Next Article in Journal
The Role of EGFR Amplification in Deep Venous Thrombosis Occurrence in IDH Wild-Type Glioblastoma
Previous Article in Journal
Financial Toxicity and Out-of-Pocket Costs for Patients with Head and Neck Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Radiomics and Radiogenomics in Pelvic Oncology: Current Applications and Future Directions

by
Niall J. O’Sullivan
1,2,* and
Michael E. Kelly
1,2
1
The Trinity St. James’s Cancer Institute, D08 NHY1 Dublin, Ireland
2
School of Medicine, Trinity College Dublin, D02 PN40 Dublin, Ireland
*
Author to whom correspondence should be addressed.
Curr. Oncol. 2023, 30(5), 4936-4945; https://doi.org/10.3390/curroncol30050372
Submission received: 8 March 2023 / Revised: 19 April 2023 / Accepted: 8 May 2023 / Published: 11 May 2023

Abstract

:
Radiomics refers to the conversion of medical imaging into high-throughput, quantifiable data in order to analyse disease patterns, guide prognosis and aid decision making. Radiogenomics is an extension of radiomics that combines conventional radiomics techniques with molecular analysis in the form of genomic and transcriptomic data, serving as an alternative to costly, labour-intensive genetic testing. Data on radiomics and radiogenomics in the field of pelvic oncology remain novel concepts in the literature. We aim to perform an up-to-date analysis of current applications of radiomics and radiogenomics in the field of pelvic oncology, particularly focusing on the prediction of survival, recurrence and treatment response. Several studies have applied these concepts to colorectal, urological, gynaecological and sarcomatous diseases, with individual efficacy yet poor reproducibility. This article highlights the current applications of radiomics and radiogenomics in pelvic oncology, as well as the current limitations and future directions. Despite a rapid increase in publications investigating the use of radiomics and radiogenomics in pelvic oncology, the current evidence is limited by poor reproducibility and small datasets. In the era of personalised medicine, this novel field of research has significant potential, particularly for predicting prognosis and guiding therapeutic decisions. Future research may provide fundamental data on how we treat this cohort of patients, with the aim of reducing the exposure of high-risk patients to highly morbid procedures.

1. Background

Radiomics refers to the conversion of medical imaging into high-throughput, quantifiable data in order to analyse disease patterns, guide prognosis and aid in decision making [1,2]. Since its inception in 2012, data have been extracted and analysed from a variety of imaging modalities, such as computed tomography (CT), magnetic resonance imaging (MRI) and positron emission tomography (PET) scans, in an attempt to determine prognosis and to predict patient outcomes, particularly in the field of surgical oncology [1,3]. The process can be broadly broken down into three primary steps: the use of automated or semi-automated methods to identify the volumes of interest in a tumour, the generation of multiple quantitative features from raw imaging data regarding the region of interest (ROI), and finally the development of models that predict tumour characteristics and guide decision making [4].
Radiogenomics is an extension of radiomics that combines conventional radiomics techniques with molecular analysis in the form of genomic and transcriptomic data [5]. This particular field of medicine aims to offset the high costs and labour intensity associated with traditional genetic testing via the development of imaging surrogates that may serve as an alternative method for identifying which patients carry specific oncogenes [6]. In short, parameters derived from advanced image processing and analysis can be used to identify specific phenotypic and genotypic characteristics of the tumour, without the need for costly genetic testing [7]. The ultimate goal of radiogenomics is to offset the high costs, workload and invasiveness associated with traditional genetic testing by developing imaging surrogates that have the potential to serve as alternative methods for identifying oncogene status [6]. Its role has been assessed in several fields of oncology, including glioblastoma, breast cancer, renal cell carcinoma and colorectal cancer [8,9,10]. While several studies have demonstrated the feasibility of constructing a radiogenomic signature, most lack prospective validation with an external cohort [11,12].
Artificial intelligence (AI) is increasingly employed in medicine due to its ability to perform higher cognitive functions, such as problem solving and decision making [13]. A subfield within AI, machine learning facilitates the ability to search for data and recognize patterns, allowing the accurate prediction of results or outcomes [14,15]. This is particularly useful for identifying subtle patterns in large datasets that are often imperceptible to individual review [13,16]. The incorporation of machine learning with other new fields of research, such as radiomics and radiogenomics, has the potential to revolutionise precision medicine by predicting treatment response, prognosis and patient outcomes [17].
This review aims to shed light on the current implementation of radiomics and radiogenomics in the field of pelvic oncology, paying particular attention to individual subspecialty use and implications for the future. We provide a sample of the current literature within this field, highlighting the strengths, weaknesses and gaps in knowledge that may guide researchers in their future endeavours. Included studies deemed interesting and representative of the literature were selected after relevant articles were searched in the National Institutes of Health (NIH) PubMed database. Broad applications are highlighted in Figure 1.

2. Current Applications of Radiomics and Radiogenomics

2.1. Colorectal

(i)
Prediction of response to neoadjuvant chemotherapy
Several studies have investigated the application of radiomics and radiogenomics in colorectal surgery [18,19,20,21,22,23,24]. Nakanishi et al. developed a radiomics-based model for predicting lateral pelvic lymph node metastases (LPLNM) following neoadjuvant (chemo)radiotherapy (nCRT) for advanced low rectal cancers [24]. A retrospective study of 247 patients demonstrated that a radiomics-based prediction model was superior to the gross measurement of LPLN short-axis diameter in the prediction of LPLNM post-neoadjuvant (chemo)radiotherapy for rectal cancer [24]. This study demonstrated how a radiomics-based prediction model can be applied to avoid unnecessary exposure of patients to high morbidity procedures such as LPLN dissection in those patients deemed ‘low risk’ by the model. Recent studies have investigated the feasibility of radiomics nomograms in predicting a pathological complete response (pCR) [25,26]. Liu et al. developed a radiomics-based model using pre- and post-nCRT T2 and diffusion-weighted images (DWI) in combination with tumour length, with the ability to predict pCR with a diagnostic accuracy of 94% [25]. Similarly, Wang et al. extracted radiomic features from 183 pre-operative mpMRI scans to develop a model capable of predicting the response to nCRT [26]. Their nomogram, which included MRI T-stage, circumferential resection margin and apparent diffusion coefficient values, predicted a good response to nCRT with a specificity of 88% and a sensitivity of 71% [26].
(ii)
Prediction of mutation status
The role of fluorodeoxyglucose (18F-FDG) PET in the assessment of KRAS mutations in colorectal cancer is another topic of research in colorectal radiogenomics [18]. This radiotracer detects areas of abnormal glucose metabolism and serves to evaluate metabolic and tumour activity [27]. Several studies have demonstrated a higher standardised uptake value (SUV) of this radiotracer in patients with KRAS mutations, with a reported diagnostic accuracy of up to 75% [28,29]. MRI remains the gold standard imaging modality for rectal cancer due to its ability to assess extramural spread, accurately stage and detect local recurrence of the disease [30]. Studies have demonstrated that rectal tumours with KRAS mutations are more likely to exhibit a longer axial length as well as a greater axial: longitudinal ratio on pre-treatment MRI [31,32]. Since genomic analysis is now essential to guide therapeutic decisions in colorectal cancer, the development of radiogenomic models capable of predicting the involvement of various genetic mutations will allow for targeted therapy and improved patient outcomes [18]. Radiogenomic prediction of specific genetic aberrations has the potential to serve as a non-invasive alternative to conventional genetic testing in the future [33]. Further studies should focus on applying this novel technology to other genetic mutations as well as validating the findings of existing studies.
(iii)
Prediction of oncological outcomes
Exenterative surgery can be associated with major complications [5]. Despite advancements in surgical techniques and our understanding of advanced and recurrent pelvic malignancies, the rate of recurrence after pelvic exenteration remains unacceptably high [34]. Radiomics and radiogenomics offer the potential to offset the risk of recurrence in this cohort of patients by accurately predicting which patients are likely to experience recurrence and subsequently avoid exenterative surgery and its associated morbidity [3,35]. Badic et al. aimed to use radiomics to assess the value of contrast-enhanced CT scans as predictors of recurrence in patients with stage II and III colorectal cancer [36]. The authors used three separate machine learning models to predict disease-free survival (DFS) in this cohort of patients. A signature was developed based on clinical, histopathological and radiomic characteristics, and a predictor of recurrence showed value when compared with the traditional staging [36]. Similarly, Jayaprakasam et al. demonstrated the capabilities of MRI radiomics in predicting tumour recurrence and response to neoadjuvant chemotherapy in patients with locally advanced rectal cancer [37]. Their study exploited the hypothesis that the tumour and mesorectal fat interaction results in microscopic changes to adipocytes and subtle changes on MRI that are invisible to the naked eye [37]. In terms of predicting response, eleven radiomics features differed significantly between complete and non-complete responders. The final predictive model obtained a diagnostic accuracy of 83.9% and an area under the curve (AUC) of 0.89. Univariate analysis revealed 36 radiomics features that were significant in predicting local recurrence. The final predictive model obtained a diagnostic accuracy of 78.3% and an AUC of 0.79 [37]. These studies demonstrated the feasibility and efficacy of employing radiomics models in pelvic malignancies to predict various oncological outcomes such as response to chemotherapy and risk of recurrence, with relatively strong accuracy.

2.2. Urological

(i)
Prostate cancer; prediction of oncological outcomes
Recent years have seen a rapid increase in the number of publications applying radiomics and radiogenomics to genitourinary cancers [38]. In the realm of prostate cancer, Bourbonne et al. developed a multiparametric MRI (mpMRI)-based radiomic model to predict the risk of biochemical recurrence (BCR) and BCR-free survival post-radical prostatectomy [39]. The authors used a size-zone emphasis from apparent diffusion coefficient (ADC) maps extracted from 107 pre-therapeutic diffusion-weighted images, and demonstrated a balanced accuracy of 78% in predicting BCR post-radical prostatectomy [39]. This model was externally validated and may be used to stratify patients post-operatively by the risk of recurrence and tailor post-operative management accordingly. External validation is a labour-intensive but valuable component of radiomic nomogram construction, verifying reproducibility across other centres. The Miami MAST trial (ClinicalTrials.gov: NCT02242773), currently ongoing, aims to extract data from mpMRI-guided MRI/ultrasound (US) fusion biopsies in order to identify high-grade tumours early on in the investigations [40]. This study is estimated to be completed in 2024 and will aid physicians in their decision to commence radical treatment earlier in high-risk diseases. The current literature on radiogenomics in prostate cancer is limited, particularly regarding the detection of specific biomarkers that may be used to guide prognosis and treatment decisions [41]. The three current commercially approved genomic panels performed on prostate biopsy cores (Genomic Health’s Oncotype Dx test®®, Myriad’s Prolaris test®® and Genome Dx’s Decipher test®®) will provide a good basis for future research into prostate cancer radiogenomics [42,43,44].
(ii)
Bladder cancer; prediction of oncological outcomes
Several studies have investigated the role of radiomics and radiogenomics in predicting clinical outcomes in bladder cancer [45,46,47,48]. Lin et al. sequestered RNA sequencing data, radiomics features and clinical parameters of 62 patients with transitional cell carcinoma (TCC) of the bladder to create an integrative nomogram capable of stratifying patients into low- and high-risk groups and subsequently predicting progression-free interval (PFI) with excellent accuracy [46]. CD8A is a novel protective gene in bladder cancer and a marker of immunotherapeutic response and immune cell infiltration [48]. Low expression is associated with immunotherapeutic failure and poor oncological outcomes [48]. Zheng et al. developed a radiomics signature using pre-operative radiomics features and RNA-sequencing data of 111 bladder tumour samples to predict CD8A expression and subsequent response to immunotherapy [48]. Receiver operating characteristic (ROC) curves revealed that the nomogram had good performance in survival prediction with 1-, 3- and 5-year area AUC of 0.679, 0.722 and 0.722, respectively. This study demonstrated the ability of a radiomics signature based on nine MRI-derived radiomics features to predict the prognosis and immunotherapeutic susceptibility in patients with bladder cancer [48]. While studies aiming to predict muscle invasiveness of bladder cancer are bountiful, those aiming to construct nomograms capable of predicting oncological outcomes are scarce [49,50,51]. Future research should focus on predicting the response to neoadjuvant therapy and the long-term oncological outcomes.

2.3. Gynaecological

(i)
Ovarian cancer; prediction of BRCA status
Despite recent progress in chemotherapeutic and surgical approaches, the high morbidity and mortality associated with many gynaecological malignancies necessitate an improved understanding of how these tumours behave from a radiological and genetic perspective [52]. Few studies have described the use of radiomics and radiogenomics in gynae-oncology [52,53,54,55]. Nero et al. developed an automated machine learning pipeline model in order to identify gBRCA1/2 status based on ultrasound images of healthy ovaries, with encouraging performance [54]. With a positive predictive value (PPV) of 0.87, only 13/100 women will be exposed to unnecessary genetic testing. Despite this, the model describes a negative predictive value (NPV) of 0.67, implying that 27/100 women carrying the gene would be missed; thus, it is a major limitation of the approach [54].
(ii)
Endometrial cancer; prediction of oncological outcomes
Hoivik et al. integrated MRI radiomic features with histologic, transcriptomic and molecular biomarkers in their study of 866 patients with endometrial cancer in an attempt to identify those with aggressive tumour features [55]. In this study, a fully automated machine learning-based tumour segmentation algorithm reproduced the same radiomic prognostic groups as manual whole-volume tumour radiomic profiling by radiologists [55]. The authors identified an 11-gene high-risk signature associated with poor survival that will aid in prognosis and guide treatment decisions in patients with endometrial carcinoma. Further research is necessary to bring radiomics-based research within gynae-oncology to the same standard as currently available in the fields of colorectal and urology.

2.4. Sarcoma

The use of artificial intelligence in soft-tissue sarcomas is a relatively novel concept that aims to serve as a non-invasive method of providing information regarding the diagnosis and prognosis of tumours [56]. The employment of radiomic texture analysis in this field has resulted in the development of radiomics MRI-based models that can distinguish histotypes, determine grades and predict response and overall survival [56]. Using T1 and fat-suppressed-T2 weighted imaging, Wang et al. identified specific radiomic features that were significantly correlated with malignant soft-tissue lesions and subsequently constructed a radiomics nomogram with superior predictive performance than that of the clinical model based on the experience of radiologists [56]. Peeken et al. compared the value of MRI-based radiomics with expert-derived clinical profiling for the prediction of overall survival (OS) [57]. A total of 105 radiomic features were extracted from the images of 108 patients and subjected to three separate machine learning techniques to predict OS. The findings were compared to the semantic imaging features determined by radiologists. T2-weighted sequence and T1-weighted fat-saturated sequence radiomic models were superior to semantic imaging features in determining the prognosis of soft-tissue sarcomas [57]. To our knowledge, no studies investigating the use of radiomics or radiogenomics in the diagnosis and prognosis of intra-abdominal sarcomatous disease have been published. Future studies should focus on filling this void, paying particular focus to the prediction of oncological outcomes in patients with this aggressive disease.

3. Current Limitations

Despite significant potential in the field of pelvic oncology, current evidence is limited by variability in feature extraction and a lack of reproducibility [58,59]. Model performance is sensitive to many intrinsic variables, including heterogeneous image acquisition parameters, segmentations and feature extraction software, as well as small and mixed patient cohorts [60]. As research in this field advances, more open-source databases and software packages are being made available in an attempt to standardise models and accelerate the development and external validation of these signatures [61,62,63]. Future studies should focus on standardising the imaging protocols and radiomic techniques [18]. Standards for radiomic features need to be set in order to allow comparability between studies and reproducibility for new studies. Similarly, the stability and reproducibility of radiomic models for predicting prognosis must be externally assessed prior to their application in the clinical setting [64]. Internal validation may not be sufficient to extrapolate performance in an external setting due to relatively small datasets, as seen in most of the current studies, and external datasets should thus be validated in a large multicentre setting prior to implementation in clinical practice [65].
Multiple studies have attempted to identify reproducible radiomics features to improve the repeatability and application of radiomics and radiogenomics. Traverso et al. performed a systematic review to identify radiomics features that were repeatable and reproducible [66]. The authors found that first-order features (histogram-based), in particular entropy, had higher reproducibility than shape metrics and textural features. The Image Biomarker Standardisation Initiative (IBSI), published in 2020, aimed to standardise a set of radiomics features [67]. Over three phases, the authors achieved good to excellent reproducibility for 167 different radiomics features utilising CT, MRI and FDG-PET in 51 patients with soft-tissue sarcoma. Similarly, Pfaehler et al. developed a checklist aiming to simplify and improve the reporting of radiomic signatures, with the goal of eventually guaranteeing full replication and validation of these studies [68]. Further systematic reviews focusing on the repeatability and reproducibility of radiomics features are necessary to further improve the standardisation of results from radiomics studies [69].

4. Future Directions

The use of radiomics and radiogenomics for predicting correlations with genetic or transcriptomic abnormalities of tumours and subsequently determining prognosis and guiding treatment decisions needs much larger data studies to become a validated tool [18]. Future studies should focus on addressing the limitations outlined previously and evaluating the datasets in a multicentre, prospective setting. There is a paucity of evidence in the literature surrounding the use of radiomics and radiogenomics to identify patients at high risk of recurrence of locally advanced or locally recurrent pelvic cancers. The development of a radiomics nomogram aimed at predicting disease recurrence in this cohort of patients may offer surgeons the ability to avoid highly morbid procedures, such as exenteration in patients who are likely to recur, or at least counsel patients better regarding expected outcomes [5].
Quantification of circulating tumour DNA (ctDNA) and cell-free DNA (cfDNA) represents a novel area of research in cancer detection and evaluation of disease burden, with the potential to revolutionise our assessment of therapeutic responses and our understanding of personalised medicine as a whole [70]. Current applications include the detection of microscopic residual disease following radiotherapy, an alternative non-invasive method of genotyping, as well as early detection of tumour recurrence [71]. Research on ctDNA in the field of radiomics and radiogenomics is scarce [72]. Lafata et al. aimed to create patient-specific radiogenomic expression patterns to guide prognosis by combining CT radiomics, next-generation sequencing of ctDNA and serum cfDNA in patients with locally advanced lung cancer receiving chemoradiotherapy [72]. Two distinct radiomic signatures were identified prior to treatment, which were subsequently associated with the presence of TP53 mutations within ctDNA and changes in cfDNA two weeks post-chemoradiation. The authors found that heterogeneous and low-attenuating disease, without a detectable ctDNA TP53 mutation, was linked to an early surge in post-treatment cfDNA concentration and improved overall survival [72]. These findings highlight the feasibility and efficacy of this approach in predicting treatment response and guiding prognosis. Further studies, ideally larger randomised clinical studies, are necessary to validate these findings.
Perhaps one of the greatest obstacles to radiomic-based research is feature reliability [73]. Heterogeneity and uncertainty can arise from many areas within a complex workflow, ultimately impeding feature reproducibility, stability and validity [74,75,76,77,78,79]. In their review of 481 radiomics studies, Xue et al. attempted to define reliability using intraclass correlation coefficient (ICC) expression [73]. ICC is a reliability index widely adopted in the medical literature, which can be applied to any radiomic feature that has continuous values [73,80]. The authors conclude by offering several suggestions for researchers carrying out radiomics-based research to mitigate the pitfalls identified in their analysis of 481 manuscripts. Koo et al. also provide clinical researchers with a practical step-by-step guideline to select the correct form of ICC and to avoid selecting inappropriate ICC forms that may result in misleading interpretations [80].

5. Conclusions

Despite a rapid increase in publications investigating the use of radiomics and radiogenomics in pelvic oncology, current evidence is limited by poor reproducibility and small datasets [60]. In the era of personalised medicine, this novel field of research has significant potential, particularly for predicting prognosis and guiding therapeutic decisions [3,76]. Future research may provide fundamental data on how we treat this cohort of patients, with the aim of reducing the exposure of high-risk patients to highly morbid procedures. While many of the studies discussed in our narrative review are small, single-centre projects, they provide the foundation necessary and proof of efficacy required for the production of further high-impact studies in the future. Large-scale, multi-institutional studies with external validation of models are required to ultimately change clinical practice.

Author Contributions

Conceptualization, N.J.O. and M.E.K.; methodology, N.J.O.; data curation, N.J.O.; writing—original draft preparation, N.J.O. and M.E.K.; writing—review and editing, N.J.O. and M.E.K.; visualization, N.J.O.; supervision, M.E.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data was created in this review.

Conflicts of Interest

This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors. The authors report no conflicts of interest.

Abbreviations

CT: Computed Tomography, MRI: Magnetic Resonance Imaging, PET: Positron Emission Tomography, ROI: Region of Interest, AI: Artificial Intelligence, LPLNM: Lateral Pelvic Lymph Node Metastasis, nCRT: Neoadjuvant Chemoradiotherapy, pCR: Pathological Complete Response, SUV: Standard Uptake Value, FDG: Fluorodeoxyglucose, DFS: Disease-free survival.

References

  1. Guiot, J.; Vaidyanathan, A.; Deprez, L.; Zerka, F.; Danthine, D.; Frix, A.N.; Lambin, P.; Bottari, F.; Tsoutzidis, N.; Miraglio, B.; et al. A review in radiomics: Making personalized medicine a reality via routine imaging. Med. Res. Rev. 2022, 42, 426–440. [Google Scholar] [CrossRef] [PubMed]
  2. Gillies, R.J.; Kinahan, P.E.; Hricak, H. Radiomics: Images Are More than Pictures, They Are Data. Radiology 2016, 278, 563–577. [Google Scholar] [CrossRef] [PubMed]
  3. Lambin, P.; Leijenaar, R.T.H.; Deist, T.M.; Peerlings, J.; de Jong, E.E.C.; van Timmeren, J.; Sanduleanu, S.; Larue, R.; Even, A.J.G.; Jochems, A.; et al. Radiomics: The bridge between medical imaging and personalized medicine. Nat. Rev. Clin. Oncol. 2017, 14, 749–762. [Google Scholar] [CrossRef] [PubMed]
  4. Lee, G.; Lee, H.Y.; Park, H.; Schiebler, M.L.; van Beek, E.J.R.; Ohno, Y.; Seo, J.B.; Leung, A. Radiomics and its emerging role in lung cancer research, imaging biomarkers and clinical management: State of the art. Eur. J. Radiol. 2017, 86, 297–307. [Google Scholar] [CrossRef] [PubMed]
  5. PelvEx, C. Contemporary Management of Locally Advanced and Recurrent Rectal Cancer: Views from the PelvEx Collaborative. Cancers 2022, 14, 1161. [Google Scholar] [CrossRef]
  6. Lo Gullo, R.; Daimiel, I.; Morris, E.A.; Pinker, K. Combining molecular and imaging metrics in cancer: Radiogenomics. Insights Imaging 2020, 11, 1. [Google Scholar] [CrossRef]
  7. Pinker, K.; Shitano, F.; Sala, E.; Do, R.K.; Young, R.J.; Wibmer, A.G.; Hricak, H.; Sutton, E.J.; Morris, E.A. Background, current role, and potential applications of radiogenomics. J. Magn. Reson. Imaging 2018, 47, 604–620. [Google Scholar] [CrossRef]
  8. Singh, G.; Manjila, S.; Sakla, N.; True, A.; Wardeh, A.H.; Beig, N.; Vaysberg, A.; Matthews, J.; Prasanna, P.; Spektor, V. Radiomics and radiogenomics in gliomas: A contemporary update. Br. J. Cancer 2021, 125, 641–657. [Google Scholar] [CrossRef]
  9. Darvish, L.; Bahreyni-Toossi, M.-T.; Roozbeh, N.; Azimian, H. The role of radiogenomics in the diagnosis of breast cancer: A systematic review. Egypt. J. Med. Hum. Genet. 2022, 23, 99. [Google Scholar] [CrossRef]
  10. Alessandrino, F.; Shinagare, A.B.; Bosse, D.; Choueiri, T.K.; Krajewski, K.M. Radiogenomics in renal cell carcinoma. Abdom. Radiol. 2019, 44, 1990–1998. [Google Scholar] [CrossRef]
  11. Chen, Y.; Li, B.; Jiang, Z.; Li, H.; Dang, Y.; Tang, C.; Xia, Y.; Zhang, H.; Song, B.; Long, L. Multi-parameter diffusion and perfusion magnetic resonance imaging and radiomics nomogram for preoperative evaluation of aquaporin-1 expression in rectal cancer. Abdom. Radiol. 2022, 47, 1276–1290. [Google Scholar] [CrossRef] [PubMed]
  12. Oh, J.E.; Kim, M.J.; Lee, J.; Hur, B.Y.; Kim, B.; Kim, D.Y.; Baek, J.Y.; Chang, H.J.; Park, S.C.; Oh, J.H.; et al. Magnetic Resonance-Based Texture Analysis Differentiating KRAS Mutation Status in Rectal Cancer. Cancer Res. Treat. 2020, 52, 51–59. [Google Scholar] [CrossRef] [PubMed]
  13. Hashimoto, D.A.; Rosman, G.; Rus, D.; Meireles, O.R. Artificial Intelligence in Surgery: Promises and Perils. Ann. Surg. 2018, 268, 70–76. [Google Scholar] [CrossRef]
  14. Deo, R.C. Machine Learning in Medicine. Circulation 2015, 132, 1920–1930. [Google Scholar] [CrossRef] [PubMed]
  15. PelvEx, C. Predicting outcomes of pelvic exenteration using machine learning. Color. Dis. 2020, 22, 1933–1940. [Google Scholar] [CrossRef]
  16. Miller, R.A.; Pople, H.E., Jr.; Myers, J.D. Internist-1, an experimental computer-based diagnostic consultant for general internal medicine. N. Engl. J. Med. 1982, 307, 468–476. [Google Scholar] [CrossRef] [PubMed]
  17. Avanzo, M.; Wei, L.; Stancanello, J.; Vallieres, M.; Rao, A.; Morin, O.; Mattonen, S.A.; El Naqa, I. Machine and deep learning methods for radiomics. Med. Phys. 2020, 47, e185–e202. [Google Scholar] [CrossRef]
  18. Badic, B.; Tixier, F.; Cheze Le Rest, C.; Hatt, M.; Visvikis, D. Radiogenomics in Colorectal Cancer. Cancers 2021, 13, 973. [Google Scholar] [CrossRef]
  19. Bedrikovetski, S.; Dudi-Venkata, N.N.; Kroon, H.M.; Seow, W.; Vather, R.; Carneiro, G.; Moore, J.W.; Sammour, T. Artificial intelligence for pre-operative lymph node staging in colorectal cancer: A systematic review and meta-analysis. BMC Cancer 2021, 21, 1058. [Google Scholar] [CrossRef]
  20. Horvat, N.; Bates, D.D.B.; Petkovska, I. Novel imaging techniques of rectal cancer: What do radiomics and radiogenomics have to offer? A literature review. Abdom Radiol 2019, 44, 3764–3774. [Google Scholar] [CrossRef]
  21. Wang, Y.; Ma, L.Y.; Yin, X.P.; Gao, B.L. Radiomics and Radiogenomics in Evaluation of Colorectal Cancer Liver Metastasis. Front. Oncol. 2021, 11, 689509. [Google Scholar] [CrossRef] [PubMed]
  22. Stanzione, A.; Verde, F.; Romeo, V.; Boccadifuoco, F.; Mainenti, P.P.; Maurea, S. Radiomics and machine learning applications in rectal cancer: Current update and future perspectives. World J. Gastroenterol. 2021, 27, 5306–5321. [Google Scholar] [CrossRef]
  23. de la Pinta, C.; Castillo, M.E.; Collado, M.; Galindo-Pumarino, C.; Pena, C. Radiogenomics: Hunting Down Liver Metastasis in Colorectal Cancer Patients. Cancers 2021, 13, 5547. [Google Scholar] [CrossRef] [PubMed]
  24. Nakanishi, R.; Akiyoshi, T.; Toda, S.; Murakami, Y.; Taguchi, S.; Oba, K.; Hanaoka, Y.; Nagasaki, T.; Yamaguchi, T.; Konishi, T.; et al. Radiomics Approach Outperforms Diameter Criteria for Predicting Pathological Lateral Lymph Node Metastasis After Neoadjuvant (Chemo)Radiotherapy in Advanced Low Rectal Cancer. Ann. Surg. Oncol. 2020, 27, 4273–4283. [Google Scholar] [CrossRef] [PubMed]
  25. Liu, Z.; Zhang, X.Y.; Shi, Y.J.; Wang, L.; Zhu, H.T.; Tang, Z.; Wang, S.; Li, X.T.; Tian, J.; Sun, Y.S. Radiomics Analysis for Evaluation of Pathological Complete Response to Neoadjuvant Chemoradiotherapy in Locally Advanced Rectal Cancer. Clin. Cancer Res. 2017, 23, 7253–7262. [Google Scholar] [CrossRef] [PubMed]
  26. Wang, J.; Liu, X.; Hu, B.; Gao, Y.; Chen, J.; Li, J. Development and validation of an MRI-based radiomic nomogram to distinguish between good and poor responders in patients with locally advanced rectal cancer undergoing neoadjuvant chemoradiotherapy. Abdom. Radiol. 2021, 46, 1805–1815. [Google Scholar] [CrossRef] [PubMed]
  27. Almuhaideb, A.; Papathanasiou, N.; Bomanji, J. 18F-FDG PET/CT imaging in oncology. Ann. Saudi. Med. 2011, 31, 3–13. [Google Scholar] [CrossRef]
  28. Lee, J.H.; Kang, J.; Baik, S.H.; Lee, K.Y.; Lim, B.J.; Jeon, T.J.; Ryu, Y.H.; Sohn, S.K. Relationship Between 18F-Fluorodeoxyglucose Uptake and V-Ki-Ras2 Kirsten Rat Sarcoma Viral Oncogene Homolog Mutation in Colorectal Cancer Patients: Variability Depending on C-Reactive Protein Level. Medicine 2016, 95, e2236. [Google Scholar] [CrossRef]
  29. Kawada, K.; Nakamoto, Y.; Kawada, M.; Hida, K.; Matsumoto, T.; Murakami, T.; Hasegawa, S.; Togashi, K.; Sakai, Y. Relationship between 18F-fluorodeoxyglucose accumulation and KRAS/BRAF mutations in colorectal cancer. Clin. Cancer Res. 2012, 18, 1696–1703. [Google Scholar] [CrossRef]
  30. Bhoday, J.; Balyasnikova, S.; Wale, A.; Brown, G. How Should Imaging Direct/Orient Management of Rectal Cancer? Clin. Colon Rectal Surg. 2017, 30, 297–312. [Google Scholar] [CrossRef]
  31. Shin, Y.R.; Kim, K.A.; Im, S.; Hwang, S.S.; Kim, K. Prediction of KRAS Mutation in Rectal Cancer Using MRI. Anticancer Res. 2016, 36, 4799–4804. [Google Scholar] [CrossRef] [PubMed]
  32. Jo, S.J.; Kim, S.H. Association between oncogenic RAS mutation and radiologic-pathologic findings in patients with primary rectal cancer. Quant. Imaging Med. Surg. 2019, 9, 238–246. [Google Scholar] [CrossRef] [PubMed]
  33. Jansen, R.W.; van Amstel, P.; Martens, R.M.; Kooi, I.E.; Wesseling, P.; de Langen, A.J.; Menke-Van der Houven van Oordt, C.W.; Jansen, B.H.E.; Moll, A.C.; Dorsman, J.C.; et al. Non-invasive tumor genotyping using radiogenomic biomarkers, a systematic review and oncology-wide pathway analysis. Oncotarget 2018, 9, 20134–20155. [Google Scholar] [CrossRef] [PubMed]
  34. Yang, T.X.; Morris, D.L.; Chua, T.C. Pelvic exenteration for rectal cancer: A systematic review. Dis. Colon Rectum 2013, 56, 519–531. [Google Scholar] [CrossRef]
  35. Horvat, N.; Veeraraghavan, H.; Pelossof, R.A.; Fernandes, M.C.; Arora, A.; Khan, M.; Marco, M.; Cheng, C.T.; Gonen, M.; Golia Pernicka, J.S.; et al. Radiogenomics of rectal adenocarcinoma in the era of precision medicine: A pilot study of associations between qualitative and quantitative MRI imaging features and genetic mutations. Eur. J. Radiol. 2019, 113, 174–181. [Google Scholar] [CrossRef]
  36. Badic, B.; Da-Ano, R.; Poirot, K.; Jaouen, V.; Magnin, B.; Gagniere, J.; Pezet, D.; Hatt, M.; Visvikis, D. Prediction of recurrence after surgery in colorectal cancer patients using radiomics from diagnostic contrast-enhanced computed tomography: A two-center study. Eur. Radiol. 2022, 32, 405–414. [Google Scholar] [CrossRef] [PubMed]
  37. Jayaprakasam, V.S.; Paroder, V.; Gibbs, P.; Bajwa, R.; Gangai, N.; Sosa, R.E.; Petkovska, I.; Golia Pernicka, J.S.; Fuqua, J.L., 3rd; Bates, D.D.B.; et al. MRI radiomics features of mesorectal fat can predict response to neoadjuvant chemoradiation therapy and tumor recurrence in patients with locally advanced rectal cancer. Eur. Radiol. 2022, 32, 971–980. [Google Scholar] [CrossRef]
  38. Liu, L.; Yi, X.; Lu, C.; Qi, L.; Zhang, Y.; Li, M.; Xiao, Q.; Wang, C.; Zhang, L.; Pang, Y.; et al. Applications of radiomics in genitourinary tumors. Am. J. Cancer Res. 2020, 10, 2293–2308. [Google Scholar]
  39. Bourbonne, V.; Fournier, G.; Vallieres, M.; Lucia, F.; Doucet, L.; Tissot, V.; Cuvelier, G.; Hue, S.; Le Penn Du, H.; Perdriel, L.; et al. External Validation of an MRI-Derived Radiomics Model to Predict Biochemical Recurrence after Surgery for High-Risk Prostate Cancer. Cancers 2020, 12, 814. [Google Scholar] [CrossRef]
  40. ClinicalTrials.gov. NCT02242773: MRI Based Active Selection for Treatment Trial (MAST). Available online: https://clinicaltrials.gov/ct2/show/NCT02242773 (accessed on 23 February 2023).
  41. Banerjee, V.; Wang, S.; Drescher, M.; Russell, R.; Siddiqui, M.M. Radiogenomics influence on the future of prostate cancer risk stratification. Ther. Adv. Urol. 2022, 14, 17562872221125317. [Google Scholar] [CrossRef]
  42. Klein, E.A.; Cooperberg, M.R.; Magi-Galluzzi, C.; Simko, J.P.; Falzarano, S.M.; Maddala, T.; Chan, J.M.; Li, J.; Cowan, J.E.; Tsiatis, A.C.; et al. A 17-gene assay to predict prostate cancer aggressiveness in the context of Gleason grade heterogeneity, tumor multifocality, and biopsy undersampling. Eur. Urol. 2014, 66, 550–560. [Google Scholar] [CrossRef] [PubMed]
  43. Cooperberg, M.R.; Simko, J.P.; Cowan, J.E.; Reid, J.E.; Djalilvand, A.; Bhatnagar, S.; Gutin, A.; Lanchbury, J.S.; Swanson, G.P.; Stone, S.; et al. Validation of a cell-cycle progression gene panel to improve risk stratification in a contemporary prostatectomy cohort. J. Clin. Oncol. 2013, 31, 1428–1434. [Google Scholar] [CrossRef] [PubMed]
  44. Klein, E.A.; Yousefi, K.; Haddad, Z.; Choeurng, V.; Buerki, C.; Stephenson, A.J.; Li, J.; Kattan, M.W.; Magi-Galluzzi, C.; Davicioni, E. A genomic classifier improves prediction of metastatic disease within 5 years after surgery in node-negative high-risk prostate cancer patients managed by radical prostatectomy without adjuvant therapy. Eur. Urol. 2015, 67, 778–786. [Google Scholar] [CrossRef]
  45. Kozikowski, M.; Suarez-Ibarrola, R.; Osiecki, R.; Bilski, K.; Gratzke, C.; Shariat, S.F.; Miernik, A.; Dobruch, J. Role of Radiomics in the Prediction of Muscle-invasive Bladder Cancer: A Systematic Review and Meta-analysis. Eur. Urol. Focus 2022, 8, 728–738. [Google Scholar] [CrossRef]
  46. Lin, P.; Wen, D.Y.; Chen, L.; Li, X.; Li, S.H.; Yan, H.B.; He, R.Q.; Chen, G.; He, Y.; Yang, H. A radiogenomics signature for predicting the clinical outcome of bladder urothelial carcinoma. Eur. Radiol. 2020, 30, 547–557. [Google Scholar] [CrossRef]
  47. Ye, F.; Hu, Y.; Gao, J.; Liang, Y.; Liu, Y.; Ou, Y.; Cheng, Z.; Jiang, H. Radiogenomics Map Reveals the Landscape of m6A Methylation Modification Pattern in Bladder Cancer. Front. Immunol. 2021, 12, 722642. [Google Scholar] [CrossRef]
  48. Zheng, Z.; Guo, Y.; Huang, X.; Liu, J.; Wang, R.; Qiu, X.; Liu, S. CD8A as a Prognostic and Immunotherapy Predictive Biomarker Can Be Evaluated by MRI Radiomics Features in Bladder Cancer. Cancers 2022, 14, 4866. [Google Scholar] [CrossRef] [PubMed]
  49. Cui, Y.; Sun, Z.; Liu, X.; Zhang, X.; Wang, X. CT-based radiomics for the preoperative prediction of the muscle-invasive status of bladder cancer and comparison to radiologists’ assessment. Clin. Radiol. 2022, 77, e473–e482. [Google Scholar] [CrossRef]
  50. Wang, H.; Xu, X.; Zhang, X.; Liu, Y.; Ouyang, L.; Du, P.; Li, S.; Tian, Q.; Ling, J.; Guo, Y.; et al. Elaboration of a multisequence MRI-based radiomics signature for the preoperative prediction of the muscle-invasive status of bladder cancer: A double-center study. Eur. Radiol. 2020, 30, 4816–4827. [Google Scholar] [CrossRef]
  51. Zhang, G.; Wu, Z.; Zhang, X.; Xu, L.; Mao, L.; Li, X.; Xiao, Y.; Ji, Z.; Sun, H.; Jin, Z. CT-based radiomics to predict muscle invasion in bladder cancer. Eur. Radiol. 2022, 32, 3260–3268. [Google Scholar] [CrossRef]
  52. Nougaret, S.; McCague, C.; Tibermacine, H.; Vargas, H.A.; Rizzo, S.; Sala, E. Radiomics and radiogenomics in ovarian cancer: A literature review. Abdom. Radiol. 2021, 46, 2308–2322. [Google Scholar] [CrossRef] [PubMed]
  53. Michalet, M.; Azria, D.; Tardieu, M.; Tibermacine, H.; Nougaret, S. Radiomics in radiation oncology for gynecological malignancies: A review of literature. Br. J. Radiol. 2021, 94, 20210032. [Google Scholar] [CrossRef] [PubMed]
  54. Nero, C.; Ciccarone, F.; Boldrini, L.; Lenkowicz, J.; Paris, I.; Capoluongo, E.D.; Testa, A.C.; Fagotti, A.; Valentini, V.; Scambia, G. Germline BRCA 1-2 status prediction through ovarian ultrasound images radiogenomics: A hypothesis generating study (PROBE study). Sci. Rep. 2020, 10, 16511. [Google Scholar] [CrossRef]
  55. Hoivik, E.A.; Hodneland, E.; Dybvik, J.A.; Wagner-Larsen, K.S.; Fasmer, K.E.; Berg, H.F.; Halle, M.K.; Haldorsen, I.S.; Krakstad, C. A radiogenomics application for prognostic profiling of endometrial cancer. Commun. Biol. 2021, 4, 1363. [Google Scholar] [CrossRef]
  56. Fanciullo, C.; Gitto, S.; Carlicchi, E.; Albano, D.; Messina, C.; Sconfienza, L.M. Radiomics of Musculoskeletal Sarcomas: A Narrative Review. J. Imaging 2022, 8, 45. [Google Scholar] [CrossRef] [PubMed]
  57. Peeken, J.C.; Neumann, J.; Asadpour, R.; Leonhardt, Y.; Moreira, J.R.; Hippe, D.S.; Klymenko, O.; Foreman, S.C.; von Schacky, C.E.; Spraker, M.B.; et al. Prognostic Assessment in High-Grade Soft-Tissue Sarcoma Patients: A Comparison of Semantic Image Analysis and Radiomics. Cancers 2021, 13, 1929. [Google Scholar] [CrossRef]
  58. Kawada, K.; Toda, K.; Nakamoto, Y.; Iwamoto, M.; Hatano, E.; Chen, F.; Hasegawa, S.; Togashi, K.; Date, H.; Uemoto, S.; et al. Relationship Between 18F-FDG PET/CT Scans and KRAS Mutations in Metastatic Colorectal Cancer. J. Nucl. Med. 2015, 56, 1322–1327. [Google Scholar] [CrossRef] [PubMed]
  59. Mao, W.; Zhou, J.; Zhang, H.; Qiu, L.; Tan, H.; Hu, Y.; Shi, H. Relationship between KRAS mutations and dual time point (18)F-FDG PET/CT imaging in colorectal liver metastases. Abdom. Radiol. 2019, 44, 2059–2066. [Google Scholar] [CrossRef]
  60. Zhao, B. Understanding Sources of Variation to Improve the Reproducibility of Radiomics. Front. Oncol. 2021, 11, 633176. [Google Scholar] [CrossRef]
  61. van Griethuysen, J.J.M.; Fedorov, A.; Parmar, C.; Hosny, A.; Aucoin, N.; Narayan, V.; Beets-Tan, R.G.H.; Fillion-Robin, J.C.; Pieper, S.; Aerts, H. Computational Radiomics System to Decode the Radiographic Phenotype. Cancer Res. 2017, 77, e104–e107. [Google Scholar] [CrossRef]
  62. Zhang, L.; Fried, D.V.; Fave, X.J.; Hunter, L.A.; Yang, J.; Court, L.E. IBEX: An open infrastructure software platform to facilitate collaborative work in radiomics. Med. Phys. 2015, 42, 1341–1353. [Google Scholar] [CrossRef] [PubMed]
  63. Pfaehler, E.; Zwanenburg, A.; de Jong, J.R.; Boellaard, R. RaCaT: An open source and easy to use radiomics calculator tool. PLoS ONE 2019, 14, e0212223. [Google Scholar] [CrossRef]
  64. Rizzo, S.; Botta, F.; Raimondi, S.; Origgi, D.; Fanciullo, C.; Morganti, A.G.; Bellomi, M. Radiomics: The facts and the challenges of image analysis. Eur. Radiol. Exp. 2018, 2, 36. [Google Scholar] [CrossRef] [PubMed]
  65. Bleeker, S.E.; Moll, H.A.; Steyerberg, E.W.; Donders, A.R.; Derksen-Lubsen, G.; Grobbee, D.E.; Moons, K.G. External validation is necessary in prediction research: A clinical example. J. Clin. Epidemiol. 2003, 56, 826–832. [Google Scholar] [CrossRef]
  66. Traverso, A.; Wee, L.; Dekker, A.; Gillies, R. Repeatability and Reproducibility of Radiomic Features: A Systematic Review. Int. J. Radiat. Oncol. Biol. Phys. 2018, 102, 1143–1158. [Google Scholar] [CrossRef] [PubMed]
  67. Zwanenburg, A.; Vallieres, M.; Abdalah, M.A.; Aerts, H.; 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] [PubMed]
  68. Pfaehler, E.; Zhovannik, I.; Wei, L.; Boellaard, R.; Dekker, A.; Monshouwer, R.; El Naqa, I.; Bussink, J.; Gillies, R.; Wee, L.; et al. A systematic review and quality of reporting checklist for repeatability and reproducibility of radiomic features. Phys. Imaging Radiat. Oncol. 2021, 20, 69–75. [Google Scholar] [CrossRef]
  69. Lohmann, P.; Bousabarah, K.; Hoevels, M.; Treuer, H. Radiomics in radiation oncology-basics, methods, and limitations. Strahlenther. Onkol. 2020, 196, 848–855. [Google Scholar] [CrossRef]
  70. Bettegowda, C.; Sausen, M.; Leary, R.J.; Kinde, I.; Wang, Y.; Agrawal, N.; Bartlett, B.R.; Wang, H.; Luber, B.; Alani, R.M.; et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci. Transl. Med. 2014, 6, 224ra224. [Google Scholar] [CrossRef]
  71. Chaudhuri, A.A.; Binkley, M.S.; Osmundson, E.C.; Alizadeh, A.A.; Diehn, M. Predicting Radiotherapy Responses and Treatment Outcomes Through Analysis of Circulating Tumor DNA. Semin. Radiat. Oncol. 2015, 25, 305–312. [Google Scholar] [CrossRef]
  72. Lafata, K.J.; Corradetti, M.N.; Gao, J.; Jacobs, C.D.; Weng, J.; Chang, Y.; Wang, C.; Hatch, A.; Xanthopoulos, E.; Jones, G.; et al. Radiogenomic Analysis of Locally Advanced Lung Cancer Based on CT Imaging and Intratreatment Changes in Cell-Free DNA. Radiol. Imaging Cancer 2021, 3, e200157. [Google Scholar] [CrossRef] [PubMed]
  73. Xue, C.; Yuan, J.; Lo, G.G.; Chang, A.T.Y.; Poon, D.M.C.; Wong, O.L.; Zhou, Y.; Chu, W.C.W. Radiomics feature reliability assessed by intraclass correlation coefficient: A systematic review. Quant. Imaging Med. Surg. 2021, 11, 4431–4460. [Google Scholar] [CrossRef] [PubMed]
  74. Reuze, S.; Schernberg, A.; Orlhac, F.; Sun, R.; Chargari, C.; Dercle, L.; Deutsch, E.; Buvat, I.; Robert, C. Radiomics in Nuclear Medicine Applied to Radiation Therapy: Methods, Pitfalls, and Challenges. Int. J. Radiat. Oncol. Biol. Phys. 2018, 102, 1117–1142. [Google Scholar] [CrossRef] [PubMed]
  75. Yip, S.S.; Aerts, H.J. Applications and limitations of radiomics. Phys. Med. Biol. 2016, 61, R150–R166. [Google Scholar] [CrossRef]
  76. Miles, K. Radiomics for personalised medicine: The long road ahead. Br. J. Cancer 2020, 122, 929–930. [Google Scholar] [CrossRef]
  77. 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] [PubMed]
  78. Pinto Dos Santos, D.; Dietzel, M.; Baessler, B. A decade of radiomics research: Are images really data or just patterns in the noise? Eur. Radiol. 2021, 31, 1–4. [Google Scholar] [CrossRef] [PubMed]
  79. Hatt, M.; Le Rest, C.C.; Tixier, F.; Badic, B.; Schick, U.; Visvikis, D. Radiomics: Data Are Also Images. J. Nucl. Med. 2019, 60, 38S–44S. [Google Scholar] [CrossRef]
  80. Koo, T.K.; Li, M.Y. A Guideline of Selecting and Reporting Intraclass Correlation Coefficients for Reliability Research. J. Chiropr. Med. 2016, 15, 155–163. [Google Scholar] [CrossRef]
Figure 1. Applications of radiomics and radiogenomics in pelvic oncology. pCR: Pathological complete response.
Figure 1. Applications of radiomics and radiogenomics in pelvic oncology. pCR: Pathological complete response.
Curroncol 30 00372 g001
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

O’Sullivan, N.J.; Kelly, M.E. Radiomics and Radiogenomics in Pelvic Oncology: Current Applications and Future Directions. Curr. Oncol. 2023, 30, 4936-4945. https://doi.org/10.3390/curroncol30050372

AMA Style

O’Sullivan NJ, Kelly ME. Radiomics and Radiogenomics in Pelvic Oncology: Current Applications and Future Directions. Current Oncology. 2023; 30(5):4936-4945. https://doi.org/10.3390/curroncol30050372

Chicago/Turabian Style

O’Sullivan, Niall J., and Michael E. Kelly. 2023. "Radiomics and Radiogenomics in Pelvic Oncology: Current Applications and Future Directions" Current Oncology 30, no. 5: 4936-4945. https://doi.org/10.3390/curroncol30050372

Article Metrics

Back to TopTop