Generic placeholder image

Current Medical Imaging

Editor-in-Chief

ISSN (Print): 1573-4056
ISSN (Online): 1875-6603

Research Article

Differentiation of Clear Cell and Non-clear-cell Renal Cell Carcinoma through CT-based Radiomics Models and Nomogram

Author(s): Delu Cheng, Yeerxiati Abudikeranmu and Batuer Tuerdi*

Volume 19, Issue 9, 2023

Published on: 13 December, 2022

Article ID: e211122211094 Pages: 13

DOI: 10.2174/1573405619666221121164235

open_access

Abstract

Purpose: The aim of the study was to investigate the feasibility of discriminating between clear-cell renal cell carcinoma (ccRCC) and non-clear-cell renal cell carcinoma (non-ccRCC) via radiomics models and nomogram.

Methods: The retrospective study included 147 patients (ccRCC=100, non-ccRCC=47) who underwent enhanced CT before surgery. CT images of the corticomedullary phase (CMP) were collected and features from the images were extracted. The data were randomly grouped into training and validation sets according to 7:3, and then the training set was normalized to extract the normalization rule for the training set, and then the rule was applied to the validation set. First, the T-test, T'-test or Wilcoxon rank-sum test were executed in the training set data to keep the statistically different parameters, and then the optimal features were picked based on the least absolute shrinkage and selection operator (LASSO) algorithm. Five machine learning (ML) models were trained to differentiate ccRCC from noccRCC, rad+cli nomogram was constructed based on clinical factors and radscore (radiomics score), and the performance of the classifier was mainly measured by area under the curve (AUC), accuracy, sensitivity, specificity, and F1. Finally, the ROC curves and radar plots were plotted according to the five performance parameters.

Results: 1130 radiomics features were extracted, there were 736 radiomics features with statistical differences were obtained, and 4 features were finally selected after the LASSO algorithm. In the validation set of this study, three of the five ML models (logistic regression, random forest and support vector machine) had excellent performance (AUC 0.9-1.0) and two models (adaptive boosting and decision tree) had good performance (AUC 0.7-0.9), all with accuracy ≥ 0.800. The rad+cli nomogram performance was found excellent in both the training set (AUC = 0.982,0.963-1.000, accuracy=0.941) and the validation set (AUC = 0.949,0.885-1.000, accuracy=0.911). The random forest model with perfect performance (AUC = 1, accuracy=1) was found superior compared to the model performance in the training set. The rad+cli nomogram model prevailed in the comparison of the model's performance in the validation set.

Conclusion: The ML models and nomogram can be used to identify the relatively common pathological subtypes in clinic and provide some reference for clinicians.

Keywords: Radiomics, computed tomography, machine learning, nomogram, clinicians, ccRCC.

Graphical Abstract
[1]
Lambin P, Rios-Velazquez E, Leijenaar R, et al. Radiomics: extracting more information from medical images using advanced feature analysis. Eur J Cancer 2012; 48(4): 441-6.
[http://dx.doi.org/10.1016/j.ejca.2011.11.036] [PMID: 22257792]
[2]
Kumar V, Gu Y, Basu S, et al. Radiomics: the process and the challenges. Magn Reson Imaging 2012; 30(9): 1234-48.
[http://dx.doi.org/10.1016/j.mri.2012.06.010] [PMID: 22898692]
[3]
Dreyfuss LD, Abel EJ, Nystrom J, Stabo NJ, Pickhardt PJ, Lubner MG. Comparison of CT Texture Analysis Software Platforms in Renal Cell Carcinoma: Reproducibility of Numerical Values and Association With Histologic Subtype Across Platforms. AJR Am J Roentgenol 2021; 216(6): 1549-57.
[http://dx.doi.org/10.2214/AJR.20.22823] [PMID: 33852332]
[4]
Doshi AM, Tong A, Davenport MS, et al. Assessment of Renal Cell Carcinoma by Texture Analysis in Clinical Practice: A Six-Site, Six-Platform Analysis of Reliability. AJR Am J Roentgenol 2021; 217(5): 1132-40.
[http://dx.doi.org/10.2214/AJR.21.25456] [PMID: 33852355]
[5]
Zwanenburg A, Leger S, Vallières M, Löck S. Image biomarker standardisation initiative. arXiv preprint arXiv 2016; 1612.07003.
[http://dx.doi.org/10.1148/radiol.2020191145]
[6]
Lambin P, Leijenaar RTH, Deist TM, et al. Radiomics: The bridge between medical imaging and personalized medicine. Nat Rev Clin Oncol 2017; 14(12): 749-62.
[http://dx.doi.org/10.1038/nrclinonc.2017.141] [PMID: 28975929]
[7]
van Griethuysen JJM, Fedorov A, Parmar C, et al. Computational radiomics system to decode the radiographic phenotype. Cancer Res 2017; 77(21): e104-7.
[http://dx.doi.org/10.1158/0008-5472.CAN-17-0339] [PMID: 29092951]
[8]
Hsieh JJ, Purdue MP, Signoretti S, et al. Renal cell carcinoma. Nat Rev Dis Primers 2017; 3: 17009.
[http://dx.doi.org/10.1038/nrdp.2017.9] [PMID: 28276433]
[9]
Ljungberg B, Albiges L, Abu-Ghanem Y, et al. European Association of Urology Guidelines on Renal Cell Carcinoma: The 2022 Update. European Urology 2022; 82(4): 399-410.
[http://dx.doi.org/10.1016/j.eururo.2022.03.006]
[10]
Thorstenson A, Bergman M, Scherman-Plogell A-H, et al. Tumour characteristics and surgical treatment of renal cell carcinoma in Sweden 2005-2010: A population-based study from the national Swedish kidney cancer register. Scand J Urol 2014; 48(3): 231-8.
[http://dx.doi.org/10.3109/21681805.2013.864698] [PMID: 24666102]
[11]
Cheville JC, Lohse CM, Zincke H, Weaver AL, Blute ML. Comparisons of outcome and prognostic features among histologic subtypes of renal cell carcinoma. Am J Surg Pathol 2003; 27(5): 612-24.
[http://dx.doi.org/10.1097/00000478-200305000-00005] [PMID: 12717246]
[12]
Kocak B, Yardimci AH, Bektas CT, et al. Textural differences between renal cell carcinoma subtypes: Machine learning-based quantitative computed tomography texture analysis with independent external validation. Eur J Radiol 2018; 107: 149-57.
[http://dx.doi.org/10.1016/j.ejrad.2018.08.014] [PMID: 30292260]
[13]
Han S, Hwang SI, Lee HJ. The classification of renal cancer in 3-phase CT images using a deep learning method. J Digit Imaging 2019; 32(4): 638-43.
[http://dx.doi.org/10.1007/s10278-019-00230-2]
[14]
Li Z-C, Zhai G, Zhang J, et al. Differentiation of clear cell and non-clear cell renal cell carcinomas by all-relevant radiomics features from multiphase CT: A VHL mutation perspective. Eur Radiol 2019; 29(8): 3996-4007.
[http://dx.doi.org/10.1007/s00330-018-5872-6]
[15]
Wu K, Wu P, Yang K, et al. A comprehensive texture feature analysis framework of renal cell carcinoma: Pathological, prognostic, and genomic evaluation based on CT images. Eur Radiol 2022; 32(4): 2255-65.
[http://dx.doi.org/10.1007/s00330-021-08353-3]
[16]
Nazari M, Shiri I, Hajianfar G, et al. Noninvasive Fuhrman grading of clear cell renal cell carcinoma using computed tomography radiomic features and machine learning. Radiologia Medica 2020; 125(8): 754-62.
[http://dx.doi.org/10.1007/s11547-020-01169-z]
[17]
Shu J, Tang Y, Cui J, et al. Clear cell renal cell carcinoma: CT-based radiomics features for the prediction of Fuhrman grade. Eur J Radiol 2018; 109: 8-12.
[http://dx.doi.org/10.1016/j.ejrad.2018.10.005] [PMID: 30527316]
[18]
Stanzione A, Ricciardi C, Cuocolo R, et al. MRI radiomics for the prediction of fuhrman grade in clear cell renal cell carcinoma: A machine learning exploratory study. J Digit Imaging 2020; 33(4): 879-87.
[http://dx.doi.org/10.1007/s10278-020-00336-y] [PMID: 32314070]
[19]
Winham SJ, Freimuth RR, Biernacka JM. A weighted random forests approach to improve predictive performance. Stat Anal Data Min 2013; 6(6): 496-505.
[http://dx.doi.org/10.1002/sam.11196] [PMID: 24501613]
[20]
Robnik-Sikonja M. Improving random forests. 2004; pp. 359-70.
[http://dx.doi.org/10.1007/978-3-540-30115-8_34]
[21]
Amaratunga D, Cabrera J, Lee Y-S. Enriched random forests. Bioinformatics 2008; 24(18): 2010-4.
[http://dx.doi.org/10.1093/bioinformatics/btn356]
[22]
Huang Y, Liang C, He L, et al. Development and validation of a radiomics nomogram for preoperative prediction of lymph node metastasis in colorectal cancer. J Clin Oncol 2016; 34(18): 2157-64.
[23]
Tang T-Y, Jiao Y, Cui Y, et al. Penumbra-based radiomics signature as prognostic biomarkers for thrombolysis of acute ischemic stroke patients: a multicenter cohort study. J Neurol 2020; 267(5): 1454-63.
[http://dx.doi.org/10.1007/s00415-020-09713-7] [PMID: 32008072]
[24]
Meehan B, Appu S, St Croix B, Rak-Poznanska K, Klotz L, Rak J. Age-related properties of the tumour vasculature in renal cell carcinoma. BJU Int 2011; 107(3): 416-24.
[http://dx.doi.org/10.1111/j.1464-410X.2010.09569.x] [PMID: 20804487]
[25]
Jeong IG, Yoo CH, Song K, et al. Age at diagnosis is an independent predictor of small renal cell carcinoma recurrence-free survival. J Urol 2009; 182(2): 445-50.
[http://dx.doi.org/10.1016/j.juro.2009.04.013]
[26]
Canino C, Perrone L, Bosco E, et al. Targeting angiogenesis in metastatic renal cell carcinoma. Expert Rev Anticancer Ther 2019; 19(3): 245-57.
[http://dx.doi.org/10.1080/14737140.2019.1574574] [PMID: 30678509]
[27]
Wang JH, Min PQ, Wang PJ, et al. Dynamic CT evaluation of tumor vascularity in renal cell carcinoma. AJR Am J Roentgenol 2006; 186(5): 1423-30.
[http://dx.doi.org/10.2214/AJR.04.1408] [PMID: 16632740]
[28]
Sato M, Nakai Y, Nakata W, et al. Microvessel area of immature vessels is a prognostic factor in renal cell carcinoma: MVA correlates with prognosis. Int J Urol 2014; 21(2): 130-4.
[http://dx.doi.org/10.1111/iju.12231]
[29]
Van der Kwast T, Perez-Ordoñez B. Renal oncocytoma, yet another tumour that does not fit in the dualistic benign/malignant paradigm? J Clin Pathol 2007; 60(6): 585-6.
[http://dx.doi.org/10.1136/jcp.2006.044438] [PMID: 17557864]
[30]
Wu J, Zhu Q, Zhu W, Chen W, Wang S. Comparative study of CT appearances in renal oncocytoma and chromophobe renal cell carcinoma. Acta Radiol 2016; 57(4): 500-6.
[http://dx.doi.org/10.1177/0284185115585035] [PMID: 25972369]

© 2024 Bentham Science Publishers | Privacy Policy