Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1965 2023-07-20 16:51:53 |
2 only format change Meta information modification 1965 2023-07-21 03:52:56 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Distante, A.; Marandino, L.; Bertolo, R.; Ingels, A.; Pavan, N.; Pecoraro, A.; Marchioni, M.; Carbonara, U.; Erdem, S.; Amparore, D.; et al. Artificial Intelligence-Based Diagnosis of Renal Cell Carcinoma Subtypes. Encyclopedia. Available online: https://encyclopedia.pub/entry/47064 (accessed on 01 July 2024).
Distante A, Marandino L, Bertolo R, Ingels A, Pavan N, Pecoraro A, et al. Artificial Intelligence-Based Diagnosis of Renal Cell Carcinoma Subtypes. Encyclopedia. Available at: https://encyclopedia.pub/entry/47064. Accessed July 01, 2024.
Distante, Alfredo, Laura Marandino, Riccardo Bertolo, Alexandre Ingels, Nicola Pavan, Angela Pecoraro, Michele Marchioni, Umberto Carbonara, Selcuk Erdem, Daniele Amparore, et al. "Artificial Intelligence-Based Diagnosis of Renal Cell Carcinoma Subtypes" Encyclopedia, https://encyclopedia.pub/entry/47064 (accessed July 01, 2024).
Distante, A., Marandino, L., Bertolo, R., Ingels, A., Pavan, N., Pecoraro, A., Marchioni, M., Carbonara, U., Erdem, S., Amparore, D., Campi, R., Roussel, E., Caliò, A., Wu, Z., Palumbo, C., Borregales, L.D., Mulders, P., & Muselaers, C.H.J. (2023, July 20). Artificial Intelligence-Based Diagnosis of Renal Cell Carcinoma Subtypes. In Encyclopedia. https://encyclopedia.pub/entry/47064
Distante, Alfredo, et al. "Artificial Intelligence-Based Diagnosis of Renal Cell Carcinoma Subtypes." Encyclopedia. Web. 20 July, 2023.
Artificial Intelligence-Based Diagnosis of Renal Cell Carcinoma Subtypes
Edit

Renal cell carcinoma (RCC) is characterized by its diverse histopathological features, which pose possible challenges to accurate diagnosis and prognosis. The use of AI in radiology, which is also known as radiomics, has shown excellent diagnostic accuracy for detecting RCC and can even provide information regarding RCC subtyping, nuclear grade prediction, gene mutations, and gene expression-based molecular signatures.

artificial intelligence renal cell carcinoma

1. Introduction

Renal cell carcinoma (RCC) is among the top 10 most common cancers in both men and women. The incidence of RCC has gradually risen in recent years, resulting in increased time-, effort-, and cost-related demands on healthcare systems [1]. Adequate RCC diagnosis and treatment planning relies on adequate clinical data, imaging, histology, and molecular profiling [2][3].
Histological analysis, which is supported by genetic and cytogenetic analysis, is crucial for RCC diagnosis, as well as subtyping and defining features with high prognostic and therapeutic impact [4][5]. These features include tumor grade, RCC subtype, lymphovascular invasion, tumor necrosis, sarcomatoid dedifferentiation, etc. [6][7][8]. RCC histological diagnosis and classification, in particular, can be a daunting task, as it encompasses a broad spectrum of histopathological entities, which have recently been subject to changes [9][10].
Over the years, the daily clinical practice of treating patients with RCC has changed from using paper charts, analog radiographs, and light microscopes to using more modern counterparts, such as electronic health records and digitalized radiology and virtual pathology. This shift has generated an enormous amount of digital data, which can be utilized in data-characterization algorithms or artificial intelligence (AI) [11][12].
The use of AI in radiology, which is also known as radiomics, has shown excellent diagnostic accuracy for detecting RCC and can even provide information regarding RCC subtyping, nuclear grade prediction, gene mutations, and gene expression-based molecular signatures [13]. In line with AI in radiology, efforts to use AI in RCC histopathology have been undertaken in recent years. This relatively new field, which is called pathomics or computational pathology, can be used to improve efficiency, accessibility, cost-effectiveness, and time consumption, as well as enhance accuracy and reproducibility with lower subjectivity [11][14][15][16][17]. In addition, Whole Slide Imaging (WSI) technology allows machine learning in pathology by providing an enormous amount of high-quality information for training and testing AI models to identify specific features and patterns that can be complex for even the human eye to discern [12][18][19]. Ultimately, AI aims to assist pathologists in making more accurate and consistent diagnoses in shorter periods of time and is a valuable implement to undercover the above-cited information [20][21]

2. Artificial Intelligence Aided Diagnosis of RCC Subtypes

Although several advances have been made in RCC diagnostics in the last decade, especially in imaging techniques, histo-pathological diagnosis based on a pathologist’s skill and experience remains the standard clinical practice used to distinguish RCC from normal renal tissue at the microscopic level [13][22][23][24].
However, RCCs can have complicated characteristics that make the diagnosis difficult, laborious, and time consuming, even for experienced pathologists. These issues are known to lead to a moderate inter-reader agreement for the RCC subtype [25][26][27]. In addition, several studies demonstrated that computational pathology could be a solution to more uniform specimen readings and reduce intra- and inter-observer variability [28][29][30].

2.1. RCC Diagnosis and Subtyping in Biopsy Specimens

RCC varies in its biological behavior, ranging from indolent to aggressive tumors. Currently, no reliable predictive models that distinguish between different clinical types are available for use in the pre-operative setting, creating concerns about under- and over-treatment, especially in small renal masses (SRMs), which now represent up to 50% of renal lesions [31][32][33][34][35]. Therefore, this issue can lead to overdiagnosis and overtreatment. To date, there are no highly reliable biomarkers or imaging methods that can correctly differentiate between benign and malignant lesions [36][37][38] As a result, there has been a growing trend of using renal mass biopsy (RMB) to address this challenge over the past decade [39][40][41].
However, RMBs have some limitations as they are non-diagnostic in approximately 10–15% of the cases and remain intrinsically invasive [42]. The main reason for the high percentage of non-diagnostic results is inadequate sampling of tumors [43]. Another crucial issue in RMB is a fair degree of interobserver variability [44], a concern that is also found in breast, prostate, and melanoma biopsies [45][46][47].
To tackle these problems, Fenstermaker et al. developed a DL-based algorithm for RCC diagnosis, grading, and subtype assessment [48]. Their method reached a high accuracy level when using only a 100 square micrometers (µm2) patch, making it a potentially valuable tool in RMB analysis. In addition, although their method was trained on whole-mount surgical specimens, a computational method trained and tested on small tissue samples may reduce the need for repeat biopsies by decreasing insufficient tissue sampling and reducing interobserver variability.
However, this research focused on identifying the three main subtypes of RCC without considering benign tumors or oncocytomas. A significant proportion of small renal masses (SRMs) are benign, with oncocytoma being the most frequent benign contrast-enhancing renal mass found. A well-known problem faced by pathologists is differentiating oncocytomas from chromophobe RCC [49][50][51]. Zhu et al. reported favorable results in RCC subtyping in surgical resection and RMB specimens, as well as promising results in oncocytoma diagnosis in RMB [52]. The group trained and tested a model on an internal dataset of renal resections. In addition, they tested this model on 79 RCC biopsy slides, 24 of which were diagnosed as renal oncocytoma, and an external dataset, achieving good performance, as shown in Table 1.

2.2. RCC Diagnosis and Subtyping in Surgical Resection Specimens

Despite the recent increased use of RMB and enormous advances in diagnostic accuracy [53][54], approximately 73% of surveyed urologists would not perform a RMB for various reasons [55]. Currently, the standard of treatment for non-metastatic RCC is surgical resection, carried out via either a radical or partial nephrectomy; this technique was also used in some selected cases of metastatic RCC [56][57]. However, examining and analyzing the complex histological patterns of RCC surgical resection specimens under a microscope can be challenging and time consuming for pathologists for many reasons. For instance, nephrectomy specimens exhibit substantial heterogeneity, exemplifying the wide variation observed within RCC surgical resection samples [58]. Moreover, variability among different observers, and even within the same observer, has been reported [26].
Good results were obtained by Tabibu et al. in terms of distinguishing between ccRCC and chRCC and normal tissue using two pre-trained convolutional neural networks (CNN) and replacing the final layers with two output layers, which were fine-tuned using RCC data [59]. Moreover, for subtype classification, the group introduced a so-called directed acyclic graph support vector machine (DAG-SVM) on top of the deep network, obtaining good accuracy in this task. Unlike Tabibu et al.’s model, Chen et al. developed a DL algorithm to detect RCC that was externally validated on an independent dataset [60]. To accomplish this task, they used LASSO (least absolute shrinkage and selection operator), which is a method used in ML to select from a more extensive set of features, i.e., the most important in predicting outcomes. Through LASSO analysis, they identified various image features based on the “The Cancer Genome Atlas” (TCGA) cohort to distinguish between ccRCC and normal renal parenchyma, as well as ccRCC and pRCC and chRCC, obtaining high accuracy in test and external validation cohorts.
Also, Marostica et al. created a pipeline using transfer learning to identify cancerous regions from slide images and classify the three major subtypes, obtaining good performance in both the test set and two external independent datasets (Table 3) [61].
RCC classification is a challenging task not only due to the complexity of the procedure itself, but also because the classification system is subject to periodic updates [62][63]. For example, only in recent years has clear cell papillary renal cell carcinoma (ccpRCC) been recognized as a specific entity [4]. This subtype of RCC histologically resembles both ccRCC and pRCC, and it has clear cell changes. However, ccpRCC has distinct immuno-histochemical and genetic profiles compared to ccRCC and pRCC [64]. It also carries a favorable prognosis relative to the latter carcinoma; therefore, the World Health Organization recently changed its denomination to a clear cell papillary renal cell tumor [65]. Abdeltawab et al. developed a computational model that could distinguish between ccRCC and ccpRCC, obtaining an accuracy of 91% in identifying ccpRCC using the institution files and 90% in diagnosing ccRCC using an external dataset [66].
Table 1. Overview of studies of AI models for diagnosis and subtyping.
The abovementioned studies were mainly supervised and highly defined for RCC approaches, making them time consuming to conduct. However, the capability to apply knowledge gained from previous experiences to novel situations is a vital skill among human beings. For example, pathologists can use lessons learned outside of their specific subspecialty because several cancer types exhibit common hallmarks of malignancy, as demonstrated by Faust et al., who tested whether a previously trained AI system developed to recognize brain tumor features could be applied to clusters and analyze RCC specimens in an unsupervised fashion [67]. The results showed that grouping cancer regions from non-neoplastic tissue elements matched expert annotations in multiple randomly selected cases. This result, hypothetically, represents a way to demonstrate that unsupervised ML-based methods, which were built for the diagnosis of other cancers, can also be used to diagnose RCC, reducing development and work time.

References

  1. Capitanio, U.; Bensalah, K.; Bex, A.; Boorjian, S.A.; Bray, F.; Coleman, J.; Gore, J.L.; Sun, M.; Wood, C.; Russo, P. Epidemiology of Renal Cell Carcinoma. Eur. Urol. 2019, 75, 74–84.
  2. Garfield, K.; LaGrange, C.A. Renal Cell Cancer; StatPearls: Treasure Island, FL, USA, 2022.
  3. Bukavina, L.; Bensalah, K.; Bray, F.; Carlo, M.; Challacombe, B.; Karam, J.A.; Kassouf, W.; Mitchell, T.; Montironi, R.; O’Brien, T.; et al. Epidemiology of Renal Cell Carcinoma: 2022 Update. Eur. Urol. 2022, 82, 529–542.
  4. Moch, H.; Cubilla, A.L.; Humphrey, P.A.; Reuter, V.E.; Ulbright, T.M. The 2016 WHO Classification of Tumours of the Urinary System and Male Genital Organs—Part A: Renal, Penile, and Testicular Tumours. Eur. Urol. 2016, 70, 93–105.
  5. Cimadamore, A.; Caliò, A.; Marandino, L.; Marletta, S.; Franzese, C.; Schips, L.; Amparore, D.; Bertolo, R.; Muselaers, S.; Erdem, S.; et al. Hot topics in renal cancer pathology: Implications for clinical management. Expert Rev. Anticancer. Ther. 2022, 22, 1275–1287.
  6. Fuhrman, S.A.; Lasky, L.C.; Limas, C. Prognostic significance of morphologic parameters in renal cell carcinoma. Am. J. Surg. Pathol. 1982, 6, 655–664.
  7. Zhang, L.; Zha, Z.; Qu, W.; Zhao, H.; Yuan, J.; Feng, Y.; Wu, B. Tumor necrosis as a prognostic variable for the clinical outcome in patients with renal cell carcinoma: A systematic review and meta-analysis. BMC Cancer 2018, 18, 870.
  8. Sun, M.; Shariat, S.F.; Cheng, C.; Ficarra, V.; Murai, M.; Oudard, S.; Pantuck, A.J.; Zigeuner, R.; Karakiewicz, P.I. Prognostic factors and predictive models in renal cell carcinoma: A contemporary review. Eur. Urol. 2011, 60, 644–661.
  9. Hora, M.; Albiges, L.; Bedke, J.; Campi, R.; Capitanio, U.; Giles, R.H.; Ljungberg, B.; Marconi, L.; Klatte, T.; Volpe, A.; et al. European Association of Urology Guidelines Panel on Renal Cell Carcinoma Update on the New World Health Organization Classification of Kidney Tumours 2022: The Urologist’s Point of View. Eur. Urol. 2023, 83, 97–100.
  10. Mimma, R.; Anna, C.; Matteo, B.; Gaetano, P.; Carlo, G.; Guido, M.; Camillo, P. Clinico-pathological implications of the 2022 WHO Renal Cell Carcinoma classification. Cancer Treat. Rev. 2023, 116, 102558.
  11. Baidoshvili, A.; Bucur, A.; Van Leeuwen, J.; Van Der Laak, J.; Kluin, P.; Van Diest, P.J. Evaluating the benefits of digital pathology implementation: Time savings in laboratory logistics. Histopathology 2018, 73, 784–794.
  12. Shmatko, A.; Ghaffari Laleh, N.; Gerstung, M.; Kather, J.N. Artificial intelligence in histopathology: Enhancing cancer research and clinical oncology. Nat. Cancer 2022, 3, 1026–1038.
  13. Roussel, E.; Capitanio, U.; Kutikov, A.; Oosterwijk, E.; Pedrosa, I.; Rowe, S.P.; Gorin, M.A. Novel Imaging Methods for Renal Mass Characterization: A Collaborative Review. Eur. Urol. 2022, 81, 476–488.
  14. Bera, K.; Schalper, K.A.; Rimm, D.L.; Velcheti, V.; Madabhushi, A. Artificial intelligence in digital pathology—New tools for diagnosis and precision oncology. Nat. Rev. Clin. Oncol. 2019, 16, 703–715.
  15. Niazi, M.K.K.; Parwani, A.V.; Gurcan, M.N. Digital pathology and artificial intelligence. Lancet Oncol. 2019, 20, e253–e261.
  16. Colling, R.; Pitman, H.; Oien, K.; Rajpoot, N.; Macklin, P.; CM-Path AI in Histopathology Working Group; Snead, D.; Sackville, T.; Verrill, C. Artificial intelligence in digital pathology: A roadmap to routine use in clinical practice. J. Pathol. 2019, 249, 143–150.
  17. Glembin, M.; Obuchowski, A.; Klaudel, B.; Rydzinski, B.; Karski, R.; Syty, P.; Jasik, P.; Narożański, W.J. Enhancing Renal Tumor Detection: Leveraging Artificial Neural Networks in Computed Tomography Analysis. Med. Sci. Monit. 2023, 29, e939462.
  18. Volpe, A.; Patard, J.J. Prognostic factors in renal cell carcinoma. World J. Urol. 2010, 28, 319–327.
  19. Tucker, M.D.; Rini, B.I. Predicting Response to Immunotherapy in Metastatic Renal Cell Carcinoma. Cancers 2020, 12, 2662.
  20. Wang, D.; Khosla, A.; Gargeya, R.; Irshad, H.; Beck, A.H.; Israel, B. Deep Learning for Identifying Metastatic Breast Cancer. arXiv 2016, arXiv:1606.05718.
  21. Hayashi, Y. Black Box Nature of Deep Learning for Digital Pathology: Beyond Quantitative to Qualitative Algorithmic Performances. In Artificial Intelligence and Machine Learning for Digital Pathology; Lecture Notes in Computer Science (Including Subseries Lecture Notes in Artificial Intelligence and Lecture Notes in Bioinformatics); Springer: Cham, Switzerland, 2020; Volume 12090, pp. 95–101.
  22. Creighton, C.J.; Morgan, M.; Gunaratne, P.H.; Wheeler, D.A.; Gibbs, R.A.; Gordon Robertson, A.; Chu, A.; Beroukhim, R.; Cibulskis, K.; Signoretti, S.; et al. Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature 2013, 499, 43–49.
  23. Krajewski, K.M.; Pedrosa, I. Imaging Advances in the Management of Kidney Cancer. J. Clin. Oncol. 2018, 36, 3582–3590.
  24. Roussel, E.; Campi, R.; Amparore, D.; Bertolo, R.; Carbonara, U.; Erdem, S.; Ingels, A.; Kara, Ö.; Marandino, L.; Marchioni, M.; et al. Expanding the Role of Ultrasound for the Characterization of Renal Masses. J. Clin. Med. 2022, 11, 1112.
  25. Shuch, B.; Hofmann, J.N.; Merino, M.J.; Nix, J.W.; Vourganti, S.; Linehan, W.M.; Schwartz, K.; Ruterbusch, J.J.; Colt, J.S.; Purdue, M.P.; et al. Pathologic validation of renal cell carcinoma histology in the Surveillance, Epidemiology, and End Results program. Urol. Oncol. Semin. Orig. Investig. 2013, 32, 23.e9–23.e13.
  26. Al-Aynati, M.; Chen, V.; Salama, S.; Shuhaibar, H.; Treleaven, D.; Vincic, L. Interobserver and Intraobserver Variability Using the Fuhrman Grading System for Renal Cell Carcinoma. Arch. Pathol. Lab. Med. 2003, 127, 593–596.
  27. Williamson, S.R.; Rao, P.; Hes, O.; Epstein, J.I.; Smith, S.C.; Picken, M.M.; Zhou, M.; Tretiakova, M.S.; Tickoo, S.K.; Chen, Y.-B.; et al. Challenges in pathologic staging of renal cell carcinoma: A study of interobserver variability among urologic pathologists. Am. J. Surg. Pathol. 2018, 42, 1253–1261.
  28. Gavrielides, M.A.; Gallas, B.D.; Lenz, P.; Badano, A.; Hewitt, S.M. Observer variability in the interpretation of HER2/neu immunohistochemical expression with unaided and computer-aided digital microscopy. Arch. Pathol. Lab. Med. 2011, 135, 233–242.
  29. Ficarra, V.; Martignoni, G.; Galfano, A.; Novara, G.; Gobbo, S.; Brunelli, M.; Pea, M.; Zattoni, F.; Artibani, W. Prognostic Role of the Histologic Subtypes of Renal Cell Carcinoma after Slide Revision. Eur. Urol. 2006, 50, 786–794.
  30. Lang, H.; Lindner, V.; de Fromont, M.; Molinié, V.; Letourneux, H.; Meyer, N.; Martin, M.; Jacqmin, D. Multicenter determination of optimal interobserver agreement using the Fuhrman grading system for renal cell carcinoma. Cancer 2004, 103, 625–629. Available online: https://acsjournals.onlinelibrary.wiley.com/doi/full/10.1002/cncr.20812 (accessed on 1 February 2023).
  31. Smaldone, M.C.; Egleston, B.; Hollingsworth, J.M.; Hollenbeck, B.K.; Miller, D.C.; Morgan, T.M.; Kim, S.P.; Malhotra, A.; Handorf, E.; Wong, Y.-N.; et al. Understanding Treatment Disconnect and Mortality Trends in Renal Cell Carcinoma Using Tumor Registry Data. Med. Care 2017, 55, 398–404.
  32. Kutikov, A.; Smaldone, M.C.; Egleston, B.L.; Manley, B.J.; Canter, D.J.; Simhan, J.; Boorjian, S.A.; Viterbo, R.; Chen, D.Y.; Greenberg, R.E.; et al. Anatomic Features of Enhancing Renal Masses Predict Malignant and High-Grade Pathology: A Preoperative Nomogram Using the RENAL Nephrometry Score. Eur. Urol. 2011, 60, 241–248.
  33. Pierorazio, P.M.; Patel, H.D.; Johnson, M.H.; Sozio, S.; Sharma, R.; Iyoha, E.; Bass, E.; Allaf, M.E. Distinguishing malignant and benign renal masses with composite models and nomograms: A systematic review and meta-analysis of clinically localized renal masses suspicious for malignancy. Cancer 2016, 122, 3267–3276.
  34. Joshi, S.; Kutikov, A. Understanding Mutational Drivers of Risk: An Important Step Toward Personalized Care for Patients with Renal Cell Carcinoma. Eur. Urol. Focus 2016, 3, 428–429.
  35. Nguyen, M.M.; Gill, I.S.; Ellison, L.M. The Evolving Presentation of Renal Carcinoma in the United States: Trends From the Surveillance, Epidemiology, and End Results Program. J. Urol. 2006, 176, 2397–2400.
  36. Sohlberg, E.M.; Metzner, T.J.; Leppert, J.T. The Harms of Overdiagnosis and Overtreatment in Patients with Small Renal Masses: A Mini-review. Eur. Urol. Focus 2019, 5, 943–945.
  37. Campi, R.; Stewart, G.D.; Staehler, M.; Dabestani, S.; Kuczyk, M.A.; Shuch, B.M.; Finelli, A.; Bex, A.; Ljungberg, B.; Capitanio, U. Novel Liquid Biomarkers and Innovative Imaging for Kidney Cancer Diagnosis: What Can Be Implemented in Our Practice Today? A Systematic Review of the Literature. Eur. Urol. Oncol. 2021, 4, 22–41.
  38. Warren, H.; Palumbo, C.; Caliò, A.; Tran, M.G.B.; Campi, R.; European Association of Urology (EAU) Young Academic Urologists (YAU) Renal Cancer Working Group. Oncocytoma on renal mass biopsy: Why is surgery even performed? World J. Urol. 2023, 41, 1709–1710.
  39. Kutikov, A.; Smaldone, M.C.; Uzzo, R.G.; Haifler, M.; Bratslavsky, G.; Leibovich, B.C. Renal Mass Biopsy: Always, Sometimes, or Never? Eur. Urol. 2016, 70, 403–406.
  40. Lane, B.R.; Samplaski, M.K.; Herts, B.R.; Zhou, M.; Novick, A.C.; Campbell, S.C. Renal Mass Biopsy—A Renaissance? J. Urol. 2008, 179, 20–27.
  41. Sinks, A.; Miller, C.; Holck, H.; Zeng, L.; Gaston, K.; Riggs, S.; Matulay, J.; Clark, P.E.; Roy, O. Renal Mass Biopsy Mandate Is Associated With Change in Treatment Decisions. J. Urol. 2023, 210, 72–78.
  42. Marconi, L.; Dabestani, S.; Lam, T.B.; Hofmann, F.; Stewart, F.; Norrie, J.; Bex, A.; Bensalah, K.; Canfield, S.E.; Hora, M.; et al. Systematic Review and Meta-analysis of Diagnostic Accuracy of Percutaneous Renal Tumour Biopsy. Eur. Urol. 2016, 69, 660–673.
  43. Evans, A.J.; Delahunt, B.; Srigley, J.R. Issues and challenges associated with classifying neoplasms in percutaneous needle biopsies of incidentally found small renal masses. Semin. Diagn. Pathol. 2015, 32, 184–195.
  44. Kümmerlin, I.; ten Kate, F.; Smedts, F.; Horn, T.; Algaba, F.; Trias, I.; de la Rosette, J.; Laguna, M.P. Core biopsies of renal tumors: A study on diagnostic accuracy, interobserver, and intraobserver variability. Eur. Urol. 2008, 53, 1219–1227.
  45. Elmore, J.G.; Longton, G.M.; Carney, P.A.; Geller, B.M.; Onega, T.; Tosteson, A.N.A.; Nelson, H.D.; Pepe, M.S.; Allison, K.H.; Schnitt, S.J.; et al. Diagnostic concordance among pathologists interpreting breast biopsy specimens. JAMA 2015, 313, 1122–1132.
  46. Elmore, J.G.; Barnhill, R.L.; Elder, D.E.; Longton, G.M.; Pepe, M.S.; Reisch, L.M.; Carney, P.A.; Titus, L.J.; Nelson, H.D.; Onega, T.; et al. Pathologists’ diagnosis of invasive melanoma and melanocytic proliferations: Observer accuracy and reproducibility study. BMJ 2017, 357, j2813.
  47. Shah, M.D.; Parwani, A.V.; Zynger, D.L. Impact of the Pathologist on Prostate Biopsy Diagnosis and Immunohistochemical Stain Usage Within a Single Institution. Am. J. Clin. Pathol. 2017, 148, 494–501.
  48. Fenstermaker, M.; Tomlins, S.A.; Singh, K.; Wiens, J.; Morgan, T.M. Development and Validation of a Deep-learning Model to Assist With Renal Cell Carcinoma Histopathologic Interpretation. Urology 2020, 144, 152–157.
  49. van Oostenbrugge, T.J.; Fütterer, J.J.; Mulders, P.F. Diagnostic Imaging for Solid Renal Tumors: A Pictorial Review. Kidney Cancer 2018, 2, 79–93.
  50. Williams, G.M.; Lynch, D.T. Renal Oncocytoma; StatPearls: Treasure Island, FL, USA, 2022.
  51. Leone, A.R.; Kidd, L.C.; Diorio, G.J.; Zargar-Shoshtari, K.; Sharma, P.; Sexton, W.J.; Spiess, P.E. Bilateral benign renal oncocytomas and the role of renal biopsy: Single institution review. BMC Urol. 2017, 17, 6.
  52. Zhu, M.; Ren, B.; Richards, R.; Suriawinata, M.; Tomita, N.; Hassanpour, S. Development and evaluation of a deep neural network for histologic classification of renal cell carcinoma on biopsy and surgical resection slides. Sci. Rep. 2021, 11, 7080.
  53. Volpe, A.; Mattar, K.; Finelli, A.; Kachura, J.R.; Evans, A.J.; Geddie, W.R.; Jewett, M.A. Contemporary results of percutaneous biopsy of 100 small renal masses: A single center experience. J. Urol. 2008, 180, 2333–2337.
  54. Wang, R.; Wolf, J.S.; Wood, D.P.; Higgins, E.J.; Hafez, K.S. Accuracy of Percutaneous Core Biopsy in Management of Small Renal Masses. Urology 2009, 73, 586–590.
  55. Barwari, K.; de la Rosette, J.J.; Laguna, M.P. The penetration of renal mass biopsy in daily practice: A survey among urologists. J. Endourol. 2012, 26, 737–747.
  56. Escudier, B. Emerging immunotherapies for renal cell carcinoma. Ann. Oncol. 2012, 23, viii35–viii40.
  57. Yanagisawa, T.; Schmidinger, M.; Fajkovic, H.; Karakiewicz, P.I.; Kimura, T.; Shariat, S.F. What is the role of cytoreductive nephrectomy in patients with metastatic renal cell carcinoma? Expert Rev. Anticancer Ther. 2023, 23, 455–459.
  58. Bertolo, R.; Pecoraro, A.; Carbonara, U.; Amparore, D.; Diana, P.; Muselaers, S.; Marchioni, M.; Mir, M.C.; Antonelli, A.; Badani, K.; et al. Resection Techniques During Robotic Partial Nephrectomy: A Systematic Review. Eur. Urol. Open Sci. 2023, 52, 7–21.
  59. Tabibu, S.; Vinod, P.K.; Jawahar, C.V. Pan-Renal Cell Carcinoma classification and survival prediction from histopathology images using deep learning. Sci. Rep. 2019, 9, 10509.
  60. Chen, S.; Zhang, N.; Jiang, L.; Gao, F.; Shao, J.; Wang, T.; Zhang, E.; Yu, H.; Wang, X.; Zheng, J. Clinical use of a machine learning histopathological image signature in diagnosis and survival prediction of clear cell renal cell carcinoma. Int. J. Cancer 2020, 148, 780–790.
  61. Marostica, E.; Barber, R.; Denize, T.; Kohane, I.S.; Signoretti, S.; Golden, J.A.; Yu, K.-H. Development of a Histopathology Informatics Pipeline for Classification and Prediction of Clinical Outcomes in Subtypes of Renal Cell Carcinoma. Clin. Cancer Res. 2021, 27, 2868–2878.
  62. Pathology Outlines—WHO Classification. Available online: https://www.pathologyoutlines.com/topic/kidneytumorWHOclass.html (accessed on 24 January 2023).
  63. Cimadamore, A.; Cheng, L.; Scarpelli, M.; Massari, F.; Mollica, V.; Santoni, M.; Lopez-Beltran, A.; Montironi, R.; Moch, H. Towards a new WHO classification of renal cell tumor: What the clinician needs to know—A narrative review. Transl. Androl. Urol. 2021, 10, 1506–1520.
  64. Weng, S.; DiNatale, R.G.; Silagy, A.; Mano, R.; Attalla, K.; Kashani, M.; Weiss, K.; Benfante, N.E.; Winer, A.G.; Coleman, J.A.; et al. The Clinicopathologic and Molecular Landscape of Clear Cell Papillary Renal Cell Carcinoma: Implications in Diagnosis and Management. Eur. Urol. 2020, 79, 468–477.
  65. Williamson, S.R.; Eble, J.N.; Cheng, L.; Grignon, D.J. Clear cell papillary renal cell carcinoma: Differential diagnosis and extended immunohistochemical profile. Mod. Pathol. 2013, 26, 697–708.
  66. Abdeltawab, H.A.; Khalifa, F.A.; Ghazal, M.A.; Cheng, L.; El-Baz, A.S.; Gondim, D.D. A deep learning framework for automated classification of histopathological kidney whole-slide images. J. Pathol. Inform. 2022, 13, 100093.
  67. Faust, K.; Roohi, A.; Leon, A.J.; Leroux, E.; Dent, A.; Evans, A.J.; Pugh, T.J.; Kalimuthu, S.N.; Djuric, U.; Diamandis, P. Unsupervised Resolution of Histomorphologic Heterogeneity in Renal Cell Carcinoma Using a Brain Tumor–Educated Neural Network. JCO Clin. Cancer Inform. 2020, 4, 811–821.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , , , , , , , , , , , , ,
View Times: 241
Revisions: 2 times (View History)
Update Date: 21 Jul 2023
1000/1000
Video Production Service