1
|
Zheng Y, Shi H, Fu S, Wang H, Li X, Li Z, Hai B, Zhang J. Development and validation of a radiomics-based nomogram for predicting pathological grade of upper urinary tract urothelial carcinoma. BMC Cancer 2024; 24:1546. [PMID: 39696125 DOI: 10.1186/s12885-024-13325-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 12/10/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Upper urinary tract urothelial carcinoma (UTUC) is a rare and highly aggressive malignancy characterized by poor prognosis, making the accurate identification of high-grade (HG) UTUC essential for subsequent treatment strategies. This study aims to develop and validate a nomogram model using computed tomography urography (CTU) images to predict HG UTUC. METHODS A retrospective cohort study was conducted to include patients with UTUC who underwent radical nephroureterectomy and received a CTU examination prior to surgery. In the CTU images, tumor lesions located in the renal calyces, renal pelvis and ureter were segmented, and radiomics features from the unenhanced, medullary, and excretory phases were extracted. The maximum relevance minimum redundancy algorithm, least absolute shrinkage and selection operator, and various machine learning (ML) algorithms-including random forest, support vector machine, and eXtreme gradient boosting-were employed to select radiomics features and calculate radiomics scores. Logistic regression (LR) analysis was performed to identify the independent influencing factors of clinical baseline characteristics. Multiple datasets of radiomics features were constructed by integrating single-phase radiomics features with the most significant independent factor. Both LR and ML algorithms were utilized to develop predictive models. The area under the receiver operating characteristic curve (AUC values), accuracy, sensitivity, and specificity were assessed for model performance evaluation. Decision curve analysis was conducted to evaluate the clinical net benefits. RESULTS A total of 167 patients were enrolled in this study. Among them, 56 were diagnosed with low-grade UTUC (including papillary urothelial neoplasms with low malignant potential and low-grade urothelial carcinoma) as confirmed by postoperative pathological examination results, and 111 were of HG. These patients were randomly allocated to the training set and the validation set at a ratio of 7:3. The training set comprised 116 patients with a mean age of 63.5 ± 9.38 years and 38 males. The validation set comprised 51 patients with a mean age of 65.6 ± 11.1 years and 18 males. Hydronephrosis was identified as the most significant independent factor in the clinical baseline features. Models that include mixed-phase development achieve better performance compared to models that rely simply on single-phase development. The nomogram model had excellent predictive ability for HG UTUC, with AUC values of 0.844 and an accuracy of 0.793 in the validation sets. The nomogram model can enhance accuracy by 14.1% (79.3% vs. 65.2%) and sensitivity by 32.8% (93.2% vs. 60.4%) compared to urinary cytology. CONCLUSIONS This study developed a nomogram model, which significantly improved the diagnostic ability for HG UTUC compared to urinary cytology. Furthermore, the results of the decision curve analysis showed that the model had a net benefit and could provide a non-invasive and potentially diagnostic reference tool for HG UTUC.
Collapse
Affiliation(s)
- Yanghuang Zheng
- Department of Urology, The 2nd Affiliated Hospital of Kunming Medical University, No. 374 Dianmian Road, Kunming, Yunnan, 650101, People's Republic of China
- Department of Urology, The Second Hospital & Clinical Medical School, No. 82 Cui Ying Gate, Cheng Guan District, Lanzhou, Gansu, 730030, People's Republic of China
| | - Hongjin Shi
- Department of Urology, The 2nd Affiliated Hospital of Kunming Medical University, No. 374 Dianmian Road, Kunming, Yunnan, 650101, People's Republic of China
| | - Shi Fu
- Department of Urology, The 2nd Affiliated Hospital of Kunming Medical University, No. 374 Dianmian Road, Kunming, Yunnan, 650101, People's Republic of China
| | - Haifeng Wang
- Department of Urology, The 2nd Affiliated Hospital of Kunming Medical University, No. 374 Dianmian Road, Kunming, Yunnan, 650101, People's Republic of China
| | - Xin Li
- Department of Urology, The Cancer Hospital of Yunnan Province, No. 157 Jinbi Road, Kunming, Yunnan, 650118, People's Republic of China
| | - Zhi Li
- Department of Radiology, The First People's Hospital of Yunnan Province, No. 519 Kunzhou Road, Kunming, Yunnan, 650032, People's Republic of China
| | - Bing Hai
- Department of Respiratory Medicine, The 2nd Affiliated Hospital of Kunming Medical University, No. 374 Dianmian Road, Kunming, Yunnan, 650101, People's Republic of China.
| | - Jinsong Zhang
- Department of Urology, The 2nd Affiliated Hospital of Kunming Medical University, No. 374 Dianmian Road, Kunming, Yunnan, 650101, People's Republic of China.
| |
Collapse
|
2
|
Du J, Yang L, Zheng T, Liu D. Radiomics-based predictive model for preoperative risk classification of gastrointestinal stromal tumors using multiparametric magnetic resonance imaging: a retrospective study. RADIOLOGIE (HEIDELBERG, GERMANY) 2024; 64:166-176. [PMID: 39545983 DOI: 10.1007/s00117-024-01393-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 10/22/2024] [Indexed: 11/17/2024]
Abstract
OBJECTIVE The aim of this study was to develop and assess a radiomics model utilizing multiparametric magnetic resonance imaging (MRI) for the prediction of preoperative risk assessment in gastrointestinal stromal tumors (GISTs). MATERIAL AND METHODS An analysis was performed retrospectively on a group of 121 patients who received a histological diagnosis of GIST. They were then divided into two sets, with 85 in the training set and 36 in the validation set through random partitioning. Radiomics features from five MRI sequences, totaling 600 per patient, were extracted and subjected to feature selection utilizing a random forest algorithm. The discriminatory efficacy of the models was evaluated through receiver operating characteristic (ROC) and precision-recall (P-R) curve analyses. Model calibration was assessed via calibration curves. Subgroup analysis was performed on GISTs with a pathological maximum diameter equal to or less than 5 cm. Furtherly, Kaplan-Meier (K-M) curves and log-rank tests were used to compare the differences in survival status among different groups. Cox regression analysis was employed to identify independent prognostic factors and to construct a prognostic prediction model. RESULTS The clinical model (ModelC) displayed limited predictive efficacy in the context of GIST. Conversely, a radiomics model (ModelR) incorporating five parameters exhibited robust discriminative capabilities across both the training and validation sets, yielding area under the ROC curve (AUC) values of 0.893 (95% confidence interval [CI]: 0.807-0.949) and 0.855 (95% CI: 0.732-0.978), respectively. The F1max scores derived from the P‑R curves were 0.741 and 0.842 for the training and validation sets, respectively. Noteworthy was the exclusion of the two-dimensional tumor diameter and tumor location when constructing a hybrid model (ModelCR) that amalgamated radiomics and clinical features. ModelR demonstrated a substantially enhanced discriminative capacity in the training set compared with ModelC (p < 0.005). The net reclassification improvement (NRI) corroborated the superior performance of ModelR over ModelC, thereby enhancing diagnostic accuracy and clinical applicability. Patients in the high-risk group had significantly worse recurrence-free survival (RFS, p < 0.001) and overall survival (OS, p = 0.004), and the radiomics signature is an independent risk factor for RFS. The extended model incorporating the radiomics signature outperformed the baseline model in terms of risk assessment accuracy (p < 0.001). CONCLUSION Our investigation underscores the value of integrating radiomics analysis in conjunction with machine learning algorithms for prognostic risk stratification in GIST, presenting promising implications for informing clinical decision-making processes as well as optimizing management strategies.
Collapse
Affiliation(s)
- Juan Du
- Department of Medical Imaging, The First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Linsha Yang
- Department of Medical Imaging, The First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Tao Zheng
- Department of Medical Imaging, The First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Defeng Liu
- Department of Medical Imaging, The First Hospital of Qinhuangdao, Qinhuangdao, China.
| |
Collapse
|
3
|
Yang L, Zhang D, Zheng T, Liu D, Fang Y. Predicting the progression-free survival of gastrointestinal stromal tumors after imatinib therapy through multi-sequence magnetic resonance imaging. Abdom Radiol (NY) 2024; 49:801-813. [PMID: 38006414 DOI: 10.1007/s00261-023-04093-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/06/2023] [Accepted: 10/09/2023] [Indexed: 11/27/2023]
Abstract
PURPOSE Identify radiomics features associated with progression-free survival (PFS) and develop a predictive model for accurate PFS prediction in liver metastatic gastrointestinal stromal tumor patients (GIST). METHODS This multi-center retrospective study involved a comprehensive review of clinical and imaging data pertaining to 211 patients with gastrointestinal stromal tumors (GIST) from Center A and B. A total of 147 patients with hepatic metastatic GIST were included, with 102 cases as the training set and 45 cases as the external validation set. Radiomics features were extracted from non-enhanced MR images, specifically T2WI, DWI, and ADC, and relevant features were selected through LASSO-Cox regression. A radiomics nomogram model was then constructed using multivariable Cox regression analysis to effectively predict PFS. The models performance were evaluated with the concordance index (C-index). RESULTS The median age of the patients was 53 years, with 82 males and 65 females. A total of 21 radiomics features were selected to generate the radiomics signature. Radiomics signature slightly outperformed the clinical model but without significant difference (P > 0.05). Integrated radiomics signature with clinical features to build a nomogram, which exhibited high predictive performance in both training (C-index 0.757, 95% CI 0.692-0.822) and validation cohorts (C-index 0.718, 95% CI 0.618-0.818). Nomogram significantly outperformed the clinical model (P = 0.002 for training cohort, P < 0.001 for validation cohort). Stable long-term predictions shown by time-dependent ROC analysis (AUC 0.765-0.919 in training, 0.766-0.893 in validation). Multivariable Cox regression confirmed radiomics signature as an independent prognostic factor for preoperative survival prediction in hepatic metastatic GIST patients (HR = 3.973). CONCLUSION Radiomics signature is valuable for predicting PFS in metastatic GIST patients. Integrating imaging features and clinical factors into a comprehensive nomogram improves accuracy and effectiveness of survival prognosis, guiding personalized treatment strategies.
Collapse
Affiliation(s)
- Linsha Yang
- Department of Medical Imaging, The First Hospital of Qinhuangdao, Qinhuangdao, People's Republic of China
| | - Duo Zhang
- Department of Medical Imaging, Baoding No. 1 Central Hospital, Baoding, People's Republic of China
| | - Tao Zheng
- Department of Medical Imaging, The First Hospital of Qinhuangdao, Qinhuangdao, People's Republic of China
| | - Defeng Liu
- Department of Medical Imaging, The First Hospital of Qinhuangdao, Qinhuangdao, People's Republic of China.
| | - Yuan Fang
- Medical Imaging Center, Chongqing Yubei District People's Hospital, Chongqing, People's Republic of China.
| |
Collapse
|
4
|
Ye JY, Fang P, Peng ZP, Huang XT, Xie JZ, Yin XY. A radiomics-based interpretable model to predict the pathological grade of pancreatic neuroendocrine tumors. Eur Radiol 2024; 34:1994-2005. [PMID: 37658884 PMCID: PMC10873440 DOI: 10.1007/s00330-023-10186-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 07/22/2023] [Accepted: 08/04/2023] [Indexed: 09/05/2023]
Abstract
OBJECTIVES To develop a computed tomography (CT) radiomics-based interpretable machine learning (ML) model to predict the pathological grade of pancreatic neuroendocrine tumors (pNETs) in a non-invasive manner. METHODS Patients with pNETs who underwent contrast-enhanced abdominal CT between 2010 and 2022 were included in this retrospective study. Radiomics features were extracted, and five radiomics-based ML models, namely logistic regression (LR), random forest (RF), support vector machine (SVM), XGBoost, and GaussianNB, were developed. The performance of these models was evaluated using a time-independent testing set, and metrics such as sensitivity, specificity, accuracy, and the area under the receiver operating characteristic curve (AUC) were calculated. The accuracy of the radiomics model was compared to that of needle biopsy. The Shapley Additive Explanation (SHAP) tool and the correlation between radiomics and biological features were employed to explore the interpretability of the model. RESULTS A total of 122 patients (mean age: 50 ± 14 years; 53 male) were included in the training set, whereas 100 patients (mean age: 48 ± 13 years; 50 male) were included in the testing set. The AUCs for LR, SVM, RF, XGBoost, and GaussianNB were 0.758, 0.742, 0.779, 0.744, and 0.745, respectively, with corresponding accuracies of 73.0%, 70.0%, 77.0%, 71.9%, and 72.9%. The SHAP tool identified two features of the venous phase as the most significant, which showed significant differences among the Ki-67 index or mitotic count subgroups (p < 0.001). CONCLUSIONS An interpretable radiomics-based RF model can effectively differentiate between G1 and G2/3 of pNETs, demonstrating favorable interpretability. CLINICAL RELEVANCE STATEMENT The radiomics-based interpretable model developed in this study has significant clinical relevance as it offers a non-invasive method for assessing the pathological grade of pancreatic neuroendocrine tumors and holds promise as an important complementary tool to traditional tissue biopsy. KEY POINTS • A radiomics-based interpretable model was developed to predict the pathological grade of pNETs and compared with preoperative needle biopsy in terms of accuracy. • The model, based on CT radiomics, demonstrated favorable interpretability. • The radiomics model holds potential as a valuable complementary technique to preoperative needle biopsy; however, it should not be considered a replacement for biopsy.
Collapse
Affiliation(s)
- Jing-Yuan Ye
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-Sen University, No.58 Zhongshan Er Road, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Peng Fang
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-Sen University, No.58 Zhongshan Er Road, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Zhen-Peng Peng
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, No.58 Zhongshan Er Road, Guangzhou, Guangdong, People's Republic of China
| | - Xi-Tai Huang
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-Sen University, No.58 Zhongshan Er Road, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Jin-Zhao Xie
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-Sen University, No.58 Zhongshan Er Road, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Xiao-Yu Yin
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-Sen University, No.58 Zhongshan Er Road, Guangzhou, 510080, Guangdong, People's Republic of China.
| |
Collapse
|
5
|
Zheng Y, Shi H, Fu S, Wang H, Wang J, Li X, Li Z, Hai B, Zhang J. A computed tomography urography-based machine learning model for predicting preoperative pathological grade of upper urinary tract urothelial carcinoma. Cancer Med 2024; 13:e6901. [PMID: 38174830 PMCID: PMC10807597 DOI: 10.1002/cam4.6901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024] Open
Abstract
OBJECTIVES Development and validation of a computed tomography urography (CTU)-based machine learning (ML) model for prediction of preoperative pathology grade of upper urinary tract urothelial carcinoma (UTUC). METHODS A total of 140 patients with UTUC who underwent CTU examination from January 2017 to August 2023 were retrospectively enrolled. Tumor lesions on the unenhanced, medullary, and excretory periods of CTU were used to extract Features, respectively. Feature selection was screened by the Pearson and Spearman correlation analysis, least absolute shrinkage and selection operator algorithm, random forest (RF), support vector machine (SVM), and eXtreme Gradient Boosting (XGBoost). The logistic regression (LR) was used to screen for independent influencing factors of clinical baseline characteristics. Machine learning models based on different feature datasets were constructed and validated using algorithms such as LR, RF, SVM, and XGBoost. By computing the selected features, a radiomics score was generated, and a diverse feature dataset was constructed. Based on the training set, 16 ML models were created, and their performance was evaluated using the validation set for metrics including sensitivity, specificity, accuracy, area under the receiver operating characteristic curve (AUC), and others. RESULTS The training set consisted of 98 patients (mean age: 64.5 ± 10.5 years; 30 males), whereas the validation set consisted of 42 patients (mean age: 65.3 ± 9.78 years; 17 males). Hydronephrosis was the best independent influence factor (p < 0.05). The RF model had the best performance in predicting high-grade UTUC, with AUC of 0.914 (95% Confidence Interval [95%CI] 0.852-0.977) and 0.903 (95%CI 0.809-0.997) in the training set and validation set, and accuracy of 0.878 and 0.857, respectively. CONCLUSIONS An ML model based on the RF algorithm exhibits excellent predictive performance, offering a non-invasive approach for predicting preoperative high-grade UTUC.
Collapse
Affiliation(s)
- Yanghuang Zheng
- Department of UrologyThe 2nd Affiliated Hospital of Kunming Medical UniversityKunmingYunnanPeople's Republic of China
| | - Hongjin Shi
- Department of UrologyThe 2nd Affiliated Hospital of Kunming Medical UniversityKunmingYunnanPeople's Republic of China
| | - Shi Fu
- Department of UrologyThe 2nd Affiliated Hospital of Kunming Medical UniversityKunmingYunnanPeople's Republic of China
| | - Haifeng Wang
- Department of UrologyThe 2nd Affiliated Hospital of Kunming Medical UniversityKunmingYunnanPeople's Republic of China
| | - Jincheng Wang
- Department of UrologyThe First People's Hospital of Luliang CountyLijiangYunnanPeople's Republic of China
| | - Xin Li
- Department of UrologyThe Cancer Hospital of Yunnan ProvinceKunmingYunnanPeople's Republic of China
| | - Zhi Li
- Department of RadiologyThe First People's Hospital of Yunnan ProvinceKunmingYunnanPeople's Republic of China
| | - Bing Hai
- Department of Respiratory MedicineThe 2nd Affiliated Hospital of Kunming Medical UniversityKunmingYunnanPeople's Republic of China
| | - Jinsong Zhang
- Department of UrologyThe 2nd Affiliated Hospital of Kunming Medical UniversityKunmingYunnanPeople's Republic of China
| |
Collapse
|
6
|
Courlet P, Abler D, Guidi M, Girard P, Amato F, Vietti Violi N, Dietz M, Guignard N, Wicky A, Latifyan S, De Micheli R, Jreige M, Dromain C, Csajka C, Prior JO, Venkatakrishnan K, Michielin O, Cuendet MA, Terranova N. Modeling tumor size dynamics based on real-world electronic health records and image data in advanced melanoma patients receiving immunotherapy. CPT Pharmacometrics Syst Pharmacol 2023; 12:1170-1181. [PMID: 37328961 PMCID: PMC10431051 DOI: 10.1002/psp4.12983] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 04/28/2023] [Accepted: 05/04/2023] [Indexed: 06/18/2023] Open
Abstract
The development of immune checkpoint inhibitors (ICIs) has revolutionized cancer therapy but only a fraction of patients benefits from this therapy. Model-informed drug development can be used to assess prognostic and predictive clinical factors or biomarkers associated with treatment response. Most pharmacometric models have thus far been developed using data from randomized clinical trials, and further studies are needed to translate their findings into the real-world setting. We developed a tumor growth inhibition model based on real-world clinical and imaging data in a population of 91 advanced melanoma patients receiving ICIs (i.e., ipilimumab, nivolumab, and pembrolizumab). Drug effect was modeled as an ON/OFF treatment effect, with a tumor killing rate constant identical for the three drugs. Significant and clinically relevant covariate effects of albumin, neutrophil to lymphocyte ratio, and Eastern Cooperative Oncology Group (ECOG) performance status were identified on the baseline tumor volume parameter, as well as NRAS mutation on tumor growth rate constant using standard pharmacometric approaches. In a population subgroup (n = 38), we had the opportunity to conduct an exploratory analysis of image-based covariates (i.e., radiomics features), by combining machine learning and conventional pharmacometric covariate selection approaches. Overall, we demonstrated an innovative pipeline for longitudinal analyses of clinical and imaging RWD with a high-dimensional covariate selection method that enabled the identification of factors associated with tumor dynamics. This study also provides a proof of concept for using radiomics features as model covariates.
Collapse
Affiliation(s)
- Perrine Courlet
- Precision Oncology Center, Department of OncologyLausanne University Hospital and University of LausanneLausanneSwitzerland
- Centre for Research and Innovation in Clinical Pharmaceutical SciencesLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Daniel Abler
- Precision Oncology Center, Department of OncologyLausanne University Hospital and University of LausanneLausanneSwitzerland
- Institute of Informatics, School of Management, University of Applied Sciences Western Switzerland (HES‐SO)SierreSwitzerland
| | - Monia Guidi
- Centre for Research and Innovation in Clinical Pharmaceutical SciencesLausanne University Hospital and University of LausanneLausanneSwitzerland
- Service of Clinical PharmacologyLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Pascal Girard
- Merck Institute of Pharmacometrics, Ares Trading S.A. (an affiliate of Merck KGaA, Darmstadt, Germany)LausanneSwitzerland
| | - Federico Amato
- Swiss Data Science Centre, École Polytechnique Fédérale de Lausanne (EPFL) and Eidgenössische Technische Hochschule Zurich (ETH)ZurichSwitzerland
| | - Naik Vietti Violi
- Department of Radiology and Interventional RadiologyLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Matthieu Dietz
- Nuclear Medicine and Molecular Imaging DepartmentLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Nicolas Guignard
- Department of Radiology and Interventional RadiologyLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Alexandre Wicky
- Precision Oncology Center, Department of OncologyLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Sofiya Latifyan
- Department of OncologyLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Rita De Micheli
- Department of OncologyLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Mario Jreige
- Nuclear Medicine and Molecular Imaging DepartmentLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Clarisse Dromain
- Department of Radiology and Interventional RadiologyLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Chantal Csajka
- Centre for Research and Innovation in Clinical Pharmaceutical SciencesLausanne University Hospital and University of LausanneLausanneSwitzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of GenevaUniversity of LausanneGenevaSwitzerland
- School of Pharmaceutical SciencesUniversity of GenevaGenevaSwitzerland
| | - John O. Prior
- Nuclear Medicine and Molecular Imaging DepartmentLausanne University Hospital and University of LausanneLausanneSwitzerland
| | | | - Olivier Michielin
- Precision Oncology Center, Department of OncologyLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Michel A. Cuendet
- Precision Oncology Center, Department of OncologyLausanne University Hospital and University of LausanneLausanneSwitzerland
- Swiss Institute of Bioinformatics, University of LausanneLausanneSwitzerland
- Department of Physiology and Biophysics, Weill Cornell MedicineNew YorkNew YorkUSA
| | - Nadia Terranova
- Merck Institute of Pharmacometrics, Ares Trading S.A. (an affiliate of Merck KGaA, Darmstadt, Germany)LausanneSwitzerland
| |
Collapse
|
7
|
Abler D, Courlet P, Dietz M, Gatta R, Girard P, Munafo A, Wicky A, Jreige M, Guidi M, Latifyan S, De Micheli R, Csajka C, Prior JO, Michielin O, Terranova N, Cuendet MA. Semiautomated Pipeline to Quantify Tumor Evolution From Real-World Positron Emission Tomography/Computed Tomography Imaging. JCO Clin Cancer Inform 2023; 7:e2200126. [PMID: 37146261 PMCID: PMC10281365 DOI: 10.1200/cci.22.00126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 11/04/2022] [Accepted: 02/03/2023] [Indexed: 05/07/2023] Open
Abstract
PURPOSE A semiautomated pipeline for the collection and curation of free-text and imaging real-world data (RWD) was developed to quantify cancer treatment outcomes in large-scale retrospective real-world studies. The objectives of this article are to illustrate the challenges of RWD extraction, to demonstrate approaches for quality assurance, and to showcase the potential of RWD for precision oncology. METHODS We collected data from patients with advanced melanoma receiving immune checkpoint inhibitors at the Lausanne University Hospital. Cohort selection relied on semantically annotated electronic health records and was validated using process mining. The selected imaging examinations were segmented using an automatic commercial software prototype. A postprocessing algorithm enabled longitudinal lesion identification across imaging time points and consensus malignancy status prediction. Resulting data quality was evaluated against expert-annotated ground-truth and clinical outcomes obtained from radiology reports. RESULTS The cohort included 108 patients with melanoma and 465 imaging examinations (median, 3; range, 1-15 per patient). Process mining was used to assess clinical data quality and revealed the diversity of care pathways encountered in a real-world setting. Longitudinal postprocessing greatly improved the consistency of image-derived data compared with single time point segmentation results (classification precision increased from 53% to 86%). Image-derived progression-free survival resulting from postprocessing was comparable with the manually curated clinical reference (median survival of 286 v 336 days, P = .89). CONCLUSION We presented a general pipeline for the collection and curation of text- and image-based RWD, together with specific strategies to improve reliability. We showed that the resulting disease progression measures match reference clinical assessments at the cohort level, indicating that this strategy has the potential to unlock large amounts of actionable retrospective real-world evidence from clinical records.
Collapse
Affiliation(s)
- Daniel Abler
- Department of Oncology, Precision Oncology Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Institute of Informatics, School of Management, University of Applied Sciences Western Switzerland (HES-SO), Sierre, Switzerland
| | - Perrine Courlet
- Department of Oncology, Precision Oncology Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Centre for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Matthieu Dietz
- Nuclear Medicine and Molecular Imaging Department, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- INSERM U1060, CarMeN Laboratory, University of Lyon, Lyon, France
| | - Roberto Gatta
- Department of Oncology, Precision Oncology Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Pascal Girard
- Translational Medicine, Merck Institute of Pharmacometrics, Lausanne, Switzerland, an Affiliate of Merck KGaA, Darmstadt, Germany
| | - Alain Munafo
- Translational Medicine, Merck Institute of Pharmacometrics, Lausanne, Switzerland, an Affiliate of Merck KGaA, Darmstadt, Germany
| | - Alexandre Wicky
- Department of Oncology, Precision Oncology Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Mario Jreige
- Nuclear Medicine and Molecular Imaging Department, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Monia Guidi
- Centre for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Service of Clinical Pharmacology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Sofiya Latifyan
- Service of Medical Oncology, Department of Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Rita De Micheli
- Service of Medical Oncology, Department of Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Chantal Csajka
- Centre for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, University of Lausanne, Geneva, Switzerland
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
| | - John O. Prior
- Nuclear Medicine and Molecular Imaging Department, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Olivier Michielin
- Department of Oncology, Precision Oncology Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Nadia Terranova
- Translational Medicine, Merck Institute of Pharmacometrics, Lausanne, Switzerland, an Affiliate of Merck KGaA, Darmstadt, Germany
| | - Michel A. Cuendet
- Department of Oncology, Precision Oncology Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, University of Lausanne, Lausanne, Switzerland
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY
| |
Collapse
|
8
|
Inoue A, Ota S, Yamasaki M, Batsaikhan B, Furukawa A, Watanabe Y. Gastrointestinal stromal tumors: a comprehensive radiological review. Jpn J Radiol 2022; 40:1105-1120. [PMID: 35809209 DOI: 10.1007/s11604-022-01305-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 06/08/2022] [Indexed: 11/29/2022]
Abstract
Gastrointestinal stromal tumors (GISTs) originating from the interstitial cells of Cajal in the muscularis propria are the most common mesenchymal tumor of the gastrointestinal tract. Multiple modalities, including computed tomography (CT), magnetic resonance imaging (MRI), fluorodeoxyglucose positron emission tomography, ultrasonography, digital subtraction angiography, and endoscopy, have been performed to evaluate GISTs. CT is most frequently used for diagnosis, staging, surveillance, and response monitoring during molecularly targeted therapy in clinical practice. The diagnosis of GISTs is sometimes challenging because of the diverse imaging findings, such as anatomical location (esophagus, stomach, duodenum, small bowel, colorectum, appendix, and peritoneum), growth pattern, and enhancement pattern as well as the presence of necrosis, calcification, ulceration, early venous return, and metastasis. Imaging findings of GISTs treated with antineoplastic agents are quite different from those of other neoplasms (e.g. adenocarcinomas) because only subtle changes in size are seen even in responsive lesions. Furthermore, the recurrence pattern of GISTs is different from that of other neoplasms. This review discusses the advantages and disadvantages of each imaging modality, describes imaging findings obtained before and after treatment, presents a few cases of complicated GISTs, and discusses recent investigations performed using CT and MRI to predict histological risk grade, gene mutations, and patient outcomes.
Collapse
Affiliation(s)
- Akitoshi Inoue
- Department of Radiology, Shiga University of Medical Science, Seta, Tsukinowa-cho, Otsu, Shiga, 520-2192, Japan. .,Department of Radiology, Mayo Clinic, Rochester, MN, USA.
| | - Shinichi Ota
- Department of Radiology, Nagahama Red Cross Hospital, Shiga, Japan
| | - Michio Yamasaki
- Department of Radiology, Kohka Public Hospital, Shiga, Japan
| | - Bolorkhand Batsaikhan
- Graduate School of Human Health Sciences, Department of Radiological Science, Tokyo Metropolitan University, Tokyo, Japan
| | - Akira Furukawa
- Graduate School of Human Health Sciences, Department of Radiological Science, Tokyo Metropolitan University, Tokyo, Japan
| | - Yoshiyuki Watanabe
- Department of Radiology, Shiga University of Medical Science, Seta, Tsukinowa-cho, Otsu, Shiga, 520-2192, Japan
| |
Collapse
|
9
|
Imaging Diagnosis of Primary Liver Cancer Using Magnetic Resonance Dilated Weighted Imaging and the Treatment Effect of Sorafenib. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:8586943. [PMID: 35799672 PMCID: PMC9256338 DOI: 10.1155/2022/8586943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/25/2022] [Accepted: 05/28/2022] [Indexed: 11/21/2022]
Abstract
Objective This work explores the application value of dilated weighted imaging (DWI) in the diagnosis of primary liver cancer (PLC) and the effect of sorafenib in the treatment of PLC. Methods 88 patients with PLC who were treated in The First Affiliated Hospital of Northwest University from March 2019 to March 2021 were selected and randomly rolled into an experimental group and a control group, with 44 cases in each group. Patients in both groups were treated with transcatheter arterial chemoembolization (TACE), and the patients in the experimental group were treated with oral sorafenib on the basis of TACE. The indicators of complications, short-term efficacy (STE), and long-term efficacy (LTE) of the two groups were observed. All patients received DWI and magnetic resonance (MR) plain scan. The diagnostic accuracy and misdiagnosis rate of the two methods in diagnosing the PLC were compared. Results The accuracy, specificity, and sensitivity of MR plain scan were 68%, 88%, and 89%, respectively, while those of DWI were 96%, 95%, and 94.2%, respectively. It indicated that the accuracy, specificity, and sensitivity of DWI in diagnosing lesions were better than those of MR plain scan, especially the diagnostic accuracy (P < 0.05). The objective response rate (ORR) and disease control rate (DCR) of the STE in the experimental group were 30% and 97%, respectively, and those in the control group were 6% and 54.5%, respectively. The experimental group's mean progression-free survival (mPFS) and mean overall survival (mOS) were 12 and 25 months, respectively, while the control group's were 8 and 19 months, respectively. It was concluded that the mPFS and mOS of patients receiving TACE combined with oral sorafenib were much higher than those receiving TACE only (P < 0.05). Conclusion DWI and TACE combined with sorafenib had high application value in the diagnosis and treatment of PLC.
Collapse
|
10
|
Prediction of the Ki-67 expression level and prognosis of gastrointestinal stromal tumors based on CT radiomics nomogram. Int J Comput Assist Radiol Surg 2022; 17:1167-1175. [PMID: 35195831 DOI: 10.1007/s11548-022-02575-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/31/2022] [Indexed: 11/05/2022]
Abstract
PURPOSE To build and validate a radiomics nomogram integrated with the radiomics signature and subjective CT characteristics to predict the Ki-67 expression level of gastrointestinal stromal tumors (GISTs). Moreover, the purpose was to compare the performance of pathological Ki-67 expression level with predicted Ki-67 expression level in estimating the prognosis of GISTs patients. METHODS According to pathological results, patients were classified into high-Ki-67 labeling index group (Ki-67 LI ≥ 5%) and low-Ki-67 LI group (Ki-67 LI < 5%). Radiomics features extracted from contrast-enhanced CT(CECT) images were selected and classified to build a radiomics signature. A combined model was built by incorporating radiomics signature and determinant subjective CT characteristics using multivariate logistic regression analysis. The diagnostic performance of the radiomics signature, subjective CT model and combined model were explored by receiver operating characteristic (ROC) curve analysis and Delong test. The model with best diagnostic performance was then set up for the prediction nomogram. Recurrence-free survival (RFS) rates were compared utilizing Kaplan-Meier curve. RESULTS The generated combined model yielded the best diagnostic performance with area under the curve (AUC) values of 0.738 [95% confidence interval (CI): 0.669-0.807] and 0.772 (95% CI 0.683-0.860) in the training set and testing set respectively. The nomogram based on the combined model demonstrated good calibration in the training set and testing set (both P > 0.05). Patients of high-Ki-67 LI group predicted by our nomogram had a poorer RFS than patients of low-Ki-67 LI group (P < 0.0001). CONCLUSION This radiomics nomogram based on CECT had a satisfactory performance in predicting both the Ki-67 expression level and prognosis noninvasively in patients with GISTs, which may serve as an effective imaging tool that can assist in guiding personalized clinical treatment.
Collapse
|
11
|
You J, Yin J. Performances of Whole Tumor Texture Analysis Based on MRI: Predicting Preoperative T Stage of Rectal Carcinomas. Front Oncol 2021; 11:678441. [PMID: 34414105 PMCID: PMC8369414 DOI: 10.3389/fonc.2021.678441] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 07/19/2021] [Indexed: 12/29/2022] Open
Abstract
Objective To determine whether there is a correlation between texture features extracted from high-resolution T2-weighted imaging (HR-T2WI) or apparent diffusion coefficient (ADC) maps and the preoperative T stage (stages T1–2 versus T3–4) in rectal carcinomas. Materials and Methods One hundred and fifty four patients with rectal carcinomas who underwent preoperative HR-T2WI and diffusion-weighted imaging were enrolled. Patients were divided into training (n = 89) and validation (n = 65) cohorts. 3D Slicer was used to segment the entire volume of interest for whole tumors based on HR-T2WI and ADC maps. The least absolute shrinkage and selection operator (LASSO) was performed to select feature. The significantly difference was tested by the independent sample t-test and Mann-Whitney U test. The support vector machine (SVM) model was used to develop classification models. The correlation between features and T stage was assessed by Spearman’s correlation analysis. Multivariate logistic regression analysis was performed to identify independent predictors of tumor invasion. The performance of classifiers was evaluated by the receiver operating characteristic (ROC) curves. Results The wavelet HHH NGTDM strength (RS = -0.364, P < 0.001) from HR-T2WI was an independent predictor of stage T3–4 tumors. The shape maximum 2D diameter column (RS = 0.431, P < 0.001), log σ = 5.0 mm 3D first-order maximum (RS = 0.276, P = 0.009), and log σ = 5.0 mm 3D first-order interquartile range (RS = -0.229, P = 0.032) from ADC maps were independent predictors. In training cohorts, the classification models from HR-T2WI, ADC maps and the combination of two achieved the area under the ROC curves (AUCs) of 0.877, 0.902 and 0.941, with the accuracy of 79.78%, 89.86% and 89.89%, respectively. In validation cohorts, the three models achieved AUCs of 0.845, 0.881 and 0.910, with the accuracy of 78.46%, 83.08% and 87.69%, respectively. Conclusions Texture analysis based on ADC maps shows more potential than HR-T2WI in identifying preoperative T stage in rectal carcinomas. The combined application of HR-T2WI and ADC maps may help to improve the accuracy of preoperative diagnosis of rectal cancer invasion.
Collapse
Affiliation(s)
- Jia You
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jiandong Yin
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
12
|
Zhou Z, Lu J, Morelli JN, Hu D, Li Z, Xiao P, Hu X, Shen Y. Utility of noncontrast MRI in the detection and risk grading of gastrointestinal stromal tumor: a comparison with contrast-enhanced CT. Quant Imaging Med Surg 2021; 11:2453-2464. [PMID: 34079715 PMCID: PMC8107337 DOI: 10.21037/qims-20-578] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 01/27/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND Recently developed adjuvant therapies for gastrointestinal stromal tumor (GIST) have been shown to improve patient survival. Guidelines currently recommend contrast-enhanced computed tomography (CECT) for GIST detection and surveillance. Patients with moderate-to-high risk GISTs require more frequent surveillance due to a higher 5-year recurrence rate. Our study aimed to compare noncontrast magnetic resonance imaging (MRI) with CECT for GIST detection, and evaluate volumetric apparent diffusion coefficients (ADCs) for risk stratification of GIST. METHODS We retrospectively enrolled 83 patients with histopathologically confirmed GISTs for lesion detection efficiency analysis between noncontrast MRI and matched CECT studies. A 5-point scale was used by two independent reviewers to determine if the lesion was present or absent. Another cohort, comprising 28 patients with pathologically confirmed primary GISTs, was further screened for risk stratification, with a comparison of volumetric ADC parameters between the pathologically very-low-to-low risk and moderate-to-high risk GIST patients. RESULTS For identifying GISTs, the sensitivity and specificity of noncontrast MRI were 83.6% and 89.3% for reader 1 respectively, and 81.8% and 92.9% for reader 2 respectively; the sensitivity and specificity of CECT were 76.4% and 89.3% for reader 1 respectively, and 76.4 and 78.6% for reader 2 respectively. Tumor volumetric ADC histogram parameters, including ADCmax, ADCstdev, 90th and 95th percentiles, inhomogeneity, and entropy, were positively correlated with a higher risk grade of GIST (r=0.421-0.758). The receiver operator characteristic curve analysis showed ADCmax achieved the highest area under the curve value of 0.938 for discriminating very-low-to-low risk versus moderate-to-high risk GISTs. CONCLUSIONS Noncontrast MRI was an efficient technique for identifying GIST patients. The combination of CECT and noncontrast MRI can improve the reliability of diagnosis. For patients with contraindications to CECT, noncontrast MRI may be a comparable alternative. Volumetric ADC histogram parameters may be useful in differentiating very-low-to-low risk from moderate-to-high risk primary GISTs.
Collapse
Affiliation(s)
- Ziling Zhou
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingyu Lu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - John N. Morelli
- Department of Radiology, St. John’s Medical Center, Tulsa, OK, USA
| | - Daoyu Hu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhen Li
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Xiao
- Biomedical Engineering Department, Huazhong University of Science and Technology, Wuhan, China
| | - Xuemei Hu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yaqi Shen
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
13
|
Mao H, Zhang B, Zou M, Huang Y, Yang L, Wang C, Pang P, Zhao Z. MRI-Based Radiomics Models for Predicting Risk Classification of Gastrointestinal Stromal Tumors. Front Oncol 2021; 11:631927. [PMID: 34041017 PMCID: PMC8141866 DOI: 10.3389/fonc.2021.631927] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 04/13/2021] [Indexed: 01/04/2023] Open
Abstract
Background We conduct a study in developing and validating four MRI-based radiomics models to preoperatively predict the risk classification of gastrointestinal stromal tumors (GISTs). Methods Forty-one patients (low-risk = 17, intermediate-risk = 13, high-risk = 11) underwent MRI before surgery between September 2013 and March 2019 in this retrospective study. The Kruskal–Wallis test with Bonferonni correction and variance threshold was used to select appropriate features, and the Random Forest model (three classification model) was used to select features among the high-risk, intermediate-risk, and low-risk of GISTs. The predictive performance of the models built by the Random Forest was estimated by a 5-fold cross validation (5FCV). Their performance was estimated using the receiver operating characteristic (ROC) curve, summarized as the area under the ROC curve (AUC). Area under the curve (AUC), accuracy, sensitivity, and specificity for risk classification were reported. Linear discriminant analysis (LDA) was used to assess the discriminative ability of these radiomics models. Results The high-risk, intermediate-risk, and low-risk of GISTs were well classified by radiomics models, the micro-average of ROC curves was 0.85, 0.81, 0.87 and 0.94 for T1WI, T2WI, ADC and combined three MR sequences. And ROC curves achieved excellent AUCs for T1WI (0.85, 0.75 and 0.82), T2WI (0.69, 0.78 and 0.78), ADC (0.85, 0.77 and 0.80) and combined three MR sequences (0.96, 0.92, 0.81) for the diagnosis of high-risk, intermediate-risk, and low-risk of GISTs, respectively. In addition, LDA demonstrated the different risk of GISTs were correctly classified by radiomics analysis (61.0% for T1WI, 70.7% for T2WI, 83.3% for ADC, and 78.9% for the combined three MR sequences). Conclusions Radiomics models based on a single sequence and combined three MR sequences can be a noninvasive method to evaluate the risk classification of GISTs, which may help the treatment of GISTs patients in the future.
Collapse
Affiliation(s)
- Haijia Mao
- Department of Radiology, Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, China
| | - Bingqian Zhang
- Department of Radiology, Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, China
| | - Mingyue Zou
- Department of Radiology, Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, China
| | - Yanan Huang
- Department of Radiology, Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, China
| | - Liming Yang
- Department of Radiology, Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, China
| | - Cheng Wang
- Department of Pathology, Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, China
| | - PeiPei Pang
- Department of Pharmaceuticals Diagnosis, GE Healthcare, Hangzhou, China
| | - Zhenhua Zhao
- Department of Radiology, Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, China
| |
Collapse
|
14
|
Zhang S, Sun H, Su X, Yang X, Wang W, Wan X, Tan Q, Chen N, Yue Q, Gong Q. Automated machine learning to predict the co-occurrence of isocitrate dehydrogenase mutations and O 6 -methylguanine-DNA methyltransferase promoter methylation in patients with gliomas. J Magn Reson Imaging 2021; 54:197-205. [PMID: 33393131 DOI: 10.1002/jmri.27498] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 02/05/2023] Open
Abstract
Combining isocitrate dehydrogenase mutation (IDHmut) with O6 -methylguanine-DNA methyltransferase promoter methylation (MGMTmet) has been identified as a critical prognostic molecular marker for gliomas. The aim of this study was to determine the ability of glioma radiomics features from magnetic resonance imaging (MRI) to predict the co-occurrence of IDHmut and MGMTmet by applying the tree-based pipeline optimization tool (TPOT), an automated machine learning (autoML) approach. This was a retrospective study, in which 162 patients with gliomas were evaluated, including 58 patients with co-occurrence of IDHmut and MGMTmet and 104 patients with other status comprising: IDH wildtype and MGMT unmethylated (n = 67), IDH wildtype and MGMTmet (n = 36), and IDHmut and MGMT unmethylated (n = 1). Three-dimensional (3D) T1-weighted images, gadolinium-enhanced 3D T1-weighted images (Gd-3DT1WI), T2-weighted images, and fluid-attenuated inversion recovery (FLAIR) images acquired at 3.0 T were used. Radiomics features were extracted from FLAIR and Gd-3DT1WI images. The TPOT was employed to generate the best machine learning pipeline, which contains both feature selector and classifier, based on input feature sets. A 4-fold cross-validation was used to evaluate the performance of automatically generated models. For each iteration, the training set included 121 subjects, while the test set included 41 subjects. Student's t-test or a chi-square test was applied on different clinical characteristics between two groups. Sensitivity, specificity, accuracy, kappa score, and AUC were used to evaluate the performance of TPOT-generated models. Finally, we compared the above metrics of TPOT-generated models to identify the best-performing model. Patients' ages and grades between two groups were significantly different (p = 0.002 and p = 0.000, respectively). The 4-fold cross-validation showed that gradient boosting classifier trained on shape and textual features from the Laplacian-of-Gaussian-filtered Gd-3DT1 achieved the best performance (average sensitivity = 81.1%, average specificity = 94%, average accuracy = 89.4%, average kappa score = 0.76, average AUC = 0.951). Using autoML based on radiomics features from MRI, a high discriminatory accuracy was achieved for predicting co-occurrence of IDHmut and MGMTmet in gliomas. LEVEL OF EVIDENCE: 3 TECHNICAL EFFICACY STAGE: 3.
Collapse
Affiliation(s)
- Simin Zhang
- Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology, West China Hospital of Sichuan University, Chengdu, China.,Huaxi Glioma Center, West China Hospital of Sichuan University, Chengdu, China
| | - Huaiqiang Sun
- Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Xiaorui Su
- Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology, West China Hospital of Sichuan University, Chengdu, China.,Huaxi Glioma Center, West China Hospital of Sichuan University, Chengdu, China
| | - Xibiao Yang
- Huaxi Glioma Center, West China Hospital of Sichuan University, Chengdu, China.,Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Weina Wang
- Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Xinyue Wan
- Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Qiaoyue Tan
- Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology, West China Hospital of Sichuan University, Chengdu, China.,Division of Radiation Physics, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Ni Chen
- Department of Pathology, West China Hospital of Sichuan University, Chengdu, China
| | - Qiang Yue
- Huaxi Glioma Center, West China Hospital of Sichuan University, Chengdu, China.,Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
15
|
Cannella R, La Grutta L, Midiri M, Bartolotta TV. New advances in radiomics of gastrointestinal stromal tumors. World J Gastroenterol 2020; 26:4729-4738. [PMID: 32921953 PMCID: PMC7459199 DOI: 10.3748/wjg.v26.i32.4729] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/16/2020] [Accepted: 08/01/2020] [Indexed: 02/06/2023] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are uncommon neoplasms of the gastrointestinal tract with peculiar clinical, genetic, and imaging characteristics. Preoperative knowledge of risk stratification and mutational status is crucial to guide the appropriate patients’ treatment. Predicting the clinical behavior and biological aggressiveness of GISTs based on conventional computed tomography (CT) and magnetic resonance imaging (MRI) evaluation is challenging, unless the lesions have already metastasized at the time of diagnosis. Radiomics is emerging as a promising tool for the quantification of lesion heterogeneity on radiological images, extracting additional data that cannot be assessed by visual analysis. Radiomics applications have been explored for the differential diagnosis of GISTs from other gastrointestinal neoplasms, risk stratification and prediction of prognosis after surgical resection, and evaluation of mutational status in GISTs. The published researches on GISTs radiomics have obtained excellent performance of derived radiomics models on CT and MRI. However, lack of standardization and differences in study methodology challenge the application of radiomics in clinical practice. The purpose of this review is to describe the new advances of radiomics applied to CT and MRI for the evaluation of gastrointestinal stromal tumors, discuss the potential clinical applications that may impact patients’ management, report limitations of current radiomics studies, and future directions.
Collapse
Affiliation(s)
- Roberto Cannella
- Section of Radiology - BiND, University Hospital “Paolo Giaccone”, Palermo 90127, Italy
| | - Ludovico La Grutta
- Section of Radiology - BiND, University Hospital “Paolo Giaccone”, Palermo 90127, Italy
| | - Massimo Midiri
- Section of Radiology - BiND, University Hospital “Paolo Giaccone”, Palermo 90127, Italy
| | - Tommaso Vincenzo Bartolotta
- Section of Radiology - BiND, University Hospital “Paolo Giaccone”, Palermo 90127, Italy
- Department of Radiology, Fondazione Istituto Giuseppe Giglio, Ct.da Pietrapollastra, Cefalù (Palermo) 90015, Italy
| |
Collapse
|
16
|
Wang ZL, Mao LL, Zhou ZG, Si L, Zhu HT, Chen X, Zhou MJ, Sun YS, Guo J. Pilot Study of CT-Based Radiomics Model for Early Evaluation of Response to Immunotherapy in Patients With Metastatic Melanoma. Front Oncol 2020; 10:1524. [PMID: 32984000 PMCID: PMC7479823 DOI: 10.3389/fonc.2020.01524] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/16/2020] [Indexed: 12/17/2022] Open
Abstract
Objective: Determine the performance of a computed tomography (CT) -based radiomics model in predicting early response to immunotherapy in patients with metastatic melanoma. Methods: This retrospective study examined 50 patients with metastatic melanoma who received immunotherapy treatment in our hospital with an anti-programmed cell death-1 (PD-1) agent or an inhibitor of cytotoxic T lymphocyte antigen-4 (CTLA-4). Thirty-four patients who received an anti-PD-1 agent were in the training sample and 16 patients who received a CTLA-4 inhibitor were in the validation sample. Patients with true progressive disease (PD) were in the poor response group, and those with pseudoprogression, complete response (CR), partial response (PR), or stable disease (SD) were in the good response group. CT images were examined at baseline and after the first and second cycles of treatment, and the imaging data were extracted for radiomics modeling. Results: The radiomics model based on pre-treatment, post-treatment, and delta features provided the best results for predicting response to immunotherapy. Receiver operating characteristic (ROC) analysis for good response indicated an area under the curve (AUC) of 0.882 for the training group and an AUC of 0.857 for the validation group. The sensitivity, specificity, and accuracy of model were 85.70% (6/7), 66.70% (6/9), and 75% (12/16) for predicting a good response. Conclusion: A CT-based radiomics model for metastatic melanoma has the potential to predict early response to immunotherapy and to identify pseudoprogression.
Collapse
Affiliation(s)
- Zhi-Long Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Li-Li Mao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhi-Guo Zhou
- School of Computer Science and Mathematics, University of Central Missouri, Warrensburg, MO, United States
| | - Lu Si
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hai-Tao Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xi Chen
- School of Information and Communication Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Mei-Juan Zhou
- School of Information and Communication Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Ying-Shi Sun
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jun Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|