1
|
Ellmann S, von Rohr F, Komina S, Bayerl N, Amann K, Polifka I, Hartmann A, Sikic D, Wullich B, Uder M, Bäuerle T. Tumor grade-titude: XGBoost radiomics paves the way for RCC classification. Eur J Radiol 2025; 188:112146. [PMID: 40334367 DOI: 10.1016/j.ejrad.2025.112146] [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: 01/27/2025] [Revised: 04/21/2025] [Accepted: 04/28/2025] [Indexed: 05/09/2025]
Abstract
This study aimed to develop and evaluate a non-invasive XGBoost-based machine learning model using radiomic features extracted from pre-treatment CT images to differentiate grade 4 renal cell carcinoma (RCC) from lower-grade tumours. A total of 102 RCC patients who underwent contrast-enhanced CT scans were included in the analysis. Radiomic features were extracted, and a two-step feature selection methodology was applied to identify the most relevant features for classification. The XGBoost model demonstrated high performance in both training (AUC = 0.87) and testing (AUC = 0.92) sets, with no significant difference between the two (p = 0.521). The model also exhibited high sensitivity, specificity, positive predictive value, and negative predictive value. The selected radiomic features captured both the distribution of intensity values and spatial relationships, which may provide valuable insights for personalized treatment decision-making. Our findings suggest that the XGBoost model has the potential to be integrated into clinical workflows to facilitate personalized adjuvant immunotherapy decision-making, ultimately improving patient outcomes. Further research is needed to validate the model in larger, multicentre cohorts and explore the potential of combining radiomic features with other clinical and molecular data.
Collapse
Affiliation(s)
- Stephan Ellmann
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; Radiologisch-Nuklearmedizinisches Zentrum (RNZ), Martin-Richter-Straße 43, 90489 Nürnberg, Germany.
| | - Felicitas von Rohr
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Selim Komina
- Institute of Pathology, Faculty of Medicine, Ss Cyril and Methodius University ul. 50 Divizija bb 1000 Skopje, North Macedonia
| | - Nadine Bayerl
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Kerstin Amann
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; BZKF: Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Iris Polifka
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; Humanpathologie Dr. Weiß MVZ GmbH, Am Weichselgarten 30a, 91058 Erlangen-Tennenlohe, Germany
| | - Arndt Hartmann
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; Comprehensive Cancer Center Erlangen - EMD, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; BZKF: Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Danijel Sikic
- Clinic of Urology and Pediatric Urology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; BZKF: Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Bernd Wullich
- Clinic of Urology and Pediatric Urology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; Comprehensive Cancer Center Erlangen - EMD, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; BZKF: Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Michael Uder
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; Comprehensive Cancer Center Erlangen - EMD, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; BZKF: Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Tobias Bäuerle
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; University Medical Center of Johannes Gutenberg-University Mainz, Department of Diagnostic and Interventional Radiology, Langenbeckstr. 1, 55131 Mainz, Germany
| |
Collapse
|
2
|
Yin D, Wang K, Xu H, Guo Y, Qian B, Duan D, Li Y, Zhang W, Li Z, Zhao Y. Contrast-Enhanced CT-Based Radiomics Nomogram for Prediction of Pathologic T3a Upstaging in Clinical T1 RCC. Diagnostics (Basel) 2025; 15:443. [PMID: 40002594 PMCID: PMC11854503 DOI: 10.3390/diagnostics15040443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 01/31/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: To develop a nomogram for the preoperative prediction of pathologic T3a (pT3a) upstaging in patients with clinical T1(cT1) renal cell carcinoma (RCC). Methods: A total of 169 cT1 patients with RCC with preoperative contrast-enhanced CT (CECT) and clinical data were enrolled in this study. Afterwards, the sample was split randomly into training and testing sets in a 7:3 ratio. Radiomics features were extracted and selected from the whole primary tumor on CECT images to develop radiomics signatures. The nomogram was constructed using the obtained radiomics signature and clinical risk factors. The predictive performance of different models was evaluated and visualized using receiver operator characteristic (ROC) curves. Results: In total, 26 radiomics features were selected for the radiomics signature construction. The radiomics signature yielded area under the curve (AUC) values of 0.945 and 0.873 in the training and testing sets, respectively. The nomogram integrating radiomics signature and predictive clinical factors, including tumor size and neutrophil-lymphocyte ratio (NLR), achieved higher predictive performance in the training [AUC, 0.958; 95% confidence interval (CI): 0.921, 0.995] and testing (AUC, 0.913; 95% CI: 0.814, 1.000) sets. Good calibration was achieved for the nomogram in both the training and testing sets (Brier score = 0.082 and 0.098). Decision curve analysis (DCA) demonstrated that the nomogram was clinically useful in predicting pT3a upstaging, with a corresponding net benefit of 0.378. Conclusions: The proposed nomogram can preoperatively predict pT3a upstaging in cT1 RCC and serve as a non-invasive imaging marker to guide individualized treatment.
Collapse
Affiliation(s)
- Di Yin
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China; (D.Y.); (H.X.); (Y.G.); (D.D.); (Y.L.); (W.Z.); (Z.L.)
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China;
| | - Keruo Wang
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China;
| | - Hongyi Xu
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China; (D.Y.); (H.X.); (Y.G.); (D.D.); (Y.L.); (W.Z.); (Z.L.)
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China;
| | - Yunfei Guo
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China; (D.Y.); (H.X.); (Y.G.); (D.D.); (Y.L.); (W.Z.); (Z.L.)
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China;
| | - Baoxin Qian
- Huiying Medical Technology (Beijing), Beijing 100192, China;
| | - Dengyi Duan
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China; (D.Y.); (H.X.); (Y.G.); (D.D.); (Y.L.); (W.Z.); (Z.L.)
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China;
| | - Yiming Li
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China; (D.Y.); (H.X.); (Y.G.); (D.D.); (Y.L.); (W.Z.); (Z.L.)
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China;
| | - Wenyi Zhang
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China; (D.Y.); (H.X.); (Y.G.); (D.D.); (Y.L.); (W.Z.); (Z.L.)
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China;
| | - Zhengyang Li
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China; (D.Y.); (H.X.); (Y.G.); (D.D.); (Y.L.); (W.Z.); (Z.L.)
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China;
| | - Yang Zhao
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China; (D.Y.); (H.X.); (Y.G.); (D.D.); (Y.L.); (W.Z.); (Z.L.)
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China;
| |
Collapse
|
3
|
Wang Y, Zhang X, Wang S, Shi H, Zhao X, Chen Y. Predicting first-line VEGFR-TKI resistance and survival in metastatic clear cell renal cell carcinoma using a clinical-radiomic nomogram. Cancer Imaging 2024; 24:151. [PMID: 39529158 PMCID: PMC11552170 DOI: 10.1186/s40644-024-00792-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND This study aims to construct predicting models using radiomic and clinical features in predicting first-line vascular endothelial growth factor receptor-tyrosine kinase inhibitor (VEGFR-TKI) early resistance in metastatic clear cell renal cell carcinoma (mccRCC) patients. We also aim to explore the correlation of predicting models with short and long-term survival of mccRCC patients. MATERIALS AND METHODS In this retrospective study, 110 mccRCC patients from 2009 to 2019 were included and assigned into training and test sets. Radiomic features were extracted from tumor 3D-ROI of baseline enhanced CT images. Radiomic features were selected by Lasso method to construct a radiomic score. A combined nomogram was established using the combination of radiomic score and clinical factors. The discriminative abilities of the radiomic, clinical and combined nomogram were quantified using ROC curve. Cox regression analysis was used to test the correlation of nomogram score with progression-free survival (PFS) and overall survival (OS). PFS and OS were compared between different risk groups by log-rank test. RESULTS The radiomic, clinical and combined nomogram demonstrated AUCs of 0.81, 0.75, and 0.83 in training set; 0.79, 0.77, and 0.88 in test set. Nomogram score ≥ 1.18 was an independent prognostic factor of PFS (HR 0.22 (0.10, 0.47), p < 0.001) and OS (HR 0.38 (0.20, 0.71), p = 0.002), in training set. PFS in low-risk group were significantly longer than high-risk group in training (p < 0.001) and test (p < 0.001) set, respectively. OS in low-risk group were significantly longer than high-risk group in training (p = 0.003) and test (p = 0.009) set, respectively. CONCLUSION A nomogram combining baseline radiomic signature and clinical factors helped detecting first-line VEGFR-TKI early resistance and predicting short and long-term prognosis in mccRCC patients.
Collapse
Affiliation(s)
- Yichen Wang
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjianyuannanli No.17, Chaoyang District, Beijing, 100021, China
| | - Xinxin Zhang
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjianyuannanli No.17, Chaoyang District, Beijing, 100021, China
| | | | - Hongzhe Shi
- Department of Urology, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xinming Zhao
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjianyuannanli No.17, Chaoyang District, Beijing, 100021, China
| | - Yan Chen
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjianyuannanli No.17, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
4
|
Bigot P, Boissier R, Khene ZE, Albigès L, Bernhard JC, Correas JM, De Vergie S, Doumerc N, Ferragu M, Ingels A, Margue G, Ouzaïd I, Pettenati C, Rioux-Leclercq N, Sargos P, Waeckel T, Barthelemy P, Rouprêt M. French AFU Cancer Committee Guidelines - Update 2024-2026: Management of kidney cancer. THE FRENCH JOURNAL OF UROLOGY 2024; 34:102735. [PMID: 39581661 DOI: 10.1016/j.fjurol.2024.102735] [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: 08/04/2024] [Revised: 09/01/2024] [Accepted: 09/02/2024] [Indexed: 11/26/2024]
Abstract
OBJECTIVE To update the French recommendations for the management of kidney cancer. METHODS A systematic review of the literature was conducted for the period from 2014 to 2024. The most relevant articles concerning the diagnosis, classification, surgical treatment, medical treatment, and follow-up of kidney cancer were selected and incorporated into the recommendations. The recommendations have been updated specifying the level of evidence (strong or weak). RESULTS Kidney cancer following prolonged occupational exposure to trichloroethylene should be considered an occupational disease. The reference examination for the diagnosis and staging of kidney cancer is the contrast-enhanced thoraco-abdominal CT scan. PET scans are not indicated in the staging of kidney cancer. Percutaneous biopsy is recommended in situations where its results will influence therapeutic decisions. It should be used to reduce the number of surgeries for benign tumors, particularly avoiding unnecessary radical nephrectomies. Kidney tumors should be classified according to the pTNM 2017 classification, the WHO 2022 classification, and the ISUP nucleolar grade. Metastatic kidney cancers should be classified according to IMDC criteria. Surveillance of tumors smaller than 2cm should be prioritized and can be offered regardless of patient age. Robot-assisted laparoscopic partial nephrectomy is the reference surgical treatment for T1 tumors. Ablative therapies and surveillance are options for elderly patients with comorbidities for tumors larger than 2cm. Stereotactic radiotherapy is an option to discuss for treating localized kidney tumors in patients not eligible for other treatments. Radical nephrectomy is the first-line treatment for locally advanced localized cancers. Pembrolizumab is recommended for patients at high risk of recurrence after surgery for localized kidney cancer. In metastatic patients, cytoreductive nephrectomy can be immediate in cases of good prognosis, delayed in cases of intermediate or poor prognosis for patients stabilized by medical treatment, or as "consolidation" in patients with complete or major partial response at metastatic sites after systemic treatment. Surgical or local treatment of metastases can be proposed for single lesions or oligometastases. Recommended first-line drugs for metastatic clear cell renal carcinoma are combinations of axitinib/pembrolizumab, nivolumab/ipilimumab, nivolumab/cabozantinib, and lenvatinib/pembrolizumab. Patients with non-clear cell metastatic kidney cancer should be presented to the CARARE Network and prioritized for inclusion in clinical trials. CONCLUSION These updated recommendations are a reference that will enable French and French-speaking practitioners to optimize their management of kidney cancer.
Collapse
Affiliation(s)
- Pierre Bigot
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology, Angers University Hospital, Angers, France.
| | - Romain Boissier
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology and Kidney Transplantation, Conception University Hospital, Aix-Marseille University, AP-HM, Marseille, France
| | - Zine-Eddine Khene
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology, Rennes University Hospital, Rennes, France
| | - Laurence Albigès
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Cancer Medicine, Gustave-Roussy, Paris-Saclay University, Villejuif, France
| | - Jean-Christophe Bernhard
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology, Hôpital Pellegrin, Bordeaux University Hospital, Bordeaux, France
| | - Jean-Michel Correas
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Adult Radiology, Hôpital Necker, University of Paris, AP-HP Centre, Paris, France
| | - Stéphane De Vergie
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology, Nantes University Hospital, Nantes, France
| | - Nicolas Doumerc
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology and Renal Transplantation, Toulouse University Hospital, Toulouse, France
| | - Matthieu Ferragu
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology, Angers University Hospital, Angers, France
| | - Alexandre Ingels
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology, UPEC, Hôpital Henri-Mondor, Créteil, France
| | - Gaëlle Margue
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology, Hôpital Pellegrin, Bordeaux University Hospital, Bordeaux, France
| | - Idir Ouzaïd
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology, Bichat University Hospital, AP-HP, Paris, France
| | - Caroline Pettenati
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology, Hôpital Foch, University of Versailles - Saint-Quentin-en-Yvelines, 40, rue Worth, 92150 Suresnes, France
| | - Nathalie Rioux-Leclercq
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Pathology, Rennes University Hospital, Rennes, France
| | - Paul Sargos
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Radiotherapy, Hôpital Pellegrin, Bordeaux University Hospital, Bordeaux, France
| | - Thibaut Waeckel
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Department of Urology, Caen University Hospital, Caen, France
| | - Philippe Barthelemy
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Medical Oncology, Institut de Cancérologie Strasbourg Europe, Strasbourg, France
| | - Morgan Rouprêt
- Oncology Committee of the French Urology Association, Kidney Group, Maison de l'Urologie, 11, rue Viète, 75017 Paris, France; Urology, Hôpital Pitié-Salpêtrière, Predictive Onco-Urology, GRC 5, Sorbonne University, AP-HP, 75013 Paris, France
| |
Collapse
|
5
|
Yu Y, Niu J, Yu Y, Xia S, Sun S. AI predictive modeling of survival outcomes for renal cancer patients undergoing targeted therapy. Sci Rep 2024; 14:26156. [PMID: 39478092 PMCID: PMC11525571 DOI: 10.1038/s41598-024-77638-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 10/24/2024] [Indexed: 11/02/2024] Open
Abstract
Renal clear cell cancer (RCC) is a complex disease that is challenging to predict patient outcomes. Despite improvements with targeted therapy, personalized treatment planning is still needed. Artificial intelligence (AI) can help address this challenge by developing predictive models that accurately forecast patient survival periods. With AI-powered decision support, clinicians can provide patients with tailored treatment plans, enhancing treatment efficacy and quality of life. The study analyzed 267 patients with renal clear cell carcinoma, focusing on 26 who received targeted drug therapy. The data was refined by excluding 8 patients without enhanced CT scans. The research team categorized patients into two groups based on their expected lifespan: Group 1 (over 3 years) and Group 2 (under 3 years). The UPerNet algorithm was used to extract features from CT tumor markers, validating their effectiveness. These features were then used to develop an AI-based predictive model trained on the dataset. The developed AI model demonstrated remarkable accuracy, achieving a rate of 93.66% in Group 1 and 94.14% in Group 2. In conclusion, our study demonstrates the potential of AI technology in predicting the survival time of RCC patients undergoing targeted drug therapy. The established prediction model exhibits high predictive accuracy and stability, serving as a valuable tool for clinicians to facilitate the development of more personalized treatment plans for patients. This study highlights the importance of integrating AI technology in clinical decision-making, enabling patients to receive more effective and targeted treatment plans that enhance their overall quality of life.
Collapse
Affiliation(s)
- Yaoqi Yu
- Department of Urology, Heilongjiang Provincial Hospital, Heilongjiang Provincial Hospital, Harbin Institute of Technology, Harbin, 150036, China
| | - Jirui Niu
- Department of Urology, Heilongjiang Provincial Hospital, Heilongjiang Provincial Hospital, Harbin Institute of Technology, Harbin, 150036, China
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, No.37 Yiyuan Street, Nangang District, Harbin, 150001, Heilongjiang, China
- Bio-Bank of Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, No.37 Yiyuan Street, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Yin Yu
- Department of Urology, Heilongjiang Provincial Hospital, Heilongjiang Provincial Hospital, Harbin Institute of Technology, Harbin, 150036, China
| | - Silong Xia
- Department of Urology, Heilongjiang Provincial Hospital, Heilongjiang Provincial Hospital, Harbin Institute of Technology, Harbin, 150036, China
| | - Shiheng Sun
- Department of Urology, Heilongjiang Provincial Hospital, Heilongjiang Provincial Hospital, Harbin Institute of Technology, Harbin, 150036, China.
| |
Collapse
|
6
|
Woon D, Qin S, Al-Khanaty A, Perera M, Lawrentschuk N. Imaging in Renal Cell Carcinoma Detection. Diagnostics (Basel) 2024; 14:2105. [PMID: 39335784 PMCID: PMC11431198 DOI: 10.3390/diagnostics14182105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
INTRODUCTION Imaging in renal cell carcinoma (RCC) is a constantly evolving landscape. The incidence of RCC has been rising over the years with the improvement in image quality and sensitivity in imaging modalities resulting in "incidentalomas" being detected. We aim to explore the latest advances in imaging for RCC. METHODS A literature search was conducted using Medline and Google Scholar, up to May 2024. For each subsection of the manuscript, a separate search was performed using a combination of the following key terms "renal cell carcinoma", "renal mass", "ultrasound", "computed tomography", "magnetic resonance imaging", "18F-Fluorodeoxyglucose PET/CT", "prostate-specific membrane antigen PET/CT", "technetium-99m sestamibi SPECT/CT", "carbonic anhydrase IX", "girentuximab", and "radiomics". Studies that were not in English were excluded. The reference lists of selected manuscripts were checked manually for eligible articles. RESULTS The main imaging modalities for RCC currently are ultrasound, computed tomography (CT) and magnetic resonance imaging (MRI). Contrast-enhanced US (CEUS) has emerged as an alternative to CT or MRI for the characterisation of renal masses. Furthermore, there has been significant research in molecular imaging in recent years, including FDG PET, PSMA PET/CT, 99mTc-Sestamibi, and anti-carbonic anhydrase IX monoclonal antibodies/peptides. Radiomics and the use of AI in radiology is a growing area of interest. CONCLUSIONS There will be significant change in the field of imaging in RCC as molecular imaging becomes increasingly popular, which reflects a shift in management to a more conservative approach, especially for small renal masses (SRMs). There is the hope that the improvement in imaging will result in less unnecessary invasive surgeries or biopsies being performed for benign or indolent renal lesions.
Collapse
Affiliation(s)
- Dixon Woon
- Department of Urology, Austin Health, Heidelberg, VIC 3084, Australia
- Department of Surgery, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Shane Qin
- Department of Urology, Austin Health, Heidelberg, VIC 3084, Australia
| | | | - Marlon Perera
- Department of Urology, Austin Health, Heidelberg, VIC 3084, Australia
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Nathan Lawrentschuk
- Department of Surgery, The University of Melbourne, Melbourne, VIC 3010, Australia
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Department of Urology, Royal Melbourne Hospital, Parkville, VIC 3052, Australia
| |
Collapse
|
7
|
Alqahtani A, Bhattacharjee S, Almopti A, Li C, Nabi G. Radiomics-Based Computed Tomography Urogram Approach for the Prediction of Survival and Recurrence in Upper Urinary Tract Urothelial Carcinoma. Cancers (Basel) 2024; 16:3119. [PMID: 39335090 PMCID: PMC11429600 DOI: 10.3390/cancers16183119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 08/30/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
Upper tract urothelial carcinoma (UTUC) is a rare and aggressive malignancy with a poor prognosis. The accurate prediction of survival and recurrence in UTUC is crucial for effective risk stratification and guiding therapeutic decisions. Models combining radiomics and clinicopathological data features derived from computed tomographic urograms (CTUs) can be a way to predict survival and recurrence in UTUC. Thus, preoperative CTUs and clinical data were analyzed from 106 UTUC patients who underwent radical nephroureterectomy. Radiomics features were extracted from segmented tumors, and the Least Absolute Shrinkage and Selection Operator (LASSO) method was used to select the most relevant features. Multivariable Cox models combining radiomics features and clinical factors were developed to predict the survival and recurrence. Harrell's concordance index (C-index) was applied to evaluate the performance and survival distribution analyses were assessed by a Kaplan-Meier analysis. The significant outcome predictors were identified by multivariable Cox models. The combined model achieved a superior predictive accuracy (C-index: 0.73) and higher recurrence prediction (C-index: 0.84). The Kaplan-Meier analysis showed significant differences in the survival (p < 0.0001) and recurrence (p < 0.002) probabilities for the combined datasets. The CTU-based radiomics models effectively predicted survival and recurrence in the UTUC patients, and enhanced the prognostic performance by combining radiomics features with clinical factors.
Collapse
Affiliation(s)
- Abdulsalam Alqahtani
- School of Medicine, Centre for Medical Engineering and Technology, University of Dundee, Dundee DD1 9SY, UK
- Radiology Department, College of Applied Medical Sciences, Najran University, Najran 55461, Saudi Arabia
| | - Sourav Bhattacharjee
- School of Veterinary Medicine, University College Dublin, D04 W6F6 Dublin, Ireland
| | - Abdulrahman Almopti
- School of Medicine, Centre for Medical Engineering and Technology, University of Dundee, Dundee DD1 9SY, UK
| | - Chunhui Li
- School of Science and Engineering, University of Dundee, Dundee DD1 4HN, UK
| | - Ghulam Nabi
- School of Medicine, Centre for Medical Engineering and Technology, University of Dundee, Dundee DD1 9SY, UK
| |
Collapse
|
8
|
Schawkat K, Krajewski KM. Insights into Renal Cell Carcinoma with Novel Imaging Approaches. Hematol Oncol Clin North Am 2023; 37:863-875. [PMID: 37302934 DOI: 10.1016/j.hoc.2023.05.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
This article presents a comprehensive overview of new imaging approaches and techniques for improving the assessment of renal masses and renal cell carcinoma. The Bosniak classification, version 2019, as well as the clear cell likelihood score, version 2.0, will be discussed as new imaging algorithms using established techniques. Additionally, newer modalities, such as contrast-enhanced ultrasound, dual energy computed tomography, and molecular imaging, will be discussed in conjunction with emerging radiomics and artificial intelligence techniques. Current diagnostic algorithms combined with newer approaches may be an effective way to overcome existing limitations in renal mass and RCC characterization.
Collapse
Affiliation(s)
- Khoschy Schawkat
- Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA; Harvard Medical School
| | - Katherine M Krajewski
- Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA; Harvard Medical School; Dana-Farber Cancer Institute, 440 Brookline Avenue, Building MA Floor L1 Room 04AC, Boston, MA 02215, USA.
| |
Collapse
|
9
|
Liu X, Shi J, Li Z, Huang Y, Zhang Z, Zhang C. The Present and Future of Artificial Intelligence in Urological Cancer. J Clin Med 2023; 12:4995. [PMID: 37568397 PMCID: PMC10419644 DOI: 10.3390/jcm12154995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/10/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
Artificial intelligence has drawn more and more attention for both research and application in the field of medicine. It has considerable potential for urological cancer detection, therapy, and prognosis prediction due to its ability to choose features in data to complete a particular task autonomously. Although the clinical application of AI is still immature and faces drawbacks such as insufficient data and a lack of prospective clinical trials, AI will play an essential role in individualization and the whole management of cancers as research progresses. In this review, we summarize the applications and studies of AI in major urological cancers, including tumor diagnosis, treatment, and prognosis prediction. Moreover, we discuss the current challenges and future applications of AI.
Collapse
Affiliation(s)
| | | | | | | | - Zhihong Zhang
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China; (X.L.)
| | - Changwen Zhang
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China; (X.L.)
| |
Collapse
|
10
|
Nolazco JI, Soerensen SJC, Chung BI. Biomarkers for the Detection and Surveillance of Renal Cancer. Urol Clin North Am 2023; 50:191-204. [PMID: 36948666 DOI: 10.1016/j.ucl.2023.01.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
Renal cell carcinoma (RCC) is a heterogeneous disease characterized by a broad spectrum of disorders in terms of genetics, molecular and clinical characteristics. There is an urgent need for noninvasive tools to stratify and select patients for treatment accurately. In this review, we analyze serum, urinary, and imaging biomarkers that have the potential to detect malignant tumors in patients with RCC. We discuss the characteristics of these numerous biomarkers and their ability to be used routinely in clinical practice. The development of biomarkers continues to evolve with promising prospects.
Collapse
Affiliation(s)
- José Ignacio Nolazco
- Division of Urological Surgery, Brigham and Women's Hospital, Harvard Medical School, 45 Francis Street, Boston, MA 02115, USA; Servicio de Urología, Hospital Universitario Austral, Universidad Austral, Av Juan Domingo Perón 1500, B1629AHJ Pilar, Argentina.
| | - Simon John Christoph Soerensen
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA; Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, USA
| | - Benjamin I Chung
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
11
|
Ferro M, Crocetto F, Barone B, del Giudice F, Maggi M, Lucarelli G, Busetto GM, Autorino R, Marchioni M, Cantiello F, Crocerossa F, Luzzago S, Piccinelli M, Mistretta FA, Tozzi M, Schips L, Falagario UG, Veccia A, Vartolomei MD, Musi G, de Cobelli O, Montanari E, Tătaru OS. Artificial intelligence and radiomics in evaluation of kidney lesions: a comprehensive literature review. Ther Adv Urol 2023; 15:17562872231164803. [PMID: 37113657 PMCID: PMC10126666 DOI: 10.1177/17562872231164803] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 03/04/2023] [Indexed: 04/29/2023] Open
Abstract
Radiomics and artificial intelligence (AI) may increase the differentiation of benign from malignant kidney lesions, differentiation of angiomyolipoma (AML) from renal cell carcinoma (RCC), differentiation of oncocytoma from RCC, differentiation of different subtypes of RCC, to predict Fuhrman grade, to predict gene mutation through molecular biomarkers and to predict treatment response in metastatic RCC undergoing immunotherapy. Neural networks analyze imaging data. Statistical, geometrical, textural features derived are giving quantitative data of contour, internal heterogeneity and gray zone features of lesions. A comprehensive literature review was performed, until July 2022. Studies investigating the diagnostic value of radiomics in differentiation of renal lesions, grade prediction, gene alterations, molecular biomarkers and ongoing clinical trials have been analyzed. The application of AI and radiomics could lead to improved sensitivity, specificity, accuracy in detecting and differentiating between renal lesions. Standardization of scanner protocols will improve preoperative differentiation between benign, low-risk cancers and clinically significant renal cancers and holds the premises to enhance the diagnostic ability of imaging tools to characterize renal lesions.
Collapse
Affiliation(s)
| | - Felice Crocetto
- Department of Neurosciences and Reproductive
Sciences and Odontostomatology, University of Naples Federico II, Naples,
Italy
| | - Biagio Barone
- Department of Neurosciences and Reproductive
Sciences and Odontostomatology, University of Naples Federico II, Naples,
Italy
| | - Francesco del Giudice
- Department of Maternal Infant and Urologic
Sciences, Policlinico Umberto I Hospital, Sapienza University of Rome, Rome,
Italy
| | - Martina Maggi
- Department of Maternal Infant and Urologic
Sciences, Policlinico Umberto I Hospital, Sapienza University of Rome, Rome,
Italy
| | - Giuseppe Lucarelli
- Urology, Andrology and Kidney Transplantation
Unit, Department of Emergency and Organ Transplantation, University of Bari,
Bari, Italy
| | - Gian Maria Busetto
- Department of Urology and Organ
Transplantation, University of Foggia, Foggia, Italy
| | | | - Michele Marchioni
- Department of Medical, Oral and
Biotechnological Sciences, Urology Unit, SS Annunziata Hospital, G.
d’Annunzio University of Chieti, Chieti, Italy
- Department of Urology, ASL Abruzzo 2, Chieti,
Italy
| | - Francesco Cantiello
- Department of Urology, Magna Graecia
University of Catanzaro, Catanzaro, Italy
| | - Fabio Crocerossa
- Department of Urology, Magna Graecia
University of Catanzaro, Catanzaro, Italy
| | - Stefano Luzzago
- Department of Urology, IEO – European
Institute of Oncology, IRCCS – Istituto di Ricovero e Cura a Carattere
Scientifico, Milan, Italy
- Università degli Studi di Milano, Milan,
Italy
| | - Mattia Piccinelli
- Cancer Prognostics and Health Outcomes Unit,
Division of Urology, University of Montréal Health Center, Montréal, QC,
Canada
- Department of Urology, IEO – European
Institute of Oncology, IRCCS – Istituto di Ricovero e Cura a Carattere
Scientifico, Milan, Italy
| | - Francesco Alessandro Mistretta
- Department of Urology, IEO – European
Institute of Oncology, IRCCS – Istituto di Ricovero e Cura a Carattere
Scientifico, Milan, Italy
- Department of Oncology and
Hematology-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Marco Tozzi
- Department of Urology, IEO – European
Institute of Oncology, IRCCS – Istituto di Ricovero e Cura a Carattere
Scientifico, Milan, Italy
- Università degli Studi di Milano, Milan,
Italy
| | - Luigi Schips
- Department of Medical, Oral and
Biotechnological Sciences, Urology Unit, SS Annunziata Hospital, G.
d’Annunzio University of Chieti, Chieti, Italy
| | | | - Alessandro Veccia
- Urology Unit, Azienda Ospedaliera
Universitaria Integrata Verona, University of Verona, Verona, Italy
| | - Mihai Dorin Vartolomei
- Department of Cell and Molecular Biology,
George Emil Palade University of Medicine, Pharmacy, Science and Technology
of Târgu Mures, Târgu Mures, Romania
- Department of Urology, Medical University of
Vienna, Vienna, Austria
| | - Gennaro Musi
- Department of Urology, IEO – European
Institute of Oncology, IRCCS – Istituto di Ricovero e Cura a Carattere
Scientifico, Milan, Italy
- Department of Oncology and
Hematology-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Ottavio de Cobelli
- Department of Urology, IEO – European
Institute of Oncology, IRCCS – Istituto di Ricovero e Cura a Carattere
Scientifico, Milan, Italy
- Department of Oncology and
Hematology-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Emanuele Montanari
- Department of Urology, Foundation IRCCS Ca’
Granda – Ospedale Maggiore Policlinico, Department of Clinical Sciences and
Community Health, University of Milan, Milan, Italy
| | - Octavian Sabin Tătaru
- Institution Organizing University Doctoral
Studies (IOSUD), George Emil Palade University of Medicine, Pharmacy,
Science and Technology of Târgu Mures, Târgu Mures, Romania
| |
Collapse
|
12
|
Lin ZF, Qin LX, Chen JH. Biomarkers for response to immunotherapy in hepatobiliary malignancies. Hepatobiliary Pancreat Dis Int 2022; 21:413-419. [PMID: 35973935 DOI: 10.1016/j.hbpd.2022.08.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/29/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND The advent of immune checkpoint inhibitors (ICIs) has revolutionized the therapeutic options of hepatobiliary malignancies. However, the clinical benefit provided by immunotherapy seems limited to a small subgroup of patients with hepatobiliary malignancies. The identification of reliable predictors of the response to immunotherapy is urgently needed. DATA SOURCES Literature search was conducted in PubMed for relevant articles published up to May 2022. Information of clinical trials was obtained from https://clinicaltrials.gov/. RESULTS Biomarkers for ICI response of hepatobiliary malignancies remain in the exploration stage and lack compelling evidence. Tumor programmed death-ligand 1 (PD-L1) expression is the most widely studied biomarker in hepatocellular carcinoma (HCC) and biliary tract cancers (BTCs), but there are conflicting results on its predictive potential. Tumor mutational burden (TMB) is generally low both in HCC and BTCs, and the clinical trials of TMB are rare in hepatobiliary malignancies. Promisingly, mismatch repair deficiency (dMMR)/high microsatellite instability (MSI-H) may be a predictive biomarker of response to anti-PD-1 therapy in BTCs. Furthermore, some emerging biomarkers, such as gut microbiota, show predictive potential in the preliminary studies. Radiomics and liquid-biopsy biomarkers, including circulating tumor cells, circulating tumor DNA (ctDNA) and exosomal PD-L1 provide a quick and non-invasive approach for monitoring the ICI response, showing a new promising direction. CONCLUSIONS Multiple potential biomarkers for predicting ICI response of hepatobiliary malignancies have been explored and tried to apply in clinic. Yet there is no robust evidence to prove their clinical value in predicting immunotherapeutic response for patients with hepatobiliary malignancies. The identification of predictors for response to ICIs is an urgent need and major challenge. Further studies are warranted to validate the role of emerging biomarkers in predicting immunotherapeutic responses.
Collapse
Affiliation(s)
- Zhi-Fei Lin
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai 200040, China
| | - Lun-Xiu Qin
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai 200040, China
| | - Jin-Hong Chen
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai 200040, China.
| |
Collapse
|
13
|
Gao J, Ye F, Han F, Jiang H, Zhang J. A radiogenomics biomarker based on immunological heterogeneity for non-invasive prognosis of renal clear cell carcinoma. Front Immunol 2022; 13:956679. [PMID: 36177018 PMCID: PMC9513051 DOI: 10.3389/fimmu.2022.956679] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/19/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundTumor immunological heterogeneity potentially influences the prognostic disparities among patients with clear cell renal cell carcinoma (ccRCC); however, there is a lack of macroscopic imaging tools that can be used to predict immune-related gene expression in ccRCC.MethodsA novel non-invasive radiogenomics biomarker was constructed for immune-related gene expression in ccRCC. First, 520 ccRCC transcriptomic datasets from The Cancer Genome Atlas (TCGA) were analyzed using a non-negative matrix decomposition (NMF) clustering to identify immune-related molecular subtypes. Immune-related prognostic genes were analyzed through Cox regression and Gene Set Enrichment Analysis (GSEA). We then built a risk model based on an immune-related gene subset to predict prognosis in patients with ccRCC. CT images corresponding to the ccRCC patients in The Cancer Imaging Archive (TCIA) database were used to extract radiomic features. To stratify immune-related gene expression levels, extracted radiogenomics features were identified according to standard consecutive steps. A nomogram was built to combine radiogenomics and clinicopathological information through multivariate logistic regression to further enhance the radiogenomics model. Mann–Whitney U test and ROC curves were used to assess the effectiveness of the radiogenomics marker.ResultsNMF methods successfully clustered patients into diverse subtypes according to gene expression levels in the tumor microenvironment (TME). The relative abundance of 10 immune cell populations in each tissue was also analyzed. The immune-related genomic signature (consisting of eight genes) of the tumor was shown to be significantly associated with survival in patients with ccRCC in TCGA database. The immune-related genomic signature was delineated by grouping the signature expression as either low- or high-risk. Using TCIA database, we constructed a radiogenomics biomarker consisting of 11 radiomic features that were optimal predictors of immune-related gene signature expression levels, which demonstrated AUC (area under the ROC curve) values of 0.76 and 0.72 in the training and validation groups, respectively. The nomogram built by combining radiomics and clinical pathological information could further improve the predictive efficacy of the radiogenomics model (AUC = 0.81, 074).ConclusionsThe novel prognostic radiogenomics biomarker achieved excellent correlation with the immune-related gene expression status of patients with ccRCC and could successfully stratify the survival status of patients in TCGA database. It is anticipated that this work will assist in selecting precise clinical treatment strategies. This study may also lead to precise theranostics for patients with ccRCC in the future.
Collapse
Affiliation(s)
- Jiahao Gao
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Fangdie Ye
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Fang Han
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Haowen Jiang
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
- *Correspondence: Jiawen Zhang, ; Haowen Jiang,
| | - Jiawen Zhang
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
- *Correspondence: Jiawen Zhang, ; Haowen Jiang,
| |
Collapse
|
14
|
Jian L, Liu Y, Xie Y, Jiang S, Ye M, Lin H. MRI-Based Radiomics and Urine Creatinine for the Differentiation of Renal Angiomyolipoma With Minimal Fat From Renal Cell Carcinoma: A Preliminary Study. Front Oncol 2022; 12:876664. [PMID: 35719934 PMCID: PMC9204342 DOI: 10.3389/fonc.2022.876664] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/26/2022] [Indexed: 12/12/2022] Open
Abstract
Objectives Standard magnetic resonance imaging (MRI) techniques are different to distinguish minimal fat angiomyolipoma (mf-AML) with minimal fat from renal cell carcinoma (RCC). Here we aimed to evaluate the diagnostic performance of MRI-based radiomics in the differentiation of fat-poor AMLs from other renal neoplasms. Methods A total of 69 patients with solid renal tumors without macroscopic fat and with a pathologic diagnosis of RCC (n=50) or mf-AML (n=19) who underwent conventional MRI and intravoxel incoherent motion diffusion-weighted imaging (IVIM-DWI) were included. Clinical data including age, sex, tumor location, urine creatinine, and urea nitrogen were collected from medical records. The apparent diffusion coefficient (ADC), pure diffusion coefficient (D), pseudodiffusion coefficient (D*), and perfusion fraction (f) were measured from renal tumors. We used the ITK-SNAP software to manually delineate the regions of interest on T2-weighted imaging (T2WI) and IVIM-DWI from the largest cross-sectional area of the tumor. We extracted 396 radiomics features by the Analysis Kit software for each MR sequence. The hand-crafted features were selected by using the Pearson correlation analysis and least absolute shrinkage and selection operator (LASSO). Diagnostic models were built by logistic regression analysis. Receiver operating characteristic curve analysis was performed using five-fold cross-validation and the mean area under the curve (AUC) values were calculated and compared between the models to obtain the optimal model for the differentiation of mf-AML and RCC. Decision curve analysis (DCA) was used to evaluate the clinical utility of the models. Results Clinical model based on urine creatinine achieved an AUC of 0.802 (95%CI: 0.761-0.843). IVIM-based model based on f value achieved an AUC of 0.692 (95%CI: 0.627-0.757). T2WI-radiomics model achieved an AUC of 0.883 (95%CI: 0.852-0.914). IVIM-radiomics model achieved an AUC of 0.874 (95%CI: 0.841-0.907). Combined radiomics model achieved an AUC of 0.919 (95%CI: 0.894-0.944). Clinical-radiomics model yielded the best performance, with an AUC of 0.931 (95%CI: 0.907-0.955). The calibration curve and DCA confirmed that the clinical-radiomics model had a good consistency and clinical usefulness. Conclusion The clinical-radiomics model may be served as a noninvasive diagnostic tool to differentiate mf-AML with RCC, which might facilitate the clinical decision-making process.
Collapse
Affiliation(s)
- Lian Jian
- Department of Radiology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yan Liu
- Department of Radiology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yu Xie
- Department of Urological Surgery, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Shusuan Jiang
- Department of Urological Surgery, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Mingji Ye
- Department of Urological Surgery, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Huashan Lin
- Department of Pharmaceuticals Diagnosis, General Electric (GE) Healthcare, Changsha, China
| |
Collapse
|
15
|
Hamada K, Fujiwara R, Takemura K, Komai Y, Oguchi T, Numao N, Yamamoto S, Yonese J, Yuasa T. Tumor shrinkage patterns of nivolumab monotherapy in metastatic renal cell carcinoma. Int J Urol 2022; 29:1181-1187. [PMID: 35717138 DOI: 10.1111/iju.14964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 06/02/2022] [Indexed: 11/29/2022]
Abstract
OBJECTIVES To investigate the tumor shrinkage patterns of patients with metastatic renal cell carcinoma treated with nivolumab monotherapy. METHODS Forty-four consecutive patients with metastatic renal cell carcinoma treated with nivolumab monotherapy (81 metastatic and four primary lesions) between September 2013 and December 2020 were retrospectively analyzed. The tumor shrinkage rate of individual visceral and lymph node metastatic lesions and the primary site lesions treated with nivolumab monotherapy, as well as the association between overall survival and pretreatment tumor size, were statistically assessed. RESULTS Pretreatment tumor size for the total and individual target lesions, which included kidneys, lungs, pancreas, and lymph nodes, were not correlated with tumor shrinkage rate. The tumor shrinkage rate was found to have no significant association with pretreatment tumor size between any organ. In addition, there is no significant difference in tumor shrinkage rate between larger (>median value) and smaller (<median value) pretreatment tumor size in any organ. Finally, there was no significant difference in overall survival between larger and smaller pretreatment tumor size. CONCLUSIONS Pretreatment tumor size was not associated with the tumor shrinkage rate and overall survival in nivolumab monotherapy.
Collapse
Affiliation(s)
- Kosuke Hamada
- Department of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Ryo Fujiwara
- Department of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kosuke Takemura
- Department of Urology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Yoshinobu Komai
- Department of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Tomohiko Oguchi
- Department of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Noboru Numao
- Department of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Shinya Yamamoto
- Department of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Junji Yonese
- Department of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takeshi Yuasa
- Department of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
16
|
Ye L, Chen Y, Xu H, Wang Z, Li H, Qi J, Wang J, Yao J, Liu J, Song B. Radiomics of Contrast-Enhanced Computed Tomography: A Potential Biomarker for Pretreatment Prediction of the Response to Bacillus Calmette-Guerin Immunotherapy in Non-Muscle-Invasive Bladder Cancer. Front Cell Dev Biol 2022; 10:814388. [PMID: 35281100 PMCID: PMC8914064 DOI: 10.3389/fcell.2022.814388] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 01/10/2022] [Indexed: 11/13/2022] Open
Abstract
Background:Bacillus Calmette-Guerin (BCG) instillation is recommended postoperatively after transurethral resection of bladder cancer (TURBT) in patients with high-risk non-muscle-invasive bladder cancer (NMIBC). An accurate prediction model for the BCG response can help identify patients with NMIBC who may benefit from alternative therapy.Objective: To investigate the value of computed tomography (CT) radiomics features in predicting the response to BCG instillation among patients with primary high-risk NMIBC.Methods: Patients with pathologically confirmed high-risk NMIBC were retrospectively reviewed. Patients who underwent contrast-enhanced CT examination within one to 2 weeks before TURBT and received ≥5 BCG instillation treatments in two independent hospitals were enrolled. Patients with a routine follow-up of at least 1 year at the outpatient department were included in the final cohort. Radiomics features based on CT images were extracted from the tumor and its periphery in the training cohort, and a radiomics signature was built with recursive feature elimination. Selected features further underwent an unsupervised radiomics analysis using the newly introduced method, non-negative matrix factorization (NMF), to compute factor factorization decompositions of the radiomics matrix. Finally, a robust component, which was most associated with BCG failure in 1 year, was selected. The performance of the selected component was assessed and tested in an external validation cohort.Results: Overall, 128 patients (training cohort, n = 104; external validation cohort, n = 24) were included, including 12 BCG failures in the training cohort and 11 failures in the validation cohort each. NMF revealed five components, of which component 3 was selected for the best discrimination of BCG failure; it had an area under the curve (AUC) of .79, sensitivity of .79, and specificity of .65 in the training set. In the external validation cohort, it achieved an AUC of .68, sensitivity of .73, and specificity of .69. Survival analysis showed that patients with higher component scores had poor recurrence-free survival (RFS) in both cohorts (C-index: training cohort, .69; validation cohort, .68).Conclusion: The study suggested that radiomics components based on NMF might be a potential biomarker to predict BCG response and RFS after BCG treatment in patients with high-risk NMIBC.
Collapse
Affiliation(s)
- Lei Ye
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuntian Chen
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Hui Xu
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Zhaoxiang Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | | | - Jin Qi
- University of Electronic Science and Technology of China, Chengdu, China
| | - Jing Wang
- University of Electronic Science and Technology of China, Chengdu, China
| | - Jin Yao
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Jin Yao, ; Jiaming Liu,
| | - Jiaming Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Jin Yao, ; Jiaming Liu,
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|