1
|
Sanz Ilundain I, Hernández-Lorenzo L, Rodríguez-Antona C, García-Donas J, Ayala JL. Autoencoder techniques for survival analysis on renal cell carcinoma. PLoS One 2025; 20:e0321045. [PMID: 40373089 PMCID: PMC12080797 DOI: 10.1371/journal.pone.0321045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 02/27/2025] [Indexed: 05/17/2025] Open
Abstract
Survival is the gold standard in oncology when determining the real impact of therapies in patients outcome. Thus, identifying molecular predictors of survival (like genetic alterations or transcriptomic patterns of gene expression) is one of the most relevant fields in current research. Statistical methods and metrics to analyze time-to-event data are crucial in understanding disease progression and the effectiveness of treatments. However, in the medical field, data is often high-dimensional, complicating the application of such methodologies. In this study, we addressed this challenge by compressing the high-dimensional transcriptomic data of patients treated with immunotherapy (avelumab + axitinib) and a TKI (sunitinib) into latent, meaningful features using autoencoders. We applied a semi-parametric statistical approach based on the COX Proportional Hazards model, coupled with Breslow's estimator, to predict each patient's Progression-Free Survival (PFS) and determine survival functions. Our analysis explored various penalty configurations and their combinations. Given the complexity of transcriptomic data, we extended our model to incorporate both tabular data and its graph variant, where edges represent protein-protein interactions between genes, offering a more insightful approach. Recognizing the interpretability challenges inherent in neural networks, particularly autoencoders, we analyzed the mutual information between genes in the original data and their latent feature representations to clarify which genes are most associated with specific latent variables. The results indicate that different types of autoencoders are better suited for different tasks: denoising autoencoders excel at accurate reconstruction, while the sparse variant is more effective at producing meaningful representations. Additionally, combining these penalties enhances both reconstruction quality and the interpretability of latent features. The interpretable models identified genes such as LRP2 and ACE2 as highly relevant to renal cell carcinoma. This research underscores the utility of autoencoders in managing high-dimensional data problems.
Collapse
Affiliation(s)
| | | | | | - Jesús García-Donas
- HM CIOCC Madrid, Hospital Universitario HM Sanchinarro, HM Hospitales, Madrid, Spain
| | | |
Collapse
|
2
|
Stewart GD, Godoy A, Farquhar F, Kitt J, Cartledge J, Kimuli M, Rossi SH, Shinkins B, Burbidge S, Burge SW, Caglic I, Collins E, Crosbie PAJ, Eckert C, Fraser S, Hancock N, Iball GR, Marshall C, Masson G, Neal RD, Rogerson S, Smith A, Sharp SJ, Simmonds I, Wallace T, Ward M, Callister MEJ, Usher-Smith JA. Abdominal Noncontrast Computed Tomography Scanning to Screen for Kidney Cancer and Other Abdominal Pathology Within Community-based Computed Tomography Screening for Lung Cancer: Results of the Yorkshire Kidney Screening Trial. Eur Urol 2025; 87:561-570. [PMID: 39271419 DOI: 10.1016/j.eururo.2024.08.029] [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: 04/17/2024] [Revised: 08/08/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND AND OBJECTIVE The Yorkshire Kidney Screening Trial (YKST) assessed the feasibility of adding abdominal noncontrast computed tomography (NCCT) to lung cancer screening to screen for kidney cancer and other abdominal pathology. METHODS A prospective diagnostic study offered abdominal NCCT to 55-80-yr-old ever-smokers attending a UK randomised lung cancer screening trial (May 2021 to October 2022). The exclusion criteria were dementia, frailty, previous kidney/lung cancer, and computed tomography (CT) of the abdomen and thorax within previous 6 and 12 mo, respectively. Six-month follow-up was undertaken. KEY FINDINGS AND LIMITATIONS A total of 4438 people attended lung screening, of whom 4309 (97%) were eligible for and 4019 (93%) accepted abdominal NCCT. Only 3.9% respondents regretted participating. The additional time to conduct the YKST processes was 13.3 min. Of the participants, 2586 (64%) had a normal abdominal NCCT, whilst 787 (20%) required an abdominal NCCT imaging review but no further action and 611 (15%) required further evaluation (investigations and/or clinic). Of the participants, 211 (5.3%) had a new serious finding, including 25 (0.62%) with a renal mass/complex cyst, of whom ten (0.25%) had histologically proven kidney cancer; ten (0.25%) with other cancers; and 60 (1.5%) with abdominal aortic aneurysms (AAAs). Twenty-five (0.62%) participants had treatment with curative intent. Of the participants, 1017 (25%) had nonserious findings, most commonly benign renal cysts (727 [18%]), whereas only 259 (6.4%) had nonserious findings requiring further tests. The number needed to screen to detect one serious abdominal finding was 18; it was 93 to detect one suspicious renal lesion and 402 to detect one histologically confirmed renal cancer. Limitations of the cohort were fixed age range and being prior lung cancer screening attendees. CONCLUSIONS AND CLINICAL IMPLICATIONS In this first prospective risk-stratified screening study of abdominal NCCT offered alongside CT thorax, uptake and participant satisfaction were high. The prevalence of serious findings, cancers, and AAAs, is in the range of established screening programmes such as bowel cancer. Longer-term outcomes and cost effectiveness should now be evaluated.
Collapse
Affiliation(s)
- Grant D Stewart
- Department of Surgery, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; CRUK Cambridge Centre, Cambridge Biomedical Campus, Cambridge, UK.
| | - Angela Godoy
- CRUK Cambridge Centre, Cambridge Biomedical Campus, Cambridge, UK; Department of Oncology, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Fiona Farquhar
- Research and Innovation, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Jessica Kitt
- Department of Surgery, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; CRUK Cambridge Centre, Cambridge Biomedical Campus, Cambridge, UK
| | - Jon Cartledge
- Department of Urology, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Michael Kimuli
- Department of Urology, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Sabrina H Rossi
- Department of Surgery, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; CRUK Cambridge Centre, Cambridge Biomedical Campus, Cambridge, UK
| | - Bethany Shinkins
- Division of Health Sciences, Warwick Medical School, University of Warwick, Coventry, UK; Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | - Simon Burbidge
- Department of Radiology, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Sarah W Burge
- CRUK Cambridge Centre, Cambridge Biomedical Campus, Cambridge, UK; Department of Oncology, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Iztok Caglic
- Department of Radiology, University of Cambridge, Cambridge, UK
| | - Emma Collins
- Department of Endocrine Surgery, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Philip A J Crosbie
- Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Claire Eckert
- Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | - Sheila Fraser
- Department of Endocrine Surgery, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Neil Hancock
- Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | - Gareth R Iball
- School of AHP & Midwifery, Faculty of Health Studies, University of Bradford, Bradford, UK
| | | | | | - Richard D Neal
- Exeter Collaboration for Academic Primary Care (APEx), University of Exeter, Exeter, UK
| | - Suzanne Rogerson
- Research and Innovation, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Andrew Smith
- The Pancreas Unit, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Stephen J Sharp
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Irene Simmonds
- Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | - Tom Wallace
- Leeds Vascular Institute, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Matthew Ward
- Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | - Matthew E J Callister
- Leeds Institute of Health Sciences, University of Leeds, Leeds, UK; Department of Respiratory Medicine, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Juliet A Usher-Smith
- CRUK Cambridge Centre, Cambridge Biomedical Campus, Cambridge, UK; Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| |
Collapse
|
3
|
Chen P, Turco S, Liu Z, Widdershoven C, Oddens J, Wijkstra H, Mischi M, Zondervan P. Contrast-ultrasound dispersion imaging for renal cell carcinoma diagnostics. Ultrasound J 2025; 17:23. [PMID: 40238020 PMCID: PMC12003236 DOI: 10.1186/s13089-025-00423-7] [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: 11/10/2024] [Accepted: 03/07/2025] [Indexed: 04/18/2025] Open
Abstract
Cost-effective screening methods for Renal Cell Carcinoma (RCC) are still lacking. Angiogenesis is a recognized hallmark of cancer growth, leading to distinguishable perfusion patterns in tumors from those in normal tissue. This establishes the basis for diagnostic imaging solutions by dynamic contrast-enhanced ultrasound (DCE-US). In the past years, we have developed contrast-ultrasound dispersion imaging (CUDI) techniques to quantify prostate DCE-US acquisitions, obtaining promising results for prostate cancer localization. In this pilot study, we investigated for the first time its feasibility for RCC localization. DCE-US acquisitions of the kidney in 5 patients were used to perform CUDI analysis. With the obtained CUDI parameters and the delineated tumor and parenchyma regions, we performed pixel-based classification, from which the highest area under the receiver-operating-characteristic curve (AUC) = 0.96 was obtained for an individual patient, and an average AUC = 0.68 was obtained for the full patient dataset, showing the potential of CUDI for solid RCC localization. Further validation in a larger dataset and evaluation of the compatibility of point-of-care diagnosis are required.
Collapse
Affiliation(s)
- Peiran Chen
- Department of Electrical Engineering, Eindhoven University of Technology, De Groene Loper 19, Eindhoven, 5612 AP, the Netherlands.
| | - Simona Turco
- Department of Electrical Engineering, Eindhoven University of Technology, De Groene Loper 19, Eindhoven, 5612 AP, the Netherlands
| | - Zhaohan Liu
- Department of Electrical Engineering, Eindhoven University of Technology, De Groene Loper 19, Eindhoven, 5612 AP, the Netherlands
| | - Christiaan Widdershoven
- Department of Urology, Amsterdam University Medical Centers, De Boelelaan 1117, Amsterdam, 1081 HV, the Netherlands
| | - Jorg Oddens
- Department of Urology, Amsterdam University Medical Centers, De Boelelaan 1117, Amsterdam, 1081 HV, the Netherlands
| | - Hessel Wijkstra
- Department of Electrical Engineering, Eindhoven University of Technology, De Groene Loper 19, Eindhoven, 5612 AP, the Netherlands
| | - Massimo Mischi
- Department of Electrical Engineering, Eindhoven University of Technology, De Groene Loper 19, Eindhoven, 5612 AP, the Netherlands
| | - Patricia Zondervan
- Department of Urology, Amsterdam University Medical Centers, De Boelelaan 1117, Amsterdam, 1081 HV, the Netherlands
| |
Collapse
|
4
|
Shi T, Chen H, Wang Z, Wang H, Peng C, Huang S, Wen Y, Pu X, Liang Z, Zhong J, Wang L, Liang X, Wei W, Li T, Chen J, Chen Z, Ma X, Ruan W, Fan JB, Ding D, Liu J, Zhang X, Huang Q. A urine DNA methylation assay for early detection of renal cancer. Oncogene 2025:10.1038/s41388-024-03268-x. [PMID: 40097805 DOI: 10.1038/s41388-024-03268-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 03/19/2025]
Abstract
Renal cancer (RC) is the most lethal urological malignancy with 30% late diagnosis. Over 50% RCs are asymptomatic and discovered incidentally. Current RC detection relies on imaging while it lacks satisfactory sensitivity for detecting small-size tumors. A sensitive and robust diagnostic tool is needed to facilitate standardized RC early detection. Herein, we performed genome-wide methylation sequencing on both tissues and urine samples for RC DNA methylation makers discovery and developed a PCR-based RC early detector (RED) using a cohort of 93 RC and 35 non-RC urine samples. RED further achieved sensitivities of 82.2% and 80.7%, and specificities of 77.1% and 75% in a testing cohort (90 RC vs. 35 non-RC) and a validation cohort (119 RC vs. 48 non-RC), respectively. Importantly, RED exhibited 89.5% sensitivity for tumors in diameter <2 cm. It can detect 83.6% clear cell renal cell carcinoma, 75.0% of translocational renal cell carcinoma, 100% of primitive neuroectodermal tumors, renal malignant masenchymomas and mucinous tubular and spindle cell carcinoma. RED showed promising performance for RC detection with early stage and small size and have potential to be used in conjunction with imaging.
Collapse
Affiliation(s)
- Tongshuai Shi
- Faculty of Urology, Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hanzhong Chen
- Department of Urology Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zhifeng Wang
- Department of Urology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Hong Wang
- AnchorDx Medical Co., Ltd., Guangzhou, China
| | - Cheng Peng
- Faculty of Urology, Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Shang Huang
- Department of Urology Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ying Wen
- AnchorDx Medical Co., Ltd., Guangzhou, China
| | - Xiaoyong Pu
- Department of Urology Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | | | | | - Lingdian Wang
- Department of Urology, Henan Provincial People's Hospital, Zhengzhou, China
| | | | - Wei Wei
- Department of Urology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Teng Li
- Department of Urology Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jiaxin Chen
- AnchorDx Medical Co., Ltd., Guangzhou, China
| | - Zhiwei Chen
- AnchorDx Medical Co., Ltd., Guangzhou, China
- AnchorDx Inc, Fremont, USA
| | - Xin Ma
- Faculty of Urology, Third Medical Center, Chinese PLA General Hospital, Beijing, China.
| | - Weimei Ruan
- AnchorDx Medical Co., Ltd., Guangzhou, China.
| | - Jian-Bing Fan
- AnchorDx Medical Co., Ltd., Guangzhou, China.
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Degang Ding
- Department of Urology, Henan Provincial People's Hospital, Zhengzhou, China.
| | - Jiumin Liu
- Department of Urology Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| | - Xu Zhang
- Faculty of Urology, Third Medical Center, Chinese PLA General Hospital, Beijing, China.
| | - Qingbo Huang
- Faculty of Urology, Third Medical Center, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
5
|
Chen Y, Zhang M, Qi Y, Lin Y, Liu S, Deng C, Jiang S, Sun N. Efficient extraction via titanium organic frameworks facilitates in-depth profiling of urinary exosome metabolite fingerprints. Anal Bioanal Chem 2025; 417:1543-1555. [PMID: 39853354 DOI: 10.1007/s00216-025-05741-2] [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: 12/11/2024] [Revised: 01/03/2025] [Accepted: 01/07/2025] [Indexed: 01/26/2025]
Abstract
Urinary exosome metabolite analysis has demonstrated notable advantages in uncovering disease status, yet its potential in decoding the intricacies of clear cell renal cell carcinoma (ccRCC) remains untapped. To address this, a core-shell magnetic titanium organic framework was designed to capture urinary exosomes and assist laser desorption/ionization mass spectrometry (LDI MS) to decipher the exosomal metabolic profile of ccRCC, with high sensitivity, throughput, and speed. A total of 492 urinary exosome metabolite fingerprints (UEMFs) from 176 samples were extracted for exploring the differences between ccRCC and healthy individuals. Leveraging machine learning algorithms, the exosomal metabolic profile was disclosed, achieving accurate differentiation and prediction of ccRCC patients versus healthy individuals, with an accuracy exceeding 97.3%. Furthermore, an optimized algorithm panel comprising five key features demonstrated consistent and high diagnosing accuracy rates of over 94.0% both in the training and blind test sets for ccRCC, underscoring the remarkable effectiveness and superiority of this strategy in ccRCC detection. This study not only refines the LDI MS method for metabolite analysis in urinary exosomes but also introduces a promising technical approach for unraveling the mysteries of ccRCC.
Collapse
Affiliation(s)
- Yijie Chen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Man Zhang
- Department of Chemistry, Fudan University, Shanghai, 200433, China
| | - Yu Qi
- Department of Urology, Zhongshan Hospital, Zhongshan Hospital Wusong Branch Fudan University, Shanghai, 200032, China
| | - Yiwen Lin
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Shasha Liu
- Department of Chemistry, Fudan University, Shanghai, 200433, China
| | - Chunhui Deng
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, 330031, China.
| | - Shuai Jiang
- Department of Urology, Zhongshan Hospital, Zhongshan Hospital Wusong Branch Fudan University, Shanghai, 200032, China.
| | - Nianrong Sun
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
6
|
Guo W, Chen W, Zhang J, Li M, Huang H, Wang Q, Fei X, Huang J, Zheng T, Fan H, Wang Y, Gu H, Ding G, Chen Y. High-throughput methylation sequencing reveals novel biomarkers for the early detection of renal cell carcinoma. BMC Cancer 2025; 25:96. [PMID: 39819319 PMCID: PMC11737265 DOI: 10.1186/s12885-024-13380-6] [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: 07/03/2024] [Accepted: 12/20/2024] [Indexed: 01/19/2025] Open
Abstract
PURPOSE Renal cell carcinoma (RCC) is a common malignancy, with patients frequently diagnosed at an advanced stage due to the absence of sufficiently sensitive detection technologies, significantly compromising patient survival and quality of life. Advances in cell-free DNA (cfDNA) methylation profiling using liquid biopsies offer a promising non-invasive diagnostic option, but robust biomarkers for early detection are current not available. This study aimed to identify methylation biomarkers for RCC and establish a DNA methylation signature-based prognostic model for this disease. METHODS High-throughput methylation sequencing was performed on peripheral blood samples obtained from 49 primarily Stage I RCC patients and 44 healthy controls. Comparative analysis and Least Absolute Shrinkage and Selection Operator (LASSO) regression methods were employed to identify RCC methylation signatures.Subsequently, methylation markers-based diagnostic and prognostic models for RCC were independently trained and validated using random forest and Cox regression methodologies, respectively. RESULTS Comparative analysis revealed 864 differentially methylated CpG islands (DMCGIs), 96.3% of which were hypermethylated. Using a training set from The Cancer Genome Atlas (TCGA) dataset of 443 early-stage RCC tumors and matched normal tissues, we applied LASSO regression and identified 23 methylation signatures. We then constructed a random forest-based diagnostic model for early-stage RCC and validated the model using two independent datasets: a TCGA set of 460 RCC tumors and controls, and a blood sample set from our study of 15 RCC cases and 29 healthy controls. For Stage I RCC tissue, the model showed excellent discrimination (AUC-ROC: 0.999, sensitivity: 98.5%, specificity: 100%). Blood sample validation also yielded commendable results (AUC-ROC: 0.852, sensitivity: 73.9%, specificity: 89.7%). Further analysis using Cox regression identified 7 of the 23 DMCGIs as prognostic markers for RCC, allowing the development of a prognostic model with strong predictive power for 1-, 3-, and 5-year survival (AUC-ROC > 0.7). CONCLUSIONS Our findings highlight the critical role of hypermethylation in RCC etiology and progression, and present these identified biomarkers as promising candidates for diagnostic and prognostic applications.
Collapse
Affiliation(s)
- Wenhao Guo
- Department of Urology, Sir Run-Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang Province, China
- Department of Urology, Shaoxing Branch of Sir Run-Run Shaw Hospital, College of Medicine, Zhejiang University, Shaoxing, 312000, Zhejiang Province, China
| | - Weiwu Chen
- Department of Urology, Sir Run-Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang Province, China
- School of Medicine, Zhejiang University, Hangzhou, 310011, Zhejiang Province, China
| | - Jie Zhang
- Department of Urology, Sir Run-Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang Province, China
| | - Mingzhe Li
- Department of Urology, Sir Run-Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang Province, China
| | - Hongyuan Huang
- Department of Urology, Jinjiang Municipal Hospital, Quanzhou, 362000, Fujian Province, China
| | - Qian Wang
- Hangzhou Shengting Medical Technology Co., Ltd., Hangzhou, 310018, Zhejiang Province, China
| | - Xiaoyi Fei
- Hangzhou Shengting Medical Technology Co., Ltd., Hangzhou, 310018, Zhejiang Province, China
| | - Jian Huang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, 350 Shushanhu Road, Hefei, 230031, Anhui Province, China
| | - Tongning Zheng
- Department of Urology, Ningbo Zhenhai People's Hospital, Ningbo, 315202, Zhejiang Province, China
| | - Haobo Fan
- Department of Urology, Sir Run-Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang Province, China
- School of Medicine, Zhejiang University, Hangzhou, 310011, Zhejiang Province, China
| | - Yunfei Wang
- Hangzhou Shengting Medical Technology Co., Ltd., Hangzhou, 310018, Zhejiang Province, China
| | - Hongcang Gu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, 350 Shushanhu Road, Hefei, 230031, Anhui Province, China.
| | - Guoqing Ding
- Department of Urology, Sir Run-Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang Province, China.
| | - Yicheng Chen
- Department of Urology, Sir Run-Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang Province, China.
| |
Collapse
|
7
|
Rajandram R, Suren Raj TL, Gobe GC, Kuppusamy S. Liquid biopsy for renal cell carcinoma. Clin Chim Acta 2025; 565:119964. [PMID: 39265757 DOI: 10.1016/j.cca.2024.119964] [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: 05/04/2024] [Revised: 09/07/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024]
Abstract
Liquid biopsies offer a less invasive alternative to tissue biopsies for diagnosis, prognosis, and determining therapeutic potential in renal cell carcinoma (RCC). Unfortunately, clinical studies using liquid biopsy biomarkers in RCC are limited. Accordingly, we examine RCC biomarkers, derived from urine, plasma, serum and feces of potential impact and clinical outcome in these patients. A PRISMA checklist was used to identify valuable liquid biopsy biomarkers for diagnosis (plasma cfDNA, serum- or urine-derived circulating RNAs, exosomes and proteins), prognosis (plasma cfDNA, plasma- or serum-derived RNAs, and proteins), and therapeutic response (plasma- and serum-derived proteins). Although other analytes have been identified, their application for routine clinical use remains unclear. In general, panels appear more effective than single biomarkers. Important considerations included proof of reproducibility. Unfortunately, many of the examined studies were insufficiently large and lacked multi-center rigor. Cost-effectiveness was also not available. Accordingly, it is clear that more standardized protocols need to be developed before liquid biopsies can be successfully integrated into clinical practice in RCC.
Collapse
Affiliation(s)
- Retnagowri Rajandram
- Division of Urology, Department of Surgery, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia.
| | - Tulsi Laxmi Suren Raj
- Division of Urology, Department of Surgery, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Glenda Carolyn Gobe
- Kidney Disease Research Collaborative, Translational Research Institute, and School of Biomedical Sciences, University of Queensland, Brisbane, Australia
| | - Shanggar Kuppusamy
- Division of Urology, Department of Surgery, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
8
|
Wang LL, Puri D, Saitta C, Liu F, Afari JA, Meagher MF, Hakimi K, Nguyen MV, Shah A, Ghassemzadeh S, Murphy JD, Javier-Desloges J, McKay RR, Derweesh IH. Trends and Outcomes in Sarcomatoid Renal Cell Carcinoma: Analysis of the National Cancer Data Base. EUR UROL SUPPL 2025; 71:96-105. [PMID: 39758852 PMCID: PMC11699467 DOI: 10.1016/j.euros.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2024] [Indexed: 01/07/2025] Open
Abstract
Background and objective Our aim was to determine the clinical characteristics, temporal trends, and survival outcomes for sarcomatoid-dedifferentiated renal cell carcinoma (sRCC), as sRCC has historically had poor prognosis and a contemporary cohort has not been well characterized in a population-based study. Methods Data for 302 630 RCC cases from 2010 to 2019 were extracted from the National Cancer Data Base, of which 4.1% (12 329) were sRCC. Trend analyses were conducted using the Cochran-Armitage test. Multivariable analyses were used to assess factors associated with sRCC diagnosis and clinicopathologic characteristics associated with all-cause mortality (ACM). Overall survival (OS) was computed via Kaplan-Meier analysis. Key findings and limitations sRCC incidence increased from 3.9% in 2010 to 4.1% in 2019 (p = 0.020). The incidence of stage I sRCC increased from 14.5% in 2010 to 19.2% in 2019 (p < 0.001). sRCC diagnosis was associated with male sex, tumor size, cN1 status, and collecting duct histology. Worse ACM in localized sRCC was associated with age, tumor size, cN1 stage, collecting duct histology, and positive surgical margins; and was inversely associated with partial nephrectomy (hazard ratio [HR] 0.61, 95% confidence interval [CI] 0.49-0.76; p < 0.001). Worse ACM in metastatic sRCC was associated with age, tumor size, cN1, collecting duct histology, positive surgical margins, and no surgery at the primary site (HR 1.66, 95% CI 1.20-2.30; p = 0.006). The 5-yr OS rates for stage I, stage II, stage III, and stage IV sRCC were 74%, 63%, 42%, and 16%, respectively (p < 0.001). Conclusions and clinical implications The proportion of sRCC cases overall and of stage I sRCC cases increased from 2010 to 2019, supporting the hypothesis of stage migration and the potential for early sarcomatoid dedifferentiation. Further studies on the causal mechanisms underpinning better survival after partial nephrectomy in localized disease and after cytoreductive surgery in metastatic disease are warranted. Patient summary We analyzed trends and outcomes for a type of aggressive kidney cancer (sarcomatoid renal cell carcinoma, sRCC) using records from the National Cancer Data Base. We found that the percentage of sRCC cases among all kidney cancers increased from 2010 to 2019. Factors such as tumor size and patient age were linked to worse survival. Surgery to remove the cancer was linked to better survival.
Collapse
Affiliation(s)
- Luke L. Wang
- Department of Urology, University of California-San Diego School of Medicine, La Jolla, CA, USA
| | - Dhruv Puri
- Department of Urology, University of California-San Diego School of Medicine, La Jolla, CA, USA
| | - Cesare Saitta
- IRCCS Humanitas Clinical and Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Franklin Liu
- Department of Medicine, University of Arizona College of Medicine, Tuscon, AZ, USA
| | - Jonathan A. Afari
- Department of Urology, University of California-San Diego School of Medicine, La Jolla, CA, USA
| | - Margaret F. Meagher
- Department of Urology, University of California-San Diego School of Medicine, La Jolla, CA, USA
| | - Kevin Hakimi
- Department of Urology, USC/Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Mimi V. Nguyen
- Department of Urology, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Aastha Shah
- Department of Urology, University of California-San Diego School of Medicine, La Jolla, CA, USA
| | - Saeed Ghassemzadeh
- Department of Urology, University of California-San Diego School of Medicine, La Jolla, CA, USA
| | - James D. Murphy
- Department of Therapeutic Radiology, University of California-San Diego School of Medicine, La Jolla, CA, USA
- Moores UCSD Cancer Center, University of California-San Diego School of Medicine, La Jolla, CA, USA
| | - Juan Javier-Desloges
- Department of Urology, University of California-San Diego School of Medicine, La Jolla, CA, USA
| | - Rana R. McKay
- Moores UCSD Cancer Center, University of California-San Diego School of Medicine, La Jolla, CA, USA
- Department of Medicine, University of California-San Diego School of Medicine, La Jolla, CA, USA
| | - Ithaar H. Derweesh
- Department of Urology, University of California-San Diego School of Medicine, La Jolla, CA, USA
- Moores UCSD Cancer Center, University of California-San Diego School of Medicine, La Jolla, CA, USA
| |
Collapse
|
9
|
Deng Z, Graff RE, Batai K, Chung BI, Langston ME, Kachuri L. Polygenic score for body mass index in relation to mortality among patients with renal cell cancer. Int J Obes (Lond) 2024; 48:1735-1740. [PMID: 39152336 DOI: 10.1038/s41366-024-01609-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 08/02/2024] [Accepted: 08/08/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND The association between body mass index (BMI) and mortality among individuals with renal cell cancer (RCC) is debated, with some observational studies suggesting a lower mortality associated with higher BMI. However, methodological issues such as confounding and reverse causation may bias these findings. Using BMI-associated genetic variants can avoid these biases and generate more valid estimates. METHODS In this prospective cohort study, we included 1264 RCC patients (446 deaths) from the UK Biobank. We created a BMI polygenic score (PGS) based on 336 BMI-associated genetic variants. The association between the PGS and mortality (all-cause and RCC-specific) was evaluated by logistic regression (all RCC cases) and Cox regression (906 incident cases). For comparison, the associations of measured pre-diagnostic BMI and waist-to-hip ratio (WHR) with mortality were quantified by Cox regression among incident cases. We stratified these analyses by time between anthropometric measurement and RCC diagnosis to assess the influence of reverse causation. RESULTS We did not observe an association between the BMI PGS and all-cause mortality among RCC patients (hazard ratio (HR) per SD increase = 0.98, 95% CI: 0.88,1.10). No association was found for pre-diagnostic BMI (HR per 5 kg/m2 increase = 0.93, 95% CI: 0.83,1.04) or WHR (HR per 0.1 increase = 0.97, 95% CI: 0.83,1.13) with mortality. In patients with anthropometrics measured within 2 years before RCC diagnosis, we observed associations of higher BMI (HR per 5 kg/m2 = 0.76, 95% CI: 0.59,0.98) and WHR (HR = 0.67 per 0.1 increase, 95% CI: 0.45,0.98) with a lower risk of death. Similar patterns were observed for RCC-specific mortality. CONCLUSION We found no association between either genetic variants for high BMI or measured pre-diagnostic body adiposity and mortality among RCC patients, and our results suggested a role for reverse causation in the association of obesity with lower mortality. Future studies should be designed carefully to produce unbiased estimates that account for confounding and reverse causation.
Collapse
Affiliation(s)
- Zhengyi Deng
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA.
| | - Rebecca E Graff
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Ken Batai
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Benjamin I Chung
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| | - Marvin E Langston
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, USA
| | - Linda Kachuri
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
10
|
McParland C, Seckin M, Johnston B. Beyond Index Conditions: Applying a Multimorbidity Approach to Renal Cancer Nursing. Semin Oncol Nurs 2024; 40:151743. [PMID: 39419719 DOI: 10.1016/j.soncn.2024.151743] [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: 08/27/2024] [Revised: 09/21/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024]
Abstract
OBJECTIVE This article aims to describe the key components of renal cancer nursing and multimorbidity nursing, and reflects on how adopting a multimorbidity approach to renal cancer nursing can help nurses provide holistic patient care. METHODS This is a discussion paper drawing on relevant evidence and theory. RESULTS Renal cancer nurses have a highly specialised knowledge base and are able to use this expertise to deliver excellent care to people with cancer. However, lots of people with cancer have other conditions as well. Adopting a multimorbidity approach to nursing care provides a more holistic framework for care delivery. CONCLUSIONS Cancer nurses are ideally placed to support patients in this way, so they are able to deliver care which accounts for factors such as treatment burden and how this impacts on patients and carers. IMPLICATIONS FOR NURSING PRACTICE Nurses who care for people with renal cancer should view their patients through the lens of multimorbidity. This involves screening for other chronic conditions, considering polypharmacy, providing emotional support and continuity of care, and coordinating care in a way that accounts for the potentially burdensome nature of the patient's interactions with health care.
Collapse
Affiliation(s)
- Chris McParland
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, UK
| | - Muzeyyen Seckin
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, UK
| | - Bridget Johnston
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, UK; NHS Greater Glasgow and Clyde, Glasgow, UK.
| |
Collapse
|
11
|
Fife K, Pearson C, Knott CS, Greaves A, Stewart GD. Variability in Kidney Cancer Treatment and Survival in England: Results of a National Cohort Study. Clin Oncol (R Coll Radiol) 2024; 36:e429-e438. [PMID: 39242248 DOI: 10.1016/j.clon.2024.08.008] [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: 03/25/2024] [Revised: 06/22/2024] [Accepted: 08/01/2024] [Indexed: 09/09/2024]
Abstract
AIMS To establish whether there are geographic differences in treatments and outcomes for patients with kidney cancer (KC) in England which could potentially be improved by the creation of national guidelines. MATERIALS AND METHODS A multidisciplinary group convened by the charity Kidney Cancer UK developed Quality Performance Indicators (QPIs) for the treatment of KC. Adherence to these QPIs was reported for all patients with a histological diagnosis of KC diagnosed in England between 2017 and 2018. Utilising data extracted from national datasets, logistic and linear probability models were used to estimate geographic variation in the delivery of surgery and systemic anti-cancer therapy at Cancer Alliance and NHS trust levels. Results were adjusted for a priori confounders, including age at diagnosis, area deprivation of residence, and Charlson Comorbidity Index. Differences in overall survival are reported. RESULTS The cohort comprised 18,640 tumours in 18,421 patients. Of tumours diagnosed, median patient age was 68 (interquartile range 58-77) years and 63.4% were in males. When stratified by Cancer Alliance, the proportions of T1a/T1b/N0/M0 KC that had radical nephrectomy (RN), nephron sparing surgery or ablation ranged from 53.3% (95% CI [48.7, 57.8]) to 80.3% (95% CI [73.0, 86.0]). For stage T1b-3 cancers, the proportion that received RN ranged from 65.6% (95% CI [60.3, 70.5]) to 77.3% (95% CI [72.1, 81.7]). Patients with M0 (n = 12,365) and M1 KC (n = 3312) at diagnosis had 24-month survival of 87.5% and 25.1%, respectively. Of patients diagnosed with M1 KC, 50.3% received systemic anti-cancer therapy, ranging from 39.7% (95% CI [33.7, 46.1]) to 70.7% (95% CI [59.6, 79.8]) between Cancer Alliances. The six-month survival of these patients was 77.4% compared to 27.6% for those that did not receive SACT. CONCLUSION These major geographical differences in surgical and systemic therapy practice have led to national guideline development.
Collapse
Affiliation(s)
- K Fife
- Oncology Dept, Box 193, Cambridge University Hospitals NHSFT, Hills Road, Cambridge, CB2 0QQ, UK; CRUK Cambridge Centre, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK.
| | - C Pearson
- Health Data Insight CIC, Cambridge, CB21 5XE, UK; National Disease Registration Service, NHS England, London, UK
| | - C S Knott
- Health Data Insight CIC, Cambridge, CB21 5XE, UK; National Disease Registration Service, NHS England, London, UK
| | - A Greaves
- Kidney Cancer UK, Creation House, Suite D, Unit 1D, Merrow Business Park, Guildford, Surrey, GU4 7WA, UK
| | - G D Stewart
- Department of Surgery, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK; CRUK Cambridge Centre, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| |
Collapse
|
12
|
Zhang Y, Wang T, Mutailipu D, Li Y, Liang S, Yi Q, Zhu R, Ma J. ZBP1 as a prognostic biomarker correlated with cell proliferation in clear cell renal cell carcinoma. Heliyon 2024; 10:e39267. [PMID: 39469683 PMCID: PMC11513510 DOI: 10.1016/j.heliyon.2024.e39267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 10/02/2024] [Accepted: 10/10/2024] [Indexed: 10/30/2024] Open
Abstract
Background Z-DNA-binding protein 1 (ZBP1) is a critical Z-DNA- and Z-RNA-binding protein. However, its diagnostic and prognostic significance, as well as its functions in clear cell renal cell carcinoma (ccRCC), are not well understood. Materials and methods Paired and unpaired differential expression analyses of ZBP1 were performed in pan-cancer cells. Receiver operating characteristic (ROC) curves and survival analyses were used to evaluate the clinical significance of ZBP1. Functional enrichment and immune infiltration analyses were used to explore the mechanisms underlying ZBP1 expression. Western blotting, qRT-PCR, CCK-8, and colony formation assays were used to investigate the potential function of ZBP1 in ccRCC cells. Molecular docking was utilized to identify drugs targeting ZBP1. Results ZBP1 was highly expressed in ccRCC, demonstrating significant diagnostic and prognostic value. ZBP1 mRNA expression correlated with TNM stage, pathologic stage, and histologic grade. Functional enrichment analyses indicated that ZBP1 is involved in multiple immune processes. Moreover, ZBP1 mRNA expression was positively correlated with the infiltrating levels of T cells and cytotoxic cells, and negatively correlated with the infiltrating levels of mast cells and Th17 cells in ccRCC. Biological experiments confirmed that ZBP1 promotes ccRCC cell proliferation. Molecular docking studies identified rucaparib as a potential drug targeting ZBP1. Conclusion Our research findings suggest that targeting ZBP1 could represent a novel therapeutic approach to inhibit the progression of ccRCC.
Collapse
Affiliation(s)
- Yuqing Zhang
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Tao Wang
- Department of Urology, Soochow University Affiliated Taicang Hospital (The First People's Hospital of Taicang), Taicang, China
| | - Daniyaerjiang Mutailipu
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Yang Li
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Shengjie Liang
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Qingtong Yi
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Rujian Zhu
- Department of Urology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Junjie Ma
- Department of Urology, Soochow University Affiliated Taicang Hospital (The First People's Hospital of Taicang), Taicang, China
| |
Collapse
|
13
|
Khandpur U, Haile B, Makary MS. Early-Stage Renal Cell Carcinoma Locoregional Therapies: Current Approaches and Future Directions. Clin Med Insights Oncol 2024; 18:11795549241285390. [PMID: 39435052 PMCID: PMC11492234 DOI: 10.1177/11795549241285390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 09/03/2024] [Indexed: 10/23/2024] Open
Abstract
Renal cell carcinoma (RCC) is the most common primary renal malignancy. Prevalence of RCC in developed countries has slowly increased. Although partial or total nephrectomy has been the first-line treatment for early-stage RCC, improved or similar safety and treatment outcomes with locoregional therapies have challenged this paradigm. In this review, we explore locoregional techniques for early-stage RCC, including radiofrequency ablation, cryoablation, and microwave ablation with a focus on procedural technique, patient selection, and safety/treatment outcomes. Furthermore, we discuss future advances and novel techniques, including radiomics, combination therapy, high-intensity focused ultrasound, and catheter-directed techniques.
Collapse
Affiliation(s)
- Umang Khandpur
- Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Bereket Haile
- Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Mina S Makary
- Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
14
|
Ma W, Jiang X, Jia R, Li Y. Mechanisms of ferroptosis and targeted therapeutic approaches in urological malignancies. Cell Death Discov 2024; 10:432. [PMID: 39384767 PMCID: PMC11464522 DOI: 10.1038/s41420-024-02195-w] [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: 06/02/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/11/2024] Open
Abstract
The prevalence of urological malignancies remains a significant global health concern, particularly given the challenging prognosis for patients in advanced disease stages. Consequently, there is a pressing need to explore the molecular mechanisms that regulate the development of urological malignancies to discover novel breakthroughs in diagnosis and treatment. Ferroptosis, characterized by iron-ion-dependent lipid peroxidation, is a form of programmed cell death (PCD) distinct from apoptosis, autophagy, and necrosis. Notably, lipid, iron, and glutathione metabolism intricately regulate intracellular ferroptosis, playing essential roles in the progression of various neoplasms and drug resistance. In recent years, ferroptosis has been found to be closely related to urological malignancies. This paper provides an overview of the involvement of ferroptosis in the pathogenesis and progression of urological malignancies, elucidates the molecular mechanisms governing its regulation, and synthesizes recent breakthroughs in diagnosing and treating these malignancies. We aim to provide a new direction for the clinical treatment of urological malignancies.
Collapse
Affiliation(s)
- Wenjie Ma
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Xiaotian Jiang
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Ruipeng Jia
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China.
| | - Yang Li
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China.
| |
Collapse
|
15
|
Yang Y, Du P, Hou X, Yan K, Dai Y, Sun Z, Wu Q, Li S, Yan Y, Wang Z, Qi L, Chen M, Zheng H, Gao W, Gao M, Xue W, Zhang X. Early cancer screening surveillance in one medical center of China. PeerJ 2024; 12:e18179. [PMID: 39351369 PMCID: PMC11441387 DOI: 10.7717/peerj.18179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 09/04/2024] [Indexed: 10/04/2024] Open
Abstract
Objectives Cancer screening aims to detect and treat malignant lesions at an early stage and to prolong patients' lifetime. There is still a lack of effective cancer screening programs in China. We initiated a screening project in 2018 and this study presented the cancer screening status in China. Methods We conducted a cross-sectional study in one cancer-care medical center of China. The screening program included routine blood tests, plasma tumor markers, gastric endoscopy, colonoscopy, ultrasound, and computed tomography (CT) scans. Screening results were presented as sensitivity, specificity and positive predictive values (PPVs). Results Twenty-three (1.46%) out of 1,576 participants were eventually diagnosed with malignant tumors or high-grade intraepithelial neoplasia (HGIN). A family history of malignancy (78.26% in diagnosed cancer and HGIN vs. 46.36% in the others) was the only statistically significant parameter associated with cancer detection (p = 0.002). None of the common tumor markers were associated with the cancers screened. Except for colonoscopy (50.00%) and ultrasound for renal cancer (66.67%), the sensitivities of most screening methods were 100%. The specificities of all the screening means were above 96%. Most PPVs ranged from 30-60%. Conclusion We emphasized risk stratification for early cancer screening, such as a family history of cancer. The survey illustrated that gastric endoscopy, colonoscopy, ultrasound, and lung CT for early cancer screening had high specificity, reasonable sensitivity, and PPV. We anticipated this report would motivate larger-sample studies to estimate the risk-to-benefit ratio of cancer screening and urge the establishment of a native Chinese screening project and even guidelines.
Collapse
Affiliation(s)
- Ying Yang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), The VIP-II Gastrointestinal Cancer Division of Medical Department, Peking University Cancer Hospital & Institute, Beijing, China
| | - Peng Du
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), Department of Urology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaolu Hou
- Western Beijing Cancer Hospital, Beijing, China
| | - Kun Yan
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), Department of Ultrasonography, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ying Dai
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), Department of Ultrasonography, Peking University Cancer Hospital & Institute, Beijing, China
| | - ZhiYing Sun
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), Department of Ultrasonography, Peking University Cancer Hospital & Institute, Beijing, China
| | - Qi Wu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), Department of Endoscopy, Peking University Cancer Hospital & Institute, Beijing, China
| | - Shijie Li
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), Department of Endoscopy, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yan Yan
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), Department of Endoscopy, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhilong Wang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Liping Qi
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Mailin Chen
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hong Zheng
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), Department of Gynecologic Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Weijiao Gao
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), Department of Gynecologic Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Min Gao
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), Department of Gynecologic Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Weicheng Xue
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaodong Zhang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education, China), The VIP-II Gastrointestinal Cancer Division of Medical Department, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
16
|
Kumar SS, Khandekar N, Dani K, Bhatt SR, Duddalwar V, D’Souza A. A Scoping Review of Population Diversity in the Common Genomic Aberrations of Clear Cell Renal Cell Carcinoma. Oncology 2024; 103:341-350. [PMID: 39250899 PMCID: PMC11890214 DOI: 10.1159/000541370] [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: 02/21/2024] [Accepted: 08/27/2024] [Indexed: 09/11/2024]
Abstract
INTRODUCTION Previous literature has shown that clear cell renal cell carcinoma (ccRCC) is becoming a more prevalent diagnosis and that the incidence and mortality differ both regionally and racially. While the molecular profiles for ccRCC are studied regionally through biopsy and sequencing techniques, the genomic landscape and ccRCC diversity data are not well studied. We conducted a review of the known genomic data on 6 of the most clinically relevant DNA biomarkers in ccRCC: von Hippel-Lindau (vHL), Polybromo-1 (PBRM1), Breast Cancer Gene 1-Associated Protein 1 (BAP1), Histone-Lysine N-Methyltransferase Domain-Containing 2 (SETD2), Mammalian Target of Rapamycin (mTOR), and Lysine-Specific Demethylase 5C (KDM5C). The review compiled genomic diversity data, incidence, and risk factor differences by geographical and racial cohorts. METHODS The review methodology was created using Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) principles from articles on PubMed and Embase through July 31, 2023, written and published in English, with diagnoses of primary or metastatic ccRCC via cytology or pathology, recorded the incidence of one or more of the 6 biomarkers, explored gene aberration via sequencing, were epidemiological in nature, and/or discussed basic science research, cohort studies, or retrospective studies. RESULTS Aberrations in vHL, PBRM1, and SETD2 driving ccRCC are studied frequently, but the data are heterogeneous, whereas there is a paucity in the data regarding KDM5C, PBRM1, and mTOR mutations. CONCLUSION Studying the genetic aberrations that frequently occur in different regions gives insight into what current research lacks. When more genomic landscape research arises, precision therapy, risk calculators, and artificial intelligence may help better prognosticate and individualize treatment for those at risk for ccRCC. Provided the scarcity of existing data, and the rising prevalence of ccRCC, more studies must be conducted at the clinical level. INTRODUCTION Previous literature has shown that clear cell renal cell carcinoma (ccRCC) is becoming a more prevalent diagnosis and that the incidence and mortality differ both regionally and racially. While the molecular profiles for ccRCC are studied regionally through biopsy and sequencing techniques, the genomic landscape and ccRCC diversity data are not well studied. We conducted a review of the known genomic data on 6 of the most clinically relevant DNA biomarkers in ccRCC: von Hippel-Lindau (vHL), Polybromo-1 (PBRM1), Breast Cancer Gene 1-Associated Protein 1 (BAP1), Histone-Lysine N-Methyltransferase Domain-Containing 2 (SETD2), Mammalian Target of Rapamycin (mTOR), and Lysine-Specific Demethylase 5C (KDM5C). The review compiled genomic diversity data, incidence, and risk factor differences by geographical and racial cohorts. METHODS The review methodology was created using Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) principles from articles on PubMed and Embase through July 31, 2023, written and published in English, with diagnoses of primary or metastatic ccRCC via cytology or pathology, recorded the incidence of one or more of the 6 biomarkers, explored gene aberration via sequencing, were epidemiological in nature, and/or discussed basic science research, cohort studies, or retrospective studies. RESULTS Aberrations in vHL, PBRM1, and SETD2 driving ccRCC are studied frequently, but the data are heterogeneous, whereas there is a paucity in the data regarding KDM5C, PBRM1, and mTOR mutations. CONCLUSION Studying the genetic aberrations that frequently occur in different regions gives insight into what current research lacks. When more genomic landscape research arises, precision therapy, risk calculators, and artificial intelligence may help better prognosticate and individualize treatment for those at risk for ccRCC. Provided the scarcity of existing data, and the rising prevalence of ccRCC, more studies must be conducted at the clinical level.
Collapse
Affiliation(s)
- Sean S. Kumar
- Macon and Joan Brock Virginia Health Sciences Eastern Virginia Medical School at Old Dominion University, Norfolk, VA, USA
- Children’s Hospital Los Angeles, Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Ninad Khandekar
- Radiomics Lab, University of Southern California, Los Angeles, CA, USA
| | - Komal Dani
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Saina R. Bhatt
- Radiomics Lab, University of Southern California, Los Angeles, CA, USA
| | - Vinay Duddalwar
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Institute of Urology, University of Southern California, Los Angeles, CA, USA
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Anishka D’Souza
- Department of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
17
|
Liang D, Yang S, Ding Z, Xu X, Tang W, Wang Y, Qian K. Engineering a Bifunctional Smart Nanoplatform Integrating Nanozyme Activity and Self-Assembly for Kidney Cancer Diagnosis and Classification. ACS NANO 2024; 18:23625-23636. [PMID: 39150349 DOI: 10.1021/acsnano.4c08085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Accurate diagnosis and classification of kidney cancer are crucial for high-quality healthcare services. However, the current diagnostic platforms remain challenges in the rapid and accurate analysis of large-scale clinical biosamples. Herein, we fabricated a bifunctional smart nanoplatform based on tannic acid-modified gold nanoflowers (TA@AuNFs), integrating nanozyme catalysis for colorimetric sensing and self-assembled nanoarray-assisted LDI-MS analysis. The TA@AuNFs presented peroxidase (POD)- and glucose oxidase-like activity owing to the abundant galloyl residues on the surface of AuNFs. Combined with the colorimetric assay, the TA@AuNF-based sensing nanoplatform was used to directly detect glucose in serum for kidney tumor diagnosis. On the other hand, TA@AuNFs could self-assemble into closely packed and homogeneous two-dimensional (2D) nanoarrays at liquid-liquid interfaces by using Fe3+ as a mediator. The self-assembled TA@AuNFs (SA-TA@AuNFs) arrays were applied to assist the LDI-MS analysis of metabolites, exhibiting high ionization efficiency and excellent MS signal reproducibility. Based on the SA-TA@AuNF array-assisted LDI-MS platform, we successfully extracted metabolic fingerprints from urine samples, achieving early-stage diagnosis of kidney tumor, subtype classification, and discrimination of benign from malignant tumors. Taken together, our developed TA@AuNF-based bifunctional smart nanoplatform showed distinguished potential in clinical disease diagnosis, point-of-care testing, and biomarker discovery.
Collapse
Affiliation(s)
- Dingyitai Liang
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering and Institute of Medical Robotics, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
- Shanghai Jiao Tong University Sichuan Research Institute, Chengdu 610213, P. R. China
| | - Shouzhi Yang
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering and Institute of Medical Robotics, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
- Shanghai Jiao Tong University Sichuan Research Institute, Chengdu 610213, P. R. China
| | - Ziqi Ding
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering and Institute of Medical Robotics, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
- Shanghai Jiao Tong University Sichuan Research Institute, Chengdu 610213, P. R. China
| | - Xiaoyu Xu
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering and Institute of Medical Robotics, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
- Shanghai Jiao Tong University Sichuan Research Institute, Chengdu 610213, P. R. China
| | - Wenxuan Tang
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering and Institute of Medical Robotics, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
- Shanghai Jiao Tong University Sichuan Research Institute, Chengdu 610213, P. R. China
| | - Yuning Wang
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering and Institute of Medical Robotics, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
- Shanghai Jiao Tong University Sichuan Research Institute, Chengdu 610213, P. R. China
| | - Kun Qian
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering and Institute of Medical Robotics, Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
- Shanghai Jiao Tong University Sichuan Research Institute, Chengdu 610213, P. R. China
| |
Collapse
|
18
|
Ding LJ, Jiang X, Li T, Wang S. Role of UFMylation in tumorigenesis and cancer immunotherapy. Front Immunol 2024; 15:1454823. [PMID: 39247188 PMCID: PMC11377280 DOI: 10.3389/fimmu.2024.1454823] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 08/06/2024] [Indexed: 09/10/2024] Open
Abstract
Protein post-translational modifications (PTMs) represent a crucial aspect of cellular regulation, occurring after protein synthesis from mRNA. These modifications, which include phosphorylation, ubiquitination, acetylation, methylation, glycosylation, Sumoylation, and palmitoylation, play pivotal roles in modulating protein function. PTMs influence protein localization, stability, and interactions, thereby orchestrating a variety of cellular processes in response to internal and external stimuli. Dysregulation of PTMs is linked to a spectrum of diseases, such as cancer, inflammatory diseases, and neurodegenerative disorders. UFMylation, a type of PTMs, has recently gained prominence for its regulatory role in numerous cellular processes, including protein stability, response to cellular stress, and key signaling pathways influencing cellular functions. This review highlights the crucial function of UFMylation in the development and progression of tumors, underscoring its potential as a therapeutic target. Moreover, we discuss the pivotal role of UFMylation in tumorigenesis and malignant progression, and explore its impact on cancer immunotherapy. The article aims to provide a comprehensive overview of biological functions of UFMylation and propose how targeting UFMylation could enhance the effectiveness of cancer immunotherapy strategies.
Collapse
Affiliation(s)
- Li-Juan Ding
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xin Jiang
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Te Li
- Department of Geriatrics, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Shudong Wang
- Department of Cardiology, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
19
|
Usher-Smith JA, Masson G, Godoy A, Burge SW, Kitt J, Farquhar F, Cartledge J, Kimuli M, Burbidge S, Crosbie PAJ, Eckert C, Hancock N, Iball GR, Rogerson S, Rossi SH, Smith A, Simmonds I, Wallace T, Ward M, Callister MEJ, Stewart GD. Acceptability of adding a non-contrast abdominal CT scan to screen for kidney cancer and other abdominal pathology within a community-based CT screening programme for lung cancer: A qualitative study. PLoS One 2024; 19:e0300313. [PMID: 38950010 PMCID: PMC11216619 DOI: 10.1371/journal.pone.0300313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 02/27/2024] [Indexed: 07/03/2024] Open
Abstract
OBJECTIVES The Yorkshire Kidney Screening Trial (YKST) is a feasibility study of adding non-contrast abdominal CT scanning to screen for kidney cancer and other abdominal malignancies to community-based CT screening for lung cancer within the Yorkshire Lung Screening Trial (YLST). This study explored the acceptability of the combined screening approach to participants and healthcare professionals (HCPs) involved in the trial. METHODS We conducted semi-structured interviews with eight HCPs and 25 participants returning for the second round of scanning within YLST, 20 who had taken up the offer of the additional abdominal CT scan and five who had declined. Transcripts were analysed using thematic analysis, guided by the Theoretical Framework of Acceptability. RESULTS Overall, combining the offer of a non-contrast abdominal CT scan alongside the low-dose thoracic CT was considered acceptable to participants, including those who had declined the abdominal scan. The offer of the additional scan made sense and fitted well within the process, and participants could see benefits in terms of efficiency, cost and convenience both for themselves as individuals and also more widely for the NHS. Almost all participants made an instant decision at the point of initial invitation based more on trust and emotions than the information provided. Despite this, there was a clear desire for more time to decide whether to accept the scan or not. HCPs also raised concerns about the burden on the study team and wider healthcare system arising from additional workload both within the screening process and downstream following findings on the abdominal CT scan. CONCLUSIONS Adding a non-contrast abdominal CT scan to community-based CT screening for lung cancer is acceptable to both participants and healthcare professionals. Giving potential participants prior notice and having clear pathways for downstream management of findings will be important if it is to be offered more widely.
Collapse
Affiliation(s)
- Juliet A. Usher-Smith
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Golnessa Masson
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Angela Godoy
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
| | - Sarah W. Burge
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
| | - Jessica Kitt
- Department of Surgery, University of Cambridge, Cambridge, United Kingdom
| | - Fiona Farquhar
- Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Jon Cartledge
- Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Michael Kimuli
- Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Simon Burbidge
- Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Philip A. J. Crosbie
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Claire Eckert
- Leeds Institute of Health Science, University of Leeds, Leeds, United Kingdom
| | - Neil Hancock
- Leeds Institute of Health Science, University of Leeds, Leeds, United Kingdom
| | - Gareth R. Iball
- Faculty of Health Studies, University of Bradford, Bradford, United Kingdom
| | | | - Sabrina H. Rossi
- Department of Surgery, University of Cambridge, Cambridge, United Kingdom
| | - Andrew Smith
- Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Irene Simmonds
- Leeds Institute of Health Science, University of Leeds, Leeds, United Kingdom
| | - Tom Wallace
- Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Matthew Ward
- Leeds Institute of Health Science, University of Leeds, Leeds, United Kingdom
| | - Matthew E. J. Callister
- Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
- Leeds Institute of Health Science, University of Leeds, Leeds, United Kingdom
| | - Grant D. Stewart
- Department of Surgery, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
20
|
El-Hussieny M, Thabet DM, Tawfik HM, Gayyed MF, Toni ND. The Overexpression of NUSAP1 and GTSE1 Could Predict An Unfavourable Prognosis and Shorter Disease Free Survival in ccRenal Cell Carcinoma. Asian Pac J Cancer Prev 2024; 25:2551-2559. [PMID: 39068590 PMCID: PMC11480612 DOI: 10.31557/apjcp.2024.25.7.2551] [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: 04/16/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND Although it has been reported that NUSAP1 and GTSE1 are highly expressed in different types of tumors and associated with malignant progression and poor clinical prognosis, their significances with clinicopathological data and correlations with patients' survival in ccRCC are still poorly understood. Therefore, in our study we attempted to evaluate the link between NUSAP1 and GTSE1 in ccRCC and to correlate their immunoexpression with clinico-pathological parameters and the patients' survival to identify their significance as potential therapeutic targets, indicators for tumor progression, and patients' prognosis. METHOD NUSAP1 and GTSE1 were examined in 100 ccRCC patients by immunohistochemistry. The association between NUSAP1 and GTSE1 immunoreactivity and clinicopathological variables were evaluated. The disease free survival (DFS) was examined by the Kaplan-Meier method. The multivariate Cox regressions was estimated to detect the prognostic role of both proteins. RESULTS We detected high NUSAP1 and GTSE1 expression in 60% and 62% of the cases, respectively. A significant association was detected between NUSAP1 and GTSE1 immunoexpression and size (p=0.007 and p=0.026, respectively), Fuhrman grade (p=0.022 and p=0.004, respectively), tumor stage (p=0.003 and p=0.019, respectively), TILs (p=0.026 and p=0.04 respectively), capsular invasion (p=0.002 and p=0.009, respectively), Distant metastasis (p=0.007 and p=0.009, respectively), and DFS (p=0.007 and 0.009, respectively). Multivariate Cox regression showed that high NUSAP1 and GTSE1 expression levels were independently associated with an unfavourable poor prognosis of ccRCC cases. CONCLUSION We demonstrated that NUSAP1 and GTSE1 overexpression was closely related to the poor prognostic clinicopathological features of ccRCC and predicted an unfavorable prognosis. Therefore, NUSAP1 and GTSE1 might act together as potential futuristic prognostic indicators and therapeutic targets for ccRCC patients. However, further analysis in molecular studies on larger scale are mandatory to highlight the interactive crosstalk regulatory mechanisms between both markers and their combined effect on ccRCC.
Collapse
Affiliation(s)
| | - Dalia M. Thabet
- Department of Pathology, Faculty of Medicine, Minia University 61511, El-Minia, Egypt.
| | | | | | | |
Collapse
|
21
|
Wang Q, Chen S, Wang G, Zhang T, Gao Y. Integrated mendelian randomization analyses highlight AFF3 as a novel eQTL-mediated susceptibility gene in renal cancer and its potential mechanisms. BMC Cancer 2024; 24:739. [PMID: 38886730 PMCID: PMC11181572 DOI: 10.1186/s12885-024-12513-1] [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: 02/21/2024] [Accepted: 06/13/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUNDS A growing number of expression quantitative trait loci (eQTLs) have been found to be linked with tumorigenesis. In this article, we employed integrated Mendelian randomization (MR) analyses to identify novel susceptibility genes in renal cancer (RC) and reveal their potential mechanisms. METHODS Two-sample MR analyses were performed to infer causal relationships between eQTLs, metabolites, and RC risks through the "TwoSampleMR" R package. Sensitivity analyses, such as heterogeneity, pleiotropy, and leave-one-out analysis, were used to assess the stability of our outcomes. Summary-data-based MR (SMR) analyses were used to verify the causal relationships among cis-eQTLs and RC risks via the SMR 1.3.1 software. RESULTS Our results provided the first evidence for AFF3 eQTL elevating RC risks, suggesting its oncogenic roles (IVW method; odds ratio (OR) = 1.0005; 95% confidence interval (CI) = 1.0001-1.0010; P = 0.0285; heterogeneity = 0.9588; pleiotropy = 0.8397). Further SMR analysis validated the causal relationships among AFF3 cis-eQTLs and RC risks (P < 0.05). Moreover, the TCGA-KIRC, the ICGC-RC, and the GSE159115 datasets verified that the AFF3 gene was more highly expressed in RC tumors than normal control via scRNA-sequencing and bulk RNA-sequencing (P < 0.05). Gene set enrichment analysis (GSEA) analysis identified six potential biological pathways of AFF3 involved in RC. As for the potential mechanism of AFF3 in RC, we concluded in this article that AFF3 eQTL could negatively modulate the levels of the X-11,315 metabolite (IVW method; OR = 0.9127; 95% CI = 0.8530-0.9765; P = 0.0081; heterogeneity = 0.4150; pleiotropy = 0.8852), exhibiting preventive effects against RC risks (IVW method; OR = 0.9987; 95% CI = 0.9975-0.9999; P = 0.0380; heterogeneity = 0.5362; pleiotropy = 0.9808). CONCLUSIONS We concluded that AFF3 could serve as a novel eQTL-mediated susceptibility gene in RC and reveal its potential mechanism of elevating RC risks via negatively regulating the X-11,315 metabolite levels.
Collapse
Affiliation(s)
- Qiming Wang
- Department of Urology, Jianhu Clinical Medical College of Yangzhou University, No. 666 South Ring Road, Yancheng, Jiangsu Province, 224700, China
| | - Shaopeng Chen
- Department of Urology, Jianhu Clinical Medical College of Yangzhou University, No. 666 South Ring Road, Yancheng, Jiangsu Province, 224700, China
| | - Gang Wang
- Department of Urology, Jianhu Clinical Medical College of Yangzhou University, No. 666 South Ring Road, Yancheng, Jiangsu Province, 224700, China
| | - Tielong Zhang
- Department of Urology, Jianhu Clinical Medical College of Yangzhou University, No. 666 South Ring Road, Yancheng, Jiangsu Province, 224700, China
| | - Yulong Gao
- Department of Urology, Jianhu Clinical Medical College of Yangzhou University, No. 666 South Ring Road, Yancheng, Jiangsu Province, 224700, China.
| |
Collapse
|
22
|
Vargová D, Dargaj J, Dohál M, Fraňová S, Ľupták J, Škorňová I, Švihra J, Briš L, Slávik P, Šutovská M. Immune analysis of urine and plasma samples from patients with clear cell renal cell carcinoma. Oncol Lett 2024; 27:281. [PMID: 38736737 PMCID: PMC11082642 DOI: 10.3892/ol.2024.14414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 03/25/2024] [Indexed: 05/14/2024] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the third most common type of urological malignancy worldwide, and it is associated with a silent progression and late manifestation. Patients with a metastatic form of ccRCC have a poor prognosis; however, when the disease is diagnosed early, it is largely curable. Currently, there are no biomarkers available in clinical practice for ccRCC. Thus, the aim of the present study was to measure 27 biologically relevant cytokines in preoperative and postoperative urine samples, and in preoperative plasma samples from 34 patients with ccRCC, and to evaluate their diagnostic significance. The concentrations of cytokines were assessed by multiplex immune assay. The results showed significantly higher levels of IL-1 receptor antagonist, IL-6, IL-15, chemokine (C-C motif) ligand (CCL)2, CCL3, CCL4, C-X-C motif ligand (CXCL)10, granulocyte-macrophage colony stimulating factor (GM-CSF) and platelet-derived growth factor-BB (PDGF-BB), and lower levels of granulocyte colony stimulating factor (G-CSF) in urine samples from patients prior to surgery compared with those in the controls. Notably, the urine levels of G-CSF, IL-5 and vascular endothelial growth factor differed following tumor removal compared with the preoperative urine levels. In addition, urinary G-CSF, GM-CSF, IL-6, CXCL10, CCL5 and PDGF-BB appeared to be potential markers of tumor grade. Plasma from patients with ccRCC contained significantly higher levels of IL-6 and lower levels of CCL2 than control plasma. In conclusion, the present findings indicated that urinary and circulating cytokines may represent a promising novel tool for the early diagnosis of ccRCC and/or prediction of tumor grade.
Collapse
Affiliation(s)
- Daniela Vargová
- Department of Pharmacology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Ján Dargaj
- Department of Urology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava and University Hospital Martin, 036 01 Martin, Slovakia
| | - Matúš Dohál
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Soňa Fraňová
- Department of Pharmacology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Ján Ľupták
- Department of Urology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava and University Hospital Martin, 036 01 Martin, Slovakia
| | - Ingrid Škorňová
- Department of Hematology and Transfusiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava and University Hospital Martin, 036 01 Martin, Slovakia
| | - Ján Švihra
- Department of Urology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava and University Hospital Martin, 036 01 Martin, Slovakia
| | - Lukáš Briš
- Department of Urology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava and University Hospital Martin, 036 01 Martin, Slovakia
| | - Pavol Slávik
- Department of Pathological Anatomy, Jessenius Faculty of Medicine in Martin, Comenius University, 036 01 Martin, Slovakia
| | - Martina Šutovská
- Department of Pharmacology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| |
Collapse
|
23
|
Lin Z, Xiao S, Qi Y, Guo J, Lu L. Tumor infiltrating B lymphocytes (TIBs) associate with poor clinical outcomes, unfavorable therapeutic benefit and immunosuppressive context in metastatic clear cell renal cell carcinoma (mccRCC) patients treated with anti-PD-1 antibody plus Axitinib. J Cancer Res Clin Oncol 2024; 150:262. [PMID: 38762825 PMCID: PMC11102881 DOI: 10.1007/s00432-024-05803-5] [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: 04/27/2024] [Accepted: 05/13/2024] [Indexed: 05/20/2024]
Abstract
PURPOSE Immune checkpoint inhibitors (ICIs) plus tyrosine kinase inhibitors (TKIs) has become first-line therapy for metastatic renal cell carcinoma patients. This study aims to investigate the effect of tumor infiltrating B lymphocytes (TIBs) on the combination therapy. METHODS The retrospective analysis was conducted on the clinical records of 115 metastatic clear cell renal cell carcinoma (mccRCC) patients treated with anti-PD-1 antibody plus Axitinib between March 2020 and June 2023. Observation target: objective response rate (ORR), and overall survival (OS), progression-free survival (PFS) and immune profile. RESULTS Patients with high TIBs portended lower ORR of the combination therapy (p = 0.033). TIBs was an independent predictor for poorer OS (p = 0.013) and PFS (p = 0.021) in mccRCC patients with combination treatment. TIBs infiltration was associated with more CD4+T (p < 0.001), CD8+T (p < 0.001), M2 macrophages (p = 0.020) and regulatory T cells (Tregs) (p = 0.004). In TIBs high patients, the percentages of PD-1, CTLA-4 and TIM-3 positive rate were significantly increased in CD4+T (p = 0.038, 0.029 and 0.002 respectively) and CD8+T cells (p = 0.006, 0.026 and < 0.001 respectively). CONCLUSIONS Our study revealed TIBs infiltration predicted adverse outcomes in mccRCC patients treated with anti-PD-1 antibody plus Axitinib. As a corollary, TIBs positively associated with M2 macrophages and Tregs, leading to subsequent multiple immune checkpoints related exhaustion of T cells. Thus, only PD-1 blockade are inadequate to reverse T cells exhaustion effectively in high TIBs mccRCC patients.
Collapse
Affiliation(s)
- Zhiyuan Lin
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shuxiu Xiao
- Clinical Center for Biotherapy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yu Qi
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jianming Guo
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lili Lu
- Clinical Center for Biotherapy, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
24
|
Runarsson TG, Bergmann A, Erlingsdottir G, Petursdottir V, Heitmann LA, Johannesson A, Asbjornsson V, Axelsson T, Hilmarsson R, Gudbjartsson T. An epidemiological and clinicopathological study of type 1 vs. type 2 morphological subtypes of papillary renal cell carcinoma- results from a nation-wide study covering 50 years in Iceland. BMC Urol 2024; 24:105. [PMID: 38741053 DOI: 10.1186/s12894-024-01494-9] [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: 12/24/2023] [Accepted: 05/02/2024] [Indexed: 05/16/2024] Open
Abstract
INTRODUCTION Papillary renal cell carcinoma (pRCC) is the second most common histology of renal cell carcinoma (RCC), accounting for 10-15% of cases. Traditionally, pRCC is divided into type 1 and type 2, although this division is currently debated as a prognostic factor of survival. Our aim was to investigate the epidemiology and survival of the pRCC subtypes in a whole nation cohort of patients during a 50-year period. MATERIALS AND METHODS A Population based retrospective study including consecutive cases of RCC in Iceland from 1971-2020. Comparisons were made between histological classifications of RCC, with emphasis on pRCC subtypes (type 1 vs. 2) for outcome estimation. Changes in RCC incidence were analyzed in 5-year intervals after age standardization. The Kaplan-Meier method and Cox regression were used for outcome analysis. RESULTS A total of 1.725 cases were identified, with 74.4%, 2.1% and 9.2% having clear cell (ccRCC), chromophobe (chRCC), and pRCC, respectively. The age standardized incidence (ASI) of pRCC was 1.97/100.000 for males and 0.5/100.000 for females, and the proportion of pRCC increased from 3.7% to 11.5% between the first and last intervals of the study (p < 0.001). Age standardized cancer specific mortality (ASCSM) of pRCC was 0.6/100.000 and 0.19/100.000 for males and females, respectively. The annual average increase in ASI was 3.6% for type 1 pRCC, but the ASI for type 2 pRCC and ASCSM for both subtypes did not change significantly. Male to female ratio was 4.4 for type 1 pRCC and 2.3 for type 2. The average tumor size for type 1 and 2 was 58.8 and 73.7 mm, respectively. Metastasis at diagnosis was found in 8.7% in the type 1 pRCC, compared to 30.0% of patients with type 2 pRCC (p < 0.001). Estimated 5-year cancer-specific survival (CSS) were 94.4%, 80.7%, and 69.3% for chRCC, pRCC and ccRCC, respectively (p < 0.001). For the pRCC subtypes, type 1 was associated with better 5-year CSS than type 2 (86.3% vs. 66.0%, p < 0.001), although this difference was not significant after adjusting for cancer stage and grading. CONCLUSIONS pRCC histology was slightly less common in Iceland than in other countries. Males are more than three times more likely to be diagnosed with pRCC, compared to other RCC histologies. The subtype of pRCC was not found to be an independent risk factor for worse survival, and as suggested by the most recent WHO Classification of Urinary Tumors, grade and TNM-stage seem to be the most important factors for estimation of survival for pRCC patients.
Collapse
Affiliation(s)
| | - Andreas Bergmann
- Department of Urology and Surgery in Landspitali University Hospital, Reykjavik, Iceland
| | - Gigja Erlingsdottir
- Department of Pathology in Landspitali University Hospital, Reykjavik, Iceland
| | - Vigdis Petursdottir
- Department of Pathology in Landspitali University Hospital, Reykjavik, Iceland
| | | | - Aevar Johannesson
- Department of Statistics in University of Iceland, Reykjavik, Iceland
| | | | - Tomas Axelsson
- Department of Urology in Danderyd Hospital, Stockholm, Sweden
| | - Rafn Hilmarsson
- Department of Urology and Surgery in Landspitali University Hospital, Reykjavik, Iceland
| | - Tomas Gudbjartsson
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland.
- Department of Urology and Surgery in Landspitali University Hospital, Reykjavik, Iceland.
- Department of Surgery and Urology, Landspitali University Hospital, University of Iceland, Hringbraut IS-101, Reykjavik, Iceland.
| |
Collapse
|
25
|
Purdue MP, Dutta D, Machiela MJ, Gorman BR, Winter T, Okuhara D, Cleland S, Ferreiro-Iglesias A, Scheet P, Liu A, Wu C, Antwi SO, Larkin J, Zequi SC, Sun M, Hikino K, Hajiran A, Lawson KA, Cárcano F, Blanchet O, Shuch B, Nepple KG, Margue G, Sundi D, Diver WR, Folgueira MAAK, van Bokhoven A, Neffa F, Brown KM, Hofmann JN, Rhee J, Yeager M, Cole NR, Hicks BD, Manning MR, Hutchinson AA, Rothman N, Huang WY, Linehan WM, Lori A, Ferragu M, Zidane-Marinnes M, Serrano SV, Magnabosco WJ, Vilas A, Decia R, Carusso F, Graham LS, Anderson K, Bilen MA, Arciero C, Pellegrin I, Ricard S, Scelo G, Banks RE, Vasudev NS, Soomro N, Stewart GD, Adeyoju A, Bromage S, Hrouda D, Gibbons N, Patel P, Sullivan M, Protheroe A, Nugent FI, Fournier MJ, Zhang X, Martin LJ, Komisarenko M, Eisen T, Cunningham SA, Connolly DC, Uzzo RG, Zaridze D, Mukeria A, Holcatova I, Hornakova A, Foretova L, Janout V, Mates D, Jinga V, Rascu S, Mijuskovic M, Savic S, Milosavljevic S, Gaborieau V, Abedi-Ardekani B, McKay J, Johansson M, Phouthavongsy L, Hayman L, Li J, Lungu I, Bezerra SM, Souza AG, Sares CTG, Reis RB, Gallucci FP, Cordeiro MD, et alPurdue MP, Dutta D, Machiela MJ, Gorman BR, Winter T, Okuhara D, Cleland S, Ferreiro-Iglesias A, Scheet P, Liu A, Wu C, Antwi SO, Larkin J, Zequi SC, Sun M, Hikino K, Hajiran A, Lawson KA, Cárcano F, Blanchet O, Shuch B, Nepple KG, Margue G, Sundi D, Diver WR, Folgueira MAAK, van Bokhoven A, Neffa F, Brown KM, Hofmann JN, Rhee J, Yeager M, Cole NR, Hicks BD, Manning MR, Hutchinson AA, Rothman N, Huang WY, Linehan WM, Lori A, Ferragu M, Zidane-Marinnes M, Serrano SV, Magnabosco WJ, Vilas A, Decia R, Carusso F, Graham LS, Anderson K, Bilen MA, Arciero C, Pellegrin I, Ricard S, Scelo G, Banks RE, Vasudev NS, Soomro N, Stewart GD, Adeyoju A, Bromage S, Hrouda D, Gibbons N, Patel P, Sullivan M, Protheroe A, Nugent FI, Fournier MJ, Zhang X, Martin LJ, Komisarenko M, Eisen T, Cunningham SA, Connolly DC, Uzzo RG, Zaridze D, Mukeria A, Holcatova I, Hornakova A, Foretova L, Janout V, Mates D, Jinga V, Rascu S, Mijuskovic M, Savic S, Milosavljevic S, Gaborieau V, Abedi-Ardekani B, McKay J, Johansson M, Phouthavongsy L, Hayman L, Li J, Lungu I, Bezerra SM, Souza AG, Sares CTG, Reis RB, Gallucci FP, Cordeiro MD, Pomerantz M, Lee GSM, Freedman ML, Jeong A, Greenberg SE, Sanchez A, Thompson RH, Sharma V, Thiel DD, Ball CT, Abreu D, Lam ET, Nahas WC, Master VA, Patel AV, Bernhard JC, Freedman ND, Bigot P, Reis RM, Colli LM, Finelli A, Manley BJ, Terao C, Choueiri TK, Carraro DM, Houlston R, Eckel-Passow JE, Abbosh PH, Ganna A, Brennan P, Gu J, Chanock SJ. Multi-ancestry genome-wide association study of kidney cancer identifies 63 susceptibility regions. Nat Genet 2024; 56:809-818. [PMID: 38671320 DOI: 10.1038/s41588-024-01725-7] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 03/13/2024] [Indexed: 04/28/2024]
Abstract
Here, in a multi-ancestry genome-wide association study meta-analysis of kidney cancer (29,020 cases and 835,670 controls), we identified 63 susceptibility regions (50 novel) containing 108 independent risk loci. In analyses stratified by subtype, 52 regions (78 loci) were associated with clear cell renal cell carcinoma (RCC) and 6 regions (7 loci) with papillary RCC. Notably, we report a variant common in African ancestry individuals ( rs7629500 ) in the 3' untranslated region of VHL, nearly tripling clear cell RCC risk (odds ratio 2.72, 95% confidence interval 2.23-3.30). In cis-expression quantitative trait locus analyses, 48 variants from 34 regions point toward 83 candidate genes. Enrichment of hypoxia-inducible factor-binding sites underscores the importance of hypoxia-related mechanisms in kidney cancer. Our results advance understanding of the genetic architecture of kidney cancer, provide clues for functional investigation and enable generation of a validated polygenic risk score with an estimated area under the curve of 0.65 (0.74 including risk factors) among European ancestry individuals.
Collapse
Affiliation(s)
- Mark P Purdue
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA.
| | - Diptavo Dutta
- Integrative Tumor Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Mitchell J Machiela
- Integrative Tumor Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | | | - Timothy Winter
- Laboratory of Genetic Susceptibility, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | | | | | | | - Paul Scheet
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Aoxing Liu
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Chao Wu
- Biosample Repository, Fox Chase Cancer Center-Temple Health, Philadelphia, PA, USA
| | - Samuel O Antwi
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - James Larkin
- Department of Medical Oncology, Royal Marsden NHS Foundation Trust, London, UK
| | - Stênio C Zequi
- Department of Urology, A.C. Camargo Cancer Center, São Paulo, Brazil
- National Institute for Science and Technology in Oncogenomics and Therapeutic Innovation INCIT-INOTE, São Paulo, Brazil
- Latin American Renal Cancer Group, São Paulo, Brazil
- Department of Surgery, Division of Urology, São Paulo Federal University, São Paulo, Brazil
| | - Maxine Sun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Keiko Hikino
- Laboratory for Pharmacogenomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Ali Hajiran
- Department of Urology, Division of Urologic Oncology, West Virginia University Cancer Institute, Morgantown, WV, USA
| | - Keith A Lawson
- Department of Surgical Oncology, Division of Urology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Flavio Cárcano
- Department of Medical Oncology, Barretos Cancer Hospital, Barretos, Brazil
| | | | - Brian Shuch
- Department of Urology, UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Kenneth G Nepple
- Department of Urology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| | - Gaëlle Margue
- Department of Urology, CHU Bordeaux, Bordeaux, France
| | - Debasish Sundi
- Department of Urology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - W Ryan Diver
- Department of Population Science, American Cancer Society, Atlanta, GA, USA
| | - Maria A A K Folgueira
- Departments of Radiology and Oncology, Comprehensive Center for Precision Oncology-C2PO, Centro de Investigação Translacional em Oncologia, Instituto do Cancer do Estado de São Paulo, Hospital das Clinicas, Faculdade de Medicina Universidade de São Paulo, São Paulo, Brazil
| | - Adrie van Bokhoven
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Kevin M Brown
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Jonathan N Hofmann
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Jongeun Rhee
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Meredith Yeager
- Cancer Genomics Research Laboratory, Frederick National Laboratory, Rockville, MD, USA
| | - Nathan R Cole
- Cancer Genomics Research Laboratory, Frederick National Laboratory, Rockville, MD, USA
| | - Belynda D Hicks
- Cancer Genomics Research Laboratory, Frederick National Laboratory, Rockville, MD, USA
| | - Michelle R Manning
- Cancer Genomics Research Laboratory, Frederick National Laboratory, Rockville, MD, USA
| | - Amy A Hutchinson
- Cancer Genomics Research Laboratory, Frederick National Laboratory, Rockville, MD, USA
| | - Nathaniel Rothman
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Wen-Yi Huang
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - W Marston Linehan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Adriana Lori
- Department of Population Science, American Cancer Society, Atlanta, GA, USA
| | | | | | - Sérgio V Serrano
- Department of Medical Oncology, Barretos Cancer Hospital, Barretos, Brazil
| | | | - Ana Vilas
- Department of Pathology, Hospital Pasteur, Montevideo, Uruguay
| | - Ricardo Decia
- Department of Urology, Hospital Pasteur, Montevideo, Uruguay
| | | | - Laura S Graham
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kyra Anderson
- Oncology Clinical Research Support Team, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Mehmet A Bilen
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Cletus Arciero
- Department of Surgery, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Solène Ricard
- Department of Urology, CHU Bordeaux, Bordeaux, France
| | - Ghislaine Scelo
- Observational and Pragmatic Research Institute Pte Ltd, Singapore, Singapore
| | - Rosamonde E Banks
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Naveen S Vasudev
- Department of Oncology, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Naeem Soomro
- Department of Urology, Newcastle Hospitals NHS Foundation Trust, Newcastle, UK
| | - Grant D Stewart
- Department of Urology, Western General Hospital, NHS Lothian, Edinburgh, UK
- Department of Surgery, University of Cambridge, Cambridge, UK
| | - Adebanji Adeyoju
- Department of Urology, Stockport NHS Foundation Trust, Stockport, UK
| | - Stephen Bromage
- Department of Urology, Stockport NHS Foundation Trust, Stockport, UK
| | - David Hrouda
- Department of Urology, Imperial College Healthcare NHS Trust, London, UK
| | - Norma Gibbons
- Department of Urology, Imperial College Healthcare NHS Trust, London, UK
| | - Poulam Patel
- Division of Oncology, University of Nottingham, Nottingham, UK
| | - Mark Sullivan
- Department of Urology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Andrew Protheroe
- Department of Oncology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Francesca I Nugent
- Department of Urology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| | | | - Xiaoyu Zhang
- Department of Surgical Oncology, Division of Urology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Lisa J Martin
- Department of Surgical Oncology, Division of Urology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Maria Komisarenko
- Department of Surgical Oncology, Division of Urology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Timothy Eisen
- Department of Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Sonia A Cunningham
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Denise C Connolly
- Cancer Signaling and Microenvironment, Biosample Repository Facility, Fox Chase Cancer Center-Temple Health, Philadelphia, PA, USA
| | - Robert G Uzzo
- Department of Urology, Fox Chase Cancer Center-Temple Health, Philadelphia, PA, USA
| | - David Zaridze
- Department of Clinical Epidemiology, N.N. Blokhin National Medical Research Centre of Oncology, Moscow, Russia
| | - Anush Mukeria
- Department of Clinical Epidemiology, N.N. Blokhin National Medical Research Centre of Oncology, Moscow, Russia
| | - Ivana Holcatova
- Institute of Public Health and Preventive Medicine, Second Faculty of Medicine, Charles University, Prague, Czech Republic
- Department of Oncology, Second Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Anna Hornakova
- Institute of Hygiene and Epidemiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Lenka Foretova
- Department of Cancer Epidemiology and Genetics, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Vladimir Janout
- Faculty of Health Sciences, Palacky University, Olomouc, Czech Republic
| | - Dana Mates
- Department of Occupational Health and Toxicology, National Center for Environmental Risk Monitoring, National Institute of Public Health, Bucharest, Romania
| | - Viorel Jinga
- Urology Department, Academy of Romanian Scientists, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Stefan Rascu
- Urology Department, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Mirjana Mijuskovic
- Clinic of Nephrology, Faculty of Medicine, Military Medical Academy, Belgrade, Serbia
| | - Slavisa Savic
- Department of Urology, Clinical Hospital Center Dr Dragisa Misovic Dedinje, Belgrade, Serbia
| | - Sasa Milosavljevic
- International Organisation for Cancer Prevention and Research, Belgrade, Serbia
| | - Valérie Gaborieau
- Genomic Epidemiology Branch, International Agency for Research on Cancer, Lyon, France
| | | | - James McKay
- Genomic Epidemiology Branch, International Agency for Research on Cancer, Lyon, France
| | - Mattias Johansson
- Genomic Epidemiology Branch, International Agency for Research on Cancer, Lyon, France
| | - Larry Phouthavongsy
- Ontario Tumour Bank, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Lindsay Hayman
- Diagnostic Development Program, Tissue Portal, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Jason Li
- Diagnostic Development Program, Tissue Portal, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Ilinca Lungu
- Ontario Tumour Bank, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- Diagnostic Development Program, Tissue Portal, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | | | - Aline G Souza
- Departments of Medical Imaging, Hematology and Oncology, Division of Medical Oncology, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, Brazil
| | - Claudia T G Sares
- Departments of Surgery and Anatomy, Division of Urology, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, Brazil
| | - Rodolfo B Reis
- Departments of Surgery and Anatomy, Division of Urology, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, Brazil
| | - Fabio P Gallucci
- Surgery Department, Urology Division, Instituto do Cancer do Estado de São Paulo, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Mauricio D Cordeiro
- Surgery Department, Urology Division, Instituto do Cancer do Estado de São Paulo, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | | | - Gwo-Shu M Lee
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Matthew L Freedman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Anhyo Jeong
- Department of Urology, UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Samantha E Greenberg
- Department of Population Sciences, Genetic Counseling Shared Resource, Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Alejandro Sanchez
- Department of Surgery, Division of Urology, Huntsman Cancer Institute and University of Utah, Salt Lake City, UT, USA
| | | | - Vidit Sharma
- Department of Urology, Mayo Clinic, Rochester, MN, USA
| | - David D Thiel
- Department of Urology, Mayo Clinic, Jacksonville, FL, USA
| | - Colleen T Ball
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Diego Abreu
- Department of Urology, Hospital Pasteur, Montevideo, Uruguay
| | - Elaine T Lam
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - William C Nahas
- Surgery Department, Urology Division, Instituto do Cancer do Estado de São Paulo, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Viraj A Master
- Department of Urology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Alpa V Patel
- Department of Population Science, American Cancer Society, Atlanta, GA, USA
| | | | - Neal D Freedman
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Pierre Bigot
- Department of Urology, CHU Angers, Angers, France
| | - Rui M Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
| | - Leandro M Colli
- Departament of Medical Image, Hematology and Oncology, Division of Medical Oncology, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, Brazil
| | - Antonio Finelli
- Department of Surgical Oncology, Division of Urology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Brandon J Manley
- Genitourinary Oncology Program, Moffitt Cancer Center, Tampa, FL, USA
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Toni K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Dirce M Carraro
- Clinical and Functional Genomics Group, CIPE (International Research Center), A.C. Camargo Cancer Center, São Paulo, Brazil
| | - Richard Houlston
- Division of Genetics and Epidemiology, Institute of Cancer Research, Sutton, UK
| | | | - Philip H Abbosh
- Department of Nuclear Dynamics and Cancer, Fox Chase Cancer Center-Temple Health, Philadelphia, PA, USA
| | - Andrea Ganna
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Paul Brennan
- Genomic Epidemiology Branch, International Agency for Research on Cancer, Lyon, France
| | - Jian Gu
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stephen J Chanock
- Laboratory of Genetic Susceptibility, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA.
| |
Collapse
|
26
|
Kim YH, Chung JS, Lee HH, Park JH, Kim MK. Influence of Dietary Polyunsaturated Fatty Acid Intake on Potential Lipid Metabolite Diagnostic Markers in Renal Cell Carcinoma: A Case-Control Study. Nutrients 2024; 16:1265. [PMID: 38732512 PMCID: PMC11085891 DOI: 10.3390/nu16091265] [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: 03/29/2024] [Revised: 04/21/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Non-invasive diagnostics are crucial for the timely detection of renal cell carcinoma (RCC), significantly improving survival rates. Despite advancements, specific lipid markers for RCC remain unidentified. We aimed to discover and validate potent plasma markers and their association with dietary fats. Using lipid metabolite quantification, machine-learning algorithms, and marker validation, we identified RCC diagnostic markers in studies involving 60 RCC and 167 healthy controls (HC), as well as 27 RCC and 74 HC, by analyzing their correlation with dietary fats. RCC was associated with altered metabolism in amino acids, glycerophospholipids, and glutathione. We validated seven markers (l-tryptophan, various lysophosphatidylcholines [LysoPCs], decanoylcarnitine, and l-glutamic acid), achieving a 96.9% AUC, effectively distinguishing RCC from HC. Decreased decanoylcarnitine, due to reduced carnitine palmitoyltransferase 1 (CPT1) activity, was identified as affecting RCC risk. High intake of polyunsaturated fatty acids (PUFAs) was negatively correlated with LysoPC (18:1) and LysoPC (18:2), influencing RCC risk. We validated seven potential markers for RCC diagnosis, highlighting the influence of high PUFA intake on LysoPC levels and its impact on RCC occurrence via CPT1 downregulation. These insights support the efficient and accurate diagnosis of RCC, thereby facilitating risk mitigation and improving patient outcomes.
Collapse
Affiliation(s)
- Yeon-Hee Kim
- Cancer Epidemiology Branch, Division of Cancer Epidemiology and Prevention, National Cancer Center, 323 Ilsandong-gu, Goyang-si 10408, Republic of Korea; (Y.-H.K.); (J.-H.P.)
| | - Jin-Soo Chung
- Department of Urology, Center for Urologic Cancer, Research Institute, Hospital of National Cancer Center, 323 Ilsandong-gu, Goyang-si 10408, Republic of Korea; (J.-S.C.); (H.-H.L.)
| | - Hyung-Ho Lee
- Department of Urology, Center for Urologic Cancer, Research Institute, Hospital of National Cancer Center, 323 Ilsandong-gu, Goyang-si 10408, Republic of Korea; (J.-S.C.); (H.-H.L.)
| | - Jin-Hee Park
- Cancer Epidemiology Branch, Division of Cancer Epidemiology and Prevention, National Cancer Center, 323 Ilsandong-gu, Goyang-si 10408, Republic of Korea; (Y.-H.K.); (J.-H.P.)
| | - Mi-Kyung Kim
- Cancer Epidemiology Branch, Division of Cancer Epidemiology and Prevention, National Cancer Center, 323 Ilsandong-gu, Goyang-si 10408, Republic of Korea; (Y.-H.K.); (J.-H.P.)
| |
Collapse
|
27
|
Yang M, Cao L, Lu T, Xiao C, Wu Z, Jiang X, Wang W, Li H. Ultrasound-guided erector spinae plane block for perioperative analgesia in patients undergoing laparoscopic nephrectomy surgery: A randomized controlled trial. Heliyon 2024; 10:e26422. [PMID: 38434013 PMCID: PMC10906293 DOI: 10.1016/j.heliyon.2024.e26422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 02/12/2024] [Accepted: 02/13/2024] [Indexed: 03/05/2024] Open
Abstract
Study objective Kidney neoplasms have a high incidence, and radical nephrectomy or partial nephrectomy are the main treatment options. Our study aims to investigate the use of ultrasound-guided erector spinae plane block for perioperative analgesia in patients undergoing laparoscopic nephrectomy surgery. Design Prospective, randomized, double-blind. Setting University hospital. Patients Our study included 50 patients (ASA I-III) who underwent laparoscopic nephrectomy at the hospital of Second Affiliated Hospital of Army Medical University. Interventions The patients were divided into two groups: the ESPB group and the control group. In the ESPB group, a mixture of 10 mL of 1% lidocaine, 10 mL of 0.7% ropivacaine, 0.5 μg/kg dexmedetomidine, and 5 mg of dexamethasone was administered. In the control group, 20 mL of 0.9% saline was administered. Measurements The primary outcome measure was the total consumption of sufentanil during the intraoperative period. Secondary outcome measures included visual analogue scale (VAS) pain scores at rest and during coughing at 1 h, 6 h, 12 h, 24 h, and 48 h postoperatively, intraoperative consumption of remifentanil, frequency of rescue analgesic administration, consumption of rescue analgesia and incidence of postoperative nausea and vomiting within 48 h. Results The ESPB group exhibited lower intraoperative consumption of sufentanil, lower consumption of rescue analgesia, as well as VAS scores at rest and during coughing within the first 24 h postoperatively, compared to the control group. However, no significant differences were observed in VAS scores at 48 h postoperatively, postoperative nausea and vomiting, or the need for postoperative rescue analgesia. Conclusions Ultrasound-guided ESPB performed in patients who underwent laparoscopic nephrectomy demonstrated a substantial decrease in intraoperative opioid consumption, as well as lower VAS scores at rest and during coughing in the postoperative period.
Collapse
Affiliation(s)
- Ming Yang
- Department of Anesthesiology, Xinqiao Hospital of Chongqing, Second Affiliated Hospital of Army Medical University, PLA, Chongqing, 400037, China
| | - Lei Cao
- Department of Anesthesiology, Xinqiao Hospital of Chongqing, Second Affiliated Hospital of Army Medical University, PLA, Chongqing, 400037, China
| | - Tong Lu
- Department of Anesthesiology, Xinqiao Hospital of Chongqing, Second Affiliated Hospital of Army Medical University, PLA, Chongqing, 400037, China
| | - Cheng Xiao
- Department of Anesthesiology, Xinqiao Hospital of Chongqing, Second Affiliated Hospital of Army Medical University, PLA, Chongqing, 400037, China
| | - Zhuoxi Wu
- Department of Anesthesiology, Xinqiao Hospital of Chongqing, Second Affiliated Hospital of Army Medical University, PLA, Chongqing, 400037, China
| | - Xuetao Jiang
- Department of Anesthesiology, Xinqiao Hospital of Chongqing, Second Affiliated Hospital of Army Medical University, PLA, Chongqing, 400037, China
| | - Wei Wang
- Department of Anesthesiology, Xinqiao Hospital of Chongqing, Second Affiliated Hospital of Army Medical University, PLA, Chongqing, 400037, China
| | - Hong Li
- Department of Anesthesiology, Xinqiao Hospital of Chongqing, Second Affiliated Hospital of Army Medical University, PLA, Chongqing, 400037, China
| |
Collapse
|
28
|
Rossi SH, Harrison H, Usher-Smith JA, Stewart GD. Risk-stratified screening for the early detection of kidney cancer. Surgeon 2024; 22:e69-e78. [PMID: 37993323 DOI: 10.1016/j.surge.2023.10.010] [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: 09/27/2023] [Revised: 10/22/2023] [Accepted: 10/30/2023] [Indexed: 11/24/2023]
Abstract
Earlier detection and screening for kidney cancer has been identified as a key research priority, however the low prevalence of the disease in unselected populations limits the cost-effectiveness of screening. Risk-stratified screening for kidney cancer may improve early detection by targeting high-risk individuals whilst limiting harms in low-risk individuals, potentially increasing the cost-effectiveness of screening. A number of models have been identified which estimate kidney cancer risk based on both phenotypic and genetic data, and while several of the former have been shown to identify individuals at high-risk of developing kidney cancer with reasonable accuracy, current evidence does not support including a genetic component. Combined screening for lung cancer and kidney cancer has been proposed, as the two malignancies share some common risk factors. A modelling study estimated that using lung cancer risk models (currently used for risk-stratified lung cancer screening) could capture 25% of patients with kidney cancer, which is only slightly lower than using the best performing kidney cancer-specific risk models based on phenotypic data (27%-33%). Additionally, risk-stratified screening for kidney cancer has been shown to be acceptable to the public. The following review summarises existing evidence regarding risk-stratified screening for kidney cancer, highlighting the risks and benefits, as well as exploring the management of potential harms and further research needs.
Collapse
Affiliation(s)
- Sabrina H Rossi
- Department of Surgery, University of Cambridge, Cambridge, UK.
| | - Hannah Harrison
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Juliet A Usher-Smith
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Grant D Stewart
- Department of Surgery, University of Cambridge, Cambridge, UK
| |
Collapse
|
29
|
Leulmi Pichot S, Vemulkar T, Verheyen J, Wallis L, Jones JO, Stewart AP, Welsh SJ, Stewart GD, Cowburn RP. Lithographically defined encoded magnetic heterostructures for the targeted screening of kidney cancer. NANOSCALE ADVANCES 2023; 6:276-286. [PMID: 38125591 PMCID: PMC10729922 DOI: 10.1039/d3na00701d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/11/2023] [Indexed: 12/23/2023]
Abstract
Renal cell carcinoma (RCC) is the 7th commonest cancer in the UK and the most lethal urological malignancy; 50% of all RCC patients will die from the condition. However, if identified early enough, small RCCs are usually cured by surgery or percutaneous procedures, with 95% 10 year survival. This study describes a newly developed non-invasive urine-based assay for the early detection of RCC. Our approach uses encoded magnetically controllable heterostructures as a substrate for immunoassays. These heterostructures have molecular recognition abilities and embedded patterned codes for a rapid identification of RCC biomarkers. The magnetic heterostructures developed for this study have a magnetic configuration designed for a remote multi axial control of their orientation by external magnetic fields, this control facilitates the code readout when the heterostructures are in liquid. Furthermore, the optical encoding of each set of heterostructures provides a multiplexed analyte capture platform, as different sets of heterostructures, specific to different biomarkers can be mixed together in a patient sample. Our results show a precise magnetic control of the heterostructures with an efficient code readout during liquid immunoassays. The use of functionalised magnetic heterostructures as a substrate for immunoassay is validated for urine specimen spiked with recombinant RCC biomarkers. Initial results of the newly proposed screening method on urine samples from RCC patients, and controls with no renal disorders are presented in this study. Comprehensive optimisation cycles are in progress to validate the robustness of this technology as a novel, non-invasive screening method for RCC.
Collapse
Affiliation(s)
- Selma Leulmi Pichot
- The Cavendish Laboratory, Department of Physics, University of Cambridge Cambridge CB3 0HE UK
| | | | | | - Lauren Wallis
- Department of Surgery, University of Cambridge, Cambridge Biomedical Campus Cambridge CB2 0QQ UK
| | - James O Jones
- Department of Oncology, University of Cambridge, Cambridge Biomedical Campus Cambridge CB2 0QQ UK
| | - Andrew P Stewart
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, MRC Laboratory of Molecular Biology Cambridge Biomedical Campus Cambridge CB2 0QQ UK
| | - Sarah J Welsh
- Department of Surgery, University of Cambridge, Cambridge Biomedical Campus Cambridge CB2 0QQ UK
| | - Grant D Stewart
- Department of Surgery, University of Cambridge, Cambridge Biomedical Campus Cambridge CB2 0QQ UK
| | - Russell P Cowburn
- The Cavendish Laboratory, Department of Physics, University of Cambridge Cambridge CB3 0HE UK
| |
Collapse
|
30
|
Campi R, Rebez G, Klatte T, Roussel E, Ouizad I, Ingels A, Pavan N, Kara O, Erdem S, Bertolo R, Capitanio U, Mir MC. Effect of smoking, hypertension and lifestyle factors on kidney cancer - perspectives for prevention and screening programmes. Nat Rev Urol 2023; 20:669-681. [PMID: 37328546 DOI: 10.1038/s41585-023-00781-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/12/2023] [Indexed: 06/18/2023]
Abstract
Renal cell carcinoma (RCC) incidence has doubled over the past few decades. However, death rates have remained stable as the number of incidental renal mass diagnoses peaked. RCC has been recognized as a European health care issue, but to date, no screening programmes have been introduced. Well-known modifiable risk factors for RCC are smoking, obesity and hypertension. A direct association between cigarette consumption and increased RCC incidence and RCC-related death has been reported, but the underlying mechanistic pathways for this association are still unclear. Obesity is associated with an increased risk of RCC, but interestingly, improved survival outcomes have been reported in obese patients, a phenomenon known as the obesity paradox. Data on the association between other modifiable risk factors such as diet, dyslipidaemia and physical activity with RCC incidence are conflicting, and potential mechanisms underlying these associations remain to be elucidated.
Collapse
Affiliation(s)
- Riccardo Campi
- Department of Urology, University of Florence, Careggi Hospital, Florence, Italy
- Young Academic Urologists (YAU) Renal Cancer Working Group, Arnhem, Netherlands
| | - Giacomo Rebez
- Young Academic Urologists (YAU) Renal Cancer Working Group, Arnhem, Netherlands
- Department of Urology, Cattinara Hospital, University of Trieste, Trieste, Italy
| | - Tobias Klatte
- Young Academic Urologists (YAU) Renal Cancer Working Group, Arnhem, Netherlands
- Department of Urology, Royal Bournemouth Hospital, Bournemouth, UK
| | - Eduard Roussel
- Young Academic Urologists (YAU) Renal Cancer Working Group, Arnhem, Netherlands
- Department of Urology, KU Leuven, Leuven, Belgium
| | - Idir Ouizad
- Young Academic Urologists (YAU) Renal Cancer Working Group, Arnhem, Netherlands
- Department of Urology, Bichat-Claude Bernard Hospital, Paris, France
| | - Alexander Ingels
- Young Academic Urologists (YAU) Renal Cancer Working Group, Arnhem, Netherlands
- Department of Urology, Henri Mondor Hospital, Créteil, France
| | - Nicola Pavan
- Young Academic Urologists (YAU) Renal Cancer Working Group, Arnhem, Netherlands
- Department of Urology, Cattinara Hospital, University of Trieste, Trieste, Italy
| | - Onder Kara
- Young Academic Urologists (YAU) Renal Cancer Working Group, Arnhem, Netherlands
- Faculty of Medicine, Kocaeli University, İzmit, Turkey
| | - Selcuk Erdem
- Young Academic Urologists (YAU) Renal Cancer Working Group, Arnhem, Netherlands
- Department of Urology, Istanbul University, Istanbul, Turkey
| | - Riccardo Bertolo
- Young Academic Urologists (YAU) Renal Cancer Working Group, Arnhem, Netherlands
- Urology Unit, Department of Surgery, Tor Vergata University of Rome, Rome, Italy
| | - Umberto Capitanio
- Young Academic Urologists (YAU) Renal Cancer Working Group, Arnhem, Netherlands
- Department of Urology, San Raffaele Scientific Institute, Milan, Italy
- Division of Experimental Oncology/Unit of Urology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Maria Carmen Mir
- Young Academic Urologists (YAU) Renal Cancer Working Group, Arnhem, Netherlands.
- Department of Urology, Hospital Universitario La Ribera, Valencia, Spain.
| |
Collapse
|
31
|
Alcala K, Zahed H, Cortez Cardoso Penha R, Alcala N, Robbins HA, Smith-Byrne K, Martin RM, Muller DC, Brennan P, Johansson M. Kidney Function and Risk of Renal Cell Carcinoma. Cancer Epidemiol Biomarkers Prev 2023; 32:1644-1650. [PMID: 37668600 PMCID: PMC10618735 DOI: 10.1158/1055-9965.epi-23-0558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/13/2023] [Accepted: 08/31/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND We evaluated the temporal association between kidney function, assessed by estimated glomerular filtration rate (eGFR), and the risk of incident renal cell carcinoma (RCC). We also evaluated whether eGFR could improve RCC risk discrimination beyond established risk factors. METHODS We analyzed the UK Biobank cohort, including 463,178 participants of whom 1,447 were diagnosed with RCC during 5,696,963 person-years of follow-up. We evaluated the temporal association between eGFR and RCC risk using flexible parametric survival models, adjusted for C-reactive protein and RCC risk factors. eGFR was calculated from creatinine and cystatin C levels. RESULTS Lower eGFR, an indication of poor kidney function, was associated with higher RCC risk when measured up to 5 years prior to diagnosis. The RCC HR per SD decrease in eGFR when measured 1 year before diagnosis was 1.26 [95% confidence interval (95% CI), 1.16-1.37], and 1.11 (95% CI, 1.05-1.17) when measured 5 years before diagnosis. Adding eGFR to the RCC risk model provided a small improvement in risk discrimination 1 year before diagnosis with an AUC of 0.73 (95% CI, 0.67-0.84) compared with the published model (0.69; 95% CI, 0.63-0.79). CONCLUSIONS This study demonstrated that kidney function markers are associated with RCC risk, but the nature of these associations are consistent with reversed causality. Markers of kidney function provided limited improvements in RCC risk discrimination beyond established risk factors. IMPACT eGFR may be of potential use to identify individuals in the extremes of the risk distribution.
Collapse
Affiliation(s)
- Karine Alcala
- Genomic Epidemiology Branch, International Agency for Research on Cancer, Lyon, France
| | - Hana Zahed
- Genomic Epidemiology Branch, International Agency for Research on Cancer, Lyon, France
| | | | - Nicolas Alcala
- Genomic Epidemiology Branch, International Agency for Research on Cancer, Lyon, France
| | - Hilary A. Robbins
- Genomic Epidemiology Branch, International Agency for Research on Cancer, Lyon, France
| | - Karl Smith-Byrne
- Cancer Epidemiology Unit, Oxford Population Health, University of Oxford, Oxford, United Kingdom
| | - Richard M. Martin
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and the University of Bristol, Bristol, United Kingdom
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
| | | | - Paul Brennan
- Genomic Epidemiology Branch, International Agency for Research on Cancer, Lyon, France
| | - Mattias Johansson
- Genomic Epidemiology Branch, International Agency for Research on Cancer, Lyon, France
| |
Collapse
|
32
|
Sequeira JP, Barros-Silva D, Ferreira-Torre P, Salta S, Braga I, Carvalho J, Freitas R, Henrique R, Jerónimo C. OncoUroMiR: Circulating miRNAs for Detection and Discrimination of the Main Urological Cancers Using a ddPCR-Based Approach. Int J Mol Sci 2023; 24:13890. [PMID: 37762193 PMCID: PMC10531069 DOI: 10.3390/ijms241813890] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
The three most common genitourinary malignancies (prostate/kidney/bladder cancers) constitute a substantial proportion of all cancer cases, mainly in the elderly population. Early detection is key to maximizing the patients' survival, but the lack of highly accurate biomarkers that might be used through non-/minimally invasive methods has impaired progress in this domain. Herein, we sought to develop a minimally invasive test to detect and discriminate among those urological cancers based on miRNAs assessment through ddPCR. Plasma samples from 268 patients with renal cell (RCC; n = 119), bladder (BlCa; n = 73), and prostate (PCa; n = 76) carcinomas (UroCancer group), and 74 healthy donors were selected. Hsa-miR-126-3p, hsa-miR-141-3p, hsa-miR-153-5p, hsa-miR-155-5p, hsa-miR-182-5p, hsa-miR-205-5p, and hsa-miR-375-3p levels were assessed. UroCancer cases displayed significantly different circulating hsa-miR-182-5p/hsa-miR-375-3p levels compared to healthy donors. Importantly, the hsa-miR-155-5p/hsa-miR-375-3p panel detected RCC with a high specificity (80.54%) and accuracy (66.04%). Furthermore, the hsa-miR-126-3p/hsa-miR-375-3p panel identified BlCa with a 94.87% specificity and 76.45% NPV whereas higher hsa-miR-126-3p levels were found in PCa patients. We concluded that plasma-derived miRNAs can identify and discriminate among the main genitourinary cancers, with high analytical performance. Although validation in a larger cohort is mandatory, these findings demonstrate that circulating miRNA assessment by ddPCR might provide a new approach for early detection and risk stratification of the most common urological cancers.
Collapse
Affiliation(s)
- José Pedro Sequeira
- Cancer Biology & Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (J.P.S.); (D.B.-S.); (P.F.-T.); (S.S.); (I.B.); (J.C.); (R.F.); (R.H.)
- Doctoral Programme in Biomedical Sciences, ICBAS-School of Medicine & Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal
| | - Daniela Barros-Silva
- Cancer Biology & Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (J.P.S.); (D.B.-S.); (P.F.-T.); (S.S.); (I.B.); (J.C.); (R.F.); (R.H.)
| | - Patrícia Ferreira-Torre
- Cancer Biology & Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (J.P.S.); (D.B.-S.); (P.F.-T.); (S.S.); (I.B.); (J.C.); (R.F.); (R.H.)
| | - Sofia Salta
- Cancer Biology & Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (J.P.S.); (D.B.-S.); (P.F.-T.); (S.S.); (I.B.); (J.C.); (R.F.); (R.H.)
- Doctoral Programme in Molecular Pathology and Genetics, ICBAS-School of Medicine & Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal
| | - Isaac Braga
- Cancer Biology & Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (J.P.S.); (D.B.-S.); (P.F.-T.); (S.S.); (I.B.); (J.C.); (R.F.); (R.H.)
- Department of Urology & Urology Clinics, Portuguese Oncology Institute of Porto (IPOP), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
- Doctoral Programme in Medical Sciences, ICBAS-School of Medicine & Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal
| | - João Carvalho
- Cancer Biology & Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (J.P.S.); (D.B.-S.); (P.F.-T.); (S.S.); (I.B.); (J.C.); (R.F.); (R.H.)
- Department of Urology & Urology Clinics, Portuguese Oncology Institute of Porto (IPOP), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
- Doctoral Programme in Medical Sciences, ICBAS-School of Medicine & Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal
| | - Rui Freitas
- Cancer Biology & Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (J.P.S.); (D.B.-S.); (P.F.-T.); (S.S.); (I.B.); (J.C.); (R.F.); (R.H.)
- Department of Urology & Urology Clinics, Portuguese Oncology Institute of Porto (IPOP), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
- Doctoral Programme in Medical Sciences, ICBAS-School of Medicine & Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal
| | - Rui Henrique
- Cancer Biology & Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (J.P.S.); (D.B.-S.); (P.F.-T.); (S.S.); (I.B.); (J.C.); (R.F.); (R.H.)
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
- Department of Pathology and Molecular Immunology, ICBAS–School of Medicine and Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal
| | - Carmen Jerónimo
- Cancer Biology & Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; (J.P.S.); (D.B.-S.); (P.F.-T.); (S.S.); (I.B.); (J.C.); (R.F.); (R.H.)
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
- Department of Pathology and Molecular Immunology, ICBAS–School of Medicine and Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal
| |
Collapse
|
33
|
Uemura T, Kawashima A, Jingushi K, Motooka D, Saito T, Nesrine S, Oka T, Okuda Y, Yamamoto A, Yamamichi G, Tomiyama E, Ishizuya Y, Yamamoto Y, Kato T, Hatano K, Tsujikawa K, Wada H, Nonomura N. Bacteria-derived DNA in serum extracellular vesicles are biomarkers for renal cell carcinoma. Heliyon 2023; 9:e19800. [PMID: 37810127 PMCID: PMC10559165 DOI: 10.1016/j.heliyon.2023.e19800] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 08/25/2023] [Accepted: 09/01/2023] [Indexed: 10/10/2023] Open
Abstract
This is the first study to determine the clinical importance of circulating bacterial DNA in patients with renal cell carcinoma (RCC). We performed 16S rRNA metagenomic analysis of serum extracellular vesicles (EVs) from 88 patients with RCC and 10 healthy donors and identified three abundant bacterial DNA: Bacteroidia, TM7-1, and Sphingomonadales. Combining characteristic bacterial DNA information (three bacteria-derived DNA), a BTS index was created to diagnose patients with RCC. The BTS index showed high sensitivity not only in the discovery cohort, but also in the validation cohort, suggesting that it was useful as a screening test. Furthermore, in nivolumab treatment of RCC, patients with higher levels of Bacteroidia DNA in serum EVs had significantly poorer progression-free and overall survival than did those with lower levels. This study showed that circulating Bacteria-derived DNA could be used as a biomarker for RCC.
Collapse
Affiliation(s)
- Toshihiro Uemura
- Department of Urology, Osaka University, Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Atsunari Kawashima
- Department of Urology, Osaka University, Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kentaro Jingushi
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Daisuke Motooka
- Department of Infection Metagenomics, Genome Information Research Center, Osaka University Research Institute for Microbial Diseases, Suita, Osaka, 565-0871, Japan
| | - Takuro Saito
- Department of Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
- Department of Clinical Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Sassi Nesrine
- Department of Urology, Osaka University, Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Toshiki Oka
- Department of Urology, Osaka University, Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yohei Okuda
- Department of Urology, Osaka University, Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Akinaru Yamamoto
- Department of Urology, Osaka University, Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Gaku Yamamichi
- Department of Urology, Osaka University, Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Eisuke Tomiyama
- Department of Urology, Osaka University, Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yu Ishizuya
- Department of Urology, Osaka University, Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshiyuki Yamamoto
- Department of Urology, Osaka University, Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Taigo Kato
- Department of Urology, Osaka University, Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Koji Hatano
- Department of Urology, Osaka University, Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kazutake Tsujikawa
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hisashi Wada
- Department of Clinical Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Norio Nonomura
- Department of Urology, Osaka University, Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
34
|
Xue Y, Chen T, Hou N, Wu X, Kong W, Huang J, Zhang J, Chen Y, Zheng J, Zhai W, Xue W. Serum extracellular vesicles derived hsa-miR-320d as an indicator for progression of clear cell renal cell carcinoma. Discov Oncol 2023; 14:114. [PMID: 37380801 DOI: 10.1007/s12672-023-00730-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/15/2023] [Indexed: 06/30/2023] Open
Abstract
BACKGROUND Renal cell carcinoma (RCC) is a prevalent malignancy with a rising incidence in developing countries. Clear cell renal cell carcinoma (ccRCC) constitutes 70% of RCC cases and is prone to metastasis and recurrence, yet lacks a liquid biomarker for surveillance. Extracellular vesicles (EVs) have shown promise as biomarkers in various malignancies. In this study, we investigated the potential of serum EV-derived miRNAs as a biomarker for ccRCC metastasis and recurrence. MATERIALS AND METHODS Patients diagnosed with ccRCC between 2017 and 2020 were recruited in this study. In the discovery phase, high throughput small RNA sequencing was used to analyze RNA extracted from serum EVs derived from localized ccRCC (LccRCC) and advanced ccRCC (AccRCC). In the validation phase, qPCR was employed for quantitative detection of candidate biomarkers. Migration and invasion assays were performed on ccRCC cell line OSRC2. RESULTS Serum EVs derived hsa-miR-320d was significantly up-regulated in patients with AccRCC than in patients with LccRCC (p < 0.01). In addition, Serum EVs derived hsa-miR-320d was also significantly up-regulated in patients who experienced recurrence or metastasis (p < 0.01). Besides, hsa-miR-320d enhances the pro-metastatic phenotype of ccRCC cells in vitro. CONCLUSIONS Serum EVs derived hsa-miR-320d as a liquid biomarker exhibits significant potential for identifying the recurrence or metastasis of ccRCC, as well as hsa-miR-320d promotes ccRCC cells migration and invasion.
Collapse
Affiliation(s)
- Yizheng Xue
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160# Pu Jian Ave, Shanghai, 200127, China
| | - Tianyi Chen
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160# Pu Jian Ave, Shanghai, 200127, China
| | - Naiqiao Hou
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160# Pu Jian Ave, Shanghai, 200127, China
| | - Xiaorong Wu
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160# Pu Jian Ave, Shanghai, 200127, China
| | - Wen Kong
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160# Pu Jian Ave, Shanghai, 200127, China
| | - Jiwei Huang
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160# Pu Jian Ave, Shanghai, 200127, China
| | - Jin Zhang
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160# Pu Jian Ave, Shanghai, 200127, China
| | - Yonghui Chen
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160# Pu Jian Ave, Shanghai, 200127, China
| | - Junhua Zheng
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160# Pu Jian Ave, Shanghai, 200127, China
| | - Wei Zhai
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160# Pu Jian Ave, Shanghai, 200127, China.
- State Key Laboratory of Oncogenes and Related Genes, Department of Urology, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Wei Xue
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160# Pu Jian Ave, Shanghai, 200127, China.
| |
Collapse
|
35
|
Harrison H, Wood A, Pennells L, Rossi SH, Callister M, Cartledge J, Stewart GD, Usher-Smith JA. Estimating the Effectiveness of Kidney Cancer Screening Within Lung Cancer Screening Programmes: A Validation in UK Biobank. Eur Urol Oncol 2023; 6:351-353. [PMID: 37003861 DOI: 10.1016/j.euo.2023.02.012] [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: 10/06/2022] [Revised: 02/07/2023] [Accepted: 02/23/2023] [Indexed: 04/03/2023]
Abstract
In the absence of population-based screening, addition of screening for kidney cancer to lung cancer screening could provide an efficient and low-resource means to improve early detection. In this study, we used the UK Biobank cohort (n = 442 865) to determine the performance of the Yorkshire Lung Cancer Screening Trial (YLST) eligibility criteria for selecting individuals for kidney cancer screening. We measured the performance of two models widely used to determine eligibility for lung cancer screening (PLCO[m2012] and the Liverpool-Lung-Project-v2) and the performance of the combined YLST criteria. We found that the lung cancer models have discrimination (area under the receiver operating curve) between 0.60 and 0.68 for kidney cancer. In the UK, one in four cases (25%) of kidney cancer cases is expected to occur in those eligible for lung cancer screening, and one case of kidney cancer detected for every 200 people invited to lung cancer screening. These results suggest that adding kidney cancer screening to lung cancer screening would be an effective strategy to improve early detection rates of kidney cancer. However, most kidney cancers would not be picked up by this approach. This analysis does not address other important considerations about kidney cancer screening, such as overdiagnosis. PATIENT SUMMARY: It has been proposed that adding-on kidney cancer screening to lung cancer screening (both carried out by a computed tomography scan of the chest/abdomen) would be an easy and low-cost way of detecting cases of kidney cancer earlier, when these can be treated more easily. Lung cancer screening is usually targeted at people who are at a high risk (eg, older smokers); therefore, here we look at whether the same group of people are also at a high risk of kidney cancer. Our analysis shows that one in four people later diagnosed with kidney cancer are also at a high risk of lung cancer; hence, a combined screening programme could detect up to a quarter of kidney cancers.
Collapse
Affiliation(s)
- Hannah Harrison
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
| | - Angela Wood
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Lisa Pennells
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Sabrina H Rossi
- Department of Surgery, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Matthew Callister
- Department of Respiratory Medicine, Leeds Teaching Hospitals Trust, Leeds, UK
| | - Jon Cartledge
- St James University Hospital, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Grant D Stewart
- Department of Surgery, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Juliet A Usher-Smith
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| |
Collapse
|
36
|
Qi J, An P, Jin D, Ji Y, Wan S, Zhang X, Luo Y, Luo J, Zhang C. Food groups and urologic cancers risk: a systematic review and meta-analysis of prospective studies. Front Nutr 2023; 10:1154996. [PMID: 37266130 PMCID: PMC10231388 DOI: 10.3389/fnut.2023.1154996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/18/2023] [Indexed: 06/03/2023] Open
Abstract
BACKGROUND To assess the association between 12 food groups intake and the risk of urologic cancers. METHODS We scanned PubMed and Web of Science databases up to April 1st, 2023, and 73 publications met the inclusion criteria in the meta-analysis. We used a random effects model to estimate the summary risk ratios (RRs) and 95% confidence intervals (95% CI). RESULTS In the linear dose-response meta-analysis, an inverse association was found between each additional daily 100 g of fruits [RR: 0.89, 95%CI = (0.83, 0.97)], 100 g of vegetables [RR: 0.92, 95%CI = (0.85, 0.99)], 12 g of alcohol [RR: 0.91, 95%CI = (0.88, 0.94)] and 1 cup of coffee [RR: 0.95, 95%CI = (0.83, 0.97)] intake and the risk of renal cell carcinoma. Conversely, each additional daily 100 g of red meat intake was positively associated with renal cell carcinoma [RR: 1.41, 95%CI = (1.03, 2.10)]. Inverse associations were observed between each additional daily 50 g of egg [RR: 0.73, 95%CI = (0.62, 0.87)] and each additional daily 1 cup of tea consumption and bladder cancer risk [RR: 0.97, 95%CI = (0.94, 0.99)]. There were no significant associations for nonlinear dose-response relationships between 12 food groups and urological cancers. CONCLUSION Our meta-analysis strengthens the evidence that appropriate intake of specific food groups, such as fruits, vegetables, alcohol, tea, and coffee, is associated with the risk of renal cell carcinoma or bladder cancer. More studies are required to fill the knowledge gap on the links between various food groups and urologic cancers because the evidence was less credible in this meta-analysis. SYSTEMATIC REVIEW REGISTRATION This study was registered on PROSPERO (CRD42022340336).
Collapse
Affiliation(s)
- Jingyi Qi
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Peng An
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Dekui Jin
- Department of General Practice, The Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yuting Ji
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Sitong Wan
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Xu Zhang
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Yongting Luo
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Junjie Luo
- Department of Nutrition and Health, Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Chengying Zhang
- Department of General Practice, The Third Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
37
|
Bernstein E, Lev-Ari S, Shapira S, Leshno A, Sommer U, Al-Shamsi H, Shaked M, Segal O, Galazan L, Hay-Levy M, Sror M, Harlap-Gat A, Peer M, Moshkowitz M, Wolf I, Liberman E, Shenberg G, Gur E, Elran H, Melinger G, Mashiah J, Isakov O, Zrifin E, Gluck N, Dekel R, Kleinman S, Aviram G, Blachar A, Kessler A, Golan O, Geva R, Yossepowitch O, Neugut AI, Arber N. Data From a One-Stop-Shop Comprehensive Cancer Screening Center. J Clin Oncol 2023; 41:2503-2510. [PMID: 36669135 DOI: 10.1200/jco.22.00938] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
PURPOSE Cancer is the second leading cause of death globally. However, by implementing evidence-based prevention strategies, 30%-50% of cancers can be detected early with improved outcomes. At the integrated cancer prevention center (ICPC), we aimed to increase early detection by screening for multiple cancers during one visit. METHODS Self-referred asymptomatic individuals, age 20-80 years, were included prospectively. Clinical, laboratory, and epidemiological data were obtained by multiple specialists, and further testing was obtained based on symptoms, family history, individual risk factors, and abnormalities identified during the visit. Follow-up recommendations and diagnoses were given as appropriate. RESULTS Between January 1, 2006, and December 31, 2019, 8,618 men and 8,486 women, average age 47.11 ± 11.71 years, were screened. Of 259 cancers detected through the ICPC, 49 (19.8%) were stage 0, 113 (45.6%) stage I, 30 (12.1%) stage II, 25 (10.1%) stage III, and 31(12.5%) stage IV. Seventeen cancers were missed, six of which were within the scope of the ICPC. Compared with the Israeli registry, at the ICPC, less cancers were diagnosed at a metastatic stage for breast (none v 3.7%), lung (6.7% v 11.4%), colon (20.0% v 46.2%), prostate (5.6% v 10.5%), and cervical/uterine (none v 8.5%) cancers. When compared with the average stage of detection in the United States, detection was earlier for breast, lung, prostate, and female reproductive cancers. Patient satisfaction rate was 8.35 ± 1.85 (scale 1-10). CONCLUSION We present a proof of concept study for a one-stop-shop approach to cancer screening in a multidisciplinary outpatient clinic. We successfully detected cancers at an early stage, which has the potential to reduce morbidity and mortality as well as offer substantial cost savings.
Collapse
Affiliation(s)
- Ezra Bernstein
- Health Promotion and Integrated Cancer Prevention Center, Tel Aviv Medical Center, Tel Aviv, Israel.,NYU Langone Medical Center, New York, NY
| | - Shahar Lev-Ari
- Health Promotion and Integrated Cancer Prevention Center, Tel Aviv Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shiran Shapira
- Health Promotion and Integrated Cancer Prevention Center, Tel Aviv Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ari Leshno
- Health Promotion and Integrated Cancer Prevention Center, Tel Aviv Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Udi Sommer
- Tel Aviv University Faculty of Social Sciences, School of Political Science, Government and International Relations, Tel Aviv, Israel
| | - Humaid Al-Shamsi
- Burjeel Cancer Institute, University of Sharjah, Sharjah, United Arab Emirates
| | - Meital Shaked
- Health Promotion and Integrated Cancer Prevention Center, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Ori Segal
- Health Promotion and Integrated Cancer Prevention Center, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Lior Galazan
- Health Promotion and Integrated Cancer Prevention Center, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Mori Hay-Levy
- Health Promotion and Integrated Cancer Prevention Center, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Miri Sror
- Health Promotion and Integrated Cancer Prevention Center, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Amira Harlap-Gat
- Health Promotion and Integrated Cancer Prevention Center, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Michael Peer
- Health Promotion and Integrated Cancer Prevention Center, Tel Aviv Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Menachem Moshkowitz
- Health Promotion and Integrated Cancer Prevention Center, Tel Aviv Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ido Wolf
- Health Promotion and Integrated Cancer Prevention Center, Tel Aviv Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Oncology, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Eliezer Liberman
- Health Promotion and Integrated Cancer Prevention Center, Tel Aviv Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Gil Shenberg
- Health Promotion and Integrated Cancer Prevention Center, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Eyal Gur
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Plastic and Reconstructive Surgery, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Hanoch Elran
- Health Promotion and Integrated Cancer Prevention Center, Tel Aviv Medical Center, Tel Aviv, Israel.,Department of Neurosurgery, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Gustavo Melinger
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,The Gynecological and Obstetric Ultrasound Unit, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Jacob Mashiah
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Division of Dermatology and Venerology, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Ofer Isakov
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Institute of Gastrointestinal and Liver Diseases, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Elad Zrifin
- Health Promotion and Integrated Cancer Prevention Center, Tel Aviv Medical Center, Tel Aviv, Israel.,Oral and Maxillofacial Surgery Unit, Otolaryngology-Head and Neck Surgery, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Nathan Gluck
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Institute of Gastrointestinal and Liver Diseases, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Roy Dekel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Institute of Gastrointestinal and Liver Diseases, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Shlomi Kleinman
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Oral and Maxillofacial Surgery Unit, Otolaryngology-Head and Neck Surgery, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Galit Aviram
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Radiology, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Arye Blachar
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Radiology, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Ada Kessler
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Radiology, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Orit Golan
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Radiology, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Ravit Geva
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Oncology, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Ofer Yossepowitch
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Urology, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Alfred I Neugut
- Columbia University Medical Center, Herbert Irving Comprehensive Cancer Center, New York, NY
| | - Nadir Arber
- Health Promotion and Integrated Cancer Prevention Center, Tel Aviv Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Institute of Gastrointestinal and Liver Diseases, Tel Aviv Medical Center, Tel Aviv, Israel
| |
Collapse
|
38
|
Grammatikaki S, Katifelis H, Farooqi AA, Stravodimos K, Karamouzis MV, Souliotis K, Varvaras D, Gazouli M. An Overview of Epigenetics in Clear Cell Renal Cell Carcinoma. In Vivo 2023; 37:1-10. [PMID: 36593023 PMCID: PMC9843790 DOI: 10.21873/invivo.13049] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/11/2022] [Accepted: 12/12/2022] [Indexed: 01/04/2023]
Abstract
Renal cell carcinoma (RCC) represents a heterogenous group of cancers with complex genetic background and histological varieties, which require various clinical therapies. Clear cell RCC represents the most common form of RCC that accounts for 3 out of 4 RCC cases. Screening methods for RCC lack sensitivity and specificity, and thus biomarkers that will allow early diagnosis are crucial. The impact of epigenetics in the development and progression of cancer, including RCC, is significant. Noncoding RNAs, histone modifications and DNA methylation represent fundamental epigenetic mechanisms and have been proved to be promising biomarkers. MicroRNAs have advantageous properties that facilitate early diagnosis of RCC, while their expression profiles have been assessed in renal cancer samples (tissue, blood, and urine). Current literature reports the up-regulation of mir122, mir1271 and mir15b in RCC specimens, which induces cell proliferation via FOXP-1 and PTEN genes. However, it should be noted that conflicting results are found in urine and serum patient samples. Moreover, promoters of at least 200 genes are methylated in renal cancers leading to epigenetic dysregulation. In this review, we analyze the vast plethora of studies that have evaluated the role of epigenetic mechanisms in RCC patients and their clinical importance.
Collapse
Affiliation(s)
- Stamatiki Grammatikaki
- Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Hector Katifelis
- Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Konstantinos Stravodimos
- 1 Department of Urology, National & Kapodistrian University of Athens, Laiko Hospital, Athens, Greece
| | - Michalis V Karamouzis
- Molecular Oncology Unit, Department of Biological Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Kyriakos Souliotis
- School of Social and Education Policy, University of Peloponnese, Corinth, Greece
- Health Policy Institute, Athens, Greece
| | - Dimitrios Varvaras
- Health Policy Institute, Athens, Greece
- Tiberia Hospital-GMV Care & Research, Rome, Italy
| | - Maria Gazouli
- Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece;
| |
Collapse
|
39
|
Su P, Zhang M, Kang X. Targeting c-Met in the treatment of urologic neoplasms: Current status and challenges. Front Oncol 2023; 13:1071030. [PMID: 36959792 PMCID: PMC10028134 DOI: 10.3389/fonc.2023.1071030] [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: 10/15/2022] [Accepted: 02/23/2023] [Indexed: 03/09/2023] Open
Abstract
At present, studies have found that c-Met is mainly involved in epithelial-mesenchymal transition (EMT) of tumor tissues in urologic neoplasms. Hepatocyte growth factor (HGF) combined with c-Met promotes the mitosis of tumor cells, and then induces motility, angiogenesis, migration, invasion and drug resistance. Therefore, c-Met targeting therapy may have great potential in urologic neoplasms. Many strategies targeting c-Met have been widely used in the study of urologic neoplasms. Although the use of targeting c-Met therapy has a strong biological basis for the treatment of urologic neoplasms, the results of current clinical trials have not yielded significant results. To promote the application of c-Met targeting drugs in the clinical treatment of urologic neoplasms, it is very important to study the detailed mechanism of c-Met in urologic neoplasms and innovate c-Met targeted drugs. This paper firstly discussed the value of c-Met targeted therapy in urologic neoplasms, then summarized the related research progress, and finally explored the potential targets related to the HGF/c-Met signaling pathway. It may provide a new concept for the treatment of middle and late urologic neoplasms.
Collapse
|
40
|
Wang H, Wang Q, Wu Y, Lou J, Zhu S, Xu Y. Autophagy-related gene LAPTM4B promotes the progression of renal clear cell carcinoma and is associated with immunity. Front Pharmacol 2023; 14:1118217. [PMID: 36937841 PMCID: PMC10017457 DOI: 10.3389/fphar.2023.1118217] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 02/22/2023] [Indexed: 03/06/2023] Open
Abstract
Renal cell carcinoma (RCC) is a common urologic disease. Currently, surgery is the primary treatment for renal cancer; immunotherapy is not as effective a treatment strategy as expected. Hence, understanding the mechanism in the tumor immune microenvironment (TME) and exploring novel immunotherapeutic targets are considered important. Recent studies have demonstrated that autophagy could affect the immune environment of renal cell carcinoma and induce proliferation and apoptosis of cancer cells. By comparing lysosomal genes and regulating autophagy genes, we identified the LAPTM4B gene to be related to RCC autophagy. By analyzing the TCGA-KIRC cohort using bioinformatics, we found M2 macrophages associated with tumor metastasis to be significantly increased in the immune microenvironment of patients with high expression of LAPTM4B. GO/KEGG/GSEA/GSVA results showed significant differences in tumor autophagy- and metastasis-related pathways. Single-cell sequencing was used to compare the expression of LAPTM4B in different cell types and obtain the differences in lysosomal and autophagy pathway activities in different ccRCC cells. Subsequently, we confirmed the differential expression of LAPTM4B in renal cell carcinoma of different Fuhrman grades using western blotting. Downregulation of LAPTM4B expression significantly reduced the proliferation of renal cell carcinoma cells and promoted cell apoptosis through cell experiments. Overall, our study demonstrated that the autophagy-related gene LAPTM4B plays a critical role in the TME of RCC, and suggested that LAPTM4B is a potential therapeutic target for RCC immunotherapy.
Collapse
Affiliation(s)
- He Wang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qibo Wang
- Department of Urology, Cancer Hospital of University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Yaoyao Wu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jianmin Lou
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shaoxing Zhu
- Department of Urology, Fujian Medical University Union Hospital, Fuzhou, China
- *Correspondence: Shaoxing Zhu, ; Yipeng Xu,
| | - Yipeng Xu
- Department of Urology, Cancer Hospital of University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
- *Correspondence: Shaoxing Zhu, ; Yipeng Xu,
| |
Collapse
|
41
|
Harrison H, Li N, Saunders CL, Rossi SH, Dennis J, Griffin SJ, Stewart GD, Usher‐Smith JA. The current state of genetic risk models for the development of kidney cancer: a review and validation. BJU Int 2022; 130:550-561. [PMID: 35460182 PMCID: PMC9790357 DOI: 10.1111/bju.15752] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE To review the current state of genetic risk models for predicting the development of kidney cancer, by identifying and comparing the performance of published models. METHODS Risk models were identified from a recent systematic review and the Cancer-PRS web directory. A narrative synthesis of the models, previous validation studies and related genome-wide association studies (GWAS) was carried out. The discrimination and calibration of the identified models was then assessed and compared in the UK Biobank (UKB) cohort (cases, 452; controls, 487 925). RESULTS A total of 39 genetic models predicting the development of kidney cancer were identified and 31 were validated in the UKB. Several of the genetic-only models (seven of 25) and most of the mixed genetic-phenotypic models (five of six) had some discriminatory ability (area under the receiver operating characteristic curve >0.5) in this cohort. In general, models containing a larger number of genetic variants identified in GWAS performed better than models containing a small number of variants associated with known causal pathways. However, the performance of the included models was consistently poorer than genetic risk models for other cancers. CONCLUSIONS Although there is potential for genetic models to identify those at highest risk of developing kidney cancer, their performance is poorer than the best genetic risk models for other cancers. This may be due to the comparatively small number of genetic variants associated with kidney cancer identified in GWAS to date. The development of improved genetic risk models for kidney cancer is dependent on the identification of more variants associated with this disease. Whether these will have utility within future kidney cancer screening pathways is yet to determined.
Collapse
Affiliation(s)
- Hannah Harrison
- Department of Public Health and Primary CareUniversity of CambridgeCambridgeUK
| | - Nicole Li
- Department of Public Health and Primary CareUniversity of CambridgeCambridgeUK
- Deanary of Biomedical SciencesUniversity of EdinburghEdinburghUK
| | | | - Sabrina H. Rossi
- Department of SurgeryUniversity of CambridgeAddenbrooke’s HospitalCambridgeUK
| | - Joe Dennis
- Department of Public Health and Primary CareUniversity of CambridgeCambridgeUK
| | - Simon J. Griffin
- Department of Public Health and Primary CareUniversity of CambridgeCambridgeUK
| | - Grant D. Stewart
- Department of SurgeryUniversity of CambridgeAddenbrooke’s HospitalCambridgeUK
| | | |
Collapse
|
42
|
Fu L, Bao J, Li J, Li Q, Lin H, Zhou Y, Li J, Yan Y, Langston ME, Sun T, Guo S, Zhou X, Chen Y, Liu Y, Zhao Y, Lu J, Huang Y, Chen W, Chung BI, Luo J. Crosstalk of necroptosis and pyroptosis defines tumor microenvironment characterization and predicts prognosis in clear cell renal carcinoma. Front Immunol 2022; 13:1021935. [PMID: 36248876 PMCID: PMC9561249 DOI: 10.3389/fimmu.2022.1021935] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 09/15/2022] [Indexed: 11/13/2022] Open
Abstract
Pyroptosis and necroptosis are two recently identified forms of immunogenic cell death in the tumor microenvironment (TME), indicating a crucial involvement in tumor metastasis. However, the characteristics of necroptosis and pyroptosis that define tumor microenvironment and prognosis in ccRCC patients remain unknown. We systematically investigated the transcriptional variation and expression patterns of Necroptosis and Pyroptosis related genes (NPRGs). After screening the necroptosis-pyroptosis clusters, the potential functional annotation for clusters was explored by GSVA enrichment analysis. The Necroptosis-Pyroptosis Genes (NPG) scores were used for the prognosis model construction and validation. Then, the correlations of NPG score with clinical features, cancer stem cell (CSC) index, tumor mutation burden (TMB), TME, and Immune Checkpoint Genes (ICGs) were also individually explored to evaluate the prognosis predictive values in ccRCC. Microarray screenings identified 27 upregulated and 1 downregulated NPRGs. Ten overall survival associated NPRGs were filtered to construct the NPG prognostic model indicating a better prognostic signature for ccRCC patients with lower NPG scores (P< 0.001), which was verified using the external cohort. Univariate and multivariate analyses along with Kaplan-Meier survival analysis demonstrated that NPG score prognostic model could be applied as an independent prognostic factor, and AUC values of nomogram from 1- to 5- year overall survival with good agreement in calibration plots suggested that the proposed prognostic signature possessed good predictive capabilities in ccRCC. A high-/sNPG score is proven to be connected with tumor growth and immune-related biological processes, according to enriched GO, KEGG, and GSEA analyses. Comparing patients with a high-NPG score to those with a low-NPG score revealed significant differences in clinical characteristics, growth and recurrence of malignancies (CSC index), TME cell infiltration, and immunotherapeutic response (P< 0.005), potentially making the NPG score multifunctional in the clinical therapeutic setting. Furthermore, AIM2, CASP4, GSDMB, NOD2, and RBCK1 were also found to be highly expressed in ccRCC cell lines and tumor tissues, and GASP4 and GSDMB promote ccRCC cells’ proliferation, migration, and invasion. This study firstly suggests that targeting the NPG score feature for TME characterization may lend novel insights into its clinical applications in the prognostic prediction of ccRCC.
Collapse
Affiliation(s)
- Liangmin Fu
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jiahao Bao
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Jinhui Li
- Department of Urology, Stanford University Medical Center, Stanford, CA, United States
| | - Qiuyang Li
- Department of Obstetrics & Gynecology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hansen Lin
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yayun Zhou
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jiangbo Li
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Yixuan Yan
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Marvin E. Langston
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, United States
| | - Tianhao Sun
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, Guangdong Engineering Technology Research Center for Orthopaedic Trauma Repair, Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Songliang Guo
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Xinwei Zhou
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yuhang Chen
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yujun Liu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yiqi Zhao
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jun Lu
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yong Huang
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Department of Emergency, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wei Chen
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Benjamin I. Chung
- Department of Urology, Stanford University Medical Center, Stanford, CA, United States
- *Correspondence: Benjamin I. Chung, ; Junhang Luo,
| | - Junhang Luo
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- *Correspondence: Benjamin I. Chung, ; Junhang Luo,
| |
Collapse
|
43
|
Usher-Smith JA, Godoy A, Burge SW, Burbidge S, Cartledge J, Crosbie PAJ, Eckert C, Farquhar F, Hammond D, Hancock N, Iball GR, Kimuli M, Masson G, Neal RD, Rogerson S, Rossi SH, Sala E, Smith A, Sharp SJ, Simmonds I, Wallace T, Ward M, Callister MEJ, Stewart GD. The Yorkshire Kidney Screening Trial (YKST): protocol for a feasibility study of adding non-contrast abdominal CT scanning to screen for kidney cancer and other abdominal pathology within a trial of community-based CT screening for lung cancer. BMJ Open 2022; 12:e063018. [PMID: 36127097 PMCID: PMC9490622 DOI: 10.1136/bmjopen-2022-063018] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
INTRODUCTION Kidney cancer (renal cell cancer (RCC)) is the seventh most common cancer in the UK. As RCC is largely curable if detected at an early stage and most patients have no symptoms, there is international interest in evaluating a screening programme for RCC. The Yorkshire Kidney Screening Trial (YKST) will assess the feasibility of adding non-contrast abdominal CT scanning to screen for RCC and other abdominal pathology within the Yorkshire Lung Screening Trial (YLST), a randomised trial of community-based CT screening for lung cancer. METHODS AND ANALYSIS In YLST, ever-smokers aged 55-80 years registered with a general practice in Leeds have been randomised to a Lung Health Check assessment, including a thoracic low-dose CT (LDCT) for those at high risk of lung cancer, or routine care. YLST participants randomised to the Lung Health Check arm who attend for the second round of screening at 2 years without a history of RCC or abdominal CT scan within the previous 6 months will be invited to take part in YKST. We anticipate inviting 4700 participants. Those who consent will have an abdominal CT immediately following their YLST thoracic LDCT. A subset of participants and the healthcare workers involved will be invited to take part in a qualitative interview. Primary objectives are to quantify the uptake of the abdominal CT, assess the acceptability of the combined screening approach and pilot the majority of procedures for a subsequent randomised controlled trial of RCC screening within lung cancer screening. ETHICS AND DISSEMINATION YKST was approved by the North West-Preston Research Ethics Committee (21/NW/0021), and the Health Research Authority on 3 February 2021. Trial results will be disseminated at clinical meetings, in peer-reviewed journals and to policy-makers. Findings will be made available to participants via the study website (www.YKST.org). TRIAL REGISTRATION NUMBERS NCT05005195 and ISRCTN18055040.
Collapse
Affiliation(s)
- Juliet A Usher-Smith
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Angela Godoy
- Department of Surgery, University of Cambridge, Cambridge, UK
| | - Sarah W Burge
- Department of Oncology, University of Cambridge, Cambridge, UK
| | - Simon Burbidge
- Department of Radiology, Leeds Teaching Hospitals NHS Trust, Leeds, UK, Leeds, UK
| | - Jon Cartledge
- Department of Urology, Leeds Teaching Hospitals NHS Trust, Leeds, UK, Leeds, UK
| | - Philip A J Crosbie
- Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Claire Eckert
- Leeds Institiute of Health Sciences, University of Leeds, Leeds, UK
| | - Fiona Farquhar
- Research and Innovation, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - David Hammond
- Research and Innovation, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Neil Hancock
- Leeds Diagnosis & Screening Unit, Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | - Gareth R Iball
- Department of Medical Physics & Engineering, Leeds teaching hospitals NHS Trust, Leeds, UK
| | - Michael Kimuli
- Department of Urology, Leeds Teaching Hospitals NHS Trust, Leeds, UK, Leeds, UK
| | - Golnessa Masson
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Pitcairn Practice, Balmullo Surgery, Fife, UK
| | - Richard D Neal
- College of Medicine and Health, University of Exeter, Exeter, UK
| | - Suzanne Rogerson
- Research and Innovation, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Sabrina H Rossi
- Department of Surgery, University of Cambridge, Cambridge, UK
| | - Evis Sala
- Department of Radiology, University of Cambridge, Cambridge, UK
- Department of Radiology, Catholic University Sacro Cuore and Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | - Andrew Smith
- Upper Gastro-intestinal and Pancreas Unit, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Stephen J Sharp
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Irene Simmonds
- Leeds Institiute of Health Sciences, University of Leeds, Leeds, UK
| | - Tom Wallace
- Leeds Vascular Institute, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Matthew Ward
- Leeds Institiute of Health Sciences, University of Leeds, Leeds, UK
| | | | - Grant D Stewart
- Department of Surgery, University of Cambridge, Cambridge, UK
| |
Collapse
|
44
|
Zhou X, Lin J, Wang F, Chen X, Zhang Y, Hu Z, Jin X. Circular RNA-regulated autophagy is involved in cancer progression. Front Cell Dev Biol 2022; 10:961983. [PMID: 36187468 PMCID: PMC9515439 DOI: 10.3389/fcell.2022.961983] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 08/03/2022] [Indexed: 12/05/2022] Open
Abstract
Circular RNAs (circRNAs) are a sort of long, non-coding RNA molecules with a covalently closed continuous ring structure without 5'-3' polarity and poly-A tail. The modulative role of circRNAs in malignant diseases has been elucidated by many studies in recent years via bioinformatics and high-throughput sequencing technologies. Generally, circRNA affects the proliferative, invasive, and migrative capacity of malignant cells via various mechanisms, exhibiting great potential as novel biomarkers in the diagnoses or treatments of malignancies. Meanwhile, autophagy preserves cellular homeostasis, serving as a vital molecular process in tumor progression. Mounting studies have demonstrated that autophagy can not only contribute to cancer cell survival but can also induce autophagic cell death in specific conditions. A growing number of research studies have indicated that there existed abundant associations between circRNAs and autophagy. Herein, we systemically reviewed and discussed recent studies on this topic in different malignancies and concluded that the circRNA–autophagy axis played crucial roles in the proliferation, metastasis, invasion, and drug or radiation resistance of different tumor cells.
Collapse
|
45
|
Stewart GD, Klatte T, Cosmai L, Bex A, Lamb BW, Moch H, Sala E, Siva S, Porta C, Gallieni M. The multispeciality approach to the management of localised kidney cancer. Lancet 2022; 400:523-534. [PMID: 35868329 DOI: 10.1016/s0140-6736(22)01059-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/27/2022] [Accepted: 05/31/2022] [Indexed: 12/12/2022]
Abstract
Historically, kidney cancer was approached in a siloed single-speciality way, with urological surgeons managing the localised stages of the disease and medical oncologists caring for patients if metastases developed. However, improvements in the management of localised kidney cancer have occurred rapidly over the past two decades with greater understanding of the disease biology, diagnostic options, and innovations in curative treatments. These developments are favourable for patients but provide a substantially more complex landscape for patients and clinicians to navigate, with associated challenging decisions about who to treat, how, and when. As such, the skill sets needed to manage the various aspects of the disease and guide patients appropriately outstrips the capabilities of one particular specialist, and the evolution of a multispeciality approach to the management of kidney cancer is now essential. In this Review, we summarise the current best multispeciality practice for the management of localised kidney cancer and the areas in need of further research and development.
Collapse
Affiliation(s)
- Grant D Stewart
- Department of Surgery, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; CRUK Cambridge Centre, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; Department of Urology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| | - Tobias Klatte
- Department of Surgery, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; Department of Urology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Laura Cosmai
- Division of Nephrology and Dialysis, ASST Fatebenefratelli Sacco, Fatebenefratelli Hospital, Milan, Italy
| | - Axel Bex
- Specialist Centre for Kidney Cancer, Royal Free Hospital, London, UK; Division of Surgery and Interventional Science, University College London, London, UK; The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Benjamin W Lamb
- Department of Urology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK; School of Allied Health, Anglia Ruskin University, Cambridge, UK
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Evis Sala
- CRUK Cambridge Centre, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; Department of Radiology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Shankar Siva
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Camillo Porta
- Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Bari, Italy; Division of Medical Oncology, AOU Consorziale Policlinico di Bari, Bari, Italy
| | - Maurizio Gallieni
- Division of Nephrology and Dialysis, ASST Fatebenefratelli Sacco, Fatebenefratelli Hospital, Milan, Italy; Department of Clinical and Biomedical Sciences, Università di Milano, Milan, Italy
| |
Collapse
|
46
|
Martini A, Fallara G, Pellegrino AA, Nocera L, Larcher A, Raggi D, Campi R, Ploussard G, Malavaud B, Montorsi F, Pal SK, Spiess PE, Choueiri TK, Necchi A, Capitanio U. Multidisciplinary team referral at diagnosis for patients with non-metastatic renal cell carcinoma. Urol Oncol 2022; 40:384.e9-384.e14. [DOI: 10.1016/j.urolonc.2022.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 04/28/2022] [Accepted: 05/03/2022] [Indexed: 10/18/2022]
|
47
|
Ueki H, Hara T, Okamura Y, Bando Y, Terakawa T, Furukawa J, Harada K, Nakano Y, Fujisawa M. Association between sarcopenia based on psoas muscle index and the response to nivolumab in metastatic renal cell carcinoma: A retrospective study. Investig Clin Urol 2022; 63:415-424. [PMID: 35796138 PMCID: PMC9262481 DOI: 10.4111/icu.20220028] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/04/2022] [Accepted: 05/18/2022] [Indexed: 12/25/2022] Open
Abstract
Purpose Two methods are used to identify sarcopenia by calculating skeletal muscle area on computed tomography: the skeletal muscle index (SMI) and the psoas muscle index (PMI). Programmed death (PD)-1 inhibitors are helpful in treating metastatic renal cell carcinoma (mRCC). However, there remains insufficient information regarding a clear and easy-to-use biomarker for predicting the response to PD-1 inhibitors in patients with mRCC. Therefore, we investigated the influence of sarcopenia on clinical outcomes in patients with mRCC undergoing treatment with nivolumab. Materials and Methods This study evaluated 96 patients with RCC who received nivolumab. The SMI and PMI were calculated for each patient and normalized for stature by use of the following formulas: SMI (cm2/m2)=([skeletal muscle cross-sectional area at the level of L3]/[height]2) and PMI (cm2/m2) = ([left-right sum of the psoas muscle areas at the level of L3]/[height]2). The relationship of the clinical variables with progression-free survival and overall survival (OS) was examined using a Cox proportional hazards model. Results According to the SMI-based definition of sarcopenia, 74.0% of patients had sarcopenia. However, according to the PMI-based definition of sarcopenia, only 34.3% of patients were diagnosed with sarcopenia. Multivariate analysis identified sarcopenia based on PMI (hazard ratio [HR], 3.85; 95% confidence interval [CI], 2.04–7.26; p<0.001) and International Metastatic RCC Database Consortium poor risk status (HR, 1.90; 95% CI, 1.03–3.50; p=0.041) as significant and independent prognostic factors of OS. Conclusions PMI-based sarcopenia is a significant prognostic factor for OS in patients with RCC who receive nivolumab therapy.
Collapse
Affiliation(s)
- Hideto Ueki
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - Takuto Hara
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yasuyoshi Okamura
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yukari Bando
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomoaki Terakawa
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Junya Furukawa
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kenichi Harada
- Department of Urology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yuzo Nakano
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masato Fujisawa
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
48
|
Rossi SH, Stewart GD. Re: Clinical Validation of a Targeted Methylation-based Multi-cancer Early Detection Test Using an Independent Validation Set. Eur Urol 2022; 82:442-443. [PMID: 35779990 DOI: 10.1016/j.eururo.2022.06.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 06/17/2022] [Indexed: 12/22/2022]
Affiliation(s)
- Sabrina H Rossi
- Department of Surgery, University of Cambridge, Cambridge Biomedical Centre, Cambridge, UK.
| | - Grant D Stewart
- Department of Surgery, University of Cambridge, Cambridge Biomedical Centre, Cambridge, UK
| |
Collapse
|
49
|
Kim NY, Jung YY, Yang MH, Chinnathambi A, Govindasamy C, Narula AS, Namjoshi OA, Blough BE, Ahn KS. Tanshinone IIA exerts autophagic cell death through down-regulation of β-catenin in renal cell carcinoma cells. Biochimie 2022; 200:119-130. [PMID: 35654241 DOI: 10.1016/j.biochi.2022.05.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 05/21/2022] [Accepted: 05/26/2022] [Indexed: 12/13/2022]
Abstract
Renal cell carcinoma (RCC), also called kidney cancer, is one of the most common malignancies worldwide, including the United States and China. Because of the characteristics of RCC that are both insidious and largely insensitive to chemo-radiation, the incidence and mortality of RCC are increasing every year. However, there are few studies describing anti-cancer effects of the natural compounds on RCC as compared to other cancers. Here, we analyzed the anti-neoplastic impact of Tanshinone IIA (TSN) on RCC cells. We noted that TSN increased the expression of LC3 proteins while having little effect on PARP and Alix protein expression. We found that TSN up-regulated the expression of autophagy-related proteins such as Atg7 and Beclin-1. Moreover, TSN promoted the formation of autophagic vacuoles such as autophagosomes and autolysosomes. However, treatment with 3-Methyladenine (3-MA) or Chloroquine (CQ), slightly decreased the ability of TSN to induce autophagy, but still autophagy occurred. In addition, TSN inhibited translocation of β-catenin into the nucleus, and β-catenin deletion and TSN treatment in RCC increased the expression of LC3 protein. Overall our findings indicate that TSN can exert significant anti-tumor effects through down-regulation of β-catenin to induce autophagic cell death.
Collapse
Affiliation(s)
- Na Young Kim
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, South Korea
| | - Young Yun Jung
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, South Korea
| | - Min Hee Yang
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, South Korea
| | - Arunachalam Chinnathambi
- Department of Botany and Microbiology, College of Science, King Saud University, PO Box -2455, Riyadh, 11451, Saudi Arabia
| | - Chandramohan Govindasamy
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh, 11433, Saudi Arabia
| | | | - Ojas A Namjoshi
- Engine Biosciences, 733 Industrial Rd, San Carlos, CA, 94070, USA
| | - Bruce E Blough
- Center for Drug Discovery, RTI International, Research Triangle Park, Durham, NC, 27616, USA
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, South Korea.
| |
Collapse
|
50
|
Extracellular Vesicles—A New Potential Player in the Immunology of Renal Cell Carcinoma. J Pers Med 2022; 12:jpm12050772. [PMID: 35629194 PMCID: PMC9144962 DOI: 10.3390/jpm12050772] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/20/2022] [Accepted: 04/28/2022] [Indexed: 02/08/2023] Open
Abstract
The incidence of renal cell carcinoma (RCC) has doubled in the developed world within the last fifty years, and now it is responsible for 2–3% of diagnosed cancers. The delay in diagnosis and the not fully understood pathogenesis are the main challenges that have to be overcome. It seems that extracellular vesicles (EVs) are one of the key players in tumor development since they ensure a proper microenvironment for the tumor cells. The stimulation of angiogenesis and immunosuppression is mediated by molecules contained in EVs. It was shown that EVs derived from cancer cells can inhibit T cell proliferation, natural killer lymphocyte activation, and dendritic cell maturation by this mechanism. Moreover, EVs may be a biomarker for the response to anti-cancer treatment. In this review, we sum up the knowledge about the role of EVs in RCC pathogenesis and show their future perspectives in this field.
Collapse
|