Next Article in Journal
BDP1 Alterations Correlate with Clinical Outcomes in Breast Cancer
Previous Article in Journal
Multimodality Treatment including Surgery Related to the Type of N2 Involvement in Locally Advanced Non-Small Cell Lung Cancer
Previous Article in Special Issue
Prediction of Prostate Cancer Disease Aggressiveness Using Bi-Parametric Mri Radiomics
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Radiomics/Radiogenomics in Lung Cancer: Basic Principles and Initial Clinical Results

by
Athanasios K. Anagnostopoulos
1,*,†,
Anastasios Gaitanis
2,†,
Ioannis Gkiozos
3,
Emmanouil I. Athanasiadis
4,
Sofia N. Chatziioannou
5,
Konstantinos N. Syrigos
3,
Dimitris Thanos
4,
Achilles N. Chatziioannou
6,‡ and
Nikolaos Papanikolaou
7,8,9,10,11,‡
1
Division of Biotechnology, Center of Systems Biology, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11525 Athens, Greece
2
Clinical and Translational Research, Center of Experimental Surgery, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece
3
Third Department of Internal Medicine, “Sotiria” Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
4
Greek Genome Centre, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece
5
Nuclear Medicine Division, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece
6
First Department of Radiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
7
Computational Clinical Imaging Group, Centre for the Unknown, Champalimaud Foundation, 1400-038 Lisbon, Portugal
8
Machine Learning Group, The Royal Marsden, London SM2 5MG, UK
9
The Institute of Cancer Research, London SM2 5MG, UK
10
Karolinska Institutet, 14186 Stockholm, Sweden
11
Institute of Computer Science, FORTH, 70013 Heraklion, Greece
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
These authors contributed equally to this work.
Cancers 2022, 14(7), 1657; https://doi.org/10.3390/cancers14071657
Submission received: 11 February 2022 / Revised: 14 March 2022 / Accepted: 16 March 2022 / Published: 25 March 2022
(This article belongs to the Special Issue Radiomics/Radiogenomics in Cancer)

Abstract

:

Simple Summary

Radiogenomics is a promising new approach in cancer assessment, providing an evaluation of the molecular basis of imaging phenotypes after establishing associations between radiological features and molecular features at the genomic–transcriptomic–proteomic level. This review focuses on describing key aspects of radiogenomics while discussing limitations of translatability to the clinic and possible solutions to these challenges, providing the clinician with an up-to-date handbook on how to use this new tool.

Abstract

Lung cancer is the leading cause of cancer-related deaths worldwide, and elucidation of its complicated pathobiology has been traditionally targeted by studies incorporating genomic as well other high-throughput approaches. Recently, a collection of methods used for cancer imaging, supplemented by quantitative aspects leading towards imaging biomarker assessment termed “radiomics”, has introduced a novel dimension in cancer research. Integration of genomics and radiomics approaches, where identifying the biological basis of imaging phenotypes is feasible due to the establishment of associations between molecular features at the genomic–transcriptomic–proteomic level and radiological features, has recently emerged termed radiogenomics. This review article aims to briefly describe the main aspects of radiogenomics, while discussing its basic limitations related to lung cancer clinical applications for clinicians, researchers and patients.

1. Introduction

Cancer is a heterogeneous disease mainly caused by the accumulation of genetic and epigenetic changes, leading to aberrant cell proliferation. One of the fundamental aspects of understanding cancer pathobiology is correlating genomic alterations to cancer phenotypes. In this regard, the advent of DNA and RNA landscaping, further amplified by the human genome project and Cancer Genome Atlas, has directly linked genomic data to a vast array of tumour types [1,2]. Cancer imaging, which has traditionally been the cornerstone of disease diagnosis, staging, radiation planning and monitoring, has been lately supplemented by quantitative aspects, leading to image biomarker assessment called “radiomics” [3]. Combining the above modalities with underlying molecular features at the genomic–transcriptomic–proteomic level is known as “radiogenomics”, a novel approach that identifies the biological basis of imaging phenotypes [4].
Given the high availability of imaging data in patients with lung cancer, this specific cancer type has been “targeted” since the early days of radiomics and radiogenomics studies. In the current review, we will briefly describe the basic aspects of radiomics and radiogenomics and focus on clinical applications attracting considerable attention for reasons that we will extensively refer to in the following sections. In addition, we will discuss the challenges and problems of radiomics regarding their limited translation to the clinic and propose potential solutions to overcome these problems and therefore create added value for the management of lung cancer patients.

2. Radiomics Pipeline

With the development of texture analysis techniques, medical images are transformed into minable data by applying mathematical transformations on selected areas of tissues. A typical radiomics pipeline involves many steps [5]. Initially, a domain expert, such as a medical oncologist, needs to define the clinical use case, in other words, to formulate what the clinicians are expecting from the model to infer. Then, the relevant data source must be identified, and a bioimaging specialist will ensure that raw data are adequate for further analysis. Depending on data size, there are two options; in the case where the number of patients is a few dozens to a few hundred, the radiomics with classical machine learning (ML) algorithms should be explored first. However, if there are thousands of patients to be examined, deep neural networks could be the first choice. Following model training, validation should be made with data similar to the training cohort, the so-called internal validation, and independent data originating from other hospitals, the so-called external validation. In the following paragraphs, each pipeline step will be briefly discussed.

2.1. Defining the Problem at the Clinical Level

A successful ML application is primarily dependent on a solid, clinically meaningful formulation of the clinical use case. It is of paramount importance when considering different use cases to weigh the final decision not only on their clinical soundness, but instead on a realistic consideration of an adequate amount of data required for efficient training of the machine learning algorithm to produce a robust model that will be accurate and general [6]. The latter is certainly true when considering input data such as the medical images that are dynamic and change often based on information representation and lack of universally adopted standardised acquisition protocols [7]. In addition, the ground truth that will be selected must be robust with low variability, easily obtained, and not dependent on human interpretation. If the latter is inevitable, panels of experts must provide the ground truth to address interobserver variability effects.

2.2. Identification of Proper Data Sources

Although radiomics promotes the idea of a data-driven approach, specific domain expertise must be considered to select the meaningful potential data sources that will be recruited for further analysis. When imaging is considered, there is a preference for simplistic instead of “exotic” types of images such as conventional T1, T2, FLAIR and post-contrast images in magnetic resonance imaging (MRI) instead of an advanced biomarker imaging, such as for example, microvascular fraction quantified from an intravoxel incoherent motion diffusion experiment. Standard-of-care imaging approaches are also preferable since they are the imaging milestones in routine clinical protocols and therefore can guarantee the availability of high patient numbers.

2.3. Data Acquisition and Standardisation

All modern imaging modalities are employed in radiomics. Computed tomography (CT), MRI, positron emission tomography (PET) and single photon emission computed tomography (SPECT) are some of the clinical modalities widely employed for textural features extraction. The variety of scanners allow for a wide range of acquisition and image reconstruction protocols, and thus, the standardisation of all these protocols is necessary [8]. Furthermore, defining the accepted range of values for any textural features remains an open issue. The requirement behind data standardisation is to reduce changes due to technical details that introduce numerical variations to textural features as they are falsely associated with biological effects. For this reason, significant efforts have been undertaken towards the development of guidelines related to the acquisition protocols, the segmentation of tissues and the computation of radiomic features [9,10].

Tumour Segmentation

Following image acquisition, segmentation is one of the most critical and challenging phases for calculating textural features. There exist many challenges related to segmentation, including the increased inter-reader variability and the fact that it is a time-consuming process; even in the presence of automated artificial intelligence (AI)-powered algorithms, human intervention to correct and validate the outcomes is still necessary. Due to the latter, it is recommended that two or more readers should perform segmentation to smooth out subjective opinions regarding the exact borders and remove features that are sensitive to slight differences in the definition of the segmentation masks (Figure 1).

2.4. Feature Computation

There are three main types of radiomics features related to first level, shape and texture [11]. First-level features provide information about signal intensities across the region or the volume of interest. They are similar to the histogram metrics when signal intensity is represented in the histogram. They include mean, median, standard deviation, min, max, skewness, kurtosis, etc. Shape-related features provide structural information of the lesion and inform for two- and three-dimensional morphological characteristics. A larger surface-area-to-volume ratio, for example, is indicative of a more spiculated tumour, which is known to have more malignant potential in comparison to a round mass with a smaller ratio [12]. Thus, shape features can differentiate malignant lung nodules from benign ones. Besides, volume estimation has been shown to be better in evaluating treatment response than conventional methods [13,14,15,16]. Finally, texture- and gradient-related features refer to the quantitative ability to retain spatial relationships and interactions between pixel intensities in a given local neighbourhood [17]. Texture features are known to measure tumour heterogeneity and have been identified as the most closely correlated to outcomes in lung cancer.

2.5. Feature Selection

In radiomics, as in other omics applications, one of the most critical problems that needs to be addressed is called p >> n, which means that the number of patients available is far lower than the number of imaging features we can extract from each patient. The process that identifies the perfect balance between the number of features and the corresponding model performance is called feature selection, which is an essential step in machine learning. Various methods, including filtered, wrapper and embedded, are used to address the dimensionality problem, where in principle, they aim to pick up the most stable, informative features to be considered for model training. An essential part of developing a radiomic signature relates to the fidelity of radiomic features that should be assessed in various levels, including robustness, temporal and spatial stability, and reproducibility. For that purpose, dedicated phantoms can perform quality control of radiomic features and compare features from different centres, with different equipment.
Feature selection is achieved most of the time in different waves. A typical workflow in the first phase permits only stable features to be forwarded. A zero or near-zero variance method removes useless features. A correlation analysis removes redundant features, and finally, a more sophisticated method such as mRMR or RFE is used for slimming down our radiomic signature.

2.6. Model Selection/Assessment

According to ML best practices, several algorithms should be applied, including, among others, logistic regression, naïve Bayes, random forests, support vector machines, and boosted trees, to the training set and then the appropriate algorithm that performs best on the validation set should be selected. The latter can be used to make decisions related to the structure of the model, such as how many and which features will be used, the kinds of resampling and transformations that should be applied, and finally, the fine-tuning of the algorithm hyperparameters. We conclude with a single finalised model with an estimated prediction error following this process. A model should be applied to the independent dataset originating from different institutions to assess the generalisation error. The latter served as an indication of the model’s capability to perform equally well on external data (external validation) instead of the initial validation performed with data derived from the same institution, known as “internal” validation. It is essential to mention that the metric to be used to assess the model’s performance should be appropriate to the specific requirements of the use case. For example, accuracy as a performance metric can be misleading in a highly imbalanced problem. Depending on the cost of false positives and false negatives, someone should use f1, f0.5 or f2 scores.

3. Clinical Applications of Radiomics/Radiogenomics in Lung Cancer

In principle, all available modern digital imaging modalities can be used to perform radiogenomics modelling. Specifically, in the case of lung cancer, the most common modalities are CT and PET–CT, given the leading role they have in the clinical diagnostic workup of patients with lung cancer.
Zhou et al. [18] demonstrated an existing correlation between CT Hounsfield attenuation measurements and lesion margins with cell-cycle genes. They noted that the presence of irregular borders and ground-glass opacities in the lesion correlated with EGFR expression. In a study by Rizzo et al. [19], EGFR mutation was shown to be associated with CT features such as the presence of air bronchogram, pleural retraction, small lesion size, and absence of fibrosis. In contrast, ALK mutation was associated with pleural effusion. Round shape, nodules in non-tumour lobes, and smoking were variables linked to KRAS mutation. Other, less frequent mutations such as RET and ROS1, which comprise 1–2% of all lung adenocarcinomas, have also been assessed for associations with imaging features [20].
In a study of 26 patients, Gevaert et al. [21] discovered that the presence of air bronchograms was associated with overexpression of the K-ras oncogene. Weiss et al. [22] also found that imaging features could predict the K-ras status of patients with NSCLC. As a predictive marker, KRAS has been linked to round shape, nodules in non-tumour lobes, and multiple small nodules, as well as general radiomic profiles. Some studies reported predicting EGFR, but not KRAS. ALK rearrangement was linked to pleural effusion and lobulated margins [23,24,25].
By far, the best-characterised genetic mutation in NSCLC is EGFR (both exon 19 and exon 21 mutations) linked to contrast, Laws-Energy, median Hounsfield unit (HU), SUVmax, pleural retraction, small size, speculation, irregular nodules, poorly defined margins, ground glass, emphysema, locoregional infiltration, and “normalised inverse difference moment”, as well as combined radiomic profiles [26,27,28,29,30,31,32,33].
In the case of solitary pulmonary nodules, and their evaluation using 18F FDG PET, the recently published scientific results reveal that the shape and texture analysis obtained with radiomics analysis can lead to a better discrimination between benign and malignant lung nodules. More specifically, Palumbo et al. [34] examined PET–CT images from 111 patients and found that CT- and PET-based shape and texture features increase the accuracy of the prediction models. Furthermore, Caruso et al. [35] analysed CT-guided lung biopsy images of patients that had undergone a PET–CT scan with SUV less than 2.5, and they found that CT texture analysis can provide better discriminating information even in those cases without a significant variation to glucose metabolism. Finally, Zhou et al. [36] analysed PET and CT images from 769 patients, and they found that the obtained radiomic features, combined with machine learning methods, can be a discriminator tool between primary and metastatic lung lesions.
Radiogenomics aims to study the relationship between radiological and genomic features to noninvasively reveal potential biological features closely related to clinical outcomes. In a recent study, Li et al. subjected subsolid nodules from 154 lung adenocarcinoma patients to whole exome-sequencing while collecting all patient clinical and radiological data [37]. Their analyses determined that EGFR showed the most common significant mutation followed by aberrations in RBM10, TP53 and KRAS. The latter radiogenomics study revealed remarkable genomic heterogeneity in subsolid nodules, whilst inculpating CT data presented the opportunity to more accurately predict lung adenocarcinoma’s malignant degree and mutation characteristics.
In the relevant study of Zhou et al., the gene expression profiles of 113 non-small cell cancer patients were linked to their CT imaging features aiming to create a radiogenomic map [18]. Their approach revealed ten metagenes framing several molecular pathways with a predominant role of the EGFR pathway in lung cancer pathobiology by utilising RNA-sequencing technologies. Based on these features, a detailed radiogenomic map was created depicting correlations between imaging features and metagenes, showing that specific characteristics such as nodule attenuation and ground-glass opacity are associated with late-cycle genes and genes of the EGFR pathway, respectively.
Furthermore, Li et al. evaluated potential radiogenomics associations in solid lung adenocarcinoma by studying quantitative parameters obtained by dual-energy spectral CT combined with rearrangements in the three genes most involved in lung cancer molecular background [38]. A total of 96 patients were assessed for EGFR, KRAS and ALK mutations and rearrangements. These parameters were associated with quantitative parameters such as water and iodine concentration CT value at 70 keV. It was shown that spectral CT has the potential to predict KRAS and EGFR mutations as well as ALK reordering in solid lung adenocarcinoma, thus accurately reflecting molecular features of this tumour.
Gevaert et al. [21] succeeded in identifying prognostic imaging biomarkers in non–small cell lung cancer (NSCLC) by correlating clusters of co-expressed genes (metagenes) with PET–CT imaging features. This radiogenomics strategy was applied to a cohort of 26 patients with NSCLC, including (a) gene expressions, (b) a set of 180 computed tomography (CT) and positron emission tomography (PET) image features. They concluded that there exist 243 statistically significant pairwise correlations between image features and metagenes with an accuracy of 59–83%, while when the predicted image features were mapped to a public gene expression dataset with survival endpoints, several features including tumour size, edge shape, and sharpness ranked the highest for prognostic significance.

4. Definition of the Clinical Problem

Lung cancer is the most aggressive cancer regarding overall survival amongst all other cancer types globally. The choice of treatment depends on the stage of the NSCLC, the performance status, the comorbidities, the histological type and the molecular status, all of which are crucial factors determining the treatment choice.
Thoracic surgery is the recommended treatment for patients with stage I–II non-small-cell lung cancer (NSCLC), with a 5-year survival of 77–92% for clinical stage IA, 68% for stage IB, 60% for stage IIA, and 53% for clinical stage IIB. Even if an early stage of NSCLC appears simple to handle, physicians consider two issues when treating patients. First, there are patients with early-stage NSCLC that have a higher risk of recurrence based on tumour size, e.g., >4 cm, local spread, or other factors, e.g., pure solid type on high-resolution computed tomography (HRCT) [39], for which adjuvant therapy after surgery could lower the risk of recurrence. However, there is no direct correlation to the patient profile that could be benefited by adjuvant treatment. In addition, there are patients with early clinical stage NSCLC who have medical contraindications to surgical resection or who refuse surgery. For these patients, stereotactic body radiation therapy (SBRT) can lead to high local tumour control, >85% at five years, and low toxicity [40]. In this case, radiogenomics can offer additional means by providing individual information rather than population information of identifying and selecting patients best fit for adjuvant therapy or providing insights for prognosis of survival after treatment.
Grove et al. [41] found that measures of heterogeneity such as spiculation and entropy gradients are strong prognostic indicators of overall survival (OS) in patients with early-stage lung cancer. Aerts et al. [42] described a combination of features (size, shape, texture and wavelets) that could predict outcomes in patients with lung cancer. Studies from Huang et al. [43], Raghunath et al. [44], and Depeursinge et al. [45] found a correlation between their radiomic biomarker on CT and disease-free survival (DFS). Parmar et al. [46] found that their marker (comprising size, intensity, shape, texture and wavelet features) is associated with lung cancer prognosis, stage and histology, and Coroller et al. [47] provided a signature that correlated to distant metastasis.

5. Stage II NSCLC

During the last two decades, it has become clear that in early-stage II NSCLC adjuvant therapy applied after surgery is of benefit for patients with N1 and N2 disease (stage II and III), resulting overall in 4–5% absolute survival improvement at five years [38,48]. These results were obtained by administering cisplatin-based doublets, delivering at least three to four cycles. Newer adding agents, such as docetaxel, gemcitabine or pemetrexed, showed comparable efficacy.
NSCLC is only moderately sensitive to chemotherapy, with single-agent response rates in the range of 15% or better. Agents (e.g., gemcitabine, pemetrexed, docetaxel, vinorelbine) have shown promising single-agent activity, with response rates from 20–25% [49]. Patient selection criteria, such as proper recovery from surgery, the interval between surgery and the start of therapy, and the absence of significant comorbidities, are essential in awaiting the treatment benefit [48].
In this realm, radiomics is already making progress in predicting responses to adjuvant therapy. Two independent studies have suggested a potential role for radiomics in predicting pathological response to neoadjuvant chemotherapy before surgery, based on pre-treatment CT images of NSCLC [50,51]. It remains to be established whether radiogenomics can provide additional or complementary data to basic clinical decisions to select the proper patients for the appropriate adjuvant setting.

6. Locally Advanced-Stage III and Metastatic-Stage IV NSCLC

Treatment for stage III NSCLC requires a multidisciplinary approach involving a thoracic surgeon, a radiotherapist, a pulmonologist, a radiologist and an oncologist. Treatment options depend on the size of the tumour, the location of lesions, which lymph nodes it has spread to, the patient performance status, comorbidities, and how well it is foreseen to tolerate treatment. At this stage, treatment may help the patients live longer and feel better by relieving symptoms, but it is not likely to cure the disease. In most cases, relapse occurs within 1–2 years; thus, radio genomics could individualise the treatment approach by identifying certain imaging factors that could affect each patient’s response rate and overall survival depending on the genomic features of individual tumours. In our days, there is also the option for immunotherapy (a fully human, immunoglobulin G1 kappa (IgG1κ) monoclonal antibody that selectively blocks the interaction of PD-L1 with PD-1 and CD80). Immunotherapy (Durvalumab) is given for a year, as a consolidation therapy, after the completion of concurrent chemo-radiotherapy for unresectable stage III NSCLC. Thus, the knowledge of radiomics/radio genomics could improve the prognostic and predictive value of NSCLC stage III is crucial.
Unfortunately, most cases of NSCLC are diagnosed at stage IV. From this starting point, the hardships for the clinicians are many.
First, the primary and metastatic lesions coexist in the imaging appearance. There is extended heterogeneity within the lesion and among lesions in terms of imaging characteristics and mutation index. Not all lesions can be biopsied to identify the primary and secondary mutations.
Next, genotyping can be possible or not for insurance reimbursement issues. Even where possible, it will often be reimbursable for one gene/mutation. At the same time, it is recommended that screening for a specific set of mutations is what best identifies the patient to benefit most from targeted therapies [49].
Third, immunotherapy for lung cancer (mainly immune checkpoint inhibitors) may potentially achieve durable responses, however, only in a subset of patients. Still, the majority of NSLC patients do not respond to immunotherapy. Notably, the pattern of responses to immunotherapy varies, including in the standard RECIST criteria, pseudoprogression and hyperprogression. Reported rates of pseudoprogression are <10%, and rapid progressions, called hyperprogression, are reported to range from 4% to 29%. Some patients may present dissociated responses, with some lesions shrinking and others growing. In such cases, local treatment with surgery or radiotherapy for growing lesions may be considered [52]. Last, immunotherapy comes along with toxicity. A total of 73.4% of patients present any immunotherapy-related adverse event, and 26.6% present with a grade 3 or higher adverse event [53]. Monitoring for immunotherapy related pneumonitis, which when untreated can be life-threatening, is mandatory to safeguard NSCLC patients.
In this context, radiogenomics studies are anticipated to shed light and support monitoring patients, allocating them to the most suitable therapeutic option available and predicting response to treatment, the prognosis of survival, or monitoring disease control.
Computed tomography (CT) studies have provided evidence supporting the clinical decision. As mentioned above, the best-characterised genetic mutation in NSCLC was EGFR (both exon 19 and exon 21 mutations) linked to contrast [28], Laws-Energy [29], median Hounsfield unit (HU) [30], SUVmax [54], pleural retraction [14], small size [14,23], speculation [23], irregular nodules [18], poorly defined margins [18], ground glass [18,31], emphysema [32], locoregional infiltration [32], and “normalised inverse difference moment” [26], as well as combined radiomic profiles [24,25,33,55,56].
As a predictive marker, KRAS has been linked to a round shape [14], nodules in non-tumour lobes [14], and multiple small nodules [23], as well as general radiomic profiles [24,25]. ALK rearrangement was linked to pleural effusion [14] and lobulated margin [57]. Halpenny’s [58] study showed that ALK-positive tumours tended to be more prominent with more solid consistency and involved more thoracic lymphadenopathy. This study showed no correlation between ALK mutation and the presence of pleural effusion on CT. HER2, a gene often amplified during acquired resistance to EGFR-targeting therapy, was also studied [57].
Most studies derived radiomics signatures in radiotherapy planning or diagnostic images acquired before therapy. Nearly all studies evaluated patients undergoing treatment with cytotoxic chemo-radiotherapy. More recently, several studies have assessed the potential of radiomics to improve patient stratification for targeted therapies and immunotherapy agents [29]. For example, Tang and colleagues linked radiomic features to a tumour immune phenotype in patients with stage I–III NSCLC, finding patients with heterogeneous tumours, which correlated with low PD-L1 and high CD3 cell count, had better prognosis [59].
There are 24 CT studies evaluating how radiomic signatures of NSCLC relate to genomics [24,29,60,61,62,63,64,65,66], signalling pathways [67] and histopathology [67,68,69,70,71,72,73,74,75,76,77]. For example, Rios Velazquez and colleagues found distinct imaging phenotypes for EGFR and KRAS mutations from CT images of patients with NSCLC [24]. Some studies that relate radiomics to patient outcomes also relate their radiomic signature to genomics [45] or biological markers.
Collectively, these 64 studies present a positive and encouraging view of the potential for radiomics signatures to deliver personalised medicine. However, two important limitations are readily apparent. First, while nearly all studies report at least one positive association between CT radiomic signature and outcome (OS, PFS, recurrence or toxicity) or tumour biology (genomic or pathology biomarkers and signalling pathways), the particular radiomic signature derived from these analyses varies substantially between studies. Consequently, few study signatures are directly comparable with one another, and thus the literature does not identify specific candidate radiomic signatures suitable for further extensive multicentre evaluation.
In conclusion, from the radiologists and clinicians perspective, radiomics/radiogenomics could better determine the accuracy of malignancy of pulmonary nodules, which have been detected by CT scan in order to treat curatively, select patients with early-stage lung cancer who are appropriate for post-surgical treatment, and determine patients with stage III NSCLC who can tolerate immunotherapy as consolidation therapy after concurrent treatment with chemotherapy-radiation therapy. Moreover, radiomics/radiogenomics could support treatment selection by excluding NSCLC patients sensitive to hyperprogression in immunotherapy, thus avoiding an expensive and without survival benefit treatment option. They could also predict oncological outcomes such as response rate, progression-free survival and overall survival according to the baseline imaging appearance. All the above medical achievements declare that radio genomics could guide more personalized patient care.

7. Discussion

7.1. Challenges

Although radiomics appears to be straightforward and simple, many challenges should be addressed. Three essential concepts need to be carefully considered regarding the data: quantity, quality, and diversity. Radiomics is a data-hungry methodology, and in the presence of regulatory directives such as GDPR or HIPPA, easy unobstructed flow of data between institutions is difficult. High quality curated data has fundamental value for every radiomics project. The concept “garbage in garbage out” applies here; thus, we need to make an effort to gather, clean, transform and curate the data to be eligible for value creation. There are some considerations regarding the number of patients that should be used in these studies. The smaller the patient sample, the less robust the developed models; therefore, they quickly become overfitted. This is a phenomenon when the model explicitly learns the patterns found on the training data, failing to make accurate predictions on slightly different data than the training set used to build the model. In addition, the smaller the dataset used, the less representative it can be on the real-world data. It is vital to allow the ML algorithms to be exposed to diverse datasets to extract robust and reproducible patterns.
Today, there is a clear translation gap of radiomics to clinical practice due to the lack of adequate data and limited interpretability of current models. Thus, it is difficult to conceptualise how a model makes a specific prediction, compromising patient safety. In the first phase of radiomics (Radiomics 1.0), the focus was on maximising the model’s performance, often using models that are black boxes, such as neural networks. The latter harmed the capability of these models to be trusted by their end users since it was impossible to explain the inner mechanics of decision making of such models. However, several recent methods, known as explainable AI (xAI), have been developed and gradually explored and integrated into radiomics. SHAP and LIME are among the most popular methods that satisfy, to a certain extent, the high performance that black box models offer with both local and global explainability capabilities. SHAP is based on game theory, where each feature in the model has a different contribution influencing the final prediction. SHAP values reflect the latter concept and represent valuable tools to understand the decision-making process at a global level.

7.2. How to Move to Radiomics 2.0

Another common misconception of Radiomics 1.0 was that all efforts regarding model development were finalised as long as the training and validation of a model were completed. A growing number of investigators criticising the norm was based on a combination of utilising historical, retrospective data for training a model and a small finite independent cohort often called the “external” to test the model. People are shifting in the somewhat overlooked area of model deployment. Instead of training and testing the model with “outdated” retrospective data, “live” real-world data should also participate in both tasks through a continual or incremental learning scheme. Thus, using static instead of adaptive learning strategies in AI is somewhat contradictory. Quoting Stephen Hawking, “Intelligence is the ability to adapt to change”. It is well known that medical images have been designed to produce eye-pleasing pictures rather than to satisfy any basic rule that all quantitative methods respect. In addition, imaging modalities are constantly updated at a hardware and software level, causing an interesting effect that can be observed only after deploying the model. The latter is called “data drift”, and in principle, it can be responsible for the gradual deterioration of the model’s performance. There is a great need to monitor the model’s behaviour and possibly organise retraining sessions with contemporary data to keep the model in good “health” and deliver its promise. Another area that has also been significantly neglected is model usability. When designing a radiomics project, the first thing is to ask the end users and gather their input on how they need to interact with the model, obtain its predictions and exploit them in a meaningful way. Failure to identify all these concepts will significantly reduce the possibility of using the model; therefore, minimal clinical value will be generated.

8. Conclusions

Radiomics/radiogenomics could support treatment selection, excluding NSCLC patients sensitive to hyperprogression to immunotherapy, thus avoiding expensive and nonsurvivable benefit treatment options by predicting oncological outcomes such as response rate, progression-free survival and overall survival. All the above achievements declare that radiogenomics could guide more personalised patient care. However, there is still a long way until radiomics will be integrated into clinics and delivered to the patient, caregiver, or health authorities and deliver what they have promised. Inevitably, similar to all the other disruptive technologies, it must go through a maturation process. In our view, we are already there, prepared to face the challenges, since at least we may acknowledge them.

Author Contributions

Design and conception, N.P. and A.N.C.; data collection: A.K.A., A.G., I.G., E.I.A. and S.N.C.; data analysis and interpretation, all authors; drafting of the article, A.K.A., A.G. and I.G.; overall supervision, N.P., A.N.C., K.N.S. and D.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare that there exist no conflicts of interest.

References

  1. Weinstein, J.N.; Collisson, E.A.; Mills, G.B.; Shaw, K.R.; Ozenberger, B.A.; Ellrott, K.; Shmulevich, I.; Sander, C.; Stuart, J.M.; Cancer Genome Atlas Research Network. The Cancer Genome Atlas Pan-Cancer analysis project. Nat. Genet. 2013, 45, 1113–1120. [Google Scholar] [CrossRef] [PubMed]
  2. Nakagawa, H.; Fujita, M. Whole genome sequencing analysis for cancer genomics and precision medicine. Cancer Sci. 2018, 109, 513–522. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Gillies, R.J.; Kinahan, P.E.; Hricak, H. Radiomics: Images Are More than Pictures, They Are Data. Radiology 2016, 278, 563–577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Story, M.D.; Durante, M. Radiogenomics. Med. Phys. 2018, 45, e1111–e1122. [Google Scholar] [CrossRef]
  5. Papanikolaou, N.; Matos, C.; Koh, D.M. How to develop a meaningful radiomic signature for clinical use in oncologic patients. Cancer Imaging 2020, 20, 33. [Google Scholar] [CrossRef]
  6. de Farias, E.C.; di Noia, C.; Han, C.; Sala, E.; Castelli, M.; Rundo, L. Impact of GAN-based lesion-focused medical image super-resolution on the robustness of radiomic features. Sci. Rep. 2021, 11, 21361. [Google Scholar] [CrossRef] [PubMed]
  7. Santinha, J.; Matos, C.; Figueiredo, M.; Papanikolaou, N. Improving performance and generalizability in radiogenomics: A pilot study for prediction of IDH1/2 mutation status in gliomas with multicentric data. J. Med. Imaging (Bellingham) 2021, 8, 031905. [Google Scholar] [CrossRef]
  8. Galavis, P.E.; Hollensen, C.; Jallow, N.; Paliwal, B.; Jeraj, R. Variability of textural features in FDG PET images due to different acquisition modes and reconstruction parameters. Acta Oncol. 2010, 49, 1012–1016. [Google Scholar] [CrossRef] [Green Version]
  9. Cook, G.J.R.; Azad, G.; Owczarczyk, K.; Siddique, M.; Goh, V. Challenges and Promises of PET Radiomics. Int. J. Radiat. Oncol. Biol. Phys. 2018, 102, 1083–1089. [Google Scholar] [CrossRef] [Green Version]
  10. Incoronato, M.; Aiello, M.; Infante, T.; Cavaliere, C.; Grimaldi, A.M.; Mirabelli, P.; Monti, S.; Salvatore, M. Radiogenomic Analysis of Oncological Data: A Technical Survey. Int. J. Mol. Sci. 2017, 18, 805. [Google Scholar] [CrossRef] [Green Version]
  11. Rizzo, S.; Botta, F.; Raimondi, S.; Origgi, D.; Fanciullo, C.; Morganti, A.G.; Bellomi, M. Radiomics: The facts and the challenges of image analysis. Eur. Radiol. Exp. 2018, 2, 36. [Google Scholar] [CrossRef] [PubMed]
  12. Lee, G.; Lee, H.Y.; Park, H.; Schiebler, M.L.; van Beek, E.J.R.; Ohno, Y.; Seo, J.B.; Leung, A. Radiomics and its emerging role in lung cancer research, imaging biomarkers and clinical management: State of the art. Eur. J. Radiol. 2017, 86, 297–307. [Google Scholar] [CrossRef] [PubMed]
  13. Labby, Z.E.; Armato, S.G.; Dignam, J.J.; Straus, C.; Kindler, H.L.; Nowak, A.K. Lung volume measurements as a surrogate marker for patient response in malignant pleural mesothelioma. J. Thorac. Oncol. 2013, 8, 478–486. [Google Scholar] [CrossRef] [Green Version]
  14. Rizzo, S.; Petrella, F.; Passaro, A.; de Marinis, F.; Bellomi, M. Proposals for revisions of the classification of lung cancers with multiple pulmonary sites: The radiologist’s, thoracic surgeon’s and oncologist’s point of view. J. Thorac. Dis. 2016, 8, E805–E808. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Lee, H.Y.; Hyun, S.H.; Lee, K.S.; Kim, B.T.; Kim, J.; Shim, Y.M.; Ahn, M.J.; Kim, T.S.; Yi, C.A.; Chung, M.J. Volume-based parameter of 18)F-FDG PET/CT in malignant pleural mesothelioma: Prediction of therapeutic response and prognostic implications. Ann. Surg. Oncol. 2010, 17, 2787–2794. [Google Scholar] [CrossRef] [PubMed]
  16. Lee, J.H.; Lee, H.Y.; Ahn, M.J.; Park, K.; Ahn, J.S.; Sun, J.M.; Lee, K.S. Volume-based growth tumor kinetics as a prognostic biomarker for patients with EGFR mutant lung adenocarcinoma undergoing EGFR tyrosine kinase inhibitor therapy: A case control study. Cancer Imaging 2016, 16, 5. [Google Scholar] [CrossRef] [Green Version]
  17. Bhargava, R.; Madabhushi, A. Emerging Themes in Image Informatics and Molecular Analysis for Digital Pathology. Annu. Rev. Biomed. Eng. 2016, 18, 387–412. [Google Scholar] [CrossRef] [Green Version]
  18. Zhou, M.; Leung, A.; Echegaray, S.; Gentles, A.; Shrager, J.B.; Jensen, K.C.; Berry, G.J.; Plevritis, S.K.; Rubin, D.L.; Napel, S.; et al. Non-Small Cell Lung Cancer Radiogenomics Map Identifies Relationships between Molecular and Imaging Phenotypes with Prognostic Implications. Radiology 2018, 286, 307–315. [Google Scholar] [CrossRef]
  19. Rizzo, S.; Petrella, F.; Buscarino, V.; De Maria, F.; Raimondi, S.; Barberis, M.; Fumagalli, C.; Spitaleri, G.; Rampinelli, C.; De Marinis, F.; et al. CT Radiogenomic Characterization of EGFR, K-RAS, and ALK Mutations in Non-Small Cell Lung Cancer. Eur. Radiol. 2016, 26, 32–42. [Google Scholar] [CrossRef]
  20. Lo Gullo, R.; Daimiel, I.; Morris, E.A.; Pinker, K. Combining molecular and imaging metrics in cancer: Radiogenomics. Insights Imaging 2020, 11, 1. [Google Scholar] [CrossRef]
  21. Gevaert, O.; Xu, J.; Hoang, C.D.; Leung, A.N.; Xu, Y.; Quon, A.; Rubin, D.L.; Napel, S.; Plevritis, S.K. Non-small cell lung cancer: Identifying prognostic imaging biomarkers by leveraging public gene expression microarray data--methods and preliminary results. Radiology 2012, 264, 387–396. [Google Scholar] [CrossRef] [PubMed]
  22. Weiss, G.J.; Ganeshan, B.; Miles, K.A.; Campbell, D.H.; Cheung, P.Y.; Frank, S.; Korn, R.L. Noninvasive image texture analysis differentiates K-ras mutation from pan-wildtype NSCLC and is prognostic. PLoS ONE 2014, 9, e100244. [Google Scholar] [CrossRef]
  23. Lv, J.; Zhang, H.; Ma, J.; Ma, Y.; Gao, G.; Song, Z.; Yang, Y. Comparison of CT radiogenomic and clinical characteristics between EGFR and KRAS mutations in lung adenocarcinomas. Clin. Radiol. 2018, 73, 590.e591–590.e598. [Google Scholar] [CrossRef] [PubMed]
  24. Rios Velazquez, E.; Parmar, C.; Liu, Y.; Coroller, T.P.; Cruz, G.; Stringfield, O.; Ye, Z.; Makrigiorgos, M.; Fennessy, F.; Mak, R.H.; et al. Somatic Mutations Drive Distinct Imaging Phenotypes in Lung Cancer. Cancer Res. 2017, 77, 3922–3930. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Yip, S.S.F.; Parmar, C.; Kim, J.; Huynh, E.; Mak, R.H.; Aerts, H.J.W.L. Impact of experimental design on PET radiomics in predicting somatic mutation status. Eur. J. Radiol. 2017, 97, 8–15. [Google Scholar] [CrossRef]
  26. Yip, S.S.; Kim, J.; Coroller, T.P.; Parmar, C.; Velazquez, E.R.; Huynh, E.; Mak, R.H.; Aerts, H.J. Associations Between Somatic Mutations and Metabolic Imaging Phenotypes in Non-Small Cell Lung Cancer. J. Nucl. Med. 2017, 58, 569–576. [Google Scholar] [CrossRef] [PubMed]
  27. Kim, T.J.; Lee, C.T.; Jheon, S.H.; Park, J.S.; Chung, J.H. Radiologic Characteristics of Surgically Resected Non-Small Cell Lung Cancer With ALK Rearrangement or EGFR Mutations. Ann. Thorac. Surg. 2016, 101, 473–480. [Google Scholar] [CrossRef] [Green Version]
  28. Ozkan, E.; West, A.; Dedelow, J.A.; Chu, B.F.; Zhao, W.; Yildiz, V.O.; Otterson, G.A.; Shilo, K.; Ghosh, S.; King, M.; et al. CT Gray-Level Texture Analysis as a Quantitative Imaging Biomarker of Epidermal Growth Factor Receptor Mutation Status in Adenocarcinoma of the Lung. AJR Am. J. Roentgenol. 2015, 205, 1016–1025. [Google Scholar] [CrossRef] [PubMed]
  29. Aerts, H.J.; Grossmann, P.; Tan, Y.; Oxnard, G.R.; Rizvi, N.; Schwartz, L.H.; Zhao, B. Defining a Radiomic Response Phenotype: A Pilot Study using targeted therapy in NSCLC. Sci. Rep. 2016, 6, 33860. [Google Scholar] [CrossRef] [Green Version]
  30. de Jong, E.E.C.; van Elmpt, W.; Rizzo, S.; Colarieti, A.; Spitaleri, G.; Leijenaar, R.T.H.; Jochems, A.; Hendriks, L.E.L.; Troost, E.G.C.; Reymen, B.; et al. Applicability of a prognostic CT-based radiomic signature model trained on stage I-III non-small cell lung cancer in stage IV non-small cell lung cancer. Lung Cancer 2018, 124, 6–11. [Google Scholar] [CrossRef] [Green Version]
  31. Gevaert, O.; Echegaray, S.; Khuong, A.; Hoang, C.D.; Shrager, J.B.; Jensen, K.C.; Berry, G.J.; Guo, H.H.; Lau, C.; Plevritis, S.K.; et al. Predictive radiogenomics modeling of EGFR mutation status in lung cancer. Sci. Rep. 2017, 7, 41674. [Google Scholar] [CrossRef] [PubMed]
  32. Sacconi, B.; Anzidei, M.; Leonardi, A.; Boni, F.; Saba, L.; Scipione, R.; Anile, M.; Rengo, M.; Longo, F.; Bezzi, M.; et al. Analysis of CT features and quantitative texture analysis in patients with lung adenocarcinoma: A correlation with EGFR mutations and survival rates. Clin. Radiol. 2017, 72, 443–450. [Google Scholar] [CrossRef] [PubMed]
  33. Nakajima, E.C.; Frankland, M.P.; Johnson, T.F.; Antic, S.L.; Chen, H.; Chen, S.C.; Karwoski, R.A.; Walker, R.; Landman, B.A.; Clay, R.D.; et al. Assessing the inter-observer variability of Computer-Aided Nodule Assessment and Risk Yield (CANARY) to characterise lung adenocarcinomas. PLoS ONE 2018, 13, e0198118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Palumbo, B.; Bianconi, F.; Palumbo, I.; Fravolini, M.L.; Minestrini, M.; Nuvoli, S.; Stazza, M.L.; Rondini, M.; Spanu, A. Value of shape and texture features from 18F-FDG PET/CT to discriminate between benign and malignant solitary pulmonary nodules: An experimental evaluation. Diagnostics 2020, 10, 696. [Google Scholar] [CrossRef]
  35. Caruso, D.; Zerunian, M.; Daffina, J.; Polici, M.; Polidori, M.; Tipaldi, M.A.; Ronconi, E.; Pucciarelli, F.; Lucertini, E.; Rossi, M.; et al. Radiomics and functional imaging in lung cancer: The importance of radiological heterogeneity beyond FDG PET/CT and lung biopsy. Eur. J. Radiol. 2021, 142, 109874. [Google Scholar] [CrossRef]
  36. Zhou, Y.; Ma, X.L.; Zhang, T.; Wang, J.; Zhang, T.; Tian, R. Use of radiomics based on (18)F-FDG PET/CT and machine learning methods to aid clinical decision-making in the classification of solitary pulmonary lesions: An innovative approach. Eur. J. Nucl. Med. Mol. Imaging 2021, 48, 2904–2913. [Google Scholar] [CrossRef]
  37. Li, Y.; Li, X.; Li, H.; Zhao, Y.; Liu, Z.; Sun, K.; Zhu, X.; Qi, Q.; An, B.; Shen, D.; et al. Genomic characterisation of pulmonary subsolid nodules: Mutational landscape and radiological features. Eur. Respir. J. 2020, 55, 1901409. [Google Scholar] [CrossRef]
  38. Li, M.; Zhang, L.; Tang, W.; Ma, P.Q.; Zhou, L.N.; Jin, Y.J.; Qi, L.L.; Wu, N. Quantitative features of dual-energy spectral computed tomography for solid lung adenocarcinoma with. Transl. Lung Cancer Res. 2019, 8, 401–412. [Google Scholar] [CrossRef]
  39. Tsutani, Y.; Shimada, Y.; Ito, H.; Miyata, Y.; Ikeda, N.; Nakayama, H.; Okada, M. Identification of High-Risk of Recurrence in Clinical Stage I Non-Small Cell Lung Cancer. Front. Oncol. 2021, 11, 622742. [Google Scholar] [CrossRef]
  40. Hirsch, F.R.; Scagliotti, G.V.; Mulshine, J.L.; Kwon, R.; Curran, W.J.; Wu, Y.L.; Paz-Ares, L. Lung cancer: Current therapies and new targeted treatments. Lancet 2017, 389, 299–311. [Google Scholar] [CrossRef]
  41. Grove, O.; Berglund, A.E.; Schabath, M.B.; Aerts, H.J.W.L.; Dekker, A.; Wang, H.; Velazquez, E.R.; Lambin, P.; Gu, Y.; Balagurunathan, Y.; et al. Correction: Quantitative Computed Tomographic Descriptors Associate Tumor Shape Complexity and Intratumor Heterogeneity with Prognosis in Lung Adenocarcinoma. PLoS ONE 2021, 16, e0248541. [Google Scholar] [CrossRef] [PubMed]
  42. Aerts, H.J.; Velazquez, E.R.; Leijenaar, R.T.; Parmar, C.; Grossmann, P.; Carvalho, S.; Cavalho, S.; Bussink, J.; Monshouwer, R.; Haibe-Kains, B.; et al. Decoding tumour phenotype by noninvasive imaging using a quantitative radiomics approach. Nat. Commun. 2014, 5, 4006. [Google Scholar] [CrossRef] [PubMed]
  43. Huang, Y.; Liu, Z.; He, L.; Chen, X.; Pan, D.; Ma, Z.; Liang, C.; Tian, J. Radiomics Signature: A Potential Biomarker for the Prediction of Disease-Free Survival in Early-Stage (I or II) Non-Small Cell Lung Cancer. Radiology 2016, 281, 947–957. [Google Scholar] [CrossRef] [PubMed]
  44. Raghunath, S.; Maldonado, F.; Rajagopalan, S.; Karwoski, R.A.; DePew, Z.S.; Bartholmai, B.J.; Peikert, T.; Robb, R.A. Noninvasive risk stratification of lung adenocarcinoma using quantitative computed tomography. J. Thorac. Oncol. 2014, 9, 1698–1703. [Google Scholar] [CrossRef] [Green Version]
  45. Depeursinge, A.; Yanagawa, M.; Leung, A.N.; Rubin, D.L. Predicting adenocarcinoma recurrence using computational texture models of nodule components in lung CT. Med. Phys. 2015, 42, 2054–2063. [Google Scholar] [CrossRef] [Green Version]
  46. Parmar, C.; Leijenaar, R.T.; Grossmann, P.; Rios Velazquez, E.; Bussink, J.; Rieveld, D.; Rietbergen, M.M.; Haibe-Kains, B.; Lambin, P.; Aerts, H.J. Radiomic feature clusters and prognostic signatures specific for Lung and Head & Neck cancer. Sci. Rep. 2015, 5, 11044. [Google Scholar] [CrossRef]
  47. Coroller, T.P.; Grossmann, P.; Hou, Y.; Rios Velazquez, E.; Leijenaar, R.T.; Hermann, G.; Lambin, P.; Haibe-Kains, B.; Mak, R.H.; Aerts, H.J. CT-based radiomic signature predicts distant metastasis in lung adenocarcinoma. Radiother. Oncol. 2015, 114, 345–350. [Google Scholar] [CrossRef]
  48. Coroller, T.P.; Agrawal, V.; Narayan, V.; Hou, Y.; Grossmann, P.; Lee, S.W.; Mak, R.H.; Aerts, H.J. Radiomic phenotype features predict pathological response in non-small cell lung cancer. Radiother. Oncol. 2016, 119, 480–486. [Google Scholar] [CrossRef] [Green Version]
  49. Coroller, T.P.; Agrawal, V.; Huynh, E.; Narayan, V.; Lee, S.W.; Mak, R.H.; Aerts, H.J.W.L. Radiomic-Based Pathological Response Prediction from Primary Tumors and Lymph Nodes in NSCLC. J. Thorac. Oncol. 2017, 12, 467–476. [Google Scholar] [CrossRef] [Green Version]
  50. Borcoman, E.; Kanjanapan, Y.; Champiat, S.; Kato, S.; Servois, V.; Kurzrock, R.; Goel, S.; Bedard, P.; Le Tourneau, C. Novel patterns of response under immunotherapy. Ann. Oncol. 2019, 30, 385–396. [Google Scholar] [CrossRef]
  51. Haanen, J.B.A.G.; Carbonnel, F.; Robert, C.; Kerr, K.M.; Peters, S.; Larkin, J.; Jordan, K.; Committee, E.G. Management of toxicities from immunotherapy: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2017, 28, iv119–iv142. [Google Scholar] [CrossRef] [PubMed]
  52. Moon, H.; Choi, J.H.; Park, J.H.; Byun, B.H.; Lim, I.; Kim, B.I.; Choi, C.W.; Lim, S.M. Differences of 18F-FDG PET tumor texture parameters according to the presence of epidermal growth factor receptor mutation in non-small cell lung cancer. J. Nucl. Med. Conf. Soc. Nucl. Med. Mol. Imaging Annu. Meet. 2015, 56 (Suppl. 3), 1393. [Google Scholar]
  53. Sorensen, J.; Erasmus, J., Jr.; Shroff, G.; Rao, A.; Stingo, F.; Court, L.; Gold, K.A.; Lee, J.; Heymach, J.V.; Swisher, S.; et al. Combining CT texture analysis with semantic imaging descriptions for the radiogenomic detection of EGFR and KRAS mutations in NSCLC. J. Thorac. Oncol. 2015, 2, S700. [Google Scholar]
  54. Jia, T.; Xiong, J.; Li, X.; Ma, J.; Ren, Y.; Xu, Z.; Cai, X.; Zhang, J.; Zhao, J.; Fu, X. Detecting epidermal growth factor receptor mutation status in patients with lung adenocarcinoma using radiomics and random forest. J. Thorac. Oncol. 2017, 12, S1860. [Google Scholar] [CrossRef] [Green Version]
  55. Clay, R.; Johnson, T.; Bartholmai, B.J.; Karwoski, R.; Maldonado, F.; Rajagopalan, S.; Peikert, T. Canary (computer aided nodule analysis and risk yield) as a radiologic biomarker to predict tumor response to EGFR-targeted therapy in adenocarcinoma of the lung. Am. J. Respir. Crit. Care Med. 2018, 197, A4684. [Google Scholar]
  56. Zhang, L.; Chen, B.; Liu, X.; Song, J.; Fang, M.; Hu, C.; Dong, D.; Li, W.; Tian, J. Quantitative Biomarkers for Prediction of Epidermal Growth Factor Receptor Mutation in Non-Small Cell Lung Cancer. Transl. Oncol. 2018, 11, 94–101. [Google Scholar] [CrossRef]
  57. Sawan, P.; Plodkowski, A.J.; Li, A.E.; Li, B.T.; Drilon, A.; Capanu, M.; Ginsberg, M.S. CT features of HER2-mutant lung adenocarcinomas. Clin. Imaging 2018, 51, 279–283. [Google Scholar] [CrossRef]
  58. Halpenny, D.F.; Riely, G.J.; Hayes, S.; Yu, H.; Zheng, J.; Moskowitz, C.S.; Ginsberg, M.S. Are there imaging characteristics associated with lung adenocarcinomas harboring ALK rearrangements? Lung Cancer 2014, 86, 190–194. [Google Scholar] [CrossRef] [Green Version]
  59. Tang, C.; Hobbs, B.; Amer, A.; Li, X.; Behrens, C.; Canales, J.R.; Cuentas, E.P.; Villalobos, P.; Fried, D.; Chang, J.Y.; et al. Development of an Immune-Pathology Informed Radiomics Model for Non-Small Cell Lung Cancer. Sci. Rep. 2018, 8, 1922. [Google Scholar] [CrossRef]
  60. Jia, T.Y.; Xiong, J.F.; Li, X.Y.; Yu, W.; Xu, Z.Y.; Cai, X.W.; Ma, J.C.; Ren, Y.C.; Larsson, R.; Zhang, J.; et al. Identifying EGFR mutations in lung adenocarcinoma by noninvasive imaging using radiomics features and random forest modeling. Eur. Radiol. 2019, 29, 4742–4750. [Google Scholar] [CrossRef]
  61. Li, S.; Ding, C.; Zhang, H.; Song, J.; Wu, L. Radiomics for the prediction of EGFR mutation subtypes in non-small cell lung cancer. Med. Phys. 2019, 46, 4545–4552. [Google Scholar] [CrossRef] [PubMed]
  62. Tu, W.; Sun, G.; Fan, L.; Wang, Y.; Xia, Y.; Guan, Y.; Li, Q.; Zhang, D.; Liu, S.; Li, Z. Radiomics signature: A potential and incremental predictor for EGFR mutation status in NSCLC patients, comparison with CT morphology. Lung Cancer 2019, 132, 28–35. [Google Scholar] [CrossRef] [PubMed]
  63. Yang, X.; Dong, X.; Wang, J.; Li, W.; Gu, Z.; Gao, D.; Zhong, N.; Guan, Y. Computed Tomography-Based Radiomics Signature: A Potential Indicator of Epidermal Growth Factor Receptor Mutation in Pulmonary Adenocarcinoma Appearing as a Subsolid Nodule. Oncologist 2019, 24, e1156–e1164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Wang, X.; Kong, C.; Xu, W.; Yang, S.; Shi, D.; Zhang, J.; Du, M.; Wang, S.; Bai, Y.; Zhang, T.; et al. Decoding tumor mutation burden and driver mutations in early stage lung adenocarcinoma using CT-based radiomics signature. Thorac. Cancer 2019, 10, 1904–1912. [Google Scholar] [CrossRef] [Green Version]
  65. Bak, S.H.; Park, H.; Lee, H.Y.; Kim, Y.; Kim, H.L.; Jung, S.H.; Kim, H.; Kim, J.; Park, K. Imaging genotyping of functional signaling pathways in lung squamous cell carcinoma using a radiomics approach. Sci. Rep. 2018, 8, 3284. [Google Scholar] [CrossRef] [PubMed]
  66. Patil, R.; Mahadevaiah, G.; Dekker, A. An Approach Toward Automatic Classification of Tumor Histopathology of Non-Small Cell Lung Cancer Based on Radiomic Features. Tomography 2016, 2, 374–377. [Google Scholar] [CrossRef]
  67. Wu, W.; Parmar, C.; Grossmann, P.; Quackenbush, J.; Lambin, P.; Bussink, J.; Mak, R.; Aerts, H.J. Exploratory Study to Identify Radiomics Classifiers for Lung Cancer Histology. Front. Oncol. 2016, 6, 71. [Google Scholar] [CrossRef] [Green Version]
  68. Zhu, X.; Dong, D.; Chen, Z.; Fang, M.; Zhang, L.; Song, J.; Yu, D.; Zang, Y.; Liu, Z.; Shi, J.; et al. Radiomic signature as a diagnostic factor for histologic subtype classification of non-small cell lung cancer. Eur. Radiol. 2018, 28, 2772–2778. [Google Scholar] [CrossRef]
  69. Linning, E.; Lu, L.; Li, L.; Yang, H.; Schwartz, L.H.; Zhao, B. Radiomics for Classifying Histological Subtypes of Lung Cancer Based on Multiphasic Contrast-Enhanced Computed Tomography. J. Comput. Assist. Tomogr. 2019, 43, 300–306. [Google Scholar] [CrossRef]
  70. Liu, J.; Cui, J.; Liu, F.; Yuan, Y.; Guo, F.; Zhang, G. Multi-subtype classification model for non-small cell lung cancer based on radiomics: SLS model. Med. Phys. 2019, 46, 3091–3100. [Google Scholar] [CrossRef]
  71. Zhou, B.; Xu, J.; Tian, Y.; Yuan, S.; Li, X. Correlation between radiomic features based on contrast-enhanced computed tomography images and Ki-67 proliferation index in lung cancer: A preliminary study. Thorac. Cancer 2018, 9, 1235–1240. [Google Scholar] [CrossRef] [PubMed]
  72. Gu, Q.; Feng, Z.; Liang, Q.; Li, M.; Deng, J.; Ma, M.; Wang, W.; Liu, J.; Liu, P.; Rong, P. Machine learning-based radiomics strategy for prediction of cell proliferation in non-small cell lung cancer. Eur. J. Radiol. 2019, 118, 32–37. [Google Scholar] [CrossRef] [PubMed]
  73. Song, S.H.; Park, H.; Lee, G.; Lee, H.Y.; Sohn, I.; Kim, H.S.; Lee, S.H.; Jeong, J.Y.; Kim, J.; Lee, K.S.; et al. Imaging Phenotyping Using Radiomics to Predict Micropapillary Pattern within Lung Adenocarcinoma. J. Thorac. Oncol. 2017, 12, 624–632. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Chen, X.; Fang, M.; Dong, D.; Wei, X.; Liu, L.; Xu, X.; Jiang, X.; Tian, J.; Liu, Z. A Radiomics Signature in Preoperative Predicting Degree of Tumor Differentiation in Patients with Non-small Cell Lung Cancer. Acad. Radiol. 2018, 25, 1548–1555. [Google Scholar] [CrossRef] [PubMed]
  75. She, Y.; Zhang, L.; Zhu, H.; Dai, C.; Xie, D.; Xie, H.; Zhang, W.; Zhao, L.; Zou, L.; Fei, K.; et al. The predictive value of CT-based radiomics in differentiating indolent from invasive lung adenocarcinoma in patients with pulmonary nodules. Eur. Radiol. 2018, 28, 5121–5128. [Google Scholar] [CrossRef]
  76. Yang, B.; Guo, L.; Lu, G.; Shan, W.; Duan, L.; Duan, S. Radiomic signature: A non-invasive biomarker for discriminating invasive and non-invasive cases of lung adenocarcinoma. Cancer Manag. Res. 2019, 11, 7825–7834. [Google Scholar] [CrossRef] [Green Version]
  77. Fornacon-Wood, I.; Faivre-Finn, C.; O’Connor, J.P.B.; Price, G.J. Radiomics as a personalised medicine tool in lung cancer: Separating the hope from the hype. Lung Cancer 2020, 146, 197–208. [Google Scholar] [CrossRef]
Figure 1. Typical workflow of a radiomic analysis. Initially, the proper medical imaging modality is selected (PET–CT in our example. Then, the radiologist segments the tissue of interest in all slices, resulting in a volume of interest. Consequently, radiomic features are computed using only the tissues included in the volume of interest. Such features can reflect shape information, signal intensities of tissues inside the VOI, and texture-related information that can reflect tissue heterogeneity. Finally, machine learning models are trained and validated to predict clinical outcomes or to classify patients according to genomic or molecular characteristics.
Figure 1. Typical workflow of a radiomic analysis. Initially, the proper medical imaging modality is selected (PET–CT in our example. Then, the radiologist segments the tissue of interest in all slices, resulting in a volume of interest. Consequently, radiomic features are computed using only the tissues included in the volume of interest. Such features can reflect shape information, signal intensities of tissues inside the VOI, and texture-related information that can reflect tissue heterogeneity. Finally, machine learning models are trained and validated to predict clinical outcomes or to classify patients according to genomic or molecular characteristics.
Cancers 14 01657 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Anagnostopoulos, A.K.; Gaitanis, A.; Gkiozos, I.; Athanasiadis, E.I.; Chatziioannou, S.N.; Syrigos, K.N.; Thanos, D.; Chatziioannou, A.N.; Papanikolaou, N. Radiomics/Radiogenomics in Lung Cancer: Basic Principles and Initial Clinical Results. Cancers 2022, 14, 1657. https://doi.org/10.3390/cancers14071657

AMA Style

Anagnostopoulos AK, Gaitanis A, Gkiozos I, Athanasiadis EI, Chatziioannou SN, Syrigos KN, Thanos D, Chatziioannou AN, Papanikolaou N. Radiomics/Radiogenomics in Lung Cancer: Basic Principles and Initial Clinical Results. Cancers. 2022; 14(7):1657. https://doi.org/10.3390/cancers14071657

Chicago/Turabian Style

Anagnostopoulos, Athanasios K., Anastasios Gaitanis, Ioannis Gkiozos, Emmanouil I. Athanasiadis, Sofia N. Chatziioannou, Konstantinos N. Syrigos, Dimitris Thanos, Achilles N. Chatziioannou, and Nikolaos Papanikolaou. 2022. "Radiomics/Radiogenomics in Lung Cancer: Basic Principles and Initial Clinical Results" Cancers 14, no. 7: 1657. https://doi.org/10.3390/cancers14071657

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop