
[26] T.A. Chan, M. Yarchoan, E. Jaffee, C. Swanton, S.A. Quezada, A. Stenzinger, Systemic Regimens - Full Text View - ClinicalTrials.gov, n.d. 〈https://www.
S. Peters, Development of tumor mutation burden as an immunotherapy clinicaltrials.gov/ct2/show/NCT01721759〉 (accessed 18 July 2020).
biomarker: Utility for the oncology clinic, Ann. Oncol. 30 (2019) 44–56, https:// [48] W. Zhao, Y. Wu, Y. Xu, Y. Sun, P. Gao, M. Tan, W. Ma, C. Li, L. Jin, Y. Hua, J. Liu,
doi.org/10.1093/annonc/mdy495. M. Li, The potential of radiomics nomogram in non-invasively prediction of
[27] A. Costantini, P. Takam Kamga, C. Dumenil, T. Chinet, J.-F. Emile, E. epidermal growth factor receptor mutation status and subtypes in lung
Giroux Leprieur, Plasma biomarkers and immune checkpoint inhibitors in non- adenocarcinoma, Front. Oncol. 9 (2019) 1485, https://doi.org/10.3389/
small cell lung cancer: new tools for better patient selection? Cancers (Basel) 11 fonc.2019.01485.
(2019) https://doi.org/10.3390/cancers11091269. [49] B. He, W. Zhao, J.-Y. Pi, D. Han, Y.-M. Jiang, Z.-G. Zhang, W. Zhao, A biomarker
[28] G. Marceddu, T. Dallavilla, G. Guerri, A. Zulian, C. Marinelli, M. Bertelli, Analysis basing on radiomics for the prediction of overall survival in non-small cell lung
of machine learning algorithms as integrative tools for validation of next cancer patients, Respir. Res. 19 (2018) 199, https://doi.org/10.1186/s12931-
generation sequencing data, Eur. Rev. Med. Pharmacol. Sci. 23 (2019) 018-0887-8.
8139–8147, https://doi.org/10.26355/eurrev_201909_19034. [50] J.K.R. Nair, U.A. Saeed, C.C. McDougall, A. Sabri, B. Kovacina, B.V.S. Raidu, R.
[29] S. Behjati, P.S. Tarpey, What is next generation sequencing? Arch. Dis. Child. A. Khokhar, S. Probst, V. Hirsh, J. Chankowsky, L.C. Van Kempen, J. Taylor,
Educ. Pract. Ed. 98 (2013) 236–238, https://doi.org/10.1136/archdischild-2013- Radiogenomic models using machine learning techniques to predict EGFR
304340. mutations in non-small cell lung cancer, Can. Assoc. Radiol. J. (2020), https://
[30] J.A. Vendrell, F.T. Mau-Them, B. Béganton, S. Godreuil, P. Coopman, J. Solassol, doi.org/10.1177/0846537119899526.
Circulating cell free tumor DNA detection as a routine tool forlung cancer patient [51] S. Rizzo, F. Petrella, V. Buscarino, F. De Maria, S. Raimondi, M. Barberis,
management, IJMS 18 (2017) 264, https://doi.org/10.3390/ijms18020264. C. Fumagalli, G. Spitaleri, C. Rampinelli, F. De Marinis, L. Spaggiari, M. Bellomi,
[31] Q.S. Chu, Targeting non-small cell lung cancer: driver mutation beyond CT radiogenomic characterization of EGFR, K-RAS, and ALK mutations in non-
epidermal growth factor mutation and anaplastic lymphoma kinase fusion, Ther. small cell lung cancer, Eur. Radiol. 26 (2016) 32–42, https://doi.org/10.1007/
Adv. Med. Oncol. 12 (2020), 1758835919895756, https://doi.org/10.1177/ s00330-015-3814-0.
1758835919895756. [52] Y. Liu, J. Kim, Y. Balagurunathan, Q. Li, A.L. Garcia, O. Stringfield, Z. Ye, R.
[32] F. Skoulidis, J.V. Heymach, Co-occurring genomic alterations in non-small-cell J. Gillies, Radiomic features are associated with EGFR mutation status in lung
lung cancer biology and therapy, Nat. Rev. Cancer 19 (2019) 495–509, https:// adenocarcinomas, Clin. Lung Cancer 17 (2016) 441–448.e6, https://doi.org/
doi.org/10.1038/s41568-019-0179-8. 10.1016/j.cllc.2016.02.001.
[33] M. Mina, F. Raynaud, D. Tavernari, E. Battistello, S. Sungalee, S. Saghafinia, [53] O. Gevaert, S. Echegaray, A. Khuong, C.D. Hoang, J.B. Shrager, K.C. Jensen, G.
T. Laessle, F. Sanchez-Vega, N. Schultz, E. Oricchio, G. Ciriello, Conditional J. Berry, H.H. Guo, C. Lau, S.K. Plevritis, D.L. Rubin, S. Napel, A.N. Leung,
selection of genomic alterations dictates cancer evolution and oncogenic Predictive radiogenomics modeling of EGFR mutation status in lung cancer, Sci.
dependencies, Cancer Cell 32 (2017) 155–168.e6, https://doi.org/10.1016/j. Rep. 7 (2017) 41674, https://doi.org/10.1038/srep41674.
ccell.2017.06.010. [54] S.R. Digumarthy, A.M. Padole, R. Lo Gullo, L.V. Sequist, M.K. Kalra, Can CT
[34] S.S.F. Yip, Y. Liu, C. Parmar, Q. Li, S. Liu, F. Qu, Z. Ye, R.J. Gillies, H.J.W.L. Aerts, radiomic analysis in NSCLC predict histology and EGFR mutation status?
Associations between radiologist-defined semantic and automatically computed Medicine (Baltimore) 98 (2019) 13963, https://doi.org/10.1097/
radiomic features in non-small cell lung cancer, Sci. Rep. 7 (2017) 3519, https:// MD.0000000000013963.
doi.org/10.1038/s41598-017-02425-5. [55] T.-Y. Jia, J.-F. Xiong, X.-Y. Li, W. Yu, Z.-Y. Xu, X.-W. Cai, J.-C. Ma, Y.-C. Ren,
[35] R.J. Gillies, P.E. Kinahan, H. Hricak, Radiomics: images are more than pictures, R. Larsson, J. Zhang, J. Zhao, X.-L. Fu, Identifying EGFR mutations in lung
they are data, Radiology 278 (2016) 563–577, https://doi.org/10.1148/ adenocarcinoma by noninvasive imaging using radiomics features and random
radiol.2015151169. forest modeling, Eur. Radiol. 29 (2019) 4742–4750, https://doi.org/10.1007/
[36] P. Lambin, E. Rios-Velazquez, R. Leijenaar, S. Carvalho, R.G.P.M. van Stiphout, s00330-019-06024-y.
P. Granton, C.M.L. Zegers, R. Gillies, R. Boellard, A. Dekker, H.J.W.L. Aerts, [56] X. Yang, X. Dong, J. Wang, W. Li, Z. Gu, D. Gao, N. Zhong, Y. Guan, Computed
Radiomics: extracting more information from medical images using advanced tomography-based radiomics signature: a potential indicator of epidermal growth
feature analysis, Eur. J. Cancer 48 (2012) 441–446, https://doi.org/10.1016/j. factor receptor mutation in pulmonary adenocarcinoma appearing as a subsolid
ejca.2011.11.036. nodule, Oncologist 24 (2019) e1156–e1164, https://doi.org/10.1634/
[37] V. Kumar, Y. Gu, S. Basu, A. Berglund, S.A. Eschrich, M.B. Schabath, K. Forster, H. theoncologist.2018-0706.
J.W.L. Aerts, A. Dekker, D. Fenstermacher, D.B. Goldgof, L.O. Hall, P. Lambin, [57] S. Li, C. Ding, H. Zhang, J. Song, L. Wu, Radiomics for the prediction of EGFR
Y. Balagurunathan, R.A. Gatenby, R.J. Gillies, Radiomics: the process and the mutation subtypes in non-small cell lung cancer, Med. Phys. 46 (2019)
4545–4552, https://doi.org/10.1002/mp.13747.

11
F. Cucchiara et al. Pharmacological Research 169 (2021) 105643

[58] D. Hong, K. Xu, L. Zhang, X. Wan, Y. Guo, Radiomics signature as a predictive DNA methylomes, Nature 563 (2018) 579–583, https://doi.org/10.1038/s41586-
factor for EGFR mutations in advanced lung adenocarcinoma, Front. Oncol. 10 018-0703-0.
(2020) 28, https://doi.org/10.3389/fonc.2020.00028. [78] L. Yang, Y. Ma, W. Han, W. Li, L. Cui, X. Zhao, Y. Tian, Z. Zhou, W. Wang,
[59] H.J. Yoon, I. Sohn, J.H. Cho, H.Y. Lee, J.-H. Kim, Y.-L. Choi, H. Kim, G. Lee, K. H. Wang, Proteinase-activated receptor 2 promotes cancer cell migration through
S. Lee, J. Kim, Decoding tumor phenotypes for ALK, ROS1, and RET fusions in RNA methylation-mediated repression of miR-125b, J. Biol. Chem. 290 (2015)
lung adenocarcinoma using a radiomics approach, Medicine (Baltimore) 94 26627–26637, https://doi.org/10.1074/jbc.M115.667717.
(2015) 1753, https://doi.org/10.1097/MD.0000000000001753. [79] J. Shen, N.W. Todd, H. Zhang, L. Yu, X. Lingxiao, Y. Mei, M. Guarnera, J. Liao,
[60] S. Yamamoto, R.L. Korn, R. Oklu, C. Migdal, M.B. Gotway, G.J. Weiss, A.J. Iafrate, A. Chou, C.L. Lu, Z. Jiang, H. Fang, R.L. Katz, F. Jiang, Plasma microRNAs as
D.-W. Kim, M.D. Kuo, ALK molecular phenotype in non–small cell lung cancer: CT potential biomarkers for non-small-cell lung cancer, Lab. Invest. 91 (2011)
radiogenomic characterization, Radiology 272 (2014) 568–576, https://doi.org/ 579–587, https://doi.org/10.1038/labinvest.2010.194.
10.1148/radiol.14140789. [80] M.B. Wozniak, G. Scelo, D.C. Muller, A. Mukeria, D. Zaridze, P. Brennan,
[61] L. Song, Z. Zhu, L. Mao, X. Li, W. Han, H. Du, H. Wu, W. Song, Z. Jin, Clinical, Circulating MicroRNAs as non-invasive biomarkers for early detection of non-
conventional CT and radiomic feature-based machine learning models for small-cell lung cancer, PLoS One 10 (2015), 0125026, https://doi.org/10.1371/
predicting ALK rearrangement status in lung adenocarcinoma patients, Front. journal.pone.0125026.
Oncol. 10 (2020) 369, https://doi.org/10.3389/fonc.2020.00369. [81] A. Bagheri, H.R.K. Khorshid, M. Tavallaie, S.J. Mowla, M. Sherafatian,
[62] M. Zhou, A. Leung, S. Echegaray, A. Gentles, J.B. Shrager, K.C. Jensen, G.J. Berry, M. Rashidi, M. Zargari, M.E. Boroujeni, S.M. Hosseini, A panel of noncoding RNAs
S.K. Plevritis, D.L. Rubin, S. Napel, O. Gevaert, Non–small cell lung cancer in non-small-cell lung cancer, J. Cell. Biochem. (2018), https://doi.org/10.1002/
radiogenomics map identifies relationships between molecular and imaging jcb.28111.
phenotypes with prognostic implications, Radiology 286 (2017) 307–315, [82] L. Niu, X. Song, N. Wang, L. Xue, X. Song, L. Xie, Tumor-derived exosomal
https://doi.org/10.1148/radiol.2017161845. proteins as diagnostic biomarkers in non-small cell lung cancer, Cancer Sci. 110
[63] E. Rios Velazquez, C. Parmar, Y. Liu, T.P. Coroller, G. Cruz, O. Stringfield, Z. Ye, (2019) 433–442, https://doi.org/10.1111/cas.13862.
M. Makrigiorgos, F. Fennessy, R.H. Mak, R. Gillies, J. Quackenbush, H.J.W. [83] M. Roś-Mazurczyk, A. Wojakowska, Ł. Marczak, K. Polański, M. Pietrowska,
L. Aerts, Somatic mutations drive distinct imaging phenotypes in lung cancer, J. Polanska, R. Dziadziuszko, J. Jassem, W. Rzyman, P. Widlak, Panel of serum
Cancer Res. 77 (2017) 3922–3930, https://doi.org/10.1158/0008-5472.CAN-17- metabolites discriminates cancer patients and healthy participants of lung cancer
0122. screening − a pilot study. Acta Biochim. Pol. 64 (2017) 513–518, https://doi.org/
[64] S. Bakr, O. Gevaert, S. Echegaray, K. Ayers, M. Zhou, M. Shafiq, H. Zheng, J. 10.18388/abp.2017_1517.
A. Benson, W. Zhang, A.N.C. Leung, M. Kadoch, C.D. Hoang, J. Shrager, A. Quon, [84] J.F. Fahrmann, D. Grapov, B.C. DeFelice, S. Taylor, K. Kim, K. Kelly, W.R. Wikoff,
D.L. Rubin, S.K. Plevritis, S. Napel, A radiogenomic dataset of non-small cell lung H. Pass, W.N. Rom, O. Fiehn, S. Miyamoto, Serum phosphatidylethanolamine
cancer, Sci. Data 5 (2018), 180202, https://doi.org/10.1038/sdata.2018.202. levels distinguish benign from malignant solitary pulmonary nodules and
[65] M.A. Mazurowski, Radiogenomics: what it is and why it is important, J. Am. Coll. represent a potential diagnostic biomarker for lung cancer, Cancer Biomark. 16
Radiol. 12 (2015) 862–866, https://doi.org/10.1016/j.jacr.2015.04.019. (2016) 609–617, https://doi.org/10.3233/CBM-160602.
[66] A.E. Revelo, A. Martin, R. Velasquez, P.C. Kulandaisamy, J. Bustamante, [85] B.T. Pan, K. Teng, C. Wu, M. Adam, R.M. Johnstone, Electron microscopic
S. Keshishyan, G. Otterson, Liquid biopsy for lung cancers: an update on recent evidence for externalization of the transferrin receptor in vesicular form in sheep
developments, Ann. Transl. Med. 7 (2019) 349, https://doi.org/10.21037/ reticulocytes. J. Cell Biol. 101 (1985) 942–948, https://doi.org/10.1083/
atm.2019.03.28. jcb.101.3.942.
[67] B. Yang, A. Qin, K. Zhang, H. Ren, S. Liu, X. Liu, X. Pan, G. Yu, Circulating tumor [86] C. Théry, A. Regnault, J. Garin, J. Wolfers, L. Zitvogel, P. Ricciardi-Castagnoli,
cells predict prognosis following tyrosine kinase inhibitor treatment in EGFR- G. Raposo, S. Amigorena, Molecular characterization of dendritic cell-derived
mutant non-small cell lung cancer patients, Oncol. Res. 25 (2017) 1601–1606, exosomes. Selective accumulation of the heat shock protein hsc73, J. Cell Biol.
https://doi.org/10.3727/096504017X14928634401178. 147 (1999) 599–610, https://doi.org/10.1083/jcb.147.3.599.
[68] M. Provencio, M. Torrente, V. Calvo, D. Pérez-Callejo, L. Gutiérrez, F. Franco, [87] N. Blanchard, D. Lankar, F. Faure, A. Regnault, C. Dumont, G. Raposo, C. Hivroz,
C. Pérez-Barrios, M. Barquín, A. Royuela, F. García-García, C. Bueno, A. Garcia- TCR activation of human T cells induces the production of exosomes bearing the
Grande, C. Camps, B. Massuti, E. Sotomayor, A. Romero, Prognostic value of TCR/CD3/zeta complex, J. Immunol. 168 (2002) 3235–3241, https://doi.org/
quantitative ctDNA levels in non small cell lung cancer patients, Oncotarget 9 10.4049/jimmunol.168.7.3235.
(2018) 488–494, https://doi.org/10.18632/oncotarget.22470. [88] G. Raposo, H.W. Nijman, W. Stoorvogel, R. Liejendekker, C.V. Harding, C.
[69] C. Zhang, B. Wei, P. Li, K. Yang, Z. Wang, J. Ma, Y. Guo, Prognostic value of J. Melief, H.J. Geuze, B lymphocytes secrete antigen-presenting vesicles, J. Exp.
plasma EGFR ctDNA in NSCLC patients treated with EGFR-TKIs, PLoS One 12 Med. 183 (1996) 1161–1172, https://doi.org/10.1084/jem.183.3.1161.
(2017), 0173524, https://doi.org/10.1371/journal.pone.0173524. [89] J. Wolfers, A. Lozier, G. Raposo, A. Regnault, C. Théry, C. Masurier, C. Flament,
[70] Y. Song, S. Ma, M. Shi, X. Xu, X. Chen, Y. Wang, Z. Yang, T. Zhang, M. Li, H. Li, S. Pouzieux, F. Faure, T. Tursz, E. Angevin, S. Amigorena, L. Zitvogel, Tumor-
L. Zhang, X. Mao, Predictive and prognostic values of circulating tumor DNA derived exosomes are a source of shared tumor rejection antigens for CTL cross-
(ctDNA) clearance in osimertinib treated advanced non-small cell lung cancer priming, Nat. Med. 7 (2001) 297–303, https://doi.org/10.1038/85438.
cohort, JCO 37 (2019) 3036, https://doi.org/10.1200/JCO.2019.37.15_ [90] M. Ostrowski, N.B. Carmo, S. Krumeich, I. Fanget, G. Raposo, A. Savina, C.
suppl.3036. F. Moita, K. Schauer, A.N. Hume, R.P. Freitas, B. Goud, P. Benaroch, N. Hacohen,
[71] Y. Song, C. Hu, Z. Xie, L. Wu, Z. Zhu, C. Rao, L. Liu, Y. Chen, N. Liang, J. Chen, M. Fukuda, C. Desnos, M.C. Seabra, F. Darchen, S. Amigorena, L.F. Moita,
C. Hu, N. Yang, J. Hu, W. Zhao, G. Tong, X. Dong, D. Zheng, M. Jin, J. Chen, C. Thery, Rab27a and Rab27b control different steps of the exosome secretion
M. Huang, Y. He, R. Rosell, G. Lippi, M. Mino-Kenudson, H. Han-Zhang, X. Mao, pathway, Nat. Cell Biol. 12 (2010) 13–19, https://doi.org/10.1038/ncb2000.
L. Zhang, H. Liu, J.K. Field, S. Chuai, J. Ye, Y. Han, S. Lu, Circulating tumor DNA [91] H. Zheng, Y. Zhan, S. Liu, J. Lu, J. Luo, J. Feng, S. Fan, The roles of tumor-derived
clearance predicts prognosis across treatment regimen in a large real-world exosomes in non-small cell lung cancer and their clinical implications, J. Exp.
longitudinally monitored advanced non-small cell lung cancer cohort, Transl. Clin. Cancer Res. 37 (2018) 226, https://doi.org/10.1186/s13046-018-0901-5.
Lung Cancer Res 9 (2020) 269–279, https://doi.org/10.21037/tlcr.2020.03.17. [92] G. Rabinowits, C. Gerçel-Taylor, J.M. Day, D.D. Taylor, G.H. Kloecker, Exosomal
[72] Y. Lee, S. Park, W.S. Kim, J.C. Lee, S.J. Jang, J. Choi, C.-M. Choi, Correlation MicroRNA: a diagnostic marker for lung cancer, Clin. Lung Cancer 10 (2009)
between progression-free survival, tumor burden, and circulating tumor DNA in 42–46, https://doi.org/10.3816/CLC.2009.n.006.
the initial diagnosis of advanced-stage EGFR-mutated non-small cell lung cancer, [93] B. Sandfeld-Paulsen, N. Aggerholm-Pedersen, R. Bæk, K.R. Jakobsen,
Thorac. Cancer 9 (2018) 1104–1110, https://doi.org/10.1111/1759- P. Meldgaard, B.H. Folkersen, T.R. Rasmussen, K. Varming, M.M. Jørgensen, B.
7714.12793. S. Sorensen, Exosomal proteins as prognostic biomarkers in non-small cell lung
[73] S. Cargnin, P.L. Canonico, A.A. Genazzani, S. Terrazzino, Quantitative analysis of cancer, Mol. Oncol. 10 (2016) 1595–1602, https://doi.org/10.1016/j.
circulating cell-free DNA for correlation with lung cancer survival: a systematic molonc.2016.10.003.
review and meta-analysis, J. Thorac. Oncol. Publ. Int. Assoc. Study Lung Cancer [94] K.R. Jakobsen, B.S. Paulsen, R. Bæk, K. Varming, B.S. Sorensen, M.M. Jørgensen,
12 (2017) 43–53, https://doi.org/10.1016/j.jtho.2016.08.002. Exosomal proteins as potential diagnostic markers in advanced non-small cell
[74] A. Szpechcinski, P. Rudzinski, W. Kupis, R. Langfort, T. Orlowski, lung carcinoma, J. Extracell. Vesicles 4 (2015) 26659, https://doi.org/10.3402/
J. Chorostowska-Wynimko, Plasma cell-free DNA levels and integrity in patients jev.v4.26659.
with chest radiological findings: NSCLC versus benign lung nodules, Cancer Lett. [95] L. Montermini, B. Meehan, D. Garnier, W.J. Lee, T.H. Lee, A. Guha, K. Al-Nedawi,
374 (2016) 202–207, https://doi.org/10.1016/j.canlet.2016.02.002. J. Rak, Inhibition of oncogenic epidermal growth factor receptor kinase triggers
[75] W. Gai, K. Sun, Epigenetic biomarkers in cell-free DNA and applications in liquid release of exosome-like extracellular vesicles and impacts their phosphoprotein
biopsy, Genes 10 (2019) 32, https://doi.org/10.3390/genes10010032. and DNA content, J. Biol. Chem. 290 (2015) 24534–24546, https://doi.org/
[76] G. Weiss, A. Schlegel, D. Kottwitz, T. König, R. Tetzner, Validation of the SHOX2/ 10.1074/jbc.M115.679217.
PTGER4 DNA methylation marker panel for plasma-based discrimination [96] R. Kalluri, V.S. LeBleu, Discovery of double-stranded genomic DNA in circulating
between patients with malignant and nonmalignant lung disease, J. Thorac. exosomes, Cold Spring Harb. Symp. Quant. Biol. 81 (2016) 275–280, https://doi.
Oncol. Publ. Int. Assoc. Study Lung Cancer 12 (2017) 77–84, https://doi.org/ org/10.1101/sqb.2016.81.030932.
10.1016/j.jtho.2016.08.123. [97] N. Hollings, P. Shaw, Diagnostic imaging of lung cancer, Eur. Respir. J. 19 (2002)
[77] S.Y. Shen, R. Singhania, G. Fehringer, A. Chakravarthy, M.H.A. Roehrl, 722–742, https://doi.org/10.1183/09031936.02.00280002.
D. Chadwick, P.C. Zuzarte, A. Borgida, T.T. Wang, T. Li, O. Kis, Z. Zhao, [98] P.K. Nguyen, J.C. Wu, Radiation exposure from imaging tests: is there an
A. Spreafico, T. da, S. Medina, Y. Wang, D. Roulois, I. Ettayebi, Z. Chen, S. Chow, increased cancer risk? Expert Rev. Cardiovasc. Ther. 9 (2011) 177–183, https://
T. Murphy, A. Arruda, G.M. O’Kane, J. Liu, M. Mansour, J.D. McPherson, doi.org/10.1586/erc.10.184.
C. O’Brien, N. Leighl, P.L. Bedard, N. Fleshner, G. Liu, M.D. Minden, S. Gallinger, [99] K. Trudgen, N.H. Khattar, E. Bensadoun, S. Arnold, A.J. Stromberg, E.
A. Goldenberg, T.J. Pugh, M.M. Hoffman, S.V. Bratman, R.J. Hung, D.D. A. Hirschowitz, Autoantibody profiling for lung cancer screening longitudinal
De Carvalho, Sensitive tumour detection and classification using plasma cell-free

12
F. Cucchiara et al. Pharmacological Research 169 (2021) 105643

retrospective analysis of CT screening cohorts, PLoS One 9 (2014) 87947, https:// [120] T.S. Mok, Y.-L. Wu, M.-J. Ahn, M.C. Garassino, H.R. Kim, S.S. Ramalingam, F.
doi.org/10.1371/journal.pone.0087947. A. Shepherd, Y. He, H. Akamatsu, W.S.M.E. Theelen, C.K. Lee, M. Sebastian,
[100] C.I. Bayarri-Lara, D. de Miguel Pérez, A. Cueto Ladrón de Guevara, A. Rodriguez A. Templeton, H. Mann, M. Marotti, S. Ghiorghiu, V.A. Papadimitrakopoulou,
Fernández, J.L. Puche, A. Sánchez-Palencia Ramos, J. Ruiz Zafra, C.F. Giraldo Osimertinib or platinum-pemetrexed in EGFR T790M-positive lung cancer,
Ospina, M. Delgado-Rodríguez, M. Expósito Ruiz, M.J. Moyano Rodriguez, J. N. Engl. J. Med. 376 (2017) 629–640, https://doi.org/10.1056/
A. Lorente, M.J. Serrano, Association of circulating tumour cells with early NEJMoa1612674.
relapse and 18F-fluorodeoxyglucose positron emission tomography uptake in [121] A. Passaro, E. Guerini-Rocco, A. Pochesci, D. Vacirca, G. Spitaleri, C.M. Catania,
resected non-small-cell lung cancers. Eur. J. Cardio-Thorac. Surg. J. Eur. Assoc. A. Rappa, M. Barberis, F. de Marinis, Targeting EGFR T790M mutation in NSCLC:
Cardio-Thorac. Surg. 52 (2017) 55–62, https://doi.org/10.1093/ejcts/ezx049. from biology to evaluation and treatment, Pharmacol. Res. 117 (2017) 406–415,
[101] S. Hawkins, H. Wang, Y. Liu, A. Garcia, O. Stringfield, H. Krewer, Q. Li, https://doi.org/10.1016/j.phrs.2017.01.003.
D. Cherezov, R.A. Gatenby, Y. Balagurunathan, D. Goldgof, M.B. Schabath, [122] A. Passaro, T. Mok, S. Peters, S. Popat, M.-J. Ahn, F. de Marinis, Recent advances
L. Hall, R.J. Gillies, Predicting malignant nodules from screening CT scans, on the role of EGFR tyrosine kinase inhibitors in the management of NSCLC with
J. Thorac. Oncol. Publ. Int. Assoc. Study Lung Cancer 11 (2016) 2120–2128, uncommon, non exon 20 insertions, EGFR mutations, J. Thorac. Oncol. Publ. Int.
https://doi.org/10.1016/j.jtho.2016.07.002. Assoc. Study Lung Cancer (2020), https://doi.org/10.1016/j.jtho.2020.12.002.
[102] W. Wu, H. Hu, J. Gong, X. Li, G. Huang, S. Nie, Malignant-benign classification of [123] H. Akamatsu, Y. Koh, I. Okamoto, D. Fujimoto, A. Bessho, K. Azuma, S. Morita,
pulmonary nodules based on random forest aided by clustering analysis, Phys. N. Yamamoto, K. Nakagawa, Clinical significance of monitoring EGFR mutation
Med. Biol. 64 (2019), 035017, https://doi.org/10.1088/1361-6560/aafab0. in plasma using multiplexed digital PCR in EGFR mutated patients treated with
[103] S. Suo, J. Cheng, M. Cao, Q. Lu, Y. Yin, J. Xu, H. Wu, Assessment of heterogeneity afatinib (West Japan Oncology Group 8114LTR study), Lung Cancer 131 (2019)
difference between edge and core by using texture analysis: differentiation of 128–133, https://doi.org/10.1016/j.lungcan.2019.03.021.
malignant from inflammatory pulmonary nodules and masses, Acad. Radiol. 23 [124] Y. Kuang, A. Rogers, B.Y. Yeap, L. Wang, M. Makrigiorgos, K. Vetrand, S. Thiede,
(2016) 1115–1122, https://doi.org/10.1016/j.acra.2016.04.009. R.J. Distel, P.A. Jänne, Noninvasive detection of EGFR T790M in gefitinib or
[104] J.R.J. Ferreira, M.C. Oliveira, P.M. de Azevedo-Marques, Characterization of erlotinib resistant non-small cell lung cancer. Clin. Cancer Res. J. Am. Assoc.
pulmonary nodules based on features of margin sharpness and texture, J. Digit. Cancer Res. 15 (2009) 2630–2636, https://doi.org/10.1158/1078-0432.CCR-08-
Imaging 31 (2018) 451–463, https://doi.org/10.1007/s10278-017-0029-8. 2592.
[105] W. Wu, L.A. Pierce, Y. Zhang, S.N.J. Pipavath, T.W. Randolph, K.J. Lastwika, P. [125] T.K. Sundaresan, L.V. Sequist, J.V. Heymach, G.J. Riely, P.A. Jänne, W.H. Koch, J.
D. Lampe, A.M. Houghton, H. Liu, L. Xia, P.E. Kinahan, Comparison of prediction P. Sullivan, D.B. Fox, R. Maher, A. Muzikansky, A. Webb, H.T. Tran, U. Giri,
models with radiological semantic features and radiomics in lung cancer M. Fleisher, H.A. Yu, W. Wei, B.E. Johnson, T.A. Barber, J.R. Walsh, J.
diagnosis of the pulmonary nodules: a case-control study, Eur. Radiol. 29 (2019) A. Engelman, S.L. Stott, R. Kapur, S. Maheswaran, M. Toner, D.A. Haber,
6100–6108, https://doi.org/10.1007/s00330-019-06213-9. Detection of T790M, the acquired resistance EGFR mutation, by tumor biopsy
[106] Y. Balagurunathan, M.B. Schabath, H. Wang, Y. Liu, R.J. Gillies, Quantitative versus noninvasive blood-based analyses, Clin. Cancer Res. J. Am. Assoc. Cancer
imaging features improve discrimination of malignancy in pulmonary nodules, Res. 22 (2016) 1103–1110, https://doi.org/10.1158/1078-0432.CCR-15-1031.
Sci. Rep. 9 (2019) 8528, https://doi.org/10.1038/s41598-019-44562-z. [126] G.R. Oxnard, K.S. Thress, R.S. Alden, R. Lawrance, C.P. Paweletz, M. Cantarini, J.
[107] L. Brunese, B. Greco, F.R. Setola, F. Lassandro, M.R. Guarracino, M. De Rimini, C.-H. Yang, J.C. Barrett, P.A. Jänne, Association between plasma genotyping and
S. Piccolo, N. De Rosa, R. Muto, A. Bianco, P. Muto, R. Grassi, A. Rotondo, Non- outcomes of treatment with osimertinib (AZD9291) in advanced non–small-cell
small cell lung cancer evaluated with quantitative contrast-enhanced CT and PET- lung cancer, J. Clin. Oncol. 34 (2016) 3375–3382, https://doi.org/10.1200/
CT: net enhancement and standardized uptake values are related to tumour size JCO.2016.66.7162.
and histology. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 19 (2013) 95–101, [127] G.R. Oxnard, The cellular origins of drug resistance in cancer, Nat. Med. 22
https://doi.org/10.12659/msm.883759. (2016) 232–234, https://doi.org/10.1038/nm.4058.
[108] F. Bianconi, I. Palumbo, M.L. Fravolini, R. Chiari, M. Minestrini, L. Brunese, [128] K. Suda, I. Murakami, K. Sakai, H. Mizuuchi, S. Shimizu, K. Sato, K. Tomizawa,
B. Palumbo, Texture analysis on [(18)F]FDG PET/CT in non-small-cell lung S. Tomida, Y. Yatabe, K. Nishio, T. Mitsudomi, Small cell lung cancer
cancer: correlations between PET features, CT features, and histological types, transformation and T790M mutation: complimentary roles in acquired resistance
Mol. Imaging Biol. 21 (2019) 1200–1209, https://doi.org/10.1007/s11307-019- to kinase inhibitors in lung cancer, Sci. Rep. 5 (2015) 14447, https://doi.org/
01336-3. 10.1038/srep14447.
[109] T. Fukui, T. Taniguchi, K. Kawaguchi, K. Fukumoto, S. Nakamura, Y. Sakao, [129] H. Izumi, A. Yamasaki, Y. Ueda, T. Sumikawa, H. Maeta, S. Nakamoto, E. Shimizu,
K. Yokoi, Comparisons of the clinicopathological features and survival outcomes Squamous cell carcinoma transformation from EGFR-mutated lung
between lung cancer patients with adenocarcinoma and squamous cell adenocarcinoma: a case report and literature review, Clin. Lung Cancer 19 (2018)
carcinoma, Gen. Thorac. Cardiovasc. Surg. 63 (2015) 507–513, https://doi.org/ e63–e66, https://doi.org/10.1016/j.cllc.2017.10.005.
10.1007/s11748-015-0564-5. [130] Z. Zhang, S. Yang, Q. Wang, Impact of MET alterations on targeted therapy with
[110] S. Rinaldi, R. Berardi, Lung cancer prognosis: can histological patterns and EGFR-tyrosine kinase inhibitors for EGFR-mutant lung cancer, Biomark. Res. 7
morphological features have a role in the management of lung cancer patients? (2019) 27, https://doi.org/10.1186/s40364-019-0179-6.
Ann. Transl. Med. 5 (2017) 353, https://doi.org/10.21037/atm.2017.05.18. [131] G.S. Herter-Sprie, H. Greulich, K.-K. Wong, Activating mutations in ERBB2 and
[111] W. Wu, C. Parmar, P. Grossmann, J. Quackenbush, P. Lambin, J. Bussink, R. Mak, their impact on diagnostics and treatment, Front. Oncol. 3 (2013) 86, https://doi.
H.J.W.L. Aerts, Exploratory study to identify radiomics classifiers for lung cancer org/10.3389/fonc.2013.00086.
histology, Front. Oncol. 6 (2016) 71, https://doi.org/10.3389/fonc.2016.00071. [132] J. Eng, K.M. Woo, C.S. Sima, A. Plodkowski, M.D. Hellmann, J.E. Chaft, M.G. Kris,
[112] M. Saad, T.-S. Choi, Computer-assisted subtyping and prognosis for non-small cell M.E. Arcila, M. Ladanyi, A. Drilon, Impact of concurrent PIK3CA mutations on
lung cancer patients with unresectable tumor, Comput. Med. Imaging Graph. 67 response to EGFR tyrosine kinase inhibition in EGFR-mutant lung cancers and on
(2018) 1–8, https://doi.org/10.1016/j.compmedimag.2018.04.003. prognosis in oncogene-driven lung adenocarcinomas, J. Thorac. Oncol. Off. Publ.
[113] D.S. Zander, Primary vs metastatic pulmonary adenocarcinoma: toward a fuller Int. Assoc. Study Lung Cancer 10 (2015) 1713–1719, https://doi.org/10.1097/
understanding of truth, Chest 137 (2010) 3–4, https://doi.org/10.1378/chest.09- JTO.0000000000000671.
1514. [133] J. Xu, J. Wang, S. Zhang, Mechanisms of resistance to irreversible epidermal
[114] W. Pao, J. Chmielecki, Rational, biologically based treatment of EGFR-mutant growth factor receptor tyrosine kinase inhibitors and therapeutic strategies in
non-small-cell lung cancer, Nat. Rev. Cancer 10 (2010) 760–774, https://doi.org/ non-small cell lung cancer, Oncotarget 8 (2017) 90557–90578, https://doi.org/
10.1038/nrc2947. 10.18632/oncotarget.21164.
[115] M.H. Cohen, J.R. Johnson, Y.-F. Chen, R. Sridhara, R. Pazdur, FDA drug approval [134] K.S. Thress, C.P. Paweletz, E. Felip, B.C. Cho, D. Stetson, B. Dougherty, Z. Lai,
summary: erlotinib (Tarceva) tablets, Oncologist 10 (2005) 461–466, https://doi. A. Markovets, A. Vivancos, Y. Kuang, D. Ercan, S.E. Matthews, M. Cantarini, J.
org/10.1634/theoncologist.10-7-461. C. Barrett, P.A. Jänne, G.R. Oxnard, Acquired EGFR C797S mutation mediates
[116] M. Maemondo, A. Inoue, K. Kobayashi, S. Sugawara, S. Oizumi, H. Isobe, resistance to AZD9291 in non-small cell lung cancer harboring EGFR T790M, Nat.
A. Gemma, M. Harada, H. Yoshizawa, I. Kinoshita, Y. Fujita, S. Okinaga, Med. 21 (2015) 560–562, https://doi.org/10.1038/nm.3854.
H. Hirano, K. Yoshimori, T. Harada, T. Ogura, M. Ando, H. Miyazawa, T. Tanaka, [135] S. Cui, W. Zhang, L. Xiong, F. Pan, Y. Niu, T. Chu, H. Wang, Y. Zhao, L. Jiang, Use
Y. Saijo, K. Hagiwara, S. Morita, T. Nukiwa, Gefitinib or chemotherapy for non- of capture-based next-generation sequencing to detect ALK fusion in plasma cell-
small-cell lung cancer with mutated EGFR, N. Engl. J. Med. 362 (2010) free DNA of patients with non-small-cell lung cancer, Oncotarget 8 (2017)
2380–2388, https://doi.org/10.1056/NEJMoa0909530. 2771–2780, https://doi.org/10.18632/oncotarget.13741.
[117] B. Ricciuti, S. Baglivo, A. De Giglio, R. Chiari, Afatinib in the first-line treatment [136] I. Dagogo-Jack, A.R. Brannon, L.A. Ferris, C.D. Campbell, J.J. Lin, K.R. Schultz,
of patients with non-small cell lung cancer: clinical evidence and experience. J. Ackil, S. Stevens, L. Dardaei, S. Yoda, H. Hubbeling, S.R. Digumarthy,
Ther. Adv. Respir. Dis. 12 (2018), 1753466618808659 https://doi.org/10.1177/ M. Riester, A.N. Hata, L.V. Sequist, I.T. Lennes, A.J. Iafrate, R.S. Heist, C.
1753466618808659. G. Azzoli, A.F. Farago, J.A. Engelman, J.K. Lennerz, C.H. Benes, R.J. Leary, A.
[118] J.-C. Soria, Y. Ohe, J. Vansteenkiste, T. Reungwetwattana, B. Chewaskulyong, K. T. Shaw, J.F. Gainor, Tracking the evolution of resistance to ALK tyrosine kinase
H. Lee, A. Dechaphunkul, F. Imamura, N. Nogami, T. Kurata, I. Okamoto, C. Zhou, inhibitors through longitudinal analysis of circulating tumor DNA., JCO precis,
B.C. Cho, Y. Cheng, E.K. Cho, P.J. Voon, D. Planchard, W.-C. Su, J.E. Gray, S.- Oncol (2018) (2018), https://doi.org/10.1200/PO.17.00160.
M. Lee, R. Hodge, M. Marotti, Y. Rukazenkov, S.S. Ramalingam, Osimertinib in [137] P. Bordi, M. Tiseo, E. Rofi, I. Petrini, G. Restante, R. Danesi, M. Del Re, Detection
untreated EGFR-mutated advanced non-small-cell lung cancer, N. Engl. J. Med. of ALK and KRAS mutations in circulating tumor DNA of patients with advanced
378 (2018) 113–125, https://doi.org/10.1056/NEJMoa1713137. ALK-positive NSCLC with disease progression during crizotinib treatment, Clin.
[119] D. Westover, J. Zugazagoitia, B.C. Cho, C.M. Lovly, L. Paz-Ares, Mechanisms of Lung Cancer 18 (2017) 692–697, https://doi.org/10.1016/j.cllc.2017.04.013.
acquired resistance to first-and second-generation EGFR tyrosine kinase [138] C.E. McCoach, C.M. Blakely, K.C. Banks, B. Levy, B.M. Chue, V.M. Raymond, A.
inhibitors, Ann. Oncol. 29 (2018) i10–i19, https://doi.org/10.1093/annonc/ T. Le, C.E. Lee, J. Diaz, S.N. Waqar, W.T. Purcell, D.L. Aisner, K.D. Davies, R.
mdx703. B. Lanman, A.T. Shaw, R.C. Doebele, Clinical utility of cell-free DNA for the

13
F. Cucchiara et al. Pharmacological Research 169 (2021) 105643

detection of ALK fusions and genomic mechanisms of ALK inhibitor resistance in H. Sakai, I. Albert, A. Vergnenegre, S. Peters, K. Syrigos, F. Barlesi, M. Reck,
non-small cell lung cancer, Clin. Cancer Res. J. Am. Assoc. Cancer Res. 24 (2018) H. Borghaei, J.R. Brahmer, K.J. O’Byrne, W.J. Geese, P. Bhagavatheeswaran, S.
2758–2770, https://doi.org/10.1158/1078-0432.CCR-17-2588. K. Rabindran, R.S. Kasinathan, F.E. Nathan, S.S. Ramalingam, Nivolumab plus
[139] S.M. Gadgeel, T.S.K. Mok, S. Peters, J.A.A. Alexander, N.B. Leighl, ipilimumab in advanced non–small-cell lung cancer, N. Engl. J. Med. 381 (2019)
V. Sriuranpong, M. Perol, G. De Castro, E. Nadal, F. De Marinis, J.-Y. Han, M. Yan, 2020–2031, https://doi.org/10.1056/NEJMoa1910231.
T. Riehl, E. Schleifman, S.M. Paul, S. Mocci, D. Shames, M.S. Mathisen, [159] F.S. Hodi, V. Chiarion-Sileni, R. Gonzalez, J.-J. Grob, P. Rutkowski, C.L. Cowey,
R. Dziadziuszko, Phase II/III blood first assay screening trial (BFAST) in patients C.D. Lao, D. Schadendorf, J. Wagstaff, R. Dummer, P.F. Ferrucci, M. Smylie,
(pts) with treatment-naïve NSCLC: Initial results from the ALK+ cohort, Ann. A. Hill, D. Hogg, I. Marquez-Rodas, J. Jiang, J. Rizzo, J. Larkin, J.D. Wolchok,
Oncol. 30 (2019) v918, https://doi.org/10.1093/annonc/mdz394.079. Nivolumab plus ipilimumab or nivolumab alone versus ipilimumab alone in
[140] N.B. Leighl, R.D. Page, V.M. Raymond, D.B. Daniel, S.G. Divers, K.L. Reckamp, M. advanced melanoma (CheckMate 067): 4-year outcomes of a multicentre,
A. Villalona-Calero, D. Dix, J.I. Odegaard, R.B. Lanman, V. randomised, phase 3 trial, Lancet Oncol. 19 (2018) 1480–1492, https://doi.org/
A. Papadimitrakopoulou, Clinical utility of comprehensive cell-free DNA analysis 10.1016/S1470-2045(18)30700-9.
to identify genomic biomarkers in patients with newly diagnosed metastatic non- [160] M. Reck, D. Rodríguez-Abreu, A.G. Robinson, R. Hui, T. Csőszi, A. Fülöp,
small cell lung cancer, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res 25 (2019) M. Gottfried, N. Peled, A. Tafreshi, S. Cuffe, M. O’Brien, S. Rao, K. Hotta, M.
4691–4700, https://doi.org/10.1158/1078-0432.CCR-19-0624. A. Leiby, G.M. Lubiniecki, Y. Shentu, R. Rangwala, J.R. Brahmer, M. O’Brien,
[141] O. Gevaert, J. Xu, C.D. Hoang, A.N. Leung, Y. Xu, A. Quon, D.L. Rubin, S. Napel, S. S. Rao, K. Hotta, M.A. Leiby, G.M. Lubiniecki, Y. Shentu, R. Rangwala, J.
K. Plevritis, Non-small cell lung cancer: identifying prognostic imaging R. Brahmer, KEYNOTE-024 investigators, pembrolizumab versus chemotherapy
biomarkers by leveraging public gene expression microarray data–methods and for PD-L1–positive non–small-cell lung cancer, N. Engl. J. Med. 375 (2016)
preliminary results, Radiology 264 (2012) 387–396, https://doi.org/10.1148/ 1823–1833, https://doi.org/10.1056/NEJMoa1606774.
radiol.12111607. [161] E.B. Garon, N.A. Rizvi, R. Hui, N. Leighl, A.S. Balmanoukian, J.P. Eder,
[142] D.F. Halpenny, G.J. Riely, S. Hayes, H. Yu, J. Zheng, C.S. Moskowitz, M. A. Patnaik, C. Aggarwal, M. Gubens, L. Horn, E. Carcereny, M.J. Ahn, E. Felip, J.
S. Ginsberg, Are there imaging characteristics associated with lung S. Lee, M.D. Hellmann, O. Hamid, J.W. Goldman, J.C. Soria, M. Dolled-Filhart, R.
adenocarcinomas harboring ALK rearrangements? Lung Cancer 86 (2014) Z. Rutledge, J. Zhang, J.K. Lunceford, R. Rangwala, G.M. Lubiniecki, C. Roach,
190–194, https://doi.org/10.1016/j.lungcan.2014.09.007. K. Emancipator, L. Gandhi, Pembrolizumab for the treatment of non-small-cell
[143] E. Ozkan, A. West, J.A. Dedelow, B.F. Chu, W. Zhao, V.O. Yildiz, G.A. Otterson, lung cancer, N. Engl. J. Med. 372 (2015) 2018–2028, https://doi.org/10.1056/
K. Shilo, S. Ghosh, M. King, R.D. White, B.S. Erdal, CT gray-level texture analysis NEJMoa1501824.
as a quantitative imaging biomarker of epidermal growth factor receptor [162] T.S.K. Mok, Y.-L. Wu, I. Kudaba, D.M. Kowalski, B.C. Cho, H.Z. Turna, G.J. Castro,
mutation status in adenocarcinoma of the lung, Ajr. Am. J. Roentgenol. 205 V. Srimuninnimit, K.K. Laktionov, I. Bondarenko, K. Kubota, G.M. Lubiniecki,
(2015) 1016–1025, https://doi.org/10.2214/AJR.14.14147. J. Zhang, D. Kush, G. Lopes, Pembrolizumab versus chemotherapy for previously
[144] H.J.W.L. Aerts, P. Grossmann, Y. Tan, G.R. Oxnard, N. Rizvi, L.H. Schwartz, untreated, PD-L1-expressing, locally advanced or metastatic non-small-cell lung
B. Zhao, Defining a radiomic response phenotype: a pilot study using targeted cancer (KEYNOTE-042): a randomised, open-label, controlled, phase 3 trial,
therapy in NSCLC, Sci. Rep. 6 (2016) 33860, https://doi.org/10.1038/srep33860. Lancet (London, England) 393 (2019) 1819–1830, https://doi.org/10.1016/
[145] E.E.C. De Jong, W. Van Elmpt, L.E.L. Hendriks, R.T.H. Leijenaar, A.M. S0140-6736(18)32409-7.
C. Dingemans, P. Lambin, OC-0609: radiomic CT features for evaluation of EGFR [163] M.A. Socinski, R.M. Jotte, F. Cappuzzo, F. Orlandi, D. Stroyakovskiy, N. Nogami,
and KRAS mutation status in patients with advanced NSCLC, Radiother. Oncol. D. Rodríguez-Abreu, D. Moro-Sibilot, C.A. Thomas, F. Barlesi, G. Finley, C. Kelsch,
119 (2016) S290–S291, https://doi.org/10.1016/s0167-8140(16)31859-x. A. Lee, S. Coleman, Y. Deng, Y. Shen, M. Kowanetz, A. Lopez-Chavez, A. Sandler,
[146] J. Guan, N.J. Xiao, M. Chen, W.L. Zhou, Y.W. Zhang, S. Wang, Y.M. Dai, L. Li, M. Reck, Atezolizumab for first-line treatment of metastatic nonsquamous NSCLC,
Y. Zhang, Q.Y. Li, X.Z. Li, M. Yang, H.B. Wu, L.H. Chen, L.Y. Liu, 18F-FDG uptake N. Engl. J. Med. 378 (2018) 2288–2301, https://doi.org/10.1056/
for prediction EGFR mutation status in non-small cell lung cancer, Medicine NEJMoa1716948.
(Baltimore) 95 (2016) 4421, https://doi.org/10.1097/MD.0000000000004421. [164] H. West, M. McCleod, M. Hussein, A. Morabito, A. Rittmeyer, H.J. Conter, H.-
[147] J. Lv, H. Zhang, J. Ma, Y. Ma, G. Gao, Z. Song, Y. Yang, Comparison of CT G. Kopp, D. Daniel, S. McCune, T. Mekhail, A. Zer, N. Reinmuth, A. Sadiq,
radiogenomic and clinical characteristics between EGFR and KRAS mutations in A. Sandler, W. Lin, T. Ochi Lohmann, V. Archer, L. Wang, M. Kowanetz,
lung adenocarcinomas, Clin. Radiol. 73 (2018) 590.e1–590.e8, https://doi.org/ F. Cappuzzo, Atezolizumab in combination with carboplatin plus nab-paclitaxel
10.1016/j.crad.2018.01.009. chemotherapy compared with chemotherapy alone as first-line treatment for
[148] M. Zhou, A. Leung, S. Echegaray, A. Gentles, J.B. Shrager, K.C. Jensen, G.J. Berry, metastatic non-squamous non-small-cell lung cancer (IMpower130): a
S.K. Plevritis, D.L. Rubin, S. Napel, O. Gevaert, Non–small cell lung cancer multicentre, randomised, open-label, phase 3 tri, Lancet Oncol. 20 (2019)
radiogenomics map identifies relationships between molecular and imaging 924–937, https://doi.org/10.1016/S1470-2045(19)30167-6.
phenotypes with prognostic implications, Radiology 286 (2017) 307–315, [165] S.J. Antonia, A. Villegas, D. Daniel, D. Vicente, S. Murakami, R. Hui, T. Kurata,
https://doi.org/10.1148/radiol.2017161845. A. Chiappori, K.H. Lee, M. de Wit, B.C. Cho, M. Bourhaba, X. Quantin, T. Tokito,
[149] B. Sacconi, M. Anzidei, A. Leonardi, F. Boni, L. Saba, R. Scipione, M. Anile, T. Mekhail, D. Planchard, Y.-C. Kim, C.S. Karapetis, S. Hiret, G. Ostoros,
M. Rengo, F. Longo, M. Bezzi, F. Venuta, A. Napoli, A. Laghi, C. Catalano, Analysis K. Kubota, J.E. Gray, L. Paz-Ares, J. de Castro Carpeño, C. Faivre-Finn, M. Reck,
of CT features and quantitative texture analysis in patients with lung J. Vansteenkiste, D.R. Spigel, C. Wadsworth, G. Melillo, M. Taboada, P.A. Dennis,
adenocarcinoma: a correlation with EGFR mutations and survival rates, Clin. M. Özgüroğlu, Overall survival with durvalumab after chemoradiotherapy in
Radiol. 72 (2017) 443–450, https://doi.org/10.1016/j.crad.2017.01.015. stage III NSCLC, N. Engl. J. Med. 379 (2018) 2342–2350, https://doi.org/
[150] S.S.F. Yip, J. Kim, T.P. Coroller, C. Parmar, E.R. Velazquez, E. Huynh, R.H. Mak, 10.1056/NEJMoa1809697.
H.J.W.L. Aerts, Associations between somatic mutations and metabolic imaging [166] S.J. Antonia, A. Villegas, D. Daniel, D. Vicente, S. Murakami, R. Hui, T. Yokoi,
phenotypes in non-small cell lung cancer, J. Nucl. Med. 58 (2017) 569–576, A. Chiappori, K.H. Lee, M. de Wit, B.C. Cho, M. Bourhaba, X. Quantin, T. Tokito,
https://doi.org/10.2967/jnumed.116.181826. T. Mekhail, D. Planchard, Y.-C. Kim, C.S. Karapetis, S. Hiret, G. Ostoros,
[151] L. Zhang, B. Chen, X. Liu, J. Song, M. Fang, C. Hu, D. Dong, W. Li, J. Tian, K. Kubota, J.E. Gray, L. Paz-Ares, J. de Castro Carpeño, C. Wadsworth, G. Melillo,
Quantitative biomarkers for prediction of epidermal growth factor receptor H. Jiang, Y. Huang, P.A. Dennis, M. Özgüroğlu, Durvalumab after
mutation in non-small cell lung cancer, Transl. Oncol. 11 (2018) 94–101, https:// chemoradiotherapy in stage III non–small-cell lung cancer, N. Engl. J. Med. 377
doi.org/10.1016/j.tranon.2017.10.012. (2017) 1919–1929, https://doi.org/10.1056/NEJMoa1709937.
[152] T.J. Kim, C.-T. Lee, S.H. Jheon, J.-S. Park, J.-H. Chung, Radiologic characteristics [167] S.C. Wei, C.R. Duffy, J.P. Allison, Fundamental mechanisms of immune
of surgically resected non-small cell lung cancer with ALK rearrangement or EGFR checkpoint blockade therapy, Cancer Discov. 8 (2018) 1069–1086, https://doi.
mutations, Ann. Thorac. Surg. 101 (2016) 473–480, https://doi.org/10.1016/j. org/10.1158/2159-8290.CD-18-0367.
athoracsur.2015.07.062. [168] M. Ilié, E. Szafer-Glusman, V. Hofman, E. Chamorey, S. Lalvée, E. Selva, S. Leroy,
[153] R. Berenguer, M.D.R. Pastor-Juan, J. Canales-Vazquez, M. Castro-Garcia, M. C.-H. Marquette, M. Kowanetz, P. Hedge, E. Punnoose, P. Hofman, Detection of
V. Villas, F. Mansilla Legorburo, S. Sabater, Radiomics of CT features may be PD-L1 in circulating tumor cells and white blood cells from patients with
nonreproducible and redundant: influence of CT acquisition parameters, advanced non-small-cell lung cancer, Ann. Oncol. J. Eur. Soc. Med. Oncol. 29
Radiology 288 (2018) 407–415, https://doi.org/10.1148/radiol.2018172361. (2018) 193–199, https://doi.org/10.1093/annonc/mdx636.
[154] P. Grossmann, O. Stringfield, N. El-Hachem, M.M. Bui, E. Rios Velazquez, [169] D.L. Adams, D.K. Adams, J. He, N. Kalhor, M. Zhang, T. Xu, H. Gao, J.M. Reuben,
C. Parmar, R.T. Leijenaar, B. Haibe-Kains, P. Lambin, R.J. Gillies, H.J. Aerts, Y. Qiao, R. Komaki, Z. Liao, M.J. Edelman, C.-M. Tang, S.H. Lin, Sequential
Defining the biological basis of radiomic phenotypes in lung cancer, Elife 6 tracking of PD-L1 expression and RAD50 induction in circulating tumor and
(2017), https://doi.org/10.7554/eLife.23421. stromal cells of lung cancer patients undergoing radiotherapy, Clin. Cancer Res. J.
[155] E. Neri, M. Del Re, F. Paiar, P. Erba, P. Cocuzza, D. Regge, R. Danesi, Radiomics Am. Assoc. Cancer Res. 23 (2017) 5948–5958, https://doi.org/10.1158/1078-
and liquid biopsy in oncology: the holons of systems medicine, Insights Imaging 9 0432.CCR-17-0802.
(2018) 915–924, https://doi.org/10.1007/s13244-018-0657-7. [170] D.J. Boffa, R.P. Graf, M.C. Salazar, J. Hoag, D. Lu, R. Krupa, J. Louw, L. Dugan,
[156] N. Emaminejad, W. Qian, Y. Guan, M. Tan, Y. Qiu, H. Liu, B. Zheng, Fusion of Y. Wang, M. Landers, M. Suraneni, S.B. Greene, M. Magaña, S. Makani,
quantitative image and genomic biomarkers to improve prognosis assessment of L. Bazhenova, R.V. Dittamore, J. Nieva, Cellular expression of PD-L1 in the
early stage lung cancer patients, IEEE Trans. Biomed. Eng. 63 (2016) 1034–1043, peripheral blood of lung cancer patients is associated with worse survival, Cancer
https://doi.org/10.1109/TBME.2015.2477688. Epidemiol. Biomark. Prev. a Publ. Am. Assoc. Cancer Res. Cosponsored Am. Soc.
[157] A.C. Chiang, R.S. Herbst, Frontline immunotherapy for NSCLC — the tale of the Prev. Oncol. 26 (2017) 1139–1145, https://doi.org/10.1158/1055-9965.EPI-17-
tail, Nat. Rev. Clin. Oncol. 17 (2020) 73–74, https://doi.org/10.1038/s41571- 0120.
019-0317-y. [171] M. Dhar, J. Wong, J. Che, M. Matsumoto, T. Grogan, D. Elashoff, E.B. Garon, J.
[158] M.D. Hellmann, L. Paz-Ares, R. Bernabe Caro, B. Zurawski, S.-W. Kim, W. Goldman, E. Sollier Christen, D. Di Carlo, R.P. Kulkarni, Evaluation of PD-L1
E. Carcereny Costa, K. Park, A. Alexandru, L. Lupinacci, E. de la Mora Jimenez,

14
F. Cucchiara et al. Pharmacological Research 169 (2021) 105643

expression on vortex-isolated circulating tumor cells in metastatic lung cancer, model for non-small cell lung cancer, Sci. Rep. 8 (2018) 1922, https://doi.org/
Sci. Rep. 8 (2018) 2592, https://doi.org/10.1038/s41598-018-19245-w. 10.1038/s41598-018-20471-5.
[172] N. Guibert, M. Delaunay, A. Lusque, N. Boubekeur, I. Rouquette, E. Clermont, [189] V. Velcheti, M. Alilou, M. Khunger, R. Thawani, A. Madabhushi, Changes in
J. Mourlanette, S. Gouin, I. Dormoy, G. Favre, J. Mazieres, A. Pradines, PD-L1 computer extracted features of vessel tortuosity on CT scans post-treatment in
expression in circulating tumor cells of advanced non-small cell lung cancer responders compared to non-responders for non-small cell lung cancer on
patients treated with nivolumab, Lung Cancer 120 (2018) 108–112, https://doi. immunotherapy, JCO 35 (2017) 11518, https://doi.org/10.1200/
org/10.1016/j.lungcan.2018.04.001. JCO.2017.35.15_suppl.11518.
[173] G. Kallergi, E.-K. Vetsika, D. Aggouraki, E. Lagoudaki, A. Koutsopoulos, F. Koinis, [190] M. Khorrami, P. Prasanna, A. Gupta, P. Patil, P.D. Velu, R. Thawani, G.
P. Katsarlinos, M. Trypaki, I. Messaritakis, C. Stournaras, V. Georgoulias, G. Corredor, M. Alilou, K. Bera, P. Fu, M. Feldman, V. Velcheti, A. Madabhushi,
A. Kotsakis, Evaluation of PD-L1/PD-1 on circulating tumor cells in patients with Changes in CT radiomic features associated with lymphocyte distribution predict
advanced non-small cell lung cancer, Ther. Adv. Med. Oncol. 10 (2018), overall survival and response to immunotherapy in non-small cell lung cancer,
1758834017750121, https://doi.org/10.1177/1758834017750121. Cancer Immunol. Res. 8 (2020) 108–119, https://doi.org/10.1158/2326-6066.
[174] C. Nicolazzo, C. Raimondi, M. Mancini, S. Caponnetto, A. Gradilone, O. Gandini, CIR-19-0476.
M. Mastromartino, G. Del Bene, A. Prete, F. Longo, E. Cortesi, P. Gazzaniga, [191] R. Sun, E.J. Limkin, M. Vakalopoulou, L. Dercle, S. Champiat, S.R. Han,
Monitoring PD-L1 positive circulating tumor cells in non-small cell lung cancer L. Verlingue, D. Brandao, A. Lancia, S. Ammari, A. Hollebecque, J.-Y. Scoazec,
patients treated with the PD-1 inhibitor Nivolumab, Sci. Rep. 6 (2016) 31726, A. Marabelle, C. Massard, J.-C. Soria, C. Robert, N. Paragios, E. Deutsch, C. Ferté,
https://doi.org/10.1038/srep31726. A radiomics approach to assess tumour-infiltrating CD8 cells and response to anti-
[175] S.-P. Li, Q.-L. Guan, D. Zhao, G.-J. Pei, H.-X. Su, L.-N. Du, J.-X. He, Z.-C. Liu, PD-1 or anti-PD-L1 immunotherapy: an imaging biomarker, retrospective
Detection of circulating tumor cells by fluorescent immunohistochemistry in multicohort study, Lancet Oncol. 19 (2018) 1180–1191, https://doi.org/
patients with esophageal squamous cell carcinoma: potential clinical applications, 10.1016/S1470-2045(18)30413-3.
Med. Sci. Monit. Int. Med. J. Exp. Clin. Res 22 (2016) 1654–1662, https://doi. [192] H.J. Yoon, J. Kang, H. Park, I. Sohn, S.-H. Lee, H.Y. Lee, Deciphering the tumor
org/10.12659/msm.898335. microenvironment through radiomics in non-small cell lung cancer: Correlation
[176] A. Asgarova, K. Asgarov, Y. Godet, P. Peixoto, A. Nadaradjane, M. Boyer-Guittaut, with immune profiles, PLoS One 15 (2020), 0231227, https://doi.org/10.1371/
J. Galaine, D. Guenat, V. Mougey, J. Perrard, J.R. Pallandre, A. Bouard, journal.pone.0231227.
J. Balland, C. Tirole, O. Adotevi, E. Hendrick, M. Herfs, P.F. Cartron, C. Borg, [193] Y. Xie, M. Khunger, R. Thawani, V. Velcheti, A. Madabhushi, Metastasis-free
E. Hervouet, PD-L1 expression is regulated by both DNA methylation and NF-kB survival is a strong surrogate of overall survival in localized prostate cancer,
during EMT signaling in non-small cell lung carcinoma, Oncoimmunology 7 J. Clin. Oncol. 35 (2017) 3097–3104, https://doi.org/10.1200/JCO.2017.35.15_
(2018), 1423170, https://doi.org/10.1080/2162402X.2017.1423170. suppl.e14534.
[177] Q.-X. Qu, Q. Huang, Y. Shen, Y.-B. Zhu, X.-G. Zhang, The increase of circulating [194] I. Tunali, Y. Tan, J.E. Gray, E. Katsoulakis, S.A. Eschrich, J. Saller, T. Boyle, J. Qi,
PD-L1-expressing CD68(+) macrophage in ovarian cancer. Tumour Biol. J. Int. A. Guvenis, R.J. Gillies, M.B. Schabath, Hypoxia-related radiomics predict
Soc. Oncodev. Biol. Med. 37 (2016) 5031–5037, https://doi.org/10.1007/ immunotherapy response: a multi-cohort study of NSCLC, BioRxiv (2020),
s13277-015-4066-y. https://doi.org/10.1101/2020.04.02.020859.
[178] J.L. Schehr, Z.D. Schultz, J.W. Warrick, D.J. Guckenberger, H.M. Pezzi, J. [195] S. Trebeschi, S.G. Drago, N.J. Birkbak, I. Kurilova, A.M. Calin, A. Delli Pizzi,
M. Sperger, E. Heninger, A. Saeed, T. Leal, K. Mattox, A.M. Traynor, T. F. Lalezari, D.M.J. Lambregts, M.W. Rohaan, C. Parmar, E.A. Rozeman, K.
C. Campbell, S.M. Berry, D.J. Beebe, J.M. Lang, High specificity in circulating J. Hartemink, C. Swanton, J.B.A.G. Haanen, C.U. Blank, E.F. Smit, R.G.H. Beets-
tumor cell identification is required for accurate evaluation of programmed Tan, H.J.W.L. Aerts, Predicting response to cancer immunotherapy using
death-ligand 1, PLoS One 11 (2016), 0159397, https://doi.org/10.1371/journal. noninvasive radiomic biomarkers. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 30
pone.0159397. (2019) 998–1004, https://doi.org/10.1093/annonc/mdz108.
[179] D.H. Kim, H. Kim, Y.J. Choi, S.Y. Kim, J.-E. Lee, K.J. Sung, Y.H. Sung, C.-G. Pack, [196] I. Tunali, J.E. Gray, J. Qi, M. Abdalah, D.K. Jeong, A. Guvenis, R.J. Gillies, M.
M. Jung, B. Han, K. Kim, W.S. Kim, S.J. Nam, C.-M. Choi, M. Yun, J.C. Lee, J. B. Schabath, Novel clinical and radiomic predictors of rapid disease progression
K. Rho, Exosomal PD-L1 promotes tumor growth through immune escape in non- phenotypes among lung cancer patients treated with immunotherapy: An early
small cell lung cancer, Exp. Mol. Med. 51 (2019) 1–13, https://doi.org/10.1038/ report, Lung Cancer 129 (2019) 75–79, https://doi.org/10.1016/j.
s12276-019-0295-2. lungcan.2019.01.010.
[180] M. Del Re, R. Marconcini, G. Pasquini, E. Rofi, C. Vivaldi, F. Bloise, G. Restante, [197] P. Lambin, R.G.P.M. van Stiphout, M.H.W. Starmans, E. Rios-Velazquez,
E. Arrigoni, C. Caparello, M.G. Bianco, S. Crucitta, I. Petrini, E. Vasile, A. Falcone, G. Nalbantov, H.J.W.L. Aerts, E. Roelofs, W. van Elmpt, P.C. Boutros, P. Granone,
R. Danesi, M. Grazia Bianco, S. Crucitta, I. Petrini, E. Vasile, A. Falcone, R. Danesi, V. Valentini, A.C. Begg, D. De Ruysscher, A. Dekker, Predicting outcomes in
PD-L1 mRNA expression in plasma-derived exosomes is associated with response radiation oncology–multifactorial decision support systems, Nat. Rev. Clin. Oncol.
to anti-PD-1 antibodies in melanoma and NSCLC, Br. J. Cancer 118 (2018) 10 (2013) 27–40, https://doi.org/10.1038/nrclinonc.2012.196.
820–824, https://doi.org/10.1038/bjc.2018.9. [198] R. Joober, N. Schmitz, L. Annable, P. Boksa, Publication bias: what are the
[181] M. Del Re, R.H.N. van Schaik, S. Fogli, R.H.J. Mathijssen, F. Cucchiara, challenges and can they be overcome? J. Psychiatry Neurosci. 37 (2012)
A. Capuano, C. Scavone, G.W. Jenster, R. Danesi, Blood-based PD-L1 analysis in 149–152, https://doi.org/10.1503/jpn.120065.
tumor-derived extracellular vesicles: applications for optimal use of anti-PD-1/ [199] I. Buvat, F. Orlhac, The dark side of radiomics: on the paramount importance of
PD-L1 axis inhibitors, Biochim Biophys. Acta Rev. Cancer 1875 (2021), 188463, publishing negative results, J. Nucl. Med. 60 (2019) 1543–1544, https://doi.org/
https://doi.org/10.1016/j.bbcan.2020.188463. 10.2967/jnumed.119.235325.
[182] M.D. Hellmann, T.-E. Ciuleanu, A. Pluzanski, J.S. Lee, G.A. Otterson, C. Audigier- [200] R. Thawani, M. McLane, N. Beig, S. Ghose, P. Prasanna, V. Velcheti,
Valette, E. Minenza, H. Linardou, S. Burgers, P. Salman, H. Borghaei, S. A. Madabhushi, Radiomics and radiogenomics in lung cancer: a review for the
S. Ramalingam, J. Brahmer, M. Reck, K.J. O’Byrne, W.J. Geese, G. Green, clinician, Lung Cancer 115 (2018) 34–41, https://doi.org/10.1016/j.
H. Chang, J. Szustakowski, P. Bhagavatheeswaran, D. Healey, Y. Fu, F. Nathan, lungcan.2017.10.015.
L. Paz-Ares, Nivolumab plus ipilimumab in lung cancer with a high tumor [201] C. Hassani, B.A. Varghese, J. Nieva, V. Duddalwar, Radiomics in pulmonary
mutational burden, N. Engl. J. Med. 378 (2018) 2093–2104, https://doi.org/ lesion imaging, Ajr. Am. J. Roentgenol. 212 (2019) 497–504, https://doi.org/
10.1056/NEJMoa1801946. 10.2214/AJR.18.20623.
[183] F. Koeppel, S. Blanchard, C. Jovelet, B. Genin, C. Marcaillou, E. Martin, [202] A. Chalkidou, M.J. O’Doherty, P.K. Marsden, False discovery rates in PET and CT
E. Rouleau, E. Solary, J.-C. Soria, F. André, L. Lacroix, Whole exome sequencing studies with texture features: a systematic review, PLoS One 10 (2015), 0124165,
for determination of tumor mutation load in liquid biopsy from advanced cancer https://doi.org/10.1371/journal.pone.0124165.
patients, PLoS One 12 (2017), 0188174, https://doi.org/10.1371/journal. [203] N. Karachaliou, A.E. Sosa, M.A. Molina, M. Centelles Ruiz, R. Rosell, Possible
pone.0188174. application of circulating free tumor DNA in non-small cell lung cancer patients,
[184] F. Fenizia, R. Pasquale, C. Roma, F. Bergantino, A. Iannaccone, N. Normanno, J. Thorac. Dis. 9 (2017) S1364–S1372, https://doi.org/10.21037/jtd.2017.09.59.
Measuring tumor mutation burden in non-small cell lung cancer: tissue versus [204] P. Hofman, S. Heeke, C. Alix-Panabières, K. Pantel, Liquid biopsy in the era of
liquid biopsy, Transl. Lung Cancer Res 7 (2018) 668–677, https://doi.org/ immuno-oncology: is it ready for prime-time use for cancer patients? Ann. Oncol.
10.21037/tlcr.2018.09.23. Off. J. Eur. Soc. Med. Oncol. 30 (2019) 1448–1459, https://doi.org/10.1093/
[185] D.R. Camidge, R.C. Doebele, K.M. Kerr, Comparing and contrasting predictive annonc/mdz196.
biomarkers for immunotherapy and targeted therapy of NSCLC, Nat. Rev. Clin. [205] J. Saarenheimo, N. Eigeliene, H. Andersen, M. Tiirola, A. Jekunen, The value of
Oncol. 16 (2019) 341–355, https://doi.org/10.1038/s41571-019-0173-9. liquid biopsies for guiding therapy decisions in non-small cell lung cancer, Front.
[186] M. Ilie, J. Benzaquen, V. Hofman, S. Lassalle, N. Yazbeck, S. Leroy, S. Heeke, Oncol. 9 (2019) 129, https://doi.org/10.3389/fonc.2019.00129.
C. Bence, B. Mograbi, N. Glaichenhaus, C.-H. Marquette, P. Hofman, [206] B.A. Sisson, J. Uvalic, K. Kelly, P. Selvam, A.N. Hesse, G. Ananda, H. Chandok,
Immunotherapy in non-small cell lung cancer: biological principles and future D. Bergeron, L. Holinka, H.V. Reddi, Technical and regulatory considerations for
opportunities, Curr. Mol. Med. 17 (2017) 527–540, https://doi.org/10.2174/ taking liquid biopsy to the clinic: validation of the JAX plasmaMonitor(TM) assay,
1566524018666180222114038. Biomark. Insights 14 (2019), 1177271919826545, https://doi.org/10.1177/
[187] A. Passaro, I. Attili, S. Morganti, E. Del Signore, L. Gianoncelli, G. Spitaleri, 1177271919826545.
V. Stati, C. Catania, G. Curigliano, F. de Marinis, Clinical features affecting [207] A.W. Hahn, R.H. Nussenzveig, S.K. Pal, N. Agarwal, Blood- and tissue-based
survival in metastatic NSCLC treated with immunotherapy: a critical review of tumor genomics: a battle royale or match made in heaven? Ann. Oncol. 28 (2017)
published data, Cancer Treat. Rev. 89 (2020), 102085, https://doi.org/10.1016/j. 2333–2335, https://doi.org/10.1093/annonc/mdx418.
ctrv.2020.102085. [208] R. Shatsky, B.A. Parker, N.Q. Bui, T. Helsten, R.B. Schwab, S.G. Boles,
[188] C. Tang, B. Hobbs, A. Amer, X. Li, C. Behrens, J.R. Canales, E.P. Cuentas, R. Kurzrock, Next-generation sequencing of tissue and circulating tumor DNA: the
P. Villalobos, D. Fried, J.Y. Chang, D.S. Hong, J.W. Welsh, B. Sepesi, L. Court, I. UC San Diego Moores Center for personalized cancer therapy experience with
I. Wistuba, E.J. Koay, Development of an immune-pathology informed radiomics

15
F. Cucchiara et al. Pharmacological Research 169 (2021) 105643

breast malignancies, Mol. Cancer Ther. 18 (2019) 1001–1011, https://doi.org/ M. Baca, B.G. Kermani, H. Eltoukhy, A. Talasaz, Analytical and clinical validation
10.1158/1535-7163.MCT-17-1038. of a digital sequencing panel for quantitative, highly accurate evaluation of cell-
[209] C. Abbosh, N.J. Birkbak, G.A. Wilson, M. Jamal-Hanjani, T. Constantin, R. Salari, free circulating tumor DNA, PLoS One 10 (2015), 0140712, https://doi.org/
J. Le Quesne, D.A. Moore, S. Veeriah, R. Rosenthal, T. Marafioti, E. Kirkizlar, T.B. 10.1371/journal.pone.0140712.
K. Watkins, N. McGranahan, S. Ward, L. Martinson, J. Riley, F. Fraioli, M. Al [211] M.W. Schmitt, S.R. Kennedy, J.J. Salk, E.J. Fox, J.B. Hiatt, L.A. Loeb, Detection of
Bakir, E. Grönroos, F. Zambrana, R. Endozo, W.L. Bi, F.M. Fennessy, N. Sponer, ultra-rare mutations by next-generation sequencing, Proc. Natl. Acad. Sci. 109
D. Johnson, J. Laycock, S. Shafi, J. Czyzewska-Khan, A. Rowan, T. Chambers, (2012) 14508–14513, https://doi.org/10.1073/pnas.1208715109.
N. Matthews, S. Turajlic, C. Hiley, S.M. Lee, M.D. Forster, T. Ahmad, M. Falzon, [212] L.M. Sholl, G.R. Oxnard, C.P. Paweletz, Traditional diagnostics versus disruptive
E. Borg, D. Lawrence, M. Hayward, S. Kolvekar, N. Panagiotopoulos, S.M. Janes, technology: the role of the pathologist in the era of liquid biopsy, Cancer Res. 80
R. Thakrar, A. Ahmed, F. Blackhall, Y. Summers, D. Hafez, A. Naik, A. Ganguly, (2020) 3197–3199, https://doi.org/10.1158/0008-5472.CAN-20-0134.
S. Kareht, R. Shah, L. Joseph, A. Marie Quinn, P.A. Crosbie, B. Naidu, [213] R.J. Gillies, A.R. Anderson, R.A. Gatenby, D.L. Morse, The biology underlying
G. Middleton, G. Langman, S. Trotter, M. Nicolson, H. Remmen, K. Kerr, molecular imaging in oncology: from genome to anatome and back again, Clin.
M. Chetty, L. Gomersall, D.A. Fennell, A. Nakas, S. Rathinam, G. Anand, S. Khan, Radiol. 65 (2010) 517–521, https://doi.org/10.1016/j.crad.2010.04.005.
P. Russell, V. Ezhil, B. Ismail, M. Irvin-Sellers, V. Prakash, J.F. Lester, [214] R. Lo Gullo, I. Daimiel, E.A. Morris, K. Pinker, Combining molecular and imaging
M. Kornaszewska, R. Attanoos, H. Adams, H. Davies, D. Oukrif, A.U. Akarca, J. metrics in cancer: radiogenomics, Insights Imaging 11 (2020) 1, https://doi.org/
A. Hartley, H.L. Lowe, S. Lock, N. Iles, H. Bell, Y. Ngai, G. Elgar, Z. Szallasi, R. 10.1186/s13244-019-0795-6.
F. Schwarz, J. Herrero, A. Stewart, S.A. Quezada, K.S. Peggs, P. Van Loo, C. Dive, [215] E.J. Limkin, R. Sun, L. Dercle, E.I. Zacharaki, C. Robert, S. Reuzé, A. Schernberg,
C.J. Lin, M. Rabinowitz, H.J.W.L. Aerts, A. Hackshaw, J.A. Shaw, B. N. Paragios, E. Deutsch, C. Ferté, Promises and challenges for the implementation
G. Zimmermann, C. Swanton, M. Jamal-Hanjani, C. Abbosh, S. Veeriah, S. Shafi, of computational medical imaging (radiomics) in oncology, Ann. Oncol. Off. J.
J. Czyzewska-Khan, D. Johnson, J. Laycock, L. Bosshard-Carter, G. Goh, Eur. Soc. Med. Oncol. 28 (2017) 1191–1206, https://doi.org/10.1093/annonc/
R. Rosenthal, P. Gorman, N. Murugaesu, R.E. Hynds, G.A. Wilson, N.J. Birkbak, T. mdx034.
B.K. Watkins, N. McGranahan, S. Horswell, M. Al Bakir, E. Grönroos, R. Mitter, [216] Precision Medicine Market Worth $85.5 Billion by 2025 | CAGR: 9.9%, n.d.
M. Escudero, A. Stewart, P. Van Loo, A. Rowan, H. Xu, S. Turajlic, C. Hiley, 〈https://www.grandviewresearch.com/press-release/global-precision-medicine-
J. Goldman, R.K. Stone, T. Denner, N. Matthews, G. Elgar, S. Ward, J. Biggs, diagnostics-therapeutics-market〉 (accessed 25 July 2020).
M. Costa, S. Begum, B. Phillimore, T. Chambers, E. Nye, S. Graca, K. Joshi, [217] J. de Leon, Teaching medical students how to think: narrative, mechanistic and
A. Furness, A. Ben Aissa, Y.N.S. Wong, A. Georgiou, S.A. Quezada, K.S. Peggs, J. mathematical thinking, Actas Esp. Psiquiatr. 46 (2018) 133–145.
A. Hartley, H.L. Lowe, J. Herrero, D. Lawrence, M. Hayward, N. Panagiotopoulos, [218] J. de Leon, C. De Las Cuevas, The art of pharmacotherapy: reflections on
S. Kolvekar, M. Falzon, E. Borg, T. Marafioti, C. Simeon, G. Hector, A. Smith, pharmacophobia, J. Clin. Psychopharmacol. 37 (2017) 131–137, https://doi.org/
M. Aranda, M. Novelli, D. Oukrif, A.U. Akarca, S.M. Janes, R. Thakrar, M. 10.1097/JCP.0000000000000675.
D. Forster, T. Ahmad, S.M. Lee, D. Papadatos-Pastos, D. Carnell, R. Mendes, [219] AMA, AMA Digital Health Study: Physicians’ motivations and requirements for
J. George, N. Navani, A. Ahmed, M. Taylor, J. Choudhary, Y. Summers, adopting digital clinical tools | Adoption and attitudinal shifts from 2016 to 2019,
R. Califano, P. Taylor, R. Shah, P. Krysiak, K. Rammohan, E. Fontaine, R. Booton, 2020; 1–37. 〈https://www.ama-assn.org/practice-management/digital〉.
M. Evison, P.A. Crosbie, S. Moss, F. Idries, L. Joseph, P. Bishop, A. Chaturvedi, A. [220] P. Lambin, R.T.H. Leijenaar, T.M. Deist, J. Peerlings, E.E.C. de Jong, J. van
M. Quinn, H. Doran, A. Leek, P. Harrison, K. Moore, R. Waddington, J. Novasio, Timmeren, S. Sanduleanu, R.T.H.M. Larue, A.J.G. Even, A. Jochems, Y. van Wijk,
F. Blackhall, J. Rogan, E. Smith, C. Dive, J. Tugwood, G. Brady, D.G. Rothwell, H. Woodruff, J. van Soest, T. Lustberg, E. Roelofs, W. van Elmpt, A. Dekker, F.
F. Chemi, J. Pierce, S. Gulati, B. Naidu, G. Langman, S. Trotter, M. Bellamy, M. Mottaghy, J.E. Wildberger, S. Walsh, Radiomics: the bridge between medical
H. Bancroft, A. Kerr, S. Kadiri, J. Webb, G. Middleton, M. Djearaman, D. imaging and personalized medicine, Nat. Rev. Clin. Oncol. 14 (2017) 749–762,
A. Fennell, J.A. Shaw, J. Le Quesne, D.A. Moore, A. Thomas, H. Walter, J. Riley, https://doi.org/10.1038/nrclinonc.2017.141.
L. Martinson, A. Nakas, S. Rathinam, W. Monteiro, H. Marshall, L. Nelson, [221] L. Leyens, M. Reumann, N. Malats, A. Brand, Use of big data for drug development
J. Bennett, L. Primrose, G. Anand, S. Khan, A. Amadi, M. Nicolson, K. Kerr, and for public and personal health and care, Genet. Epidemiol. 41 (2017) 51–60,
S. Palmer, H. Remmen, J. Miller, K. Buchan, M. Chetty, L. Gomersall, J.F. Lester, https://doi.org/10.1002/gepi.22012.
A. Edwards, F. Morgan, H. Adams, H. Davies, M. Kornaszewska, R. Attanoos, [222] C. Auffray, R. Balling, I. Barroso, L. Bencze, M. Benson, J. Bergeron, E. Bernal-
S. Lock, A. Verjee, M. MacKenzie, M. Wilcox, H. Bell, N. Iles, A. Hackshaw, Delgado, N. Blomberg, C. Bock, A. Conesa, S. Del Signore, C. Delogne, P. Devilee,
Y. Ngai, S. Smith, N. Gower, C. Ottensmeier, S. Chee, B. Johnson, A. Alzetani, A. Di Meglio, M. Eijkemans, P. Flicek, N. Graf, V. Grimm, H.-J. Guchelaar, Y.-
E. Shaw, E. Lim, P. De Sousa, M.T. Barbosa, A. Bowman, S. Jordan, A. Rice, K. Guo, I.G. Gut, A. Hanbury, S. Hanif, R.-D. Hilgers, Á. Honrado, D.R. Hose,
H. Raubenheimer, C. Proli, M.E. Cufari, J.C. Ronquillo, A. Kwayie, H. Bhayani, J. Houwing-Duistermaat, T. Hubbard, S.H. Janacek, H. Karanikas, T. Kievits,
M. Hamilton, Y. Bakar, N. Mensah, L. Ambrose, A. Devaraj, S. Buderi, J. Finch, M. Kohler, A. Kremer, J. Lanfear, T. Lengauer, E. Maes, T. Meert, W. Müller,
L. Azcarate, H. Chavan, S. Green, H. Mashinga, A.G. Nicholson, K. Lau, M. Sheaff, D. Nickel, P. Oledzki, B. Pedersen, M. Petkovic, K. Pliakos, M. Rattray, J.R. i Màs,
P. Schmid, T. Tracer. consortium, Phylogenetic ctDNA analysis depicts early-stage R. Schneider, T. Sengstag, X. Serra-Picamal, W. Spek, L.A.I. Vaas, O. van
lung cancer evolution, Nature 545 (2017) 446–451, https://doi.org/10.1038/ Batenburg, M. Vandelaer, P. Varnai, P. Villoslada, J.A. Vizcaíno, J.P.M. Wubbe,
nature22364. G. Zanetti, Making sense of big data in health research: Towards an EU action
[210] R.B. Lanman, S.A. Mortimer, O.A. Zill, D. Sebisanovic, R. Lopez, S. Blau, E. plan, Genome Med. 8 (2016) 71, https://doi.org/10.1186/s13073-016-0323-y.
A. Collisson, S.G. Divers, D.S.B. Hoon, E.S. Kopetz, J. Lee, P.G. Nikolinakos, A.

16

View publication stats

You might also like