Next Article in Journal
IgA Nephropathy: Current Understanding and Perspectives on Pathogenesis and Targeted Treatment
Next Article in Special Issue
An Isolated Intestinal Juvenile Polyp Diagnosed by Abdominal Ultrasonography and Resected by Double-Balloon Endoscopy: A Case Report and Literature Review
Previous Article in Journal
Machine Learning in Colorectal Cancer Risk Prediction from Routinely Collected Data: A Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immunotherapy Assessment: A New Paradigm for Radiologists

1
Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
2
Medical Oncology Division, Igea SpA, 80013 Naples, Italy
3
Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, 20122 Milan, Italy
4
Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131 Naples, Italy
5
Thoracic Medical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
6
Division of Epatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, 80131 Naples, Italy
*
Author to whom correspondence should be addressed.
Diagnostics 2023, 13(2), 302; https://doi.org/10.3390/diagnostics13020302
Submission received: 17 November 2022 / Revised: 31 December 2022 / Accepted: 8 January 2023 / Published: 13 January 2023
(This article belongs to the Special Issue Imaging Diagnosis in Abdomen)

Abstract

:
Immunotherapy denotes an exemplar change in an oncological setting. Despite the effective application of these treatments across a broad range of tumors, only a minority of patients have beneficial effects. The efficacy of immunotherapy is affected by several factors, including human immunity, which is strongly correlated to genetic features, such as intra-tumor heterogeneity. Classic imaging assessment, based on computed tomography (CT) or magnetic resonance imaging (MRI), which is useful for conventional treatments, has a limited role in immunotherapy. The reason is due to different patterns of response and/or progression during this kind of treatment which differs from those seen during other treatments, such as the possibility to assess the wide spectrum of immunotherapy-correlated toxic effects (ir-AEs) as soon as possible. In addition, considering the unusual response patterns, the limits of conventional response criteria and the necessity of using related immune-response criteria are clear. Radiomics analysis is a recent field of great interest in a radiological setting and recently it has grown the idea that we could identify patients who will be fit for this treatment or who will develop ir-AEs.

1. Background

Immunotherapy denotes an exemplar change in an oncological setting [1,2,3,4,5,6]. In fact, different to other therapies as conventional chemotherapy [7,8,9,10,11,12,13], such as radiation [14,15,16,17,18,19,20] or targeted therapies [21,22,23,24,25,26,27] which target the cancer, these treatments work by stimulating the patient’s immune system to obtain an immune reaction against the tumor [2,3].
Immunotherapy can be categorized as passive or active, according to the action mechanism [28,29,30,31,32,33,34]. In passive treatment [28,29,30], immunoglobulins can be administered and attached to tumor-related antigens. Otherwise, in the active treatment, there is a stimulation of the immune system to recognize antigens of the tumor and act against them [30,31,32,33,34]. Although several approaches are presently utilized in clinical and pre-clinical settings, the main means are centered on the so-called checkpoint inhibitors (ICI), that include programmed cell death–1 (PD-1) protein, PD-1′s main ligand (PD-L1), and cytotoxic T-lymphocyte–associated protein 4 (CTLA-4) [35,36,37,38,39,40,41,42,43].
In 2011, the U.S. Food and Drug Administration (FDA) approved the first agent, ipilimumab, a CTLA-4 inhibitor, for metastatic and unresectable melanoma [44]. Subsequently, different FDA approved agents, have been introduced in both routine and experimental studies to treat different categories of solid and hematologic tumors as after failure of conventional therapies or as first-line therapies [39,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69]. Moreover, these treatments may be combined with conventional treatments since they can increase the cytotoxic effect of chemotherapy [70,71,72].
Although the effective utility of these treatments is within a wide range of tumors, only a sub-group of patients show real benefits [73]. The efficacy of immunotherapy is affected by several factors, including human immunity, which is strongly correlated to genetic features [74,75,76,77], such as intra-tumor heterogeneity [78,79,80,81,82,83,84,85,86]. In fact, cancer is complex, flexible, and heterogeneous, representing the result of an innumerable number of genetic mutations that affect the regular cell functionality. However, these mutations allow the cancer cell to appear as foreign to the immune system, offering an opportunity for this treatment [78,79,80,81,82,83,84,85,86]. Clinical studies have demonstrated that even when several patients are well qualified as immunotherapy responders (e.g., high tumor PD-L1 expression), a large percentage of them (>50%) do not respond to this [87]. In addition, during treatment, some patients can have a clinical and/or radiological disease progression [88]; these events are known as “primary immune escape” and “secondary immune escape”, respectively [89]. The mechanisms implicated in these two phenomena (also known as resistance) can overlap. Multi-omics analyses obtained from immunotherapy-treated patient tissues showed as several features are correlated to immune resistance [90,91,92].
In addition, while immunotherapy has increased patient outcomes, it can cause several immune-related adverse events (ir-AEs), which can be transitory or chronic, life-threatening or mild, and could affect several organ systems, sometimes numerous organs at the same time [93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108].
Classic imaging assessment, based on computed tomography (CT) [109,110,111,112,113,114,115,116] or magnetic resonance imaging (MRI) [117,118,119,120,121,122,123,124,125,126,127], which is useful for conventional treatment, has a limited role in immunotherapy. The reason is due to different patterns of response and/or progression during this kind of treatment which differs from those seen during other treatments, such as the possibility to assess the wide spectrum of immunotherapy-related toxic effects as soon as possible [128,129,130,131,132,133,134,135,136,137,138,139]. Thus, the introduction of robust noninvasive imaging biomarkers that can allow the immunotherapy-response prediction and prognosis is crucial.
The aim of this article is to (a) explain the response pattern and response criteria utilized during immunotherapy assessments, (b) explain the wide spectrum of toxic effects due to immunotherapy, and (c) explain the potential role of radiomics features as imaging biomarkers for immunotherapy.

2. Treatment Assessment and Pattern Response

The frequently employed radiological response criteria have been introduced considering the chemotherapeutic agents’ cytotoxic results, which caused a decrease in target size when the treatment has been effective [132,133,140]. The World Health Organization (WHO) criteria assessed the tumor burden considering the sum of the products of orthogonal largest diameters of the target lesions [141], while the response evaluation criteria in solid tumors (RECIST) criteria [142] assessed the tumor burden considering a one-dimensional (the largest diameter of target lesions). These criteria are not able to evaluate functional or metabolic status, such as necrosis, that causes morphological changes on CT (density decrease) or MRI (inhomogeneous signal on conventional sequences and different pattern on functional ones) [130,143,144,145,146]. The CHOI criteria [143] were introduced to assess target therapy in gastrointestinal stromal tumors, such as the PET response criteria in solid tumors (PERCIST) to evaluate FDG uptake reduction [147,148].
In contrast to conventional treatments, immunotherapy involves a complex process that includes different phases and during each one, the immune system is activated. Such as, a number of immune cells move to the target with increasing in target volume and/or new lesion growth [139,149,150]. This process can cause an unusual response pattern known as pseudoprogression [139]. Although, it is possible to obtain conventional response patterns as a complete response (Figure 1) or a partial response (Figure 2), pseudoprogression (Figure 3), characterized by an increment in volume target, can occur in 4–10% of immunotherapy-treated patients [128]. Divergent to pseudoprogression, in which treatment is preserved, a true severe progression is hyperprogression (Figure 4 and Figure 5) [128]. During an imaging study assessment, it could be complicated differ pseudoprogression to hyperprogression [128], so a multidisciplinary team should assess complete patient status [128].
Another atypical response pattern are dissociated responses (Figure 6) [128]. In this pattern, several lesions show dimension increase and others show regression. This pattern is associated with a better survival compared to true progressions [128].
Considering these response patterns, the limits of conventional response criteria are clear. So, to overcome these limits, new criteria have been proposed. The first criteria were the immune-related response criteria (irRC), based on the WHO criteria [136]. In these criteria, two main features were introduced: (a) the first radiological progression may be confirmed after 4 weeks, so in this phase, the patient can be treated; (b) the development of new lesions is not assessed as a disease progression, but these may be included in the total tumor burden [136]. The main limit of irRC is due to the bi-dimensional measurement of target lesions. In addition, since the greater part of immunotherapy trials in development at that time have been assessed according to RECIST, the obtained results could be difficult to compare according to the new criteria [137]. Subsequently, the immune-related RECIST (irRECIST), established on the unidimensional evaluation [138], have been proposed. According to irRECIST, an increase of 20% in the total tumor burden from nadir with a minimum of 5 mm, such as the progression of non-target lesions or the appearance of a new lesion, has been defined as immune-related progression disease (irPD). Moreover, in this case, the irPD must been verified after 4 weeks, and if there is also a new unequivocal progression (UEP) in this phase, the patient is defined in disease progression [138].
To homogenize results between different trials, the RECIST working group proposed an adaptation to the immunotherapy version of RECIST1.1, the immune RECIST (iRECIST) [139]. The iRECIST identified a standard lexicon including immune complete response (iCR), immune stable disease (iSD), immune PR (iPR), and immune unconfirmed PD (iUPD) or confirmed PD (iCPD). The key element in iRECIST is that iUPD must be confirmed by imaging at least 4 weeks after the first evaluation, but no more than 8 weeks from iUPD, and that a patient may be classified as iUPD more times until there is an iCPD [139]. Regarding new lesions, these must not be involved in the sum of total tumor burden, but the patient can be classified as an iUPD. After an iUPD, the appearance of another new lesion, increase in size of target or non-target lesions, an increase in the sum of measurement of new target lesions >5 mm, or any progression of new non-target lesions can cause an iCPD [139].
Although these immunotherapy-response evaluation criteria have been proposed to allow the comparison of different clinical trial results, at the present, these are not utilized in clinical settings, such as none of these takes into account the atypical response patterns as hyperprogression or dissociated responses.

3. Immune-Related Adverse Events Assessment

If on the one hand, immunotherapy has improved oncological patient outcomes, on the other, these treatments have caused a rise in adverse events, called ir-AEs, which can involve several tissues and organs, from head to toe [93,95,96,151,152,153]. The ir-AEs are classified according to the Common Terminology Criteria for Adverse Events (CTCAE v 5.0) [94], which; moreover, permits to assess these events throughout several clinical trials. A systematic review showed that about 74% of patients treated with anti-PD-L-1 inhibitors developed ir-AEs, and among them, 14% had a grade ≥3, while the rate increased in anti-CTLA-4 inhibitors treated patients (about 89% and in this group, about 34% had a grade ≥3). When the authors evaluated patients treated with a combination of ICIs, the rate was 90%, and among them, 55% had a grade ≥3 [95].
The ir-AEs are relatively new conditions, so very little is known about them. The enhancement of the immune system is probably also responsible for an accidental autoimmune response [97,98,99,100,101,102,154,155,156,157,158,159,160]. Recombinant cytokines have a high toxicity profile, with low possibility either in terms of the target population either duration time. Toxic effects can involve several organs, as gastrointestinal tract, liver, lungs, heart, skin, endocrine, and hematologic systems [161,162,163,164]. The ir-AEs can appear early, even after the first dose. During ipilimumab treatment, the majority of ir-AEs appeared around 12 weeks, although the appearance time is due to the target: for skin, around 3 weeks; for the liver, around 3–9 weeks; for the gastrointestinal-tract, around 8 weeks; and for the endocrine system, around 7–20 weeks [165]. The majority of ir-AEs are mild or moderate, and readily reversed by stopping the treatment and starting corticosteroid therapy [128]. Otherwise, endocrinopathy related events are usually irreversible [128]. Life-threatening ir-AEs necessitate hospitalization and, in higher grade, intensive care unit admission [128]. To prevent the risk of milder forms becoming life threatening, radiologists should recognize ir-AEs and should inform the clinical team as soon as possible. Consequently, there is a rising request for radiologists to recognize ir-AEs imaging features to permit proper patient management [97,98,99,100,101,102,154,155].
Regarding ir-endocrinopathies, hypophysitis, occurring in 10–13% of ipilimumab patients, is the most frequent [99,100,101,166,167]. The typical symptoms are headache and fatigue with anterior hypopituitarism and multiple hormonal deficiencies. At MR assessment, the patient shows diffuse enlargement of the pituitary with no optic chiasm compression. It is possible to find pituitary stalk thickening, such as during post-contrast assessment pituitary enhancement [128].
With regard to gastrointestinal ir-AEs, the main symptom is diarrhea in a colitis setting [128]. A study of 162 melanoma ipilimumab treated patients showed colitis in 28 (19%) of them [168]. On CT evaluation, typical colitis features include bowel wall thickening with increased enhancement and mesenteric hyperemia, such as a fluid-filled colon. Considering the extension of the colitis, two patterns have been reported: the diffuse and segmental.
In the diffuse pattern, we find a fluid-filled distended colon with mild diffuse wall thickening and mesenteric vessel engorgement. In the segmental pattern, colitis is associated with pre-existing diverticulosis (SCAD), so that we find a moderate segmental wall thickening and pericolic fat stranding [128].
Pneumonia (Figure 7) is an ir-AE characterized by a focal or diffuse inflammation of the pulmonary parenchyma, and its incidence in immunotherapy-treated patients ranges from 0 to 10% [43]. Compared to pneumonia due to conventional chemotherapy, immunotherapy treated patients showed a greater susceptibility to develop this adverse event, showing an increased risk of high-grade pneumonia, which could cause significant morbidity, possible discontinuation of treatment, and a significant rate of mortality. However, according to previous authors, clinical and radiological diagnosis can improve patient outcomes [102,159,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183,184,185,186,187]. Imaging has a critical role in pneumonitis detection, and CT is the modality that should been chosen since this tool allows the identification of all little changes in the lung and the characterization of different sub-types [179,180,181,182,183,184,185,186,187]. Delaunay et al. [159] identified different patterns on CT studies from 64 patients, (a) organized pneumonia (OP) (occurred in about the 23% of patients), (b) hypersensitivity pneumonitis (HP) (in 16% of patients), (c) non-specific interstitial pneumonia (NSIP) (in 8% of patients), and (d) bronchiolitis (in 6% of patients), showing as in the same patients was possible to detect different patterns [159].
Usually, in OPs sub-type, the typical features are bilateral peribronchovascular and subpleural ground-glass and air space opacities, with mid- to lower-lung predominance [159]. CT features in HP pattern include diffuse and predominant ground glass centrilobular nodules in the upper lobe [159]. In NSIP pattern, there are ground glass and lattice opacities prevalently in the lower lobe. Sub-pleural sparing of the posterior and inferior lobes is a specific feature [159]. The bronchiolitis pattern is characterized by centrilobular nodularity, with a tree-in-bud pattern [159]. Acute interstitial pneumonia (AIP)–acute respiratory distress syndrome (ARDS) is not a typical pattern of ir-pneumonitis; however, it is possible to find this sub-type in extensive pulmonary involvement [43].
Granulomatosis and sarcoid-like lymphadenopathies involve mediastinal and hilar lymph nodes [128]. On CT, mediastinal and hilar lymphadenopathies have an appearance and distribution similar to sarcoidosis nodes [128], so it is critical to recognize this condition to avoid defining a PD.
Liver involvement is a rare condition (nearby 1–2% of treated patients) [128]; usually only elevated liver function test results are detected, while in severe cases of ir-hepatitis on imaging tools, it is possible to assess periportal edema, hepatomegaly, and periportal lymphadenopathies [128].

4. Radiomics and Immunotherapy

Recently, the idea that imaging studies contain a great amount of data as grey level patterns that usually are imperceptible to the human eyes has become more and more interesting [188,189,190,191,192,193,194,195,196,197,198]. These texture features, when correlated with clinical-pathological data and outcomes [199,200,201,202,203,204,205,206,207,208,209,210,211], theoretically allow for diagnostic and prognostic assessments and it could produce evidence-based clinical-decision support systems [212,213,214,215,216,217,218,219,220]. The assessment of textural characteristics, obtained using radiological images which depend on mathematical analysis as histogram analysis, is called radiomics [221,222,223,224,225,226,227,228,229,230,231,232,233]. The main objective is to combine several multimodal quantitative data with mathematical methods to provide clear and robust parameters allowing an outcome prediction. Radiomics offers outstanding benefits over qualitative imaging assessment since this is clearly limited by the subjective evaluation of radiologists. A radiomics information extension can be obtained by adding genomics data (radiogenomics); in fact, genomic markers, such as microRNA expression, have been shown to be associated with treatment response, metastatic spread, and prognosis that could offer personalized and precision medicine. This approach is captivating since it could be used to obtain molecular data from images [229,234,235,236,237,238,239,240,241,242,243,244,245] with no invasive approach to reduce costs, time, and any risk for the patient. For several tumors, radiomics has already offered a precise molecular assessment, allowing the recognition of biomarkers that are associated with the prognostic assessment [223,224,225,226,227,228,229,230,231,232,233,234,235,236,237,238,239,240].
Tumor immune phenotypes can be classified by histological and immunohistochemical analysis as immune-inflamed, immune-excluded, and immune-desert types [246]. Dense tumor-infiltrating lymphocytes (TILs) and a high tumor mutational burden characterize immune-inflamed tumors. These elements correlate strongly to favor immunotherapy response [247]. Otherwise, in immune-excluded and immune-desert types, which have low TILs and highly proliferating tumor cells, the probability of primary immune escape is high [89]. New biomarkers are needed to quantify TILs and PD-L1 expression to predict and monitor tumor immunotherapy response [248].
Several authors have assessed the radiomics features in predicting response for this treatment [39,249,250,251,252,253,254,255,256]. Granata et al. [39], in lung adenocarcinoma patients, showed that the shift in the center of mass of the lesion was significant as in prediction of overall survival (OS) as in prediction of progression free survival (PFS). By using univariate analysis, the authors obtained low diagnostic accuracy, while by using multivariate analysis, a support vector machine model showed the best results for stratifying patients based on OS (area under curve (AUC) of 0.89 and an accuracy of 81.6%). In addition, a decision tree model showed the best results for stratifying patients based on PFS time (AUC of 0.96 and an accuracy of 94.7%) [39]. Sun et al. [249], in a retrospective multicohort study, analyzed CT images and CD8 T cell RNA expression data from 135 treated patients with anti-PD-1 and anti-PD-L1 antibodies to identify and validate a radiomics signature to predict treatment response. They showed that a high baseline radiomics score was correlated with a higher objective response rate and OS [249]. Moreover, Tang et al. validated a non-small-cell lung cancer (NSCLC) radiomics signature associated with PD-L1 expression and density of TILs [250]. They showed that the radiomics signature was correlated to OS [250]. Tunali et al. [252] demonstrated that a pretreatment radiomics model was able to predict rapid disease progression phenotypes, including hyperprogression (AUCs ranging 0.804–0.865).
The results of these studies, although still in an embryonic phase, could support the hypothesis that radiomics analysis may represent an effective biomarker to select immunotherapy-responsive patients.
In addition, with great interest, we look at the possibility of applying radiomics to the identification of patients who will develop ir-AEs. Colen et al. [253], as the first group, assessed the possibility of radiomics to predict ir-pneumonitis. They obtained radiomics features from a CT study of patients who did (n = 2) and did not (n = 30) develop this event. However, the major limitation of this study was related to the number of patients and unbalanced groups (n = 2 vs. n = 30). Larger groups of patients and balanced groups are needed for radiomics analysis.
Thomas et al. [254] assessed 39 patients subjected to chemo-radiation therapy and consolidative ICI therapy for locally advanced NSCLC. They evaluated radiomics data, obtained by pre-treatment [99 mTc] MAA SPECT/CT perfusion images and clinical data. In their study population, 16/39 (41%) patients developed pneumonitis and, with regard to clinical characteristics, only the presence of baseline chronic obstructive pulmonary disease (COPD) was correlated to pneumonitis. Perfused lung radiomics texture features were correlated with lung volume, representing surrogates rather than independent predictors of pneumonitis risk. However, the major limit of this study was related to the number of patients and the lack of external validation group.
Several limitations to the clinical application of radiomics remain. The first key challenge is the use of different imaging techniques by different institutions. To ensure that the academic community can obtain high-quality radiological data resources, it is necessary to establish and promote certain imaging acquisition protocols. Second, the current research uses different software and different feature-selection methods, focuses on different feature sets, and applies different statistical and bioinformatic methods for data analysis and interpretation, which limit the reproducibility of radiomics models. Future research workflows need to be standardized. Third, many relevant radiomics studies employ single-center retrospective datasets. A quality-controlled multicenter prospective study plan is ideal. In addition, the evidence level rating reflects the feasibility of incorporating radiomics research into clinical practice. Recently published guidelines and checklists aiming to improve the quality of radiomics studies, including the radiomics quality score, modified Quality Assessment of Diagnostic Accuracy Studies tool, image biomarker standardization initiative guideline, and Transparent Reporting of a multivariable prediction model for Individual Prognosis or Diagnosis checklist, have been applied to radiomics evaluation [257,258].

5. Conclusions

Immunotherapy has a crucial role in the treatment of several tumors. Radiological assessment is essential for evaluating tumor response such as ir-AEs in immunotherapy-treated patients. Knowledge of the current response criteria and the response patterns, such as the different patterns of ir-AES, is critical for radiologists to offer valuable data for clinical providers to guide patient therapies. Radiomics analysis is a promising tool for the selection of responsive patients to immunotherapy such as in early ir-AES detections in order to avoid unnecessary treatments for unfit patients.

Author Contributions

Conceptualization, V.G., A.P. and R.F.; methodology, V.G. and S.V.S.; software, I.S. and V.V.; validation, C.P. and E.S.; formal analysis, V.G., R.F. and L.F.; investigation, L.F., C.P. and V.V.; resources, E.S., S.V.S. and M.G.V.; data curation, A.M. (Agnese Montanino), I.S. and E.S.; writing—original draft preparation, F.I., M.G.V. and A.M. (Agnese Montanino); writing—review and editing, P.A.A. and A.M. (Alessandro Morabito); visualization, A.P., A.M. (Alessandro Morabito), V.V. and F.I.; supervision, V.G.; project administration, V.G. All authors have read and agreed to the published version of the manuscript.

Funding

Funding by the Ministry of Health—Current Research 2022.

Data Availability Statement

Data are reported in the manuscript and images are reported at link https://zenodo.org/record/7521647#.Y72ceXbMK3A (accessed on 25 October 2022).

Acknowledgments

The authors are grateful to Alessandra Trocino, a librarian at the National Cancer Institute of Naples, Italy.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Riley, R.S.; June, C.H.; Langer, R.; Mitchell, M.J. Delivery technologies for cancer immunotherapy. Nat. Rev. Drug Discov. 2019, 18, 175–196. [Google Scholar] [CrossRef] [PubMed]
  2. Abbott, M.; Ustoyev, Y. Cancer and the Immune System: The History and Background of Immunotherapy. Semin. Oncol. Nurs. 2019, 35, 150923. [Google Scholar] [CrossRef] [PubMed]
  3. O’Donnell, J.S.; Teng, M.W.L.; Smyth, M.J. Cancer immunoediting and resistance to T cell-based immunotherapy. Nat. Rev. Clin. Oncol. 2019, 16, 151–167. [Google Scholar] [CrossRef] [PubMed]
  4. Zhang, Y.; Zhang, Z. The history and advances in cancer immunotherapy: Understanding the characteristics of tumor-infiltrating immune cells and their therapeutic implications. Cell. Mol. Immunol. 2020, 17, 807–821. [Google Scholar] [CrossRef]
  5. Merlotti, A.; Bruni, A.; Borghetti, P.; Ramella, S.; Scotti, V.; Trovò, M.; Chiari, R.; Lohr, F.; Ricardi, U.; Bria, E.; et al. Sequential chemo-hypofractionated RT versus concurrent standard CRT for locally advanced NSCLC: GRADE recommendation by the Italian Association of Radiotherapy and Clinical Oncology (AIRO). Radiol. Med. 2021, 126, 1117–1128. [Google Scholar] [CrossRef] [PubMed]
  6. Lesch, S.; Gill, S. The promise and perils of immunotherapy. Blood Adv. 2021, 5, 3709–3725. [Google Scholar] [CrossRef]
  7. Avallone, A.; Pecori, B.; Bianco, F.; Aloj, L.; Tatangelo, F.; Romano, C.; Granata, V.; Marone, P.; Leone, A.; Botti, G.; et al. Critical role of bevacizumab scheduling in combination with pre-surgical chemo-radiotherapy in MRI-defined high-risk locally advanced rectal cancer: Results of the BRANCH trial. Oncotarget 2015, 6, 30394–30407. [Google Scholar] [CrossRef] [Green Version]
  8. Bimonte, S.; Leongito, M.; Barbieri, A.; Del Vecchio, V.; Barbieri, M.; Albino, V.; Piccirillo, M.; Amore, A.; Di Giacomo, R.; Nasto, A.; et al. Inhibitory effect of (-)-epigallocatechin-3-gallate and bleomycin on human pancreatic cancer MiaPaca-2 cell growth. Infect. Agent Cancer 2015, 10, 22. [Google Scholar] [CrossRef]
  9. Ottaiano, A.; Scala, S.; Santorsola, M.; Trotta, A.M.; D’Alterio, C.; Portella, L.; Clemente, O.; Nappi, A.; Zanaletti, N.; De Stefano, A.; et al. Aflibercept or bevacizumab in combination with FOLFIRI as second-line treatment of mRAS metastatic colorectal cancer patients: The ARBITRATION study protocol. Ther. Adv. Med. Oncol. 2021, 13, 1758835921989223. [Google Scholar] [CrossRef]
  10. Ottaiano, A.; Caraglia, M.; Di Mauro, A.; Botti, G.; Lombardi, A.; Galon, J.; Luce, A.; D’Amore, L.; Perri, F.; Santorsola, M.; et al. Evolution of Mutational Landscape and Tumor Immune-Microenvironment in Liver Oligo-Metastatic Colorectal Cancer. Cancers 2020, 12, 3073. [Google Scholar] [CrossRef]
  11. Izzo, F.; Granata, V.; Fusco, R.; D’Alessio, V.; Petrillo, A.; Lastoria, S.; Piccirillo, M.; Albino, V.; Belli, A.; Tafuto, S.; et al. Clinical Phase I/II Study: Local Disease Control and Survival in Locally Advanced Pancreatic Cancer Treated with Electrochemotherapy. J. Clin. Med. 2021, 10, 1305. [Google Scholar] [CrossRef]
  12. Izzo, F.; Granata, V.; Fusco, R.; D’Alessio, V.; Petrillo, A.; Lastoria, S.; Piccirillo, M.; Albino, V.; Belli, A.; Nasti, G.; et al. A Multicenter Randomized Controlled Prospective Study to Assess Efficacy of Laparoscopic Electrochemotherapy in the Treatment of Locally Advanced Pancreatic Cancer. J. Clin. Med. 2021, 10, 4011. [Google Scholar] [CrossRef] [PubMed]
  13. Avallone, A.; Piccirillo, M.C.; Nasti, G.; Rosati, G.; Carlomagno, C.; Di Gennaro, E.; Romano, C.; Tatangelo, F.; Granata, V.; Cassata, A.; et al. Effect of Bevacizumab in Combination with Standard Oxaliplatin-Based Regimens in Patients with Metastatic Colorectal Cancer: A Randomized Clinical Trial. JAMA Netw. Open 2021, 4, e2118475. [Google Scholar] [CrossRef] [PubMed]
  14. Fiore, F.; Somma, F.; D’Angelo, R.; Tarotto, L.; Stoia, V. Cone beam computed tomography (CBCT) guidance is helpful in reducing dose exposure to pediatric patients undergoing radiofrequency ablation of osteoid osteoma. Radiol Med. 2022, 127, 183–190. [Google Scholar] [CrossRef] [PubMed]
  15. Falcinelli, L.; Mendichi, M.; Chierchini, S.; Tenti, M.V.; Bellavita, R.; Saldi, S.; Ingrosso, G.; Reggioli, V.; Bini, V.; Aristei, C. Pulmonary function in stereotactic body radiotherapy with helical tomotherapy for primary and metastatic lung lesions. Radiol. Med. 2021, 126, 163–169. [Google Scholar] [CrossRef] [PubMed]
  16. Arslan, A.; Aktas, E.; Sengul, B.; Tekin, B. Dosimetric evaluation of left ventricle and left anterior descending artery in left breast radiotherapy. Radiol. Med. 2021, 126, 14–21. [Google Scholar] [CrossRef]
  17. Barra, S.; Guarnieri, A.; di Monale EBastia, M.B.; Marcenaro, M.; Tornari, E.; Belgioia, L.; Magrini, S.M.; Ricardi, U.; Corvò, R. Short fractionation radiotherapy for early prostate cancer in the time of COVID-19: Long-term excellent outcomes from a multicenter Italian trial suggest a larger adoption in clinical practice. Radiol. Med. 2021, 126, 142–146. [Google Scholar] [CrossRef] [PubMed]
  18. Cellini, F.; Di Franco, R.; Manfrida, S.; Borzillo, V.; Maranzano, E.; Pergolizzi, S.; Morganti, A.G.; Fusco, V.; Deodato, F.; Santarelli, M.; et al. Palliative radiotherapy indications during the COVID-19 pandemic and in future complex logistic settings: The NORMALITY model. Radiol. Med. 2021, 126, 1619–1656. [Google Scholar] [CrossRef] [PubMed]
  19. Ottaiano, A.; Petito, A.; Santorsola, M.; Gigantino, V.; Capuozzo, M.; Fontanella, D.; Di Franco, R.; Borzillo, V.; Buonopane, S.; Ravo, V.; et al. Prospective Evaluation of Radiotherapy-Induced Immunologic and Genetic Effects in Colorectal Cancer Oligo-Metastatic Patients with Lung-Limited Disease: The PRELUDE-1 Study. Cancers 2021, 13, 4236. [Google Scholar] [CrossRef] [PubMed]
  20. Lancellotta, V.; Del Regno, L.; Di Stefani, A.; Fionda, B.; Marazzi, F.; Rossi, E.; Balducci, M.; Pampena, R.; Morganti, A.G.; Mangoni, M.; et al. The role of stereotactic radiotherapy in addition to immunotherapy in the management of melanoma brain metastases: Results of a systematic review. Radiol. Med. 2022, 127, 773–783. [Google Scholar] [CrossRef]
  21. Lev, S. Targeted therapy and drug resistance in triple-negative breast cancer: The EGFR axis. Biochem. Soc. Trans. 2020, 48, 657–665. [Google Scholar] [CrossRef]
  22. Shariati, M.; Meric-Bernstam, F. Targeting AKT for cancer therapy. Expert Opin. Investig. Drugs 2019, 28, 977–988. [Google Scholar] [CrossRef] [PubMed]
  23. Hussein, M.A.M.; Cafarelli, F.P.; Paparella, M.T.; Rennie, W.J.; Guglielmi, G. Phosphaturic mesenchymal tumors: Radiological aspects and suggested imaging pathway. Radiol. Med. 2021, 126, 1609–1618. [Google Scholar] [CrossRef]
  24. Wang, F.H.; Zheng, H.L.; Li, J.T.; Li, P.; Zheng, C.H.; Chen, Q.Y.; Huang, C.M.; Xie, J.W. Prediction of recurrence-free survival and adjuvant therapy benefit in patients with gastrointestinal stromal tumors based on radiomics features. Radiol Med. 2022, 127, 1085–1097. [Google Scholar] [CrossRef] [PubMed]
  25. Zafar, A.; Wang, W.; Liu, G.; Wang, X.; Xian, W.; McKeon, F.; Foster, J.; Zhou, J.; Zhang, R. Molecular targeting therapies for neuroblastoma: Progress and challenges. Med. Res. Rev. 2021, 41, 961–1021. [Google Scholar] [CrossRef] [PubMed]
  26. Laurelli, G.; Falcone, F.; Gallo, M.S.; Scala, F.; Losito, S.; Granata, V.; Cascella, M.; Greggi, S. Long-Term Oncologic and Reproductive Outcomes in Young Women with Early Endometrial Cancer Conservatively Treated: A Prospective Study and Literature Update. Int. J. Gynecol. Cancer 2016, 26, 1650–1657. [Google Scholar] [CrossRef] [PubMed]
  27. Colli, L.M.; Machiela, M.J.; Zhang, H.; Myers, T.A.; Jessop, L.; Delattre, O.; Yu, K.; Chanock, S.J. Landscape of Combination Immunotherapy and Targeted Therapy to Improve Cancer Management. Cancer Res. 2017, 77, 3666–3671. [Google Scholar] [CrossRef] [Green Version]
  28. Polesel, J.; Talamini, R.; Montella, M.; Maso, L.D.; Crovatto, M.; Parpinel, M.; Izzo, F.; Tommasi, L.G.; Serraino, D.; La Vecchia, C.; et al. Nutrients intake and the risk of hepatocellular carcinoma in Italy. Eur. J. Cancer 2007, 43, 2381–2387. [Google Scholar] [CrossRef]
  29. Pignata, S.; Gallo, C.; Daniele, B.; Elba, S.; Giorgio, A.; Capuano, G.; Adinolfi, L.E.; De Sio, I.; Izzo, F.; Farinati, F.; et al. Characteristics at presentation and outcome of hepatocellular carcinoma (HCC) in the elderly. A study of the Cancer of the Liver Italian Program (CLIP). Crit. Rev. Oncol. Hematol. 2006, 59, 243–249. [Google Scholar] [CrossRef] [PubMed]
  30. Baxter, D. Active and passive immunization for cancer. Hum. Vaccines Immunother. 2014, 10, 2123–2129. [Google Scholar] [CrossRef]
  31. Bregy, A.; Wong, T.M.; Shah, A.H.; Goldberg, J.M.; Komotar, R.J. Active immunotherapy using dendritic cells in the treatment of glioblastoma multiforme. Cancer Treat. Rev. 2013, 39, 891–907. [Google Scholar] [CrossRef]
  32. Foy, J.P.; Karabajakian, A.; Ortiz-Cuaran, S.; Boussageon, M.; Michon, L.; Bouaoud, J.; Fekiri, D.; Robert, M.; Baffert, K.A.; Hervé, G.; et al. Datasets for gene expression profiles of head and neck squamous cell carcinoma and lung cancer treated or not by PD1/PD-L1 inhibitors. Data Brief 2022, 44, 108556. [Google Scholar] [CrossRef]
  33. Vavolizza, R.D.; Petroni, G.R.; Mauldin, I.S.; Chianese-Bullock, K.A.; Olson, W.C.; Smith, K.T.; Dengel, L.T.; Haden, K.; Grosh, W.W.; Kaur, V.; et al. Phase I/II clinical trial of a helper peptide vaccine plus PD-1 blockade in PD-1 antibody-naïve and PD-1 antibody-experienced patients with melanoma (MEL64). J. Immunother. Cancer 2022, 10, e005424. [Google Scholar] [CrossRef] [PubMed]
  34. Yang, M.; Mahanty, A.; Jin, C.; Wong, A.N.N.; Yoo, J.S. Label-free metabolic imaging for sensitive and robust monitoring of anti-CD47 immunotherapy response in triple-negative breast cancer. J. Immunother. Cancer 2022, 10, e005199. [Google Scholar] [CrossRef] [PubMed]
  35. Gao, T.T.; Shan, J.H.; Yang, Y.X.; Zhang, Z.W.; Liu, S.L.; Xi, M.; Liu, M.Z.; Zhao, L. Comparative efficacy and safety of immunotherapy for patients with advanced or metastatic esophageal squamous cell carcinoma: A systematic review and network Meta-analysis. BMC Cancer 2022, 22, 992. [Google Scholar] [CrossRef] [PubMed]
  36. Wang, Y.; Lai, Y.; Peng, H.; Yan, S.; Liu, Z.; Tong, C.; Huang, X. Multiparametric immune profiling of advanced cervical cancer to predict response to programmed death-1 inhibitor combination therapy: An exploratory study of the CLAP trial. Clin. Transl. Oncol. 2022, 25, 256–268. [Google Scholar] [CrossRef] [PubMed]
  37. Yuan, Z.G.; Zeng, T.M.; Tao, C.J. Current and emerging immunotherapeutic approaches for biliary tract cancers. Hepatobiliary Pancreat. Dis. Int. 2022, 21, 440–449. [Google Scholar] [CrossRef]
  38. Taieb, J.; Svrcek, M.; Cohen, R.; Basile, D.; Tougeron, D.; Phelip, J.M. Deficient mismatch repair/microsatellite unstable colorectal cancer: Diagnosis, prognosis and treatment. Eur. J. Cancer 2022, 175, 136–157. [Google Scholar] [CrossRef]
  39. Granata, V.; Fusco, R.; De Muzio, F.; Cutolo, C.; Setola, S.V.; Dell’Aversana, F.; Grassi, F.; Belli, A.; Silvestro, L.; Ottaiano, A.; et al. Radiomics and machine learning analysis based on magnetic resonance imaging in the assessment of liver mucinous colorectal metastases. Radiol. Med. 2022, 127, 763–772. [Google Scholar] [CrossRef]
  40. Granata, V.; Grassi, R.; Fusco, R.; Belli, A.; Palaia, R.; Carrafiello, G.; Miele, V.; Grassi, R.; Petrillo, A.; Izzo, F. Local ablation of pancreatic tumors: State of the art and future perspectives. World J. Gastroenterol. 2021, 27, 3413–3428. [Google Scholar] [CrossRef]
  41. Cholangiocarcinoma Working Group. Italian Clinical Practice Guidelines on Cholangiocarcinoma—Part II: Treatment. Dig. Liver Dis. 2020, 52, 1430–1442. [Google Scholar] [CrossRef] [PubMed]
  42. Ottaiano, A.; de Vera d’Aragona, R.P.; Trotta, A.M.; Santorsola, M.; Napolitano, M.; Scognamiglio, G.; Tatangelo, F.; Grieco, P.; Zappavigna, S.; Granata, V.; et al. Characterization of KRAS Mutational Regression in Oligometastatic Patients. Front. Immunol. 2022, 13, 898561. [Google Scholar] [CrossRef] [PubMed]
  43. Fusco, R.; Simonetti, I.; Ianniello, S.; Villanacci, A.; Grassi, F.; Dell’Aversana, F.; Grassi, R.; Cozzi, D.; Bicci, E.; Palumbo, P.; et al. Pulmonary Lymphangitis Poses a Major Challenge for Radiologists in an Oncological Setting during the COVID-19 Pandemic. J. Pers. Med. 2022, 12, 624. [Google Scholar] [CrossRef] [PubMed]
  44. Hodi, F.S.; O’Day, S.J.; McDermott, D.F.; Weber, R.W.; Sosman, J.A.; Haanen, J.B.; Gonzalez, R.; Robert, C.; Schadendorf, D.; Hassel, J.C.; et al. Improved survival with ipilimumab in patients with metastatic melanoma. New Engl. J. Med. 2010, 363, 711–723. [Google Scholar] [CrossRef]
  45. Fushimi, Y.; Yoshida, K.; Okawa, M.; Maki, T.; Nakajima, S.; Sakata, A.; Okuchi, S.; Hinoda, T.; Kanagaki, M.; Nakamoto, Y. Vessel wall MR imaging in neuroradiology. Radiol. Med. 2022, 127, 1032–1045. [Google Scholar] [CrossRef] [PubMed]
  46. Granata, V.; Simonetti, I.; Fusco, R.; Setola, S.V.; Izzo, F.; Scarpato, L.; Vanella, V.; Festino, L.; Simeone, E.; Ascierto, P.A.; et al. Management of cutaneous melanoma: Radiologists challenging and risk assessment. Radiol. Med. 2022, 127, 899–911. [Google Scholar] [CrossRef]
  47. Michielin, O.; van Akkooi, A.C.J.; Ascierto, P.A.; Dummer, R.; Keilholz, U. Cutaneous melanoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2019, 30, 1884–1901. [Google Scholar] [CrossRef] [Green Version]
  48. Keilholz, U.; Ascierto, P.A.; Dummer, R.; Robert, C.; Lorigan, P.; van Akkooi, A.; Arance, A.; Blank, C.U.; Chiarion Sileni, V.; Donia, M.; et al. ESMO consensus conference recommendations on the management of metastatic melanoma: Under the auspices of the ESMO Guidelines Committee. Ann. Oncol. 2020, 31, 1435–1448. [Google Scholar] [CrossRef] [PubMed]
  49. Cirillo, L.; Rustici, A.; Toni, F.; Zoli, M.; Bartiromo, F.; Gramegna, L.L.; Cicala, D.; Tonon, C.; Caranci, F.; Lodi, R. Vessel Wall MRI: Clinical implementation in cerebrovascular disorders-technical aspects. Radiol. Med. 2022, 127, 645–651. [Google Scholar] [CrossRef]
  50. Tagliafico, A.S.; Campi, C.; Bianca, B.; Bortolotto, C.; Buccicardi, D.; Francesca, C.; Prost, R.; Rengo, M.; Faggioni, L. Blockchain in radiology research and clinical practice: Current trends and future directions. Radiol. Med. 2022, 127, 391–397. [Google Scholar] [CrossRef]
  51. Perrone, F.; Gallo, C.; Daniele, B.; Gaeta, G.B.; Izzo, F.; Capuano, G.; Adinolfi, L.E.; Mazzanti, R.; Farinati, F.; Elba, S.; et al. Tamoxifen in the treatment of hepatocellular carcinoma: 5-year results of the CLIP-1 multicentre randomised controlled trial. Curr. Pharm. Des. 2002, 8, 1013–1019. [Google Scholar] [CrossRef] [PubMed]
  52. Granata, V.; Fusco, R.; Salati, S.; Petrillo, A.; Di Bernardo, E.; Grassi, R.; Palaia, R.; Danti, G.; La Porta, M.; Cadossi, M.; et al. A Systematic Review about Imaging and Histopathological Findings for Detecting and Evaluating Electroporation Based Treatments Response. Int. J. Environ. Res. Public Health 2021, 18, 5592. [Google Scholar] [CrossRef]
  53. Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.J.; Rutkowski, P.; Lao, C.D.; Cowey, C.L.; Schadendorf, D.; Wagstaff, J.; Dummer, R.; et al. Five-Year Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. New Engl. J. Med. 2019, 381, 1535–1546. [Google Scholar] [CrossRef] [Green Version]
  54. Chiti, G.; Grazzini, G.; Flammia, F.; Matteuzzi, B.; Tortoli, P.; Bettarini, S.; Pasqualini, E.; Granata, V.; Busoni, S.; Messserini, L.; et al. Gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs): A radiomic model to predict tumor grade. Radiol. Med. 2022, 127, 928–938. [Google Scholar] [CrossRef]
  55. Granata, V.; Grassi, R.; Fusco, R.; Setola, S.V.; Belli, A.; Ottaiano, A.; Nasti, G.; La Porta, M.; Danti, G.; Cappabianca, S.; et al. Intrahepatic cholangiocarcinoma and its differential diagnosis at MRI: How radiologist should assess MR features. Radiol. Med. 2021, 126, 1584–1600. [Google Scholar] [CrossRef] [PubMed]
  56. Cholangiocarcinoma Working Group. Italian Clinical Practice Guidelines on Cholangiocarcinoma—Part I: Classification, diagnosis and staging. Dig. Liver Dis. 2020, 52, 1282–1293. [Google Scholar] [CrossRef] [PubMed]
  57. Granata, V.; Fusco, R.; De Muzio, F.; Cutolo, C.; Setola, S.V.; Grassi, R.; Grassi, F.; Ottaiano, A.; Nasti, G.; Tatangelo, F.; et al. Radiomics textural features by MR imaging to assess clinical outcomes following liver resection in colorectal liver metastases. Radiol. Med. 2022, 127, 461–470. [Google Scholar] [CrossRef]
  58. Fusco, R.; Setola, S.V.; Raiano, N.; Granata, V.; Cerciello, V.; Pecori, B.; Petrillo, A. Analysis of a monocentric computed tomography dosimetric database using a radiation dose index monitoring software: Dose levels and alerts before and after the implementation of the adaptive statistical iterative reconstruction on CT images. Radiol. Med. 2022, 127, 733–742. [Google Scholar] [CrossRef]
  59. Robert, C.; Long, G.V.; Brady, B.; Dutriaux, C.; Maio, M.; Mortier, L.; Hassel, J.C.; Rutkowski, P.; McNeil, C.; Kalinka-Warzocha, E.; et al. Nivolumab in previously untreated melanoma without BRAF mutation. New Engl. J. Med. 2015, 372, 320–330. [Google Scholar] [CrossRef] [Green Version]
  60. Seth, R.; Messersmith, H.; Kaur, V.; Kirkwood, J.M.; Kudchadkar, R.; McQuade, J.L.; Provenzano, A.; Swami, U.; Weber, J.; Alluri, K.C.; et al. Systemic Therapy for Melanoma: ASCO Guideline. J. Clin. Oncol. 2020, 38, 3947–3970. [Google Scholar] [CrossRef]
  61. Weber, J.; Mandala, M.; Del Vecchio, M.; Gogas, H.J.; Arance, A.M.; Cowey, C.L.; Dalle, S.; Schenker, M.; Chiarion-Sileni, V.; Marquez-Rodas, I.; et al. Adjuvant Nivolumab versus Ipilimumab in Resected Stage III or IV Melanoma. New Engl. J. Med. 2017, 377, 1824–1835. [Google Scholar] [CrossRef] [PubMed]
  62. Wolchok, J.D.; Chiarion-Sileni, V.; Gonzalez, R.; Rutkowski, P.; Grob, J.J.; Cowey, C.L.; Lao, C.D.; Wagstaff, J.; Schadendorf, D.; Ferrucci, P.F.; et al. Overall Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. New Engl. J. Med. 2017, 377, 1345–1356. [Google Scholar] [CrossRef]
  63. Ascierto, P.A.; Del Vecchio, M.; Mandalá, M.; Gogas, H.; Arance, A.M.; Dalle, S.; Cowey, C.L.; Schenker, M.; Grob, J.J.; Chiarion-Sileni, V.; et al. Adjuvant nivolumab versus ipilimumab in resected stage IIIB-C and stage IV melanoma (CheckMate 238): 4-year results from a multicentre, double-blind, randomised, controlled, phase 3 trial. Lancet Oncol. 2020, 21, 1465–1477. [Google Scholar] [CrossRef] [PubMed]
  64. Amaria, R.N.; Menzies, A.M.; Burton, E.M.; Scolyer, R.A.; Tetzlaff, M.T.; Antdbacka, R.; Ariyan, C.; Bassett, R.; Carter, B.; Daud, A.; et al. Neoadjuvant systemic therapy in melanoma: Recommendations of the International Neoadjuvant Melanoma Consortium. Lancet Oncol. 2019, 20, e378–e389. [Google Scholar] [CrossRef] [PubMed]
  65. Gutzmer, R.; Stroyakovskiy, D.; Gogas, H.; Robert, C.; Lewis, K.; Protsenko, S.; Pereira, R.P.; Eigentler, T.; Rutkowski, P.; Demidov, L.; et al. Atezolizumab, vemurafenib, and cobimetinib as first-line treatment for unresectable advanced BRAFV600mutation-positive melanoma (IMspire150): Primary analysis of the randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2020, 395, 1835–1844. [Google Scholar] [CrossRef]
  66. Barretta, M.L.; Catalano, O.; Setola, S.V.; Granata, V.; Marone, U.; D’Errico Gallipoli, A. Gallbladder metastasis: Spectrum of imaging findings. Abdom. Imaging 2011, 36, 729–734. [Google Scholar] [CrossRef]
  67. Chapman, P.B.; Hauschild, A.; Robert, C.; Haanen, J.B.; Ascierto, P.; Larkin, J.; Dummer, R.; Garbe, C.; Testori, A.; Maio, M.; et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. New Engl. J. Med. 2011, 364, 2507–2516. [Google Scholar] [CrossRef] [Green Version]
  68. Fusco, R.; Granata, V.; Sansone, M.; Rega, D.; Delrio, P.; Tatangelo, F.; Romano, C.; Avallone, A.; Pupo, D.; Giordano, M.; et al. Validation of the standardized index of shape tool to analyze DCE-MRI data in the assessment of neo-adjuvant therapy in locally advanced rectal cancer. Radiol. Med. 2021, 126, 1044–1054. [Google Scholar] [CrossRef] [PubMed]
  69. Ascierto, P.A.; Kirkwood, J.M.; Grob, J.J.; Simeone, E.; Grimaldi, A.M.; Maio, M.; Palmieri, G.; Testori, A.; Marincola, F.M.; Mozzillo, N. The role of BRAF V600 mutation in melanoma. J. Transl. Med. 2012, 10, 85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Fusco, R.; Sansone, M.; Granata, V.; Setola, S.V.; Petrillo, A. A systematic review on multiparametric MR imaging in prostate cancer detection. Infect. Agents Cancer 2017, 312, 57. [Google Scholar] [CrossRef]
  71. Granier, C.; De Guillebon, E.; Blanc, C.; Roussel, H.; Badoual, C.; Colin, E.; Saldmann, A.; Gey, A.; Oudard, S.; Tartour, E. Mechanisms of action and rationale for the use of checkpoint inhibitors in cancer. ESMO Open 2017, 2, e000213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Martínez-Lostao, L.; Anel, A.; Pardo, J. How Do Cyto-toxic Lymphocytes Kill Cancer Cells? Clin. Cancer Res. 2015, 21, 5047–5056. [Google Scholar] [CrossRef] [Green Version]
  73. Hegde, P.S.; Chen, D.S. Top 10 Challenges in Cancer Immunotherapy. Immunity 2020, 52, 17–35. [Google Scholar] [CrossRef]
  74. Zimmermannova, O.; Caiado, I.; Ferreira, A.G.; Pereira, C.F. Cell Fate Reprogramming in the Era of Cancer Immunotherapy. Front. Immunol. 2021, 12, 714822. [Google Scholar] [CrossRef]
  75. Schoenfeld, A.J.; Hellmann, M.D. Acquired Resistance to Immune Checkpoint Inhibitors. Cancer Cell 2020, 37, 443–455. [Google Scholar] [CrossRef] [PubMed]
  76. Stefanini, M.; Simonetti, G. Interventional Magnetic Resonance Imaging Suite (IMRIS): How to build and how to use. Radiol. Med. 2022, 127, 1063–1067. [Google Scholar] [CrossRef] [PubMed]
  77. Renzulli, M.; Brandi, N.; Argalia, G.; Brocchi, S.; Farolfi, A.; Fanti, S.; Golfieri, R. Morphological, dynamic and functional characteristics of liver pseudolesions and benign lesions. Radiol. Med. 2022, 127, 129–144. [Google Scholar] [CrossRef] [PubMed]
  78. Palatresi, D.; Fedeli, F.; Danti, G.; Pasqualini, E.; Castiglione, F.; Messerini, L.; Massi, D.; Bettarini, S.; Tortoli, P.; Busoni, S.; et al. Correlation of CT radiomic features for GISTs with pathological classification and molecular subtypes: Preliminary and monocentric experience. Radiol. Med. 2022, 127, 117–128. [Google Scholar] [CrossRef] [PubMed]
  79. Ledda, R.E.; Silva, M.; McMichael, N.; Sartorio, C.; Branchi, C.; Milanese, G.; Nayak, S.M.; Sverzellati, N. The diagnostic value of grey-scale inversion technique in chest radiography. Radiol. Med. 2022, 127, 294–304. [Google Scholar] [CrossRef] [PubMed]
  80. Ierardi, A.M.; Stellato, E.; Pellegrino, G.; Bonelli, C.; Cellina, M.; Renzulli, M.; Biondetti, P.; Carrafiello, G. Fluid-dynamic control microcatheter used with glue: Preliminary experience on its feasibility and safety. Radiol. Med. 2022, 127, 272–276. [Google Scholar] [CrossRef] [PubMed]
  81. Sansone, M.; Marrone, S.; Di Salvio, G.; Belfiore, M.P.; Gatta, G.; Fusco, R.; Vanore, L.; Zuiani, C.; Grassi, F.; Vietri, M.T.; et al. Comparison between two packages for pectoral muscle removal on mammographic images. Radiol. Med. 2022, 127, 848–856. [Google Scholar] [CrossRef] [PubMed]
  82. Granata, V.; Fusco, R.; Belli, A.; Danti, G.; Bicci, E.; Cutolo, C.; Petrillo, A.; Izzo, F. Diffusion weighted imaging and diffusion kurtosis imaging in abdominal oncological setting: Why and when. Infect. Agents Cancer 2022, 17, 25. [Google Scholar] [CrossRef] [PubMed]
  83. Petrillo, A.; Fusco, R.; Di Bernardo, E.; Petrosino, T.; Barretta, M.L.; Porto, A.; Granata, V.; Di Bonito, M.; Fanizzi, A.; Massafra, R.; et al. Prediction of Breast Cancer Histological Outcome by Radiomics and Artificial Intelligence Analysis in Contrast-Enhanced Mammography. Cancers 2022, 14, 2132. [Google Scholar] [CrossRef] [PubMed]
  84. Granata, V.; Fusco, R.; De Muzio, F.; Cutolo, C.; Setola, S.V.; Dell’Aversana, F.; Belli, A.; Romano, C.; Ottaiano, A.; Nasti, G.; et al. Magnetic Resonance Features of Liver Mucinous Colorectal Metastases: What the Radiologist Should Know. J. Clin. Med. 2022, 11, 2221. [Google Scholar] [CrossRef] [PubMed]
  85. Cutolo, C.; Dell’Aversana, F.; Fusco, R.; Grazzini, G.; Chiti, G.; Simonetti, I.; Bruno, F.; Palumbo, P.; Pierpaoli, L.; Valeri, T.; et al. Combined Hepatocellular-Cholangiocarcinoma: What the Multidisciplinary Team Should Know. Diagnostics 2022, 12, 890. [Google Scholar] [CrossRef]
  86. Committeri, U.; Fusco, R.; Di Bernardo, E.; Abbate, V.; Salzano, G.; Maglitto, F.; Dell’Aversana Orabona, G.; Piombino, P.; Bonavolontà, P.; Arena, A.; et al. Radiomics Metrics Combined with Clinical Data in the Surgical Management of Early-Stage (cT1-T2 N0) Tongue Squamous Cell Carcinomas: A Preliminary Study. Biology 2022, 11, 468. [Google Scholar] [CrossRef]
  87. Reck, M.; Rodríguez-Abreu, D.; Robinson, A.G.; Hui, R.; Csőszi, T.; Fülöp, A.; Gottfried, M.; Peled, N.; Tafreshi, A.; Cuffe, S.; et al. Pembrolizumab versus chemotherapy for PD-L1-positive non- small-cell lung cancer. N. Engl. J. Med. 2016, 375, 1823–1833. [Google Scholar] [CrossRef] [Green Version]
  88. Hamid, O.; Molinero, L.; Bolen, C.R.; Sosman, J.A.; Muñoz-Couselo, E.; Kluger, H.M.; McDermott, D.F.; Powderly, J.D.; Sarkar, I.; Ballinger, M.; et al. Safety, Clinical Activity, and Biological Correlates of Response in Patients with Metastatic Melanoma: Results from a Phase I Trial of Atezolizumab. Clin. Cancer Res. 2019, 25, 6061–6072. [Google Scholar] [CrossRef] [Green Version]
  89. Kim, J.M.; Chen, D.S. Immune escape to PD-L1/PD-1 blockade: Seven steps to success (or failure). Ann. Oncol. 2016, 8, 1492–1504. [Google Scholar] [CrossRef]
  90. Anagnostou, V.; Smith, K.N.; Forde, P.M.; Niknafs, N.; Bhattacharya, R.; White, J.; Zhang, T.; Adleff, V.; Phallen, J.; Wali, N.; et al. Evolution of Neoantigen Landscape during Immune Checkpoint Blockade in Non-Small Cell Lung Cancer. Cancer Discov. 2017, 7, 264–276. [Google Scholar] [CrossRef]
  91. Wang, D.Y.; Eroglu, Z.; Ozgun, A.; Leger, P.D.; Zhao, S.; Ye, F.; Luke, J.J.; Joseph, R.W.; Haq, R.; Ott, P.A.; et al. Clinical Features of Acquired Resistance to Anti-PD-1 Therapy in Advanced Melanoma. Cancer Immunol. Res. 2017, 5, 357–362. [Google Scholar] [CrossRef] [Green Version]
  92. McGranahan, N.; Rosenthal, R.; Hiley, C.T.; Rowan, A.J.; Watkins, T.B.K.; Wilson, G.A.; Birkbak, N.J.; Veeriah, S.; Van Loo, P.; Herrero, J.; et al. Allele-Specific HLA Loss and Immune Escape in Lung Cancer Evolution. Cell 2017, 171, 1259–1271.e11. [Google Scholar] [CrossRef] [Green Version]
  93. Darnell, E.P.; Mooradian, M.J.; Baruch, E.N.; Yilmaz, M.; Reynolds, K.L. Immune-Related Adverse Events (irAEs): Diagnosis, Management, and Clinical Pearls. Curr. Oncol. Rep. 2020, 22, 39. [Google Scholar] [CrossRef]
  94. Common Terminology Criteria for Adverse Events (CTCAE) V5. Available online: https://ctep.cancer.gov/protocolDevelopment/electronic_applications/ctc.htm (accessed on 25 October 2022).
  95. Arnaud-Coffin, P.; Maillet, D.; Gan, H.K.; Stelmes, J.-J.; You, B.; Dalle, S.; Péron, J. A systematic review of adverse events in randomized trials assessing immune checkpoint inhibitors. Int. J. Cancer 2019, 145, 639–648. [Google Scholar] [CrossRef]
  96. Simeone, E.; Grimaldi, A.M.; Festino, L.; Trojaniello, C.; Vitale, M.G.; Vanella, V.; Palla, M.; Ascierto, P.A. Immunotherapy in metastatic melanoma: A novel scenario of new toxicities and their management. Melanoma Manag. 2019, 6, MMT30. [Google Scholar] [CrossRef]
  97. Nishino, M.; Hatabu, H.; Hodi, F.S. Imaging of Cancer Immunotherapy: Current Approaches and Future Directions. Radiology 2019, 290, 9–22. [Google Scholar] [CrossRef]
  98. Brahmer, J.R.; Abu-Sbeih, H.; Ascierto, P.A.; Brufsky, J.; Cappelli, L.C.; Cortazar, F.B.; Gerber, D.E.; Hamad, L.; Hansen, E.; Johnson, D.B.; et al. Society for Immunotherapy of Cancer (SITC) clinical practice guideline on immune checkpoint inhibitor-related adverse events. J. Immunother. Cancer 2021, 9, e002435. [Google Scholar] [CrossRef]
  99. Comstock, D.E.; Nishino, M.; Giardino, A.A. Headache in the setting of immunotherapy treatment for metastatic melanoma. JAMA Oncol. 2017, 3, 703–704. [Google Scholar] [CrossRef]
  100. Faje, A.T.; Sullivan, R.; Lawrence, D.; Tritos, N.A.; Fadden, R.; Klibanski, A.; Nachtigall, L. Ipilimumab-induced hypophysitis: A detailed longitudinal analysis in a large cohort of patients with metastatic melanoma. J. Clin. Endocrinol. Metab. 2014, 99, 4078–4085. [Google Scholar] [CrossRef] [Green Version]
  101. Min, L.; Hodi, F.S.; Giobbie-Hurder, A.; Ott, P.A.; Luke, J.J.; Donahue, H.; Davis, M.; Carroll, R.S.; Kaiser, U.B. Systemic high-dose corticosteroid treatment does not improve the outcome of ipilimumab-related hypophysitis: A retrospective cohort study. Clin. Cancer Res. 2015, 21, 749–755. [Google Scholar] [CrossRef]
  102. Khunger, M.; Rakshit, S.; Pasupuleti, V.; Hernandez, A.V.; Mazzone, P.; Stevenson, J.; Pennell, N.A.; Velcheti, V. Incidence of Pneumonitis with Use of Programmed Death 1 and Programmed Death-Ligand 1 Inhibitors in Non-Small Cell Lung Cancer: A Systematic Review and Meta-Analysis of Trials. Chest 2017, 152, 271–281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Kalisz, K.R.; Ramaiya, N.H.; Laukamp, K.R.; Gupta, A. Immune Checkpoint Inhibitor Therapy-related Pneumonitis: Patterns and Management. Radiographics 2019, 39, 1923–1937. [Google Scholar] [CrossRef] [PubMed]
  104. Bianchi, A.; Mazzoni, L.N.; Busoni, S.; Pinna, N.; Albanesi, M.; Cavigli, E.; Cozzi, D.; Poggesi, A.; Miele, V.; Fainardi, E.; et al. Assessment of cerebrovascular disease with computed tomography in COVID-19 patients: Correlation of a novel specific visual score with increased mortality risk. Radiol. Med. 2021, 126, 570–576. [Google Scholar] [CrossRef] [PubMed]
  105. Cartocci, G.; Colaiacomo, M.C.; Lanciotti, S.; Andreoli, C.; De Cicco, M.L.; Brachetti, G.; Pugliese, S.; Capoccia, L.; Tortora, A.; Scala, A.; et al. Correction to: Chest CT for early detection and management of coronavirus disease (COVID-19): A report of 314 patients admitted to Emergency Department with suspected pneumonia. Radiol. Med. 2021, 126, 642. [Google Scholar] [CrossRef] [PubMed]
  106. Masci, G.M.; Iafrate, F.; Ciccarelli, F.; Pambianchi, G.; Panebianco, V.; Pasculli, P.; Ciardi, M.R.; Mastroianni, C.M.; Ricci, P.; Catalano, C.; et al. Tocilizumab effects in COVID-19 pneumonia: Role of CT texture analysis in quantitative assessment of response to therapy. Radiol. Med. 2021, 126, 1170–1180. [Google Scholar] [CrossRef] [PubMed]
  107. Francolini, G.; Desideri, I.; Stocchi, G.; Ciccone, L.P.; Salvestrini, V.; Garlatti, P.; Aquilano, M.; Greto, D.; Bonomo, P.; Meattini, I.; et al. Impact of COVID-19 on workload burden of a complex radiotherapy facility. Radiol. Med. 2021, 126, 717–721. [Google Scholar] [CrossRef]
  108. McKay, M.J.; Foster, R. Radiation recall reactions: An oncologic enigma. Crit. Rev. Oncol. 2021, 168, 103527. [Google Scholar] [CrossRef] [PubMed]
  109. Granata, V.; Fusco, R.; Catalano, O.; Avallone, A.; Palaia, R.; Botti, G.; Tatangelo, F.; Granata, F.; Cascella, M.; Izzo, F.; et al. Diagnostic accuracy of magnetic resonance, computed tomography and contrast enhanced ultrasound in radiological multimodality assessment of peribiliary liver metastases. PLoS ONE 2017, 12, e0179951. [Google Scholar] [CrossRef] [Green Version]
  110. Granata, V.; Fusco, R.; Catalano, O.; Avallone, A.; Leongito, M.; Izzo, F.; Petrillo, A. Peribiliary liver metastases MR findings. Med. Oncol. 2017, 34, 124. [Google Scholar] [CrossRef]
  111. Li, N.; Wakim, J.; Koethe, Y.; Huber, T.; Schenning, R.; Gade, T.P.; Hunt, S.J.; Park, B.J. Multicenter assessment of augmented reality registration methods for image-guided interventions. Radiol. Med. 2022, 127, 857–865. [Google Scholar] [CrossRef]
  112. Caruso, D.; Polici, M.; Rinzivillo, M.; Zerunian, M.; Nacci, I.; Marasco, M.; Magi, L.; Tarallo, M.; Gargiulo, S.; Iannicelli, E.; et al. CT-based radiomics for prediction of therapeutic response to Everolimus in metastatic neuroendocrine tumors. Radiol. Med. 2022, 127, 691–701. [Google Scholar] [CrossRef]
  113. Han, D.; Yu, N.; Yu, Y.; He, T.; Duan, X. Performance of CT radiomics in predicting the overall survival of patients with stage III clear cell renal carcinoma after radical nephrectomy. Radiol. Med. 2022, 127, 837–847. [Google Scholar] [CrossRef]
  114. Granata, V.; Fusco, R.; de Lutio di Castelguidone, E.; Avallone, A.; Palaia, R.; Delrio, P.; Tatangelo, F.; Botti, G.; Grassi, R.; Izzo, F.; et al. Diagnostic performance of gadoxetic acid-enhanced liver MRI versus multidetector CT in the assessment of colorectal liver metastases compared to hepatic resection. BMC Gastroenterol. 2019, 19, 129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Masci, G.M.; Ciccarelli, F.; Mattei, F.I.; Grasso, D.; Accarpio, F.; Catalano, C.; Laghi, A.; Sammartino, P.; Iafrate, F. Role of CT texture analysis for predicting peritoneal metastases in patients with gastric cancer. Radiol. Med. 2022, 127, 251–258. [Google Scholar] [CrossRef]
  116. Tafuto, S.; von Arx, C.; De Divitiis, C.; Maura, C.T.; Palaia, R.; Albino, V.; Fusco, R.; Membrini, M.; Petrillo, A.; Granata, V.; et al. Electrochemotherapy as a new approach on pancreatic cancer and on liver metastases. Int. J. Surg. 2015, 21 (Suppl. S1), S78–S82. [Google Scholar] [CrossRef] [PubMed]
  117. Zerunian, M.; Pucciarelli, F.; Caruso, D.; Polici, M.; Masci, B.; Guido, G.; De Santis, D.; Polverari, D.; Principessa, D.; Benvenga, A.; et al. Artificial intelligence based image quality enhancement in liver MRI: A quantitative and qualitative evaluation. Radiol. Med. 2022, 127, 1098–1105. [Google Scholar] [CrossRef] [PubMed]
  118. Kang, Y.J.; Cho, J.H.; Hwang, S.H. Diagnostic value of various criteria for deep lobe involvement in radiologic studies with parotid mass: A systematic review and meta-analysis. Radiol. Med. 2022, 127, 1124–1133. [Google Scholar] [CrossRef]
  119. Borgheresi, A.; De Muzio, F.; Agostini, A.; Ottaviani, L.; Bruno, A.; Granata, V.; Fusco, R.; Danti, G.; Flammia, F.; Grassi, R.; et al. Lymph Nodes Evaluation in Rectal Cancer: Where Do We Stand and Future Perspective. J. Clin. Med. 2022, 11, 2599. [Google Scholar] [CrossRef]
  120. Granata, V.; de Lutio di Castelguidone, E.; Fusco, R.; Catalano, O.; Piccirillo, M.; Palaia, R.; Izzo, F.; Gallipoli, A.D.; Petrillo, A. Irreversible electroporation of hepatocellular carcinoma: Preliminary report on the diagnostic accuracy of magnetic resonance, computer tomography, and contrast-enhanced ultrasound in evaluation of the ablated area. Radiol. Med. 2016, 121, 122–131. [Google Scholar] [CrossRef]
  121. Granata, V.; Grassi, R.; Fusco, R.; Belli, A.; Cutolo, C.; Pradella, S.; Grazzini, G.; La Porta, M.; Brunese, M.C.; De Muzio, F.; et al. Diagnostic evaluation and ablation treatments assessment in hepatocellular carcinoma. Infect. Agents Cancer 2021, 16, 53. [Google Scholar] [CrossRef]
  122. Granata, V.; Grassi, R.; Fusco, R.; Setola, S.V.; Belli, A.; Piccirillo, M.; Pradella, S.; Giordano, M.; Cappabianca, S.; Brunese, L.; et al. Abbreviated MRI Protocol for the Assessment of Ablated Area in HCC Patients. Int. J. Environ. Res. Public Health 2021, 18, 3598. [Google Scholar] [CrossRef] [PubMed]
  123. Petrillo, A.; Fusco, R.; Granata, V.; Filice, S.; Sansone, M.; Rega, D.; Delrio, P.; Bianco, F.; Romano, G.M.; Tatangelo, F.; et al. Assessing response to neo-adjuvant therapy in locally advanced rectal cancer using Intra-voxel Incoherent Motion modelling by DWI data and Standardized Index of Shape from DCE-MRI. Ther. Adv. Med. Oncol. 2018, 10, 1758835918809875. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Fusco, R.; Sansone, M.; Granata, V.; Grimm, R.; Pace, U.; Delrio, P.; Tatangelo, F.; Botti, G.; Avallone, A.; Pecori, B.; et al. Diffusion and perfusion MR parameters to assess preoperative short-course radiotherapy response in locally advanced rectal cancer: A comparative explorative study among Standardized Index of Shape by DCE-MRI, intravoxel incoherent motion- and diffusion kurtosis imaging-derived parameters. Abdom. Radiol. 2019, 44, 3683–3700. [Google Scholar] [CrossRef]
  125. Scola, E.; Desideri, I.; Bianchi, A.; Gadda, D.; Busto, G.; Fiorenza, A.; Amadori, T.; Mancini, S.; Miele, V.; Fainardi, E. Assessment of brain tumors by magnetic resonance dynamic susceptibility contrast perfusion-weighted imaging and computed tomography perfusion: A comparison study. Radiol. Med. 2022, 127, 664–672. [Google Scholar] [CrossRef]
  126. Vicini, S.; Bortolotto, C.; Rengo, M.; Ballerini, D.; Bellini, D.; Carbone, I.; Preda, L.; Laghi, A.; Coppola, F.; Faggioni, L. A narrative review on current imaging applications of artificial intelligence and radiomics in oncology: Focus on the three most common cancers. Radiol. Med. 2022, 127, 819–836. [Google Scholar] [CrossRef]
  127. Petrillo, A.; Fusco, R.; Petrillo, M.; Granata, V.; Delrio, P.; Bianco, F.; Pecori, B.; Botti, G.; Tatangelo, F.; Caracò, C.; et al. Standardized Index of Shape (DCE-MRI) and Standardized Uptake Value (PET/CT): Two quantitative approaches to discriminate chemo-radiotherapy locally advanced rectal cancer responders under a functional profile. Oncotarget 2017, 8, 8143–8153. [Google Scholar] [CrossRef] [Green Version]
  128. Wang, G.X.; Kurra, V.; Gainor, J.F.; Sullivan, R.J.; Flaherty, K.T.; Lee, S.I.; Fintelmann, F.J. Immune Checkpoint Inhibitor Cancer Therapy: Spectrum of Imaging Findings. Radiographics 2017, 37, 2132–2144. [Google Scholar] [CrossRef] [Green Version]
  129. De Cecco, C.N.; Buffa, V.; Fedeli, S.; Vallone, A.; Ruopoli, R.; Luzietti, M.; Miele, V.; Rengo, M.; Maurizi Enrici, M.; Fina, P.; et al. Preliminary experience with abdominal dual-energy CT (DECT): True versus virtual nonenhanced images of the liver. Radiol. Med. 2010, 115, 1258–1266. [Google Scholar] [CrossRef]
  130. Tirkes, T.; Hollar, M.A.; Tann, M.; Kohli, M.D.; Akisik, F.; Sandrasegaran, K. Response criteria in oncologic imaging: Review of traditional and new criteria. Radiographics 2013, 33, 1323–1341. [Google Scholar] [CrossRef]
  131. Granata, V.; Fusco, R.; Setola, S.V.; Piccirillo, M.; Leongito, M.; Palaia, R.; Granata, F.; Lastoria, S.; Izzo, F.; Petrillo, A. Early radiological assessment of locally advanced pancreatic cancer treated with electrochemotherapy. World J. Gastroenterol. 2017, 23, 4767–4778. [Google Scholar] [CrossRef]
  132. di Giacomo, V.; Trinci, M.; van der Byl, G.; Catania, V.D.; Calisti, A.; Miele, V. Ultrasound in newborns and children suffering from non-traumatic acute abdominal pain: Imaging with clinical and surgical correlation. J. Ultrasound 2014, 18, 385–393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Eisenhauer, E.; Therasse, P.; Bogaerts, J.; Schwartz, L.H.; Sargent, D.; Ford, R.; Dancey, J.; Arbuck, S.; Gwyther, S.; Mooney, M.; et al. New response evaluation criteria in solid tumours: Revised RECIST guideline (version 1.1). Eur. J. Cancer 2009, 45, 228–247. [Google Scholar] [CrossRef] [PubMed]
  134. Granata, V.; Grassi, R.; Fusco, R.; Setola, S.V.; Palaia, R.; Belli, A.; Miele, V.; Brunese, L.; Grassi, R.; Petrillo, A.; et al. Assessment of Ablation Therapy in Pancreatic Cancer: The Radiologist’s Challenge. Front. Oncol. 2020, 10, 560952. [Google Scholar] [CrossRef] [PubMed]
  135. Chiti, G.; Grazzini, G.; Cozzi, D.; Danti, G.; Matteuzzi, B.; Granata, V.; Pradella, S.; Recchia, L.; Brunese, L.; Miele, V. Imaging of Pancreatic Neuroendocrine Neoplasms. Int. J. Environ. Res. Public Health 2021, 18, 8895. [Google Scholar] [CrossRef] [PubMed]
  136. Wolchok, J.D.; Hoos, A.; O’Day, S.; Weber, J.S.; Hamid, O.; Lebbé, C.; Maio, M.; Binder, M.; Bohnsack, O.; Nichol, G.; et al. Guidelines for the evaluation of immune therapy activity in solid tumors: Immune-related response criteria. Clin. Cancer Res. 2009, 15, 7412–7420. [Google Scholar] [CrossRef] [Green Version]
  137. 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]
  138. Nishino, M.; Giobbie-Hurder, A.; Gargano, M.; Suda, M.; Ramaiya, N.H.; Hodi, F.S. Developing a common language for tumor response to immunotherapy: Immune-related response criteria using unidimensional measurements. Clin. Cancer Res. 2013, 19, 3936–3943. [Google Scholar] [CrossRef] [Green Version]
  139. Seymour, L.; Bogaerts, J.; Perrone, A.; Ford, R.; Schwartz, L.; Mandrekar, S.; Lin, N.; Litière, S.; Dancey, J.; Chen, A.; et al. iRECIST: Guidelines for response criteria for use in trials testing immunotherapeutics. Lancet Oncol. 2017, 18, e143–e152. [Google Scholar] [CrossRef] [Green Version]
  140. Regine, G.; Stasolla, A.; Miele, V. Multidetector computed tomography of the renal arteries in vascular emergencies. Eur. J. Radiol. 2007, 64, 83–91. [Google Scholar] [CrossRef]
  141. Choi, J.H.; Ahn, M.J.; Rhim, H.C.; Kim, J.W.; Lee, G.H.; Lee, Y.Y.; Kim, I.S. Comparison of WHO and RECIST criteria for response in metastatic colorectal carcinoma. Cancer Res. Treat. 2005, 37, 290–293. [Google Scholar] [CrossRef] [Green Version]
  142. Regine, G.; Atzori, M.; Miele, V.; Buffa, V.; Galluzzo, M.; Luzietti, M.; Adami, L. Second-generation sonographic contrast agents in the evaluation of renal trauma. Radiol. Med. 2007, 112, 581–587, (In English and Italian). [Google Scholar] [CrossRef]
  143. Choi, H.; Charnsangavej, C.; Faria, S.C.; Macapinlac, H.A.; Burgess, M.A.; Patel, S.R.; Chen, L.L.; Podoloff, D.A.; Benjamin, R.S. Correlation of computed tomography and positron emission tomography in patients with metastatic gastrointestinal stromal tumor treated at a single institution with imatinib mesylate: Proposal of new computed tomography response criteria. J. Clin. Oncol. 2007, 25, 1753–1759. [Google Scholar] [CrossRef]
  144. Park, S.H.; Kim, Y.S.; Choi, J. Dosimetric analysis of the effects of a temporary tissue expander on the radiotherapy technique. Radiol. Med. 2021, 126, 437–444. [Google Scholar] [CrossRef]
  145. Bozkurt, M.; Eldem, G.; Bozbulut, U.B.; Bozkurt, M.F.; Kılıçkap, S.; Peynircioğlu, B.; Çil, B.; Lay Ergün, E.; Volkan-Salanci, B. Factors affecting the response to Y-90 microsphere therapy in the cholangiocarcinoma patients. Radiol. Med. 2021, 126, 323–333. [Google Scholar] [CrossRef] [PubMed]
  146. Gregory, J.; Dioguardi Burgio, M.; Corrias, G.; Vilgrain, V.; Ronot, M. Evaluation of liver tumour response by imaging. JHEP Rep. 2020, 2, 100100. [Google Scholar] [CrossRef] [PubMed]
  147. Wahl, R.L.; Jacene, H.; Kasamon, Y.; Lodge, M.A. From RECIST to PERCIST: Evolving Considerations for PET response criteria in solid tumors. J. Nucl. Med. 2009, 50 (Suppl. S1), 122S–150S. [Google Scholar] [CrossRef] [Green Version]
  148. Skougaard, K.; Nielsen, D.; Jensen, B.V.; Hendel, H.W. Comparison of EORTC criteria and PERCIST for PET/CT response evaluation of patients with metastatic colorectal cancer treated with irinotecan and cetuximab. J. Nucl. Med. 2013, 54, 1026–1031. [Google Scholar] [CrossRef] [Green Version]
  149. Kim, N.; Lee, E.S.; Won, S.E.; Yang, M.; Lee, A.J.; Shin, Y.; Ko, Y.; Pyo, J.; Park, H.J.; Kim, K.W. Evolution of Radiological Treatment Response Assessments for Cancer Immunotherapy: From iRECIST to Radiomics and Artificial Intelligence. Korean J. Radiol. 2022, 23, 1089. [Google Scholar] [CrossRef]
  150. Ter Maat, L.S.; van Duin, I.A.J.; Elias, S.G.; van Diest, P.J.; Pluim, J.P.W.; Verhoeff, J.J.C.; de Jong, P.A.; Leiner, T.; Veta, M.; Suijkerbuijk, K.P.M. Imaging to predict checkpoint inhibitor outcomes in cancer. A systematic review. Eur. J. Cancer 2022, 175, 60–76. [Google Scholar] [CrossRef] [PubMed]
  151. Bracco, S.; Zanoni, M.; Casseri, T.; Castellano, D.; Cioni, S.; Vallone, I.M.; Gennari, P.; Mazzei, M.A.; Romano, D.G.; Piano, M.; et al. Endovascular treatment of acute ischemic stroke due to tandem lesions of the anterior cerebral circulation: A multicentric Italian observational study. Radiol. Med. 2021, 126, 804–817. [Google Scholar] [CrossRef]
  152. Nardone, V.; Boldrini, L.; Salvestrini, V.; Greco, C.; Petrianni, G.M.; Desideri, I.; De Felice, F. Are you planning to be a radiation oncologist? A survey by the young group of the Italian Association of Radiotherapy and Clinical Oncology (yAIRO). Radiol. Med. 2022; Epub ahead of print. [Google Scholar] [CrossRef]
  153. Giurazza, F.; Contegiacomo, A.; Calandri, M.; Mosconi, C.; Modestino, F.; Corvino, F.; Scrofani, A.R.; Marra, P.; Coniglio, G.; Failla, G.; et al. IVC filter retrieval: A multicenter proposal of two score systems to predict application of complex technique and procedural outcome. Radiol. Med. 2021, 126, 1007–1016. [Google Scholar] [CrossRef] [PubMed]
  154. Barile, A. Some thoughts and greetings from the new Editor-in-Chief. Radiol. Med. 2021, 126, 3–4. [Google Scholar] [CrossRef]
  155. Cappabianca, S.; Granata, V.; Di Grezia, G.; Mandato, Y.; Reginelli, A.; Di Mizio, V.; Grassi, R.; Rotondo, A. The role of nasoenteric intubation in the MR study of patients with Crohn’s disease: Our experience and literature review. Radiol. Med. 2011, 116, 389–406. [Google Scholar] [CrossRef]
  156. De Filippo, M.; Puglisi, S.; D’Amuri, F.; Gentili, F.; Paladini, I.; Carrafiello, G.; Maestroni, U.; Del Rio, P.; Ziglioli, F.; Pagnini, F. CT-guided percutaneous drainage of abdominopelvic collections: A pictorial essay. Radiol. Med. 2021, 126, 1561–1570. [Google Scholar] [CrossRef] [PubMed]
  157. De Felice, F.; Boldrini, L.; Greco, C.; Nardone, V.; Salvestrini, V.; Desideri, I. ESTRO vision 2030: The young Italian Association of Radiotherapy and Clinical Oncology (yAIRO) commitment statement. Radiol. Med. 2021, 126, 1374–1376. [Google Scholar] [CrossRef]
  158. Pecoraro, M.; Cipollari, S.; Marchitelli, L.; Messina, E.; Del Monte, M.; Galea, N.; Ciardi, M.R.; Francone, M.; Catalano, C.; Panebianco, V. Cross-sectional analysis of follow-up chest MRI and chest CT scans in patients previously affected by COVID-19. Radiol. Med. 2021, 126, 1273–1281. [Google Scholar] [CrossRef]
  159. Delaunay, M.; Cadranel, J.; Lusque, A.; Meyer, N.; Gounant, V.; Moro-Sibilot, D.; Michot, J.M.; Raimbourg, J.; Girard, N.; Guisier, F.; et al. Immune-checkpoint inhibitors associated with interstitial lung disease in cancer patients. Eur. Respir. J. 2017, 50, 1700050. [Google Scholar] [CrossRef]
  160. Piccolo, C.L.; Galluzzo, M.; Ianniello, S.; Trinci, M.; Russo, A.; Rossi, E.; Zeccolini, M.; Laporta, A.; Guglielmi, G.; Miele, V. Pediatric musculoskeletal injuries: Role of ultrasound and magnetic resonance imaging. Musculoskelet. Surg. 2017, 101 (Suppl. S1), 85–102. [Google Scholar] [CrossRef]
  161. Conte, P.; Ascierto, P.A.; Patelli, G.; Danesi, R.; Vanzulli, A.; Sandomenico, F.; Tarsia, P.; Cattelan, A.; Comes, A.; De Laurentiis, M.; et al. Drug-induced interstitial lung disease during cancer therapies: Expert opinion on diagnosis and treatment. ESMO Open 2022, 7, 100404. [Google Scholar] [CrossRef]
  162. Luke, J.J.; Rutkowski, P.; Queirolo, P.; Del Vecchio, M.; Mackiewicz, J.; Chiarion-Sileni, V.; de la Cruz Merino, L.; Khattak, M.A.; Schadendorf, D.; Long, G.V.; et al. Pembrolizumab versus placebo as adjuvant therapy in completely resected stage IIB or IIC melanoma (KEYNOTE-716): A randomised, double-blind, phase 3 trial. Lancet 2022, 399, 1718–1729. [Google Scholar] [CrossRef]
  163. Goldman, J.D.; Ascierto, P.A. Perspectives on COVID-19 and cancer immunotherapy: A review series. J. Immunother. Cancer 2021, 9, e002489. [Google Scholar] [CrossRef]
  164. Kwak, J.J.; Tirumani, S.H.; Van den Abbeele, A.D.; Koo, P.J.; Jacene, H.A. Cancer immunotherapy: Imaging assessment of novel treatment response patterns and immune-related adverse events. Radiographics 2015, 35, 424–437. [Google Scholar] [CrossRef] [Green Version]
  165. Assadsangabi, R.; Babaei, R.; Songco, C.; Ivanovic, V.; Bobinski, M.; Chen, Y.J.; Nabavizadeh, S.A. Multimodality oncologic evaluation of superficial neck and facial lymph nodes. Radiol. Med. 2021, 126, 1074–1084. [Google Scholar] [CrossRef]
  166. Petralia, G.; Zugni, F.; Summers, P.E.; Colombo, A.; Pricolo, P.; Grazioli, L.; Colagrande, S.; Giovagnoni, A.; Padhani, A.R.; Italian Working Group on Magnetic Resonance. Whole-body magnetic resonance imaging (WB-MRI) for cancer screening: Recommendations for use. Radiol. Med. 2021, 126, 1434–1450. [Google Scholar] [CrossRef]
  167. Tirumani, S.H.; Ramaiya, N.H.; Keraliya, A.; Bailey, N.D.; Ott, P.A.; Hodi, F.S.; Nishino, M. Radiographic profiling of immune-related adverse events in advanced melanoma patients treated with ipilimumab. Cancer Immunol. Res. 2015, 3, 1185–1192. [Google Scholar] [CrossRef] [Green Version]
  168. Palmucci, S.; Roccasalva, F.; Puglisi, S.; Torrisi, S.E.; Vindigni, V.; Mauro, L.A.; Ettorre, G.C.; Piccoli, M.; Vancheri, C. Clinical and radiological features of idiopathic interstitial pneumonias (IIPs): A pictorial review. Insights Imaging 2014, 5, 347–364. [Google Scholar] [CrossRef] [Green Version]
  169. Khoja, L.; Day, D.; Chen, T.W.-W.; Siu, L.L.; Hansen, A.R. Tumour-and class specific patterns of immune-related adverse events of immune checkpoint inhibitors: A systematic review. Ann. Oncol. 2017, 28, 2377–2385. [Google Scholar] [CrossRef]
  170. Suresh, K.; Voong, K.R.; Shankar, B.; Forde, P.M.; Ettinger, D.S.; Marrone, K.A.; Kelly, R.J.; Hann, C.L.; Levy, B.; Feliciano, J.L.; et al. Pneumonitis in Non–Small Cell Lung Cancer Patients Receiving Immune Checkpoint Immunotherapy: Incidence and Risk Factors. J. Thorac. Oncol. 2018, 13, 1930–1939. [Google Scholar] [CrossRef] [Green Version]
  171. Tay, R.Y.; Califano, R. Checkpoint Inhibitor Pneumonitis Real World Incidence and Risk. J. Thorac. Oncol. 2018, 13, 1812–1814. [Google Scholar] [CrossRef]
  172. Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.J.; Cowey, L.; Lao, C.D.; Schadendorf, D.; Dummer, R.; Smylie, M.; Rutkowwski, P.; et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N. Engl. J. Med. 2015, 373, 23–34. [Google Scholar] [CrossRef] [Green Version]
  173. D’Agostino, V.; Caranci, F.; Negro, A.; Piscitelli, V.; Tuccillo, B.; Fasano, F.; Sirabella, G.; Marano, I.; Granata, V.; Grassi, R.; et al. A Rare Case of Cerebral Venous Thrombosis and Disseminated Intravascular Coagulation Temporally Associated to the COVID-19 Vaccine Administration. J. Pers. Med. 2021, 11, 285. [Google Scholar] [CrossRef]
  174. Agostini, A.; Borgheresi, A.; Carotti, M.; Ottaviani, L.; Badaloni, M.; Floridi, C.; Giovagnoni, A. Third-generation iterative reconstruction on a dual-source, high-pitch, low-dose chest CT protocol with tin filter for spectral shaping at 100 kV: A study on a small series of COVID-19 patients. Radiol. Med. 2021, 126, 388–398. [Google Scholar] [CrossRef]
  175. Palmisano, A.; Scotti, G.M.; Ippolito, D.; Morelli, M.J.; Vignale, D.; Gandola, D.; Sironi, S.; De Cobelli, F.; Ferrante, L.; Spessot, M.; et al. Chest CT in the emergency department for suspected COVID-19 pneumonia. Radiol. Med. 2021, 126, 498–502. [Google Scholar] [CrossRef]
  176. Lombardi, A.F.; Afsahi, A.M.; Gupta, A.; Gholamrezanezhad, A. Severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS), influenza, and COVID-19, beyond the lungs: A review article. Radiol. Med. 2021, 126, 561–569. [Google Scholar] [CrossRef]
  177. Gabelloni, M.; Faggioni, L.; Cioni, D.; Mendola, V.; Falaschi, Z.; Coppola, S.; Corradi, F.; Isirdi, A.; Brandi, N.; Coppola, F.; et al. Extracorporeal membrane oxygenation (ECMO) in COVID-19 patients: A pocket guide for radiologists. Radiol. Med. 2022, 13, 369–382. [Google Scholar] [CrossRef]
  178. Grassi, R.; Cappabianca, S.; Urraro, F.; Feragalli, B.; Montanelli, A.; Patelli, G.; Granata, V.; Giacobbe, G.; Russo, G.M.; Grillo, A.; et al. Chest CT Computerized Aided Quantification of PNEUMONIA Lesions in COVID-19 Infection: A Comparison among Three Commercial Software. Int. J. Environ. Res. Public Health 2020, 17, 6914. [Google Scholar] [CrossRef]
  179. Fusco, R.; Grassi, R.; Granata, V.; Setola, S.V.; Grassi, F.; Cozzi, D.; Pecori, B.; Izzo, F.; Petrillo, A. Artificial Intelligence and COVID-19 Using Chest CT Scan and Chest X-ray Images: Machine Learning and Deep Learning Approaches for Diagnosis and Treatment. J. Pers. Med. 2021, 11, 993. [Google Scholar] [CrossRef]
  180. Özel, M.; Aslan, A.; Araç, S. Use of the COVID-19 Reporting and Data System (CO-RADS) classification and chest computed tomography involvement score (CT-IS) in COVID-19 pneumonia. Radiol. Med. 2021, 126, 679–687. [Google Scholar] [CrossRef]
  181. Ippolito, D.; Giandola, T.; Maino, C.; Pecorelli, A.; Capodaglio, C.; Ragusi, M.; Porta, M.; Gandola, D.; Masetto, A.; Drago, S.; et al. Acute pulmonary embolism in hospitalized patients with SARS-CoV-2-related pneumonia: Multicentric experience from Italian endemic area. Radiol. Med. 2021, 126, 669–678. [Google Scholar] [CrossRef]
  182. Moroni, C.; Cozzi, D.; Albanesi, M.; Cavigli, E.; Bindi, A.; Luvarà, S.; Busoni, S.; Mazzoni, L.N.; Grifoni, S.; Nazerian, P.; et al. Chest X-ray in the emergency department during COVID-19 pandemic descending phase in Italy: Correlation with patients’ outcome. Radiol. Med. 2021, 126, 661–668. [Google Scholar] [CrossRef]
  183. Cereser, L.; Girometti, R.; Da Re, J.; Marchesini, F.; Como, G.; Zuiani, C. Inter-reader agreement of high-resolution computed tomography findings in patients with COVID-19 pneumonia: A multi-reader study. Radiol. Med. 2021, 126, 577–584. [Google Scholar] [CrossRef] [PubMed]
  184. Rawashdeh, M.A.; Saade, C. Radiation dose reduction considerations and imaging patterns of ground glass opacities in coronavirus: Risk of over exposure in computed tomography. Radiol. Med. 2021, 126, 380–387. [Google Scholar] [CrossRef] [PubMed]
  185. Granata, V.; Ianniello, S.; Fusco, R.; Urraro, F.; Pupo, D.; Magliocchetti, S.; Albarello, F.; Campioni, P.; Cristofaro, M.; Di Stefano, F.; et al. Quantitative Analysis of Residual COVID-19 Lung CT Features: Consistency among Two Commercial Software. J. Pers. Med. 2021, 11, 1103. [Google Scholar] [CrossRef]
  186. Granata, V.; Fusco, R.; Bicchierai, G.; Cozzi, D.; Grazzini, G.; Danti, G.; De Muzio, F.; Maggialetti, N.; Smorchkova, O.; D’Elia, M.; et al. Diagnostic protocols in oncology: Workup and treatment planning: Part 1: The optimitation of CT protocol. Eur. Rev. Med. Pharmacol. Sci. 2021, 25, 6972–6994. [Google Scholar]
  187. Fusco, R.; Granata, V.; Petrillo, A. Introduction to Special Issue of Radiology and Imaging of Cancer. Cancers 2020, 12, 2665. [Google Scholar] [CrossRef] [PubMed]
  188. Scapicchio, C.; Gabelloni, M.; Barucci, A.; Cioni, D.; Saba, L.; Neri, E. A deep look into radiomics. Radiol. Med. 2021, 126, 1296–1311. [Google Scholar] [CrossRef]
  189. Neri, E.; Coppola, F.; Larici, A.R.; Sverzellati, N.; Mazzei, M.A.; Sacco, P.; Dalpiaz, G.; Feragalli, B.; Miele, V.; Grassi, R. Structured reporting of chest CT in COVID-19 pneumonia: A consensus proposal. Insights Imaging 2020, 11, 92. [Google Scholar] [CrossRef]
  190. Cellina, M.; Pirovano, M.; Ciocca, M.; Gibelli, D.; Floridi, C.; Oliva, G. Radiomic analysis of the optic nerve at the first episode of acute optic neuritis: An indicator of optic nerve pathology and a predictor of visual recovery? Radiol. Med. 2021, 126, 698–706. [Google Scholar] [CrossRef]
  191. Santone, A.; Brunese, M.C.; Donnarumma, F.; Guerriero, P.; Mercaldo, F.; Reginelli, A.; Miele, V.; Giovagnoni, A.; Brunese, L. Radiomic features for prostate cancer grade detection through formal verification. Radiol. Med. 2021, 126, 688–697. [Google Scholar] [CrossRef]
  192. Granata, V.; Fusco, R.; Barretta, M.L.; Picone, C.; Avallone, A.; Belli, A.; Patrone, R.; Ferrante, M.; Cozzi, D.; Grassi, R.; et al. Radiomics in hepatic metastasis by colorectal cancer. Infect. Agents Cancer 2021, 16, 39. [Google Scholar] [CrossRef]
  193. Granata, V.; Grassi, R.; Fusco, R.; Galdiero, R.; Setola, S.V.; Palaia, R.; Belli, A.; Silvestro, L.; Cozzi, D.; Brunese, L.; et al. Pancreatic cancer detection and characterization: State of the art and radiomics. Eur. Rev. Med. Pharmacol. Sci. 2021, 25, 3684–3699. [Google Scholar] [CrossRef] [PubMed]
  194. Agazzi, G.M.; Ravanelli, M.; Roca, E.; Medicina, D.; Balzarini, P.; Pessina, C.; Vermi, W.; Berruti, A.; Maroldi, R.; Farina, D. CT texture analysis for prediction of EGFR mutational status and ALK rearrangement in patients with non-small cell lung cancer. Radiol. Med. 2021, 126, 786–794. [Google Scholar] [CrossRef] [PubMed]
  195. Agüloğlu, N.; Aksu, A.; Akyol, M.; Katgı, N.; Doksöz, T.Ç. Importance of Pretreatment 18F-FDG PET/CT Texture Analysis in Predicting EGFR and ALK Mutation in Patients with Non-Small Cell Lung Cancer. Nuklearmedizin 2022, 61, 433–439. [Google Scholar] [CrossRef] [PubMed]
  196. Mayerhoefer, M.E.; Materka, A.; Langs, G.; Häggström, I.; Szczypiński, P.; Gibbs, P.; Cook, G. Introduction to Radiomics. J. Nucl. Med. 2020, 61, 488–495. [Google Scholar] [CrossRef]
  197. Yip, S.S.; Aerts, H.J. Applications and limitations of radiomics. Phys. Med. Biol. 2016, 61, R150–R166. [Google Scholar] [CrossRef] [Green Version]
  198. Granata, V.; Bicchierai, G.; Fusco, R.; Cozzi, D.; Grazzini, G.; Danti, G.; De Muzio, F.; Maggialetti, N.; Smorchkova, O.; D’Elia, M.; et al. Diagnostic protocols in oncology: Workup and treatment planning. Part 2: Abbreviated MR protocol. Eur. Rev. Med. Pharmacol. Sci. 2021, 25, 6499–6528. [Google Scholar] [CrossRef]
  199. Granata, V.; Fusco, R.; Catalano, O.; Setola, S.V.; de Lutio di Castelguidone, E.; Piccirillo, M.; Palaia, R.; Grassi, R.; Granata, F.; Izzo, F.; et al. Multidetector computer tomography in the pancreatic adenocarcinoma assessment: An update. Infect. Agents Cancer 2016, 11, 57. [Google Scholar] [CrossRef] [Green Version]
  200. Granata, V.; Fusco, R.; Setola, S.V.; Galdiero, R.; Picone, C.; Izzo, F.; D’Aniello, R.; Miele, V.; Grassi, R.; Grassi, R.; et al. Lymphadenopathy after BNT162b2 Covid-19 Vaccine: Preliminary Ultrasound Findings. Biology 2021, 10, 214. [Google Scholar] [CrossRef]
  201. Binczyk, F.; Prazuch, W.; Bozek, P.; Polanska, J. Radiomics and artificial intelligence in lung cancer screening. Transl. Lung Cancer Res. 2021, 10, 1186–1199. [Google Scholar] [CrossRef]
  202. Fiz, F.; Viganò, L.; Gennaro, N.; Costa, G.; La Bella, L.; Boichuk, A.; Cavinato, L.; Sollini, M.; Politi, L.S.; Chiti, A.; et al. Radiomics of Liver Metastases: A Systematic Review. Cancers 2020, 12, 2881. [Google Scholar] [CrossRef]
  203. Granata, V.; Fusco, R.; Avallone, A.; Catalano, O.; Piccirillo, M.; Palaia, R.; Nasti, G.; Petrillo, A.; Izzo, F. A radiologist’s point of view in the presurgical and intraoperative setting of colorectal liver metastases. Future Oncol. 2018, 14, 2189–2206. [Google Scholar] [CrossRef] [PubMed]
  204. Benedetti, G.; Mori, M.; Panzeri, M.M.; Barbera, M.; Palumbo, D.; Sini, C.; Muffatti, F.; Andreasi, V.; Steidler, S.; Doglioni, C.; et al. CT-derived radiomic features to discriminate histologic characteristics of pancreatic neuroendocrine tumors. Radiol. Med. 2021, 126, 745–760. [Google Scholar] [CrossRef] [PubMed]
  205. Gurgitano, M.; Angileri, S.A.; Rodà, G.M.; Liguori, A.; Pandolfi, M.; Ierardi, A.M.; Wood, B.J.; Carrafiello, G. Interventional Radiology ex-machina: Impact of Artificial Intelligence on practice. Radiol. Med. 2021, 126, 998–1006. [Google Scholar] [CrossRef] [PubMed]
  206. Granata, V.; Fusco, R.; Avallone, A.; De Stefano, A.; Ottaiano, A.; Sbordone, C.; Brunese, L.; Izzo, F.; Petrillo, A. Radiomics-Derived Data by Contrast Enhanced Magnetic Resonance in RAS Mutations Detection in Colorectal Liver Metastases. Cancers 2021, 13, 453. [Google Scholar] [CrossRef] [PubMed]
  207. Granata, V.; Fusco, R.; Costa, M.; Picone, C.; Cozzi, D.; Moroni, C.; La Casella, G.V.; Montanino, A.; Monti, R.; Mazzoni, F.; et al. Preliminary Report on Computed Tomography Radiomics Features as Biomarkers to Immunotherapy Selection in Lung Adenocarcinoma Patients. Cancers 2021, 13, 3992. [Google Scholar] [CrossRef]
  208. Taha, B.; Boley, D.; Sun, J.; Chen, C.C. State of Radiomics in Glioblastoma. Neurosurgery 2021, 89, 177–184. [Google Scholar] [CrossRef]
  209. Yu, Y.; He, Z.; Ouyang, J.; Tan, Y.; Chen, Y.; Gu, Y.; Mao, L.; Ren, W.; Wang, J.; Lin, L.; et al. Magnetic resonance imaging radiomics predicts preoperative axillary lymph node metastasis to support surgical decisions and is associated with tumor microenvironment in invasive breast cancer: A machine learning, multicenter study. eBioMedicine 2021, 69, 103460. [Google Scholar] [CrossRef]
  210. Taha, B.; Boley, D.; Sun, J.; Chen, C. Potential and limitations of radiomics in neuro-oncology. J. Clin. Neurosci. 2021, 90, 206–211. [Google Scholar] [CrossRef]
  211. Frix, A.N.; Cousin, F.; Refaee, T.; Bottari, F.; Vaidyanathan, A.; Desir, C.; Vos, W.; Walsh, S.; Occhipinti, M.; Lovinfosse, P.; et al. Radiomics in Lung Diseases Imaging: State-of-the-Art for Clinicians. J. Pers. Med. 2021, 11, 602. [Google Scholar] [CrossRef]
  212. Fusco, R.; Sansone, M.; Filice, S.; Granata, V.; Catalano, O.; Amato, D.M.; Di Bonito, M.; D’Aiuto, M.; Capasso, I.; Rinaldo, M.; et al. Integration of DCE-MRI and DW-MRI Quantitative Parameters for Breast Lesion Classification. Biomed. Res. Int. 2015, 2015, 237863. [Google Scholar] [CrossRef]
  213. Fusco, R.; Granata, V.; Grazzini, G.; Pradella, S.; Borgheresi, A.; Bruno, A.; Palumbo, P.; Bruno, F.; Grassi, R.; Giovagnoni, A.; et al. Radiomics in medical imaging: Pitfalls and challenges in clinical management. Jpn. J. Radiol. 2022, 40, 919–929. [Google Scholar] [CrossRef]
  214. Nakamura, Y.; Higaki, T.; Honda, Y.; Tatsugami, F.; Tani, C.; Fukumoto, W.; Narita, K.; Kondo, S.; Akagi, M.; Awai, K. Advanced CT techniques for assessing hepatocellular carcinoma. Radiol. Med. 2021, 126, 925–935. [Google Scholar] [CrossRef]
  215. Chianca, V.; Albano, D.; Messina, C.; Vincenzo, G.; Rizzo, S.; Del Grande, F.; Sconfienza, L.M. An update in musculoskeletal tumors: From quantitative imaging to radiomics. Radiol. Med. 2021, 126, 1095–1105. [Google Scholar] [CrossRef] [PubMed]
  216. Halefoglu, A.M.; Ozagari, A.A. Tumor grade estimation of clear cell and papillary renal cell carcinoma using contrast-enhanced MDCT and FSE T2 weighted MR imaging: Radiology-pathology correlation. Radiol. Med. 2021, 126, 1139–1148. [Google Scholar] [CrossRef] [PubMed]
  217. Granata, V.; Fusco, R.; Risi, C.; Ottaiano, A.; Avallone, A.; De Stefano, A.; Grimm, R.; Grassi, R.; Brunese, L.; Izzo, F.; et al. Diffusion-Weighted MRI and Diffusion Kurtosis Imaging to Detect RAS Mutation in Colorectal Liver Metastasis. Cancers 2020, 12, 2420. [Google Scholar] [CrossRef]
  218. Granata, V.; Fusco, R.; Setola, S.V.; Simonetti, I.; Cozzi, D.; Grazzini, G.; Grassi, F.; Belli, A.; Miele, V.; Izzo, F.; et al. An update on radiomics techniques in primary liver cancers. Infect. Agents Cancer 2022, 17, 6. [Google Scholar] [CrossRef] [PubMed]
  219. Avanzo, M.; Stancanello, J.; El Naqa, I. Beyond imaging: The promise of radiomics. Phys. Med. 2017, 38, 122–139. [Google Scholar] [CrossRef] [PubMed]
  220. Fornacon-Wood, I.; Faivre-Finn, C.; O’Connor, J.P.B.; Price, G.J. Radiomics as a personalized medicine tool in lung cancer: Separating the hope from the hype. Lung Cancer 2020, 146, 197–208. [Google Scholar] [CrossRef]
  221. Bogowicz, M.; Vuong, D.; Huellner, M.W.; Pavic, M.; Andratschke, N.; Gabrys, H.S.; Guckenberger, M.; Tanadini-Lang, S. CT radiomics and PET radiomics: Ready for clinical implementation? Q. J. Nucl. Med. Mol. Imaging 2019, 63, 355–370. [Google Scholar] [CrossRef]
  222. Granata, V.; Fusco, R.; Sansone, M.; Grassi, R.; Maio, F.; Palaia, R.; Tatangelo, F.; Botti, G.; Grimm, R.; Curley, S.; et al. Magnetic resonance imaging in the assessment of pancreatic cancer with quantitative parameter extraction by means of dynamic contrast-enhanced magnetic resonance imaging, diffusion kurtosis imaging and intravoxel incoherent motion diffusion-weighted imaging. Ther. Adv. Gastroenterol. 2020, 13, 1756284819885052. [Google Scholar] [CrossRef]
  223. Granata, V.; Fusco, R.; Setola, S.V.; Picone, C.; Vallone, P.; Belli, A.; Incollingo, P.; Albino, V.; Tatangelo, F.; Izzo, F.; et al. Microvascular invasion and grading in hepatocellular carcinoma: Correlation with major and ancillary features according to LIRADS. Abdom. Radiol. 2019, 44, 2788–2800. [Google Scholar] [CrossRef]
  224. Granata, V.; Fusco, R.; Setola, S.V.; De Muzio, F.; Dell’ Aversana, F.; Cutolo, C.; Faggioni, L.; Miele, V.; Izzo, F.; Petrillo, A. CT-Based Radiomics Analysis to Predict Histopathological Outcomes Following Liver Resection in Colorectal Liver Metastases. Cancers 2022, 14, 1648. [Google Scholar] [CrossRef]
  225. Sun, J.; Li, H.; Gao, J.; Li, J.; Li, M.; Zhou, Z.; Peng, Y. Performance evaluation of a deep learning image reconstruction (DLIR) algorithm in “double low” chest CTA in children: A feasibility study. Radiol. Med. 2021, 126, 1181–1188. [Google Scholar] [CrossRef]
  226. Granata, V.; Faggioni, L.; Grassi, R.; Fusco, R.; Reginelli, A.; Rega, D.; Maggialetti, N.; Buccicardi, D.; Frittoli, B.; Rengo, M.; et al. Structured reporting of computed tomography in the staging of colon cancer: A Delphi consensus proposal. Radiol. Med. 2022, 127, 21–29. [Google Scholar] [CrossRef] [PubMed]
  227. Granata, V.; Fusco, R.; De Muzio, F.; Cutolo, C.; Setola, S.V.; Dell’Aversana, F.; Ottaiano, A.; Nasti, G.; Grassi, R.; Pilone, V.; et al. EOB-MR Based Radiomics Analysis to Assess Clinical Outcomes following Liver Resection in Colorectal Liver Metastases. Cancers 2022, 14, 1239. [Google Scholar] [CrossRef]
  228. Arimura, H.; Soufi, M.; Kamezawa, H.; Ninomiya, K.; Yamada, M. Radiomics with artificial intelligence for precision medicine in radiation therapy. J. Radiat. Res. 2019, 60, 150–157. [Google Scholar] [CrossRef] [PubMed]
  229. Fusco, R.; Granata, V.; Mazzei, M.A.; Meglio, N.D.; Roscio, D.D.; Moroni, C.; Monti, R.; Cappabianca, C.; Picone, C.; Neri, E.; et al. Quantitative imaging decision support (QIDSTM) tool consistency evaluation and radiomic analysis by means of 594 metrics in lung carcinoma on chest CT scan. Cancer Control 2021, 28, 1073274820985786. [Google Scholar] [CrossRef] [PubMed]
  230. Zanfardino, M.; Franzese, M.; Pane, K.; Cavaliere, C.; Monti, S.; Esposito, G.; Salvatore, M.; Aiello, M. Bringing radiomics into a multi-omics framework for a comprehensive genotype-phenotype characterization of oncological diseases. J. Transl. Med. 2019, 17, 337. [Google Scholar] [CrossRef] [PubMed]
  231. Izzo, F.; Piccirillo, M.; Albino, V.; Palaia, R.; Belli, A.; Granata, V.; Setola, S.; Fusco, R.; Petrillo, A.; Orlando, R.; et al. Prospective screening increases the detection of potentially curable hepatocellular carcinoma: Results in 8,900 high-risk patients. HPB 2013, 15, 985–990. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  232. Granata, V.; Fusco, R.; De Muzio, F.; Cutolo, C.; Setola, S.V.; Dell’ Aversana, F.; Ottaiano, A.; Avallone, A.; Nasti, G.; Grassi, F.; et al. Contrast MR-Based Radiomics and Machine Learning Analysis to Assess Clinical Outcomes following Liver Resection in Colorectal Liver Metastases: A Preliminary Study. Cancers 2022, 14, 1110. [Google Scholar] [CrossRef]
  233. Liu, J.; Wang, C.; Guo, W.; Zeng, P.; Liu, Y.; Lang, N.; Yuan, H. A preliminary study using spinal MRI-based radiomics to predict high-risk cytogenetic abnormalities in multiple myeloma. Radiol. Med. 2021, 126, 1226–1235. [Google Scholar] [CrossRef]
  234. Qin, H.; Que, Q.; Lin, P.; Li, X.; Wang, X.R.; He, Y.; Chen, J.Q.; Yang, H. Magnetic resonance imaging (MRI) radiomics of papillary thyroid cancer (PTC): A comparison of predictive performance of multiple classifiers modeling to identify cervical lymph node metastases before surgery. Radiol. Med. 2021, 126, 1312–1327. [Google Scholar] [CrossRef]
  235. Fusco, R.; Di Bernardo, E.; Piccirillo, A.; Rubulotta, M.R.; Petrosino, T.; Barretta, M.L.; Mattace Raso, M.; Vallone, P.; Raiano, C.; Di Giacomo, R.; et al. Radiomic and Artificial Intelligence Analysis with Textural Metrics Extracted by Contrast-Enhanced Mammography and Dynamic Contrast Magnetic Resonance Imaging to Detect Breast Malignant Lesions. Curr. Oncol. 2022, 29, 159. [Google Scholar] [CrossRef]
  236. Neri, E.; Granata, V.; Montemezzi, S.; Belli, P.; Bernardi, D.; Brancato, B.; Caumo, F.; Calabrese, M.; Coppola, F.; Cossu, E.; et al. Structured reporting of x-ray mammography in the first diagnosis of breast cancer: A Delphi consensus proposal. Radiol. Med. 2022, 127, 471–483. [Google Scholar] [CrossRef] [PubMed]
  237. Granata, V.; Fusco, R.; De Muzio, F.; Cutolo, C.; Mattace Raso, M.; Gabelloni, M.; Avallone, A.; Ottaiano, A.; Tatangelo, F.; Brunese, M.C.; et al. Radiomics and Machine Learning Analysis Based on Magnetic Resonance Imaging in the Assessment of Colorectal Liver Metastases Growth Pattern. Diagnostics 2022, 12, 1115. [Google Scholar] [CrossRef] [PubMed]
  238. Brunese, L.; Brunese, M.C.; Carbone, M.; Ciccone, V.; Mercaldo, F.; Santone, A. Automatic PI-RADS assignment by means of formal methods. Radiol. Med. 2022, 127, 83–89. [Google Scholar] [CrossRef] [PubMed]
  239. Bellardita, L.; Colciago, R.R.; Frasca, S.; De Santis, M.C.; Gay, S.; Palorini, F.; La Rocca, E.; Valdagni, R.; Rancati, T.; Lozza, L. Breast cancer patient perspective on opportunities and challenges of a genetic test aimed to predict radio-induced side effects before treatment: Analysis of the Italian branch of the REQUITE project. Radiol. Med. 2021, 126, 1366–1373. [Google Scholar] [CrossRef]
  240. Caruso, D.; Pucciarelli, F.; Zerunian, M.; Ganeshan, B.; De Santis, D.; Polici, M.; Rucci, C.; Polidori, T.; Guido, G.; Bracci, B.; et al. Chest CT texture-based radiomics analysis in differentiating COVID-19 from other interstitial pneumonia. Radiol. Med. 2021, 126, 1415–1424. [Google Scholar] [CrossRef]
  241. Karmazanovsky, G.; Gruzdev, I.; Tikhonova, V.; Kondratyev, E.; Revishvili, A. Computed tomography-based radiomics approach in pancreatic tumors characterization. Radiol. Med. 2021, 126, 1388–1395. [Google Scholar] [CrossRef]
  242. Danti, G.; Flammia, F.; Matteuzzi, B.; Cozzi, D.; Berti, V.; Grazzini, G.; Pradella, S.; Recchia, L.; Brunese, L.; Miele, V. Gastrointestinal neuroendocrine neoplasms (GI-NENs): Hot topics in morphological, functional, and prognostic imaging. Radiol. Med. 2021, 126, 1497–1507. [Google Scholar] [CrossRef]
  243. Satake, H.; Ishigaki, S.; Ito, R.; Naganawa, S. Radiomics in breast MRI: Current progress toward clinical application in the era of artificial intelligence. Radiol. Med. 2022, 127, 39–56. [Google Scholar] [CrossRef]
  244. Chiloiro, G.; Cusumano, D.; de Franco, P.; Lenkowicz, J.; Boldrini, L.; Carano, D.; Barbaro, B.; Corvari, B.; Dinapoli, N.; Giraffa, M.; et al. Does restaging MRI radiomics analysis improve pathological complete response prediction in rectal cancer patients? A prognostic model development. Radiol. Med. 2022, 127, 11–20. [Google Scholar] [CrossRef] [PubMed]
  245. Gregucci, F.; Fiorentino, A.; Mazzola, R.; Ricchetti, F.; Bonaparte, I.; Surgo, A.; Figlia, V.; Carbonara, R.; Caliandro, M.; Ciliberti, M.P.; et al. Radiomic analysis to predict local response in locally advanced pancreatic cancer treated with stereotactic body radiation therapy. Radiol. Med. 2022, 127, 100–107. [Google Scholar] [CrossRef] [PubMed]
  246. Herbst, R.S.; Soria, J.C.; Kowanetz, M.; Fine, G.D.; Hamid, O.; Gordon, M.S.; Sosman, J.A.; McDermott, D.F.; Powderly, J.D.; Gettinger, S.N.; et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 2014, 515, 563–567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  247. Fusco, R.; Petrillo, M.; Granata, V.; Filice, S.; Sansone, M.; Catalano, O.; Petrillo, A. Magnetic Resonance Imaging Evaluation in Neoadjuvant Therapy of Locally Advanced Rectal Cancer: A Systematic Review. Radiol. Oncol. 2017, 51, 252–262. [Google Scholar] [CrossRef]
  248. Sun, R.; Limkin, E.J.; Vakalopoulou, M.; Dercle, L.; Champiat, S.; Han, S.R.; Verlingue, L.; Brandao, D.; Lancia, A.; Ammari, S.; et al. A radiomics approach to assess tumour-infiltrating CD8 cells and response to anti-PD-1 or anti-PD-L1 immunotherapy: An imaging biomarker, retrospective multicohort study. Lancet Oncol. 2018, 19, 1180–1191. [Google Scholar] [CrossRef]
  249. 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] [Green Version]
  250. Tunali, I.; Tan, Y.; Gray, J.E.; Katsoulakis, E.; Eschrich, S.A.; Saller, J.; Aerts, H.J.W.L.; Boyle, T.; Qi, J.; Guvenis, A.; et al. Hypoxia-Related Radiomics and Immunotherapy Response: A Multicohort Study of Non-Small Cell Lung Cancer. JNCI Cancer Spectr. 2021, 5, pkab048. [Google Scholar] [CrossRef]
  251. Tunali, I.; Tan, Y.; Gray, J.E.; Eschrich, S.; Guvenis, A.; Gillies, R.; Schabath, M. Clinical-radiomic models predict overall survival among non-small cell lung cancer patients treated with immunotherapy. J. Thorac. Oncol. 2019, 14, S1129. [Google Scholar] [CrossRef]
  252. Granata, V.; Cascella, M.; Fusco, R.; dell’Aprovitola, N.; Catalano, O.; Filice, S.; Schiavone, V.; Izzo, F.; Cuomo, A.; Petrillo, A. Immediate Adverse Reactions to Gadolinium-Based MR Contrast Media: A Retrospective Analysis on 10,608 Examinations. Biomed. Res. Int. 2016, 2016, 3918292. [Google Scholar] [CrossRef]
  253. Thomas, H.M.T.; Hippe, D.S.; Forouzannezhad, P.; Sasidharan, B.K.; Kinahan, P.E.; Miyaoka, R.S.; Vesselle, H.J.; Rengan, R.; Zeng, J.; Bowen, S.R. Radiation and immune checkpoint inhibitor-mediated pneumonitis risk stratification in patients with locally advanced non-small cell lung cancer: Role of functional lung radiomics? Discov. Oncol. 2022, 13, 85. [Google Scholar] [CrossRef]
  254. Granata, V.; Fusco, R.; Avallone, A.; Catalano, O.; Filice, F.; Leongito, M.; Palaia, R.; Izzo, F.; Petrillo, A. Major and ancillary magnetic resonance features of LI-RADS to assess HCC: An overview and update. Infect. Agents Cancer 2017, 12, 23. [Google Scholar] [CrossRef] [Green Version]
  255. Matsoukas, S.; Scaggiante, J.; Schuldt, B.R.; Smith, C.J.; Chennareddy, S.; Kalagara, R.; Majidi, S.; Bederson, J.B.; Fifi, J.T.; Mocco, J.; et al. Accuracy of artificial intelligence for the detection of intracranial hemorrhage and chronic cerebral microbleeds: A systematic review and pooled analysis. Radiol. Med. 2022, 127, 1106–1123. [Google Scholar] [CrossRef]
  256. De Robertis, R.; Geraci, L.; Tomaiuolo, L.; Bortoli, L.; Beleù, A.; Malleo, G.; D’Onofrio, M. Liver metastases in pancreatic ductal adenocarcinoma: A predictive model based on CT texture analysis. Radiol. Med. 2022, 127, 1079–1084. [Google Scholar] [CrossRef]
  257. Park, J.E.; Kim, D.; Kim, H.S.; Park, S.Y.; Kim, J.Y.; Cho, S.J.; Shin, J.H.; Kim, J.H. Quality of science and reporting of radiomics in oncologic studies: Room for improvement according to radiomics quality score and TRIPOD statement. Eur. Radiol. 2020, 30, 523–536. [Google Scholar] [CrossRef]
  258. Zwanenburg, A.; Vallières, M.; Abdalah, M.A.; Aerts, H.J.W.L.; Andrearczyk, V.; Apte, A.; Ashrafinia, S.; Bakas, S.; Beukinga, R.J.; Boellaard, R.; et al. The Image Biomarker Standardization Initiative: Standardized Quantitative Radiomics for High-Throughput Image-based Phenotyping. Radiology 2020, 295, 328–338. [Google Scholar] [CrossRef]
Figure 1. Complete response pattern in a mesothelioma patient treated with immunotherapy. CT scan assessment in pre-treatment phase (A) of lesion (arrow), at 3 months (B), and after 6 months (C).
Figure 1. Complete response pattern in a mesothelioma patient treated with immunotherapy. CT scan assessment in pre-treatment phase (A) of lesion (arrow), at 3 months (B), and after 6 months (C).
Diagnostics 13 00302 g001
Figure 2. Partial response pattern in non small lung cancer (NSLC) treated with immunotherapy (arrows). CT scan assessment in pre-treatment phase (A,D), at 3 months (B,E), and after 6 months (C,F).
Figure 2. Partial response pattern in non small lung cancer (NSLC) treated with immunotherapy (arrows). CT scan assessment in pre-treatment phase (A,D), at 3 months (B,E), and after 6 months (C,F).
Diagnostics 13 00302 g002
Figure 3. Pseudoprogression of brain metastasis (arrows) in an NSLC patient treated with immunotherapy. CT evaluation (A) in pre-treatment phase, after 1 month (B), and after 12 weeks of the first CT evaluation (C).
Figure 3. Pseudoprogression of brain metastasis (arrows) in an NSLC patient treated with immunotherapy. CT evaluation (A) in pre-treatment phase, after 1 month (B), and after 12 weeks of the first CT evaluation (C).
Diagnostics 13 00302 g003
Figure 4. Iperprogression in an NSLC (arrows) patient treated with immunotherapy. CT assessment in pre-treatment phase (AC) and after 3 months (DF).
Figure 4. Iperprogression in an NSLC (arrows) patient treated with immunotherapy. CT assessment in pre-treatment phase (AC) and after 3 months (DF).
Diagnostics 13 00302 g004
Figure 5. The same patient in Figure 4 (A). New lesions (arrows) in ileo-psoas muscle (B) due to iperprogression during immunotherapy.
Figure 5. The same patient in Figure 4 (A). New lesions (arrows) in ileo-psoas muscle (B) due to iperprogression during immunotherapy.
Diagnostics 13 00302 g005
Figure 6. Dissociated responses in a melanoma patient during immunotherapy. CT assessment in pre-treatment phase (A) and at 3 months (B) follow-up: black arrows show the regression of the lesion while white arrows show the increase in lesion dimensions.
Figure 6. Dissociated responses in a melanoma patient during immunotherapy. CT assessment in pre-treatment phase (A) and at 3 months (B) follow-up: black arrows show the regression of the lesion while white arrows show the increase in lesion dimensions.
Diagnostics 13 00302 g006
Figure 7. NSLC (black arrows) patient treated with immunotherapy. CT assessment in pre-treatment phase (A). During follow-up, in (B), appearance of ir-Pneumonitis (white arrow), with disease regression after corticosteroid treatment (C).
Figure 7. NSLC (black arrows) patient treated with immunotherapy. CT assessment in pre-treatment phase (A). During follow-up, in (B), appearance of ir-Pneumonitis (white arrow), with disease regression after corticosteroid treatment (C).
Diagnostics 13 00302 g007
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Granata, V.; Fusco, R.; Setola, S.V.; Simonetti, I.; Picone, C.; Simeone, E.; Festino, L.; Vanella, V.; Vitale, M.G.; Montanino, A.; et al. Immunotherapy Assessment: A New Paradigm for Radiologists. Diagnostics 2023, 13, 302. https://doi.org/10.3390/diagnostics13020302

AMA Style

Granata V, Fusco R, Setola SV, Simonetti I, Picone C, Simeone E, Festino L, Vanella V, Vitale MG, Montanino A, et al. Immunotherapy Assessment: A New Paradigm for Radiologists. Diagnostics. 2023; 13(2):302. https://doi.org/10.3390/diagnostics13020302

Chicago/Turabian Style

Granata, Vincenza, Roberta Fusco, Sergio Venanzio Setola, Igino Simonetti, Carmine Picone, Ester Simeone, Lucia Festino, Vito Vanella, Maria Grazia Vitale, Agnese Montanino, and et al. 2023. "Immunotherapy Assessment: A New Paradigm for Radiologists" Diagnostics 13, no. 2: 302. https://doi.org/10.3390/diagnostics13020302

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