Radiomics and “radi-…omics” in cancer immunotherapy: a guide for clinicians

https://doi.org/10.1016/j.critrevonc.2020.103068Get rights and content

Highlights

  • Radiomics detects imaging features not identifiable by the simple visual analysis.

  • Radiomics features can be combined with data from other “…-omics” data.

  • Radiomics creates enhanced data models for supporting medical decision.

  • These algorithms help to assess various aspects of cancer immunotherapy treatment.

Abstract

In recent years the concept of precision medicine has become a popular topic particularly in medical oncology. Besides the identification of new molecular prognostic and predictive biomarkers and the development of new targeted and immunotherapeutic drugs, imaging has started to play a central role in this new era. Terms such as “radiomics”, “radiogenomics” or “radi…-omics” are becoming increasingly common in the literature and soon they will represent an integral part of clinical practice. The use of artificial intelligence, imaging and “-omics” data can be used to develop models able to predict, for example, the features of the tumor immune microenvironment through imaging, and to monitor the therapeutic response beyond the standard radiological criteria.

The aims of this narrative review are to provide a simplified guide for clinicians to these concepts, and to summarize the existing evidence on radiomics and “radi…-omics” in cancer immunotherapy.

Introduction

In recent years the concept of precision medicine has become an area of immense interest (Burki, 2017; Ashley, 2016) in medical oncology (Le Tourneau et al., 2018). In this field, emerging mechanisms of resistance (Khan and Spicer, 2019; Lim and Ma, 2019; Narayanan et al., 2020; Kalbasi and Ribas, 2020), financial costs and toxicities (Postow et al., 2018; Porcu et al., 2020; Solinas et al., 2018; Marin-Acevedo et al., 2018; Porcu et al., 2019) are important elements to take into account for guiding treatment decisions, in order to identify ideal candidates that might benefit from a variety of novel and expensive therapies. Besides the identification of new molecular prognostic and predictive biomarkers (Dumitrescu, 2018) and the development of new targeted (Dugger et al., 2018) and immunotherapeutic drugs (Hegde and Chen, 2020; Dobosz and Dzieciątkowski, 2019), imaging has started to play a pivotal role in the evolution towards precision medicine (Acharya et al., 2018).

A fruitful collaboration between radiologists and medical oncologists is essential due to the growing dependency on imaging as a therapeutic biomarker. This is particularly relevant in this new era of cancer immunotherapy with immune checkpoint blockade (ICB) targeting a variety of immune checkpoint molecules that physiologically modulate the immune response (Solinas et al., 2019a; Solinas et al., 2019b; Solinas et al., 2019c; Solinas et al., 2020; ElTanbouly et al., 2020; Rowshanravan et al., 2018). These new drug regimens have a novel mechanism of action. Instead of directly killing tumor cells, they are able to harness the patient’s own immune response against cancer, making unique and challenging the assessment of treatment response (Litière et al., 2017; Seymour et al., 2017; Solinas et al., 2017; Porcu et al., 2018). This requires a deep knowledge for the specific features that characterize immune-related phenomena in each organ site. The contribution of imaging is also important for differential diagnosis when facing with immune related adverse events (irAEs) (Porcu et al., 2020; Solinas et al., 2018; Porcu et al., 2019).

Radiological sciences are thus moving from a dependency on basic visual features in medical images to new non-visual features buried in the pixel data that are not routinely detectable with the human eye (Gillies et al., 2016). This is obtained thanks to the improvement of computational power of informatic systems and to the use of artificial intelligence (AI) (Obermeyer and Emanuel, 2016), specifically machine learning techniques (Choy et al., 2018) such as deep learning (LeCun et al., 2015).

With their ability to combine quantitative data obtained from imaging together with those derived from genomic analyses, “radiomics” and “radiogenomics” metadata are becoming of common use in clinical research. Hopefully, they will soon be part of daily routine clinical practice with the aim to allow oncologists and radiologists to optimize patient selection and management for medical therapy and to better assess treatment responses (Lambin et al., 2012).

The aims of this narrative review are to provide a simplified guide for clinicians to these new concepts, including some important technical aspects, and to summarize the existing evidence on radiomics in cancer immunotherapy.

Section snippets

Definitions

“Radiomics” is defined as the analysis of imaging data through the use of specific algorithms aimed at identifying quantitative features otherwise not identifiable with the simple visual analysis, in order to create enhanced data models for improving medical decision support (Gillies et al., 2016; Lambin et al., 2012). Images from ultrasound (US), computed tomography (CT), magnetic resonance (MR) and positron emission tomography (PET) combined with CT (PET-CT) or MR (PET-MR) can be analyzed

Feature identification in radiomics

Radiomics is based on the discovery of imaging features not identifiable with the simple visual analysis, through the use of specific and sophisticated algorithms (Gillies et al., 2016; Lambin et al., 2012). From a technical point of view, the whole process can be divided into three steps (Gillies et al., 2016; Mazurowski, 2015) (Fig. 1):

  • 1)

    Image acquisition, database creation and data selection: all imaging examination can be “radiomically” analyzed, including MR, CT, US and PET (Gillies et al.,

The model development in radiomics and “radi…-omics”

Once radiomic and non-radiomic features (such as those derived from histologic, genomic, proteomic or transcriptomic analyses) are collected, they can be used for data mining, representing the process of discovering patterns in datasets (Gillies et al., 2016; Incoronato et al., 2017). By exploring the mutual relationships between radiomic and non-radiomic features with a selected outcome variable, AI modeling can be used to develop models that, once validated, could be used for different

Radiomics and “radi…-omics” in cancer immunotherapy

Radiomics could be a helpful tool in cancer immunotherapy for optimal patient selection (El Naqa I, Ten Haken RK, 2018). It may act as a non-invasive digital biopsy technique able to quantify tumor T-cell infiltration, to support personalized immunotherapy interventions, and to longitudinally monitor the therapeutic response above the traditional dimensional criteria (Solinas et al., 2017; Porcu et al., 2018). In particular, digital biopsy thorugh the combination of radiomics and pathomics

Current situation and future prospects

Radiomics is changing radiology and oncology (Ha, 2019; Trebeschi et al., 2019; Bera et al., 2018; Du et al., 2019; Banna et al., 2019b), but the process of translation from pure research to clinical practice is facing some difficulties (Park et al., 2020; Deutsch and Paragios, 2019). In a recent article by Sollini et al (Choi et al., 2016), authors adopted a phase classification criteria for radiomic studies similar to the one used for drug development, and the relative paucity of phase III

Conclusions

Radiomics and “radi…-omics” techniques leverage imaging information beyond those attained through visual inspection. In the field of cancer immunotherapy, radiomics has already shown value in characterizing the tumor immune-environment and to stratify prognosis by easily evaluating the baseline imaging investigations.

Even though the process of translation of these models from pure research to clinical practice is still in progress, they represent an innovative tool that once applied in clinical

Funding

This research did not receive any specific grant from funding agencies in the public, commercial, or non-profit sectors.

Declaration of Competing Interest

Matteo Lambertini acted as consultant for Roche and Lilly, and received speaker honoraria from Roche, Takeda and Theramex outside the submitted work. The other authors declare that they have no conflict of interests.

References (124)

  • M.A. Mazurowski

    Radiogenomics: what it is and why it is important

    J Am Coll Radiol.

    (2015)
  • C.E. Metz

    Basic principles of ROC analysis

    Semin Nucl Med.

    (1978)
  • S. Narayanan et al.

    Targeting the ubiquitin-proteasome pathway to overcome anti-cancer drug resistance

    Drug Resist Updat.

    (2020)
  • M. Porcu et al.

    Radiological evaluation of response to immunotherapy in brain tumors: Where are we now and where are we going?

    Crit Rev Oncol Hematol.

    (2018)
  • B. Rowshanravan et al.

    CTLA-4: a moving target in immunotherapy

    Blood

    (2018)
  • L. Saba et al.

    The present and future of deep learning in radiology

    Eur J Radiol.

    (2019)
  • L. Seymour et al.

    iRECIST: guidelines for response criteria for use in trials testing immunotherapeutics [published correction appears in Lancet Oncol. 2019 May;20(5):e242]

    Lancet Oncol.

    (2017)
  • U.R. Acharya et al.

    Towards precision medicine: from quantitative imaging to radiomics

    J Zhejiang Univ Sci B.

    (2018)
  • H.J. Aerts et al.

    Defining a Radiomic Response Phenotype: A Pilot Study using targeted therapy in NSCLC [published correction appears in Sci Rep. 2017 Feb 17;7:41197]

    Sci Rep.

    (2016)
  • A. Alberich-Bayarri et al.

    Imaging Biomarkers and Imaging Biobanks

  • M. Amadasun et al.

    Textural features corresponding to textural properties

    IEEE Trans. Syst. Man Cybern.

    (1989)
  • E.A. Ashley

    Towards precision medicine

    Nat Rev Genet.

    (2016)
  • B. Aslam et al.

    Proteomics: Technologies and Their Applications

    J Chromatogr Sci.

    (2017)
  • H.X. Bai et al.

    Imaging genomics in cancer research: limitations and promises

    Br J Radiol.

    (2016)
  • G.L. Banna et al.

    The Promise of Digital Biopsy for the Prediction of Tumor Molecular Features and Clinical Outcomes Associated With Immunotherapy

    Front Med (Lausanne).

    (2019)
  • G.L. Banna et al.

    The Promise of Digital Biopsy for the Prediction of Tumor Molecular Features and Clinical Outcomes Associated With Immunotherapy

    Front Med (Lausanne).

    (2019)
  • K. Bera et al.

    Novel Quantitative Imaging for Predicting Response to Therapy: Techniques and Clinical Applications

    Am Soc Clin Oncol Educ Book.

    (2018)
  • A. Bhatia et al.

    MRI radiomic features are associated with survival in melanoma brain metastases treated with immune checkpoint inhibitors

    Neuro Oncol.

    (2019)
  • M. Bogowicz et al.

    CT radiomics and PET radiomics: ready for clinical implementation?

    Q J Nucl Med Mol Imaging.

    (2019)
  • A.J. Buckler et al.

    Group. A collaborative enterprise for multi-stakeholder participation in the advancement of quantitative imaging

    Radiology.

    (2011)
  • A. Chaddad et al.

    Radiomics Evaluation of Histological Heterogeneity Using Multiscale Textures Derived From 3D Wavelet Transformation of Multispectral Images

    Front Oncol.

    (2018)
  • Y. Chang et al.

    An investigation of machine learning methods in delta-radiomics feature analysis

    PLoS One.

    (2019)
  • S. Chen et al.

    Pretreatment prediction of immunoscore in hepatocellular cancer: a radiomics-based clinical model based on Gd-EOB-DTPA-enhanced MRI imaging

    Eur Radiol.

    (2019)
  • Y.J. Choi et al.

    Does the Reporting Quality of Diagnostic Test Accuracy Studies, as Defined by STARD 2015, Affect Citation?

    Korean J Radiol.

    (2016)
  • G. Choy et al.

    Current Applications and Future Impact of Machine Learning in Radiology

    Radiology.

    (2018)
  • R.R. Colen et al.

    Radiomics to predict immunotherapy-induced pneumonitis: proof of concept

    Invest New Drugs.

    (2018)
  • T.D. Cook et al.

    Quasi-experimentation: design & analysis issues for field settings

    (1979)
  • A.S. Cottereau et al.

    18F-FDG PET Dissemination Features in Diffuse Large B-Cell Lymphoma Are Predictive of Outcome

    J Nucl Med.

    (2020)
  • D. Cusumano et al.

    Fractal-based radiomic approach to predict complete pathological response after chemo-radiotherapy in rectal cancer

    Radiol Med.

    (2018)
  • F. Davnall et al.

    Assessment of tumor heterogeneity: an emerging imaging tool for clinical practice?

    Insights Imaging.

    (2012)
  • J.E. Dayhoff et al.

    Artificial neural networks: opening the black box

    Cancer

    (2001)
  • R.C. Deo

    Machine Learning in Medicine

    Circulation.

    (2015)
  • M. Djekidel

    Radiogenomics and Radioproteomics

    OMICS J Radiology.

    (2013)
  • P. Dobosz et al.

    The Intriguing History of Cancer Immunotherapy

    Front Immunol.

    (2019)
  • S.A. Dugger et al.

    Drug development in the era of precision medicine

    Nat Rev Drug Discov.

    (2018)
  • R.G. Dumitrescu

    Early Epigenetic Markers for Precision Medicine

    Methods Mol Biol.

    (2018)
  • El Naqa I, Ten Haken RK

    Can radiomics personalise immunotherapy?

    Lancet Oncol.

    (2018)
  • M.A. ElTanbouly et al.

    VISTA: Coming of age as a multi-lineage immune checkpoint [published online ahead of print, 2020 Jan 13]

    Clin Exp Immunol.

    (2020)
  • X. Fave et al.

    Delta-radiomics features for the prediction of patient outcomes in non-small cell lung cancer

    Sci Rep.

    (2017)
  • L. Ferguson

    External validity, generalizability, and knowledge utilization

    J Nurs Scholarsh.

    (2004)
  • Cited by (24)

    • Radiomics signature based on CECT for non-invasive prediction of response to anti-PD-1 therapy in patients with hepatocellular carcinoma

      2023, Clinical Radiology
      Citation Excerpt :

      The radiomics features were extracted lesion-per-lesion. Electronic Supplementary Material S2 describes the detailed process of feature extraction.24 Lesion-level progressive disease (PD) was defined as an increase in the diameter of the target lesion by at least 20% compared with the baseline value according to the enhanced imaging during the arterial phase (Fig 2).

    • When artificial intelligence meets PD-1/PD-L1 inhibitors: Population screening, response prediction and efficacy evaluation

      2022, Computers in Biology and Medicine
      Citation Excerpt :

      Big data generated in clinical oncology, which may come from a variety of sources such as molecular, imaging, pharmacological, and demographic information, presents clinicians and data scientists with challenges for analysis while also provides opportunities for knowledge discovery in tumor biology and therapy [8]. Clinical oncology research is increasingly focusing on the full exploitation of clinical big data and multi-omics research represented by radiomics studies, which occupies an important aspect of prognostic analysis of immunotherapy [9]. As an important derivative of Precision Medicine in immunotherapy, medical AI in omics research allows machine learning to play an important role in therapy discovery and response prediction [10], on the basis of which numerous exciting results have been generated, among which the application of AI in anti-PD-1/PD-L1 therapy is an essential and representative aspect (Fig. 1-(B)).

    View all citing articles on Scopus
    1

    Contributed equally: co-first authors.

    View full text