1
|
Elias R, Nirschl T, Rezaee M, Yerrapragada A, Wang S, Cheaib J, Alam R, Patel S, Jing Y, Allaf M, McKean D, Klein AP, Fertig EJ, Baraban E, Ged Y, Yegnasubramanian S, Singla N. Clear-Cell Renal Cell Carcinoma Molecular Subtypes Differ by African and European Genetic Similarity. CANCER RESEARCH COMMUNICATIONS 2025; 5:743-755. [PMID: 40131247 PMCID: PMC12044083 DOI: 10.1158/2767-9764.crc-24-0624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/20/2025] [Accepted: 03/21/2025] [Indexed: 03/26/2025]
Abstract
SIGNIFICANCE Our study shows that AFR genetic similarity correlates with distinct ccRCC molecular subtypes. Further research is needed to disentangle environmental and genetic influences. Identifying these differences underscores the critical importance of including racially and ethnically diverse populations in cancer research to ensure more equitable and sustainable outcomes worldwide for all patients.
Collapse
Affiliation(s)
- Roy Elias
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Thomas Nirschl
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael Rezaee
- Brady Urologic Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Anirudh Yerrapragada
- Brady Urologic Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Shirley Wang
- Brady Urologic Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Joseph Cheaib
- Brady Urologic Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ridwan Alam
- Brady Urologic Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sunil Patel
- Brady Urologic Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Yuezhou Jing
- Brady Urologic Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mohamad Allaf
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Brady Urologic Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - David McKean
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alison P. Klein
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Epidemiology, Johns Hopkins University School of Public Health, Baltimore, Maryland
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Elana J. Fertig
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Applied Mathematics and Statistics, Johns Hopkins University Whiting School of Engineering, Baltimore, Maryland
| | - Ezra Baraban
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Yasser Ged
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Srinivasan Yegnasubramanian
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Epidemiology, Johns Hopkins University School of Public Health, Baltimore, Maryland
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- inHealth Precision Medicine program, Johns Hopkins Medicine, Baltimore, Maryland
| | - Nirmish Singla
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Brady Urologic Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
2
|
Bischoff ME, Shamsaei B, Yang J, Secic D, Vemuri B, Reisz JA, D’Alessandro A, Bartolacci C, Adamczak R, Schmidt L, Wang J, Martines A, Venkat J, Tcheuyap VT, Biesiada J, Behrmann CA, Vest KE, Brugarolas J, Scaglioni PP, Plas DR, Patra KC, Gulati S, Landero Figueroa JA, Meller J, Cunningham JT, Czyzyk-Krzeska MF. Copper Drives Remodeling of Metabolic State and Progression of Clear Cell Renal Cell Carcinoma. Cancer Discov 2025; 15:401-426. [PMID: 39476412 PMCID: PMC11803400 DOI: 10.1158/2159-8290.cd-24-0187] [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: 02/05/2024] [Revised: 09/23/2024] [Accepted: 10/30/2024] [Indexed: 11/02/2024]
Abstract
SIGNIFICANCE The work establishes a requirement for glucose-dependent coordination between energy production and redox homeostasis, which is fundamental for the survival of cancer cells that accumulate Cu and contributes to tumor growth.
Collapse
Affiliation(s)
- Megan E. Bischoff
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Behrouz Shamsaei
- Department of Biostatistics, Health Informatics and Data Sciences, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Biostatistics and Bioinformatics, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Juechen Yang
- Department of Biostatistics, Health Informatics and Data Sciences, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Biostatistics and Bioinformatics, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Dina Secic
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Bhargav Vemuri
- Department of Biostatistics, Health Informatics and Data Sciences, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Julie A. Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado
| | - Caterina Bartolacci
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Rafal Adamczak
- Institute of Engineering and Technology, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University, Torun, Poland
| | - Lucas Schmidt
- Trace Elements Group, Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jiang Wang
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Amelia Martines
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Jahnavi Venkat
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Vanina Toffessi Tcheuyap
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jacek Biesiada
- Division of Biostatistics and Bioinformatics, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Catherine A. Behrmann
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Katherine E. Vest
- Department of Molecular and Cellular Biosciences, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - James Brugarolas
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Pier Paolo Scaglioni
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - David R. Plas
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Krushna C. Patra
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Shuchi Gulati
- Division of Hematology and Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Oncology and Hematology, Department of Internal Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, California
| | - Julio A. Landero Figueroa
- Trace Elements Group, Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jarek Meller
- Department of Biostatistics, Health Informatics and Data Sciences, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Biostatistics and Bioinformatics, Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Institute of Engineering and Technology, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University, Torun, Poland
- Department of Computer Science, University of Cincinnati College of Engineering and Applied Sciences, Cincinnati, Ohio
| | - John T. Cunningham
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Maria F. Czyzyk-Krzeska
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Department of Veterans Affairs, Veteran Affairs Medical Center, Cincinnati, Ohio
- Department of Pharmacology and System Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
3
|
Sokouti B. A systematic investigation of clear cell renal cell carcinoma using meta-analysis and systems biology approaches. Mol Genet Genomics 2024; 299:87. [PMID: 39283494 DOI: 10.1007/s00438-024-02180-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 09/01/2024] [Indexed: 11/03/2024]
Abstract
Renal cell carcinoma with clear cells (ccRCC) is the most frequent kind; it accounts for almost 70% of all kidney cancers. A primary objective of current research was to find genes that may be used in ccRCC gene therapy to understand better the molecular pathways underlying the disease. Based on PubMed microarray searches and meta-analyses, we compared overall survival and recurrence-free survival rates in ccRCC patients with those in healthy samples. The technique was followed by a KEGG pathway and Gene Ontology (GO) function analyses, both performed in conjunction with the approach. Tumor immune estimate and multi-gene biomarkers validation for clinical outcomes were performed at the molecular and clinical cohort levels. Our analysis included fourteen GEO datasets based on inclusion and exclusion criteria. A meta-analysis procedure, network construction using PPIs, and four significant gene identification standard algorithms indicated that 11 genes had the most important differences. Ten genes were upregulated, and one was downregulated in the study. In order to analyze RFS and OS survival rates, 11 genes expressed in the GEPIA2 database were examined. Nearly nine of eleven significant genes have been found to beinvolved in tumor immunity. Furthermore, it was found that mRNA expression levels of these genes were significantly correlated with experimental literature studies on ccRCCs, which explained these findings. This study identified eleven gene panels associated with ccRCC growth and metastasis, as well as their immune system infiltration.
Collapse
Affiliation(s)
- Babak Sokouti
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
4
|
Gupta A, Roy AM. Racial and Ethnic Disparities in Survival Outcomes of Metastatic Renal Cell Carcinoma Patients Receiving Immunotherapy. Clin Genitourin Cancer 2024; 22:102104. [PMID: 38834500 DOI: 10.1016/j.clgc.2024.102104] [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: 02/07/2024] [Revised: 04/21/2024] [Accepted: 04/22/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICI) have significantly improved survival outcomes of metastatic renal cell carcinoma (mRCC). However, ethnic and racial minorities are often underrepresented in ICI clinical trials, leading to limited knowledge about ICI-specific survival outcomes for mRCC across different racial and ethnic groups. We investigated the impact of race and ethnicity on the ICI-specific survival outcomes of mRCC. MATERIALS We used The National Cancer Database (NCDB) to retrieve the data of 4858 mRCC patients diagnosed from 2014 to 2019 and receiving ICI-based regimens. We then compared survival outcomes using the Kaplan-Meier method and the Log-rank test. We analyzed the data using univariate and multivariable Cox regression analysis, adjusted for age, sex, comorbidity index, treatment centers, and grade. RESULTS White and Asian patients had significantly longer median overall survival (mOS) than African American (AA) patients (23.2 [95% CI 21.6, 24.7; P = .001] and 22.2 [95% CI 16.4, 55.1; P = .047] vs. 14.8 [95% CI 11.9, 19.2] months, respectively). After adjustment, White patients had significantly longer median OS (adjusted hazard ratio [HR] 0.71 [95% CI 0.58, 0.84]; P = .001). There was no significant difference in the mOS between Hispanic and non-Hispanic patients (P = .39). CONCLUSION Black race is an independent predictor of ICI-related survival in mRCC patients, independent of sociodemographics, clinicopathological, and treatment-related factors. Future research is required to understand the underlying reasons for these disparities, including potential genetic or biological differences and social and environmental factors.
Collapse
Affiliation(s)
- Amol Gupta
- Department of Medicine, The Johns Hopkins Hospital, Baltimore, MD.
| | - Arya Mariam Roy
- Department of Medicine, Division of Hematology and Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| |
Collapse
|
5
|
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
|
6
|
Vazquez LC, Xi Y, Rasmussen RG, Venzor JER, Kapur P, Zhong H, Dai JC, Morgan TN, Cadeddu JA, Pedrosa I. Characterization of Demographical Histologic Diversity in Small Renal Masses With the Clear Cell Likelihood Score. J Comput Assist Tomogr 2024; 48:370-377. [PMID: 38213063 DOI: 10.1097/rct.0000000000001567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2024]
Abstract
OBJECTIVE This study aimed to develop a diagnostic model to estimate the distribution of small renal mass (SRM; ≤4 cm) histologic subtypes for patients with different demographic backgrounds and clear cell likelihood score (ccLS) designations. MATERIALS AND METHODS A bi-institution retrospective cohort study was conducted where 347 patients (366 SRMs) underwent magnetic resonance imaging and received a ccLS before pathologic confirmation between June 2016 and November 2021. Age, sex, race, ethnicity, socioeconomic status, body mass index (BMI), and the ccLS were tabulated. The socioeconomic status for each patient was determined using the Area Deprivation Index associated with their residential address. The magnetic resonance imaging-derived ccLS assists in the characterization of SRMs by providing a likelihood of clear cell renal cell carcinoma (ccRCC). Pathological subtypes were grouped into four categories (ccRCC, papillary renal cell carcinoma, other renal cell carcinomas, or benign). Generalized estimating equations were used to estimate probabilities of the pathological subtypes across different patient subgroups. RESULTS Race and ethnicity, BMI, and ccLS were significant predictors of histology (all P < 0.001). Obese (BMI, ≥30 kg/m 2 ) Hispanic patients with ccLS of ≥4 had the highest estimated rate of ccRCC (97.1%), and normal-weight (BMI, <25 kg/m 2 ) non-Hispanic Black patients with ccLS ≤2 had the lowest (0.2%). The highest estimated rates of papillary renal cell carcinoma were found in overweight (BMI, 25-30 kg/m 2 ) non-Hispanic Black patients with ccLS ≤2 (92.3%), and the lowest, in obese Hispanic patients with ccLS ≥4 (<0.1%). CONCLUSIONS Patient race, ethnicity, BMI, and ccLS offer synergistic information to estimate the probabilities of SRM histologic subtypes.
Collapse
Affiliation(s)
| | - Yin Xi
- From the Department of Radiology, University of Texas Southwestern School of Medicine
| | - Robert G Rasmussen
- From the Department of Radiology, University of Texas Southwestern School of Medicine
| | | | | | | | | | | | | | | |
Collapse
|
7
|
Kotecha RR, Knezevic A, Arora K, Bandlamudi C, Kuo F, Carlo MI, Fitzgerald KN, Feldman DR, Shah NJ, Reznik E, Hakimi AA, Carrot-Zhang J, Mandelker D, Berger M, Lee CH, Motzer RJ, Voss MH. Genomic ancestry in kidney cancer: Correlations with clinical and molecular features. Cancer 2024; 130:692-701. [PMID: 37864521 PMCID: PMC11220722 DOI: 10.1002/cncr.35074] [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: 08/07/2023] [Revised: 09/13/2023] [Accepted: 09/18/2023] [Indexed: 10/23/2023]
Abstract
INTRODUCTION Genetic ancestry (GA) refers to population hereditary patterns that contribute to phenotypic differences seen among race/ethnicity groups, and differences among GA groups may highlight unique biological determinants that add to our understanding of health care disparities. METHODS A retrospective review of patients with renal cell carcinoma (RCC) was performed and correlated GA with clinicopathologic, somatic, and germline molecular data. All patients underwent next-generation sequencing of normal and tumor DNA using Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets, and contribution of African (AFR), East Asian (EAS), European (EUR), Native American, and South Asian (SAS) ancestry was inferred through supervised ADMIXTURE. Molecular data was compared across GA groups by Fisher exact test and Kruskal-Wallis test. RESULTS In 953 patients with RCC, the GA distribution was: EUR (78%), AFR (4.9%), EAS (2.5%), SAS (2%), Native American (0.2%), and Admixed (12.2%). GA distribution varied by tumor histology and international metastatic RCC database consortium disease risk status (intermediate-poor: EUR 58%, AFR 88%, EAS 74%, and SAS 73%). Pathogenic/likely pathogenic germline variants in cancer-predisposition genes varied (16% EUR, 23% AFR, 8% EAS, and 0% SAS), and most occurred in CHEK2 in EUR (3.1%) and FH in AFR (15.4%). In patients with clear cell RCC, somatic alteration incidence varied with significant enrichment in BAP1 alterations (EUR 17%, AFR 50%, SAS 29%; p = .01). Comparing AFR and EUR groups within The Cancer Genome Atlas, significant differences were identified in angiogenesis and inflammatory pathways. CONCLUSION Differences in clinical and molecular data by GA highlight population-specific variations in patients with RCC. Exploration of both genetic and nongenetic variables remains critical to optimize efforts to overcome health-related disparities.
Collapse
Affiliation(s)
- Ritesh R. Kotecha
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Weill Cornell Medical Center, New York, New York, USA
| | - Andrea Knezevic
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Kanika Arora
- Marie-Jose and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Chaitanya Bandlamudi
- Marie-Jose and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Fengshen Kuo
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Maria I. Carlo
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Weill Cornell Medical Center, New York, New York, USA
| | - Kelly N. Fitzgerald
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Darren R. Feldman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Weill Cornell Medical Center, New York, New York, USA
| | - Neil J. Shah
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Weill Cornell Medical Center, New York, New York, USA
| | - Ed Reznik
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Marie-Jose and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - A. Ari Hakimi
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jian Carrot-Zhang
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Marie-Jose and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Diana Mandelker
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Michael Berger
- Marie-Jose and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Chung-Han Lee
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Weill Cornell Medical Center, New York, New York, USA
| | - Robert J. Motzer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Weill Cornell Medical Center, New York, New York, USA
| | - Martin H. Voss
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Weill Cornell Medical Center, New York, New York, USA
| |
Collapse
|
8
|
Bischoff ME, Shamsaei B, Yang J, Secic D, Vemuri B, Reisz JA, D'Alessandro A, Bartolacci C, Adamczak R, Schmidt L, Wang J, Martines A, Biesiada J, Vest KE, Scaglioni PP, Plas DR, Patra KC, Gulati S, Figueroa JAL, Meller J, Cunningham JT, Czyzyk-Krzeska MF. Copper drives remodeling of metabolic state and progression of clear cell renal cell carcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.16.575895. [PMID: 38293110 PMCID: PMC10827129 DOI: 10.1101/2024.01.16.575895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Copper (Cu) is an essential trace element required for mitochondrial respiration. Late-stage clear cell renal cell carcinoma (ccRCC) accumulates Cu and allocates it to mitochondrial cytochrome c oxidase. We show that Cu drives coordinated metabolic remodeling of bioenergy, biosynthesis and redox homeostasis, promoting tumor growth and progression of ccRCC. Specifically, Cu induces TCA cycle-dependent oxidation of glucose and its utilization for glutathione biosynthesis to protect against H 2 O 2 generated during mitochondrial respiration, therefore coordinating bioenergy production with redox protection. scRNA-seq determined that ccRCC progression involves increased expression of subunits of respiratory complexes, genes in glutathione and Cu metabolism, and NRF2 targets, alongside a decrease in HIF activity, a hallmark of ccRCC. Spatial transcriptomics identified that proliferating cancer cells are embedded in clusters of cells with oxidative metabolism supporting effects of metabolic states on ccRCC progression. Our work establishes novel vulnerabilities with potential for therapeutic interventions in ccRCC. Accumulation of copper is associated with progression and relapse of ccRCC and drives tumor growth.Cu accumulation and allocation to cytochrome c oxidase (CuCOX) remodels metabolism coupling energy production and nucleotide biosynthesis with maintenance of redox homeostasis.Cu induces oxidative phosphorylation via alterations in the mitochondrial proteome and lipidome necessary for the formation of the respiratory supercomplexes. Cu stimulates glutathione biosynthesis and glutathione derived specifically from glucose is necessary for survival of Cu Hi cells. Biosynthesis of glucose-derived glutathione requires activity of glutamyl pyruvate transaminase 2, entry of glucose-derived pyruvate to mitochondria via alanine, and the glutamate exporter, SLC25A22. Glutathione derived from glucose maintains redox homeostasis in Cu-treated cells, reducing Cu-H 2 O 2 Fenton-like reaction mediated cell death. Progression of human ccRCC is associated with gene expression signature characterized by induction of ETC/OxPhos/GSH/Cu-related genes and decrease in HIF/glycolytic genes in subpopulations of cancer cells. Enhanced, concordant expression of genes related to ETC/OxPhos, GSH, and Cu characterizes metabolically active subpopulations of ccRCC cells in regions adjacent to proliferative subpopulations of ccRCC cells, implicating oxidative metabolism in supporting tumor growth.
Collapse
|
9
|
de Moraes FCA, Vilbert M, Alves VFC, de Oliveira Almeida G, Priantti JN, Madeira T, Stecca C, Fernandes MR, dos Santos NPC. Mesenchymal-Epithelial Transition Kinase Inhibitor Therapy in Patients with Advanced Papillary Renal-Cell Carcinoma: A Systematic Review and Meta-Analysis. Int J Mol Sci 2023; 24:17582. [PMID: 38139411 PMCID: PMC10744118 DOI: 10.3390/ijms242417582] [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: 10/07/2023] [Revised: 11/25/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023] Open
Abstract
Papillary subtypes of renal-cell carcinoma (pRCC) represent 10-15% of the cases and commonly have MET alterations. This systematic review and single-arm meta-analysis evaluated MET inhibitor therapy (METi) efficacy and safety in adults with confirmed advanced pRCC. The search strategy included PubMed, Web-of-science, Cochrane, and Scopus. We used the DerSimonian/Laird random effect model for all analyses; p-value < 5% was considered significant, and heterogeneity was assessed with I2. Three clinical trials and six cohort studies were included with 504 patients; 31% were MET-driven. Our pooled analysis demonstrated an objective response rate (ORR) in MET-driven, MET-independent, and overall patients of: 36% (95%CI: 10-62), 0% (95%CI: 0-3), and 21% (95%CI: 1-41), respectively. One-year disease control and progression-free survival rates were, respectively, 70% (95%CI: 52-88) and 15% (95%CI: 10-20). Twelve- and twenty-four-month survival rates were, respectively, 43% (95%CI: 23-64) and 10% (95%CI: 0-30). The prevalence of adverse events of any grade and grades 3-5 were 96% (95%CI: 91-100) and 44% (95%CI: 37-50), respectively. We suggest METi has anti-tumor activity and is tolerable in patients with advanced pRCC.
Collapse
Affiliation(s)
| | - Maysa Vilbert
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5T 2S8, Canada
| | | | | | - Jonathan N. Priantti
- School of Medicine, Federal University of Amazonas—UFAM, Manaus 69020-160, Brazil
| | - Thiago Madeira
- School of Medicine, Federal University of Minas Gerais—UFMG, Belo Horizonte 31270-901, Brazil
| | - Carlos Stecca
- Mackenzie Evangelical University Hospital, Curitiba 80710-390, Brazil
| | | | | |
Collapse
|
10
|
Cen HS, Dandamudi S, Lei X, Weight C, Desai M, Gill I, Duddalwar V. Diversity in Renal Mass Data Cohorts: Implications for Urology AI Researchers. Oncology 2023; 102:574-584. [PMID: 38104555 PMCID: PMC11178677 DOI: 10.1159/000535841] [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: 09/30/2023] [Accepted: 12/08/2023] [Indexed: 12/19/2023]
Abstract
INTRODUCTION We examine the heterogeneity and distribution of the cohort populations in two publicly used radiological image cohorts, the Cancer Genome Atlas Kidney Renal Clear Cell Carcinoma (TCIA TCGA KIRC) collection and 2019 MICCAI Kidney Tumor Segmentation Challenge (KiTS19), and deviations in real-world population renal cancer data from the National Cancer Database (NCDB) Participant User Data File (PUF) and tertiary center data. PUF data are used as an anchor for prevalence rate bias assessment. Specific gene expression and, therefore, biology of RCC differ by self-reported race, especially between the African American and Caucasian populations. AI algorithms learn from datasets, but if the dataset misrepresents the population, reinforcing bias may occur. Ignoring these demographic features may lead to inaccurate downstream effects, thereby limiting the translation of these analyses to clinical practice. Consciousness of model training biases is vital to patient care decisions when using models in clinical settings. METHODS Data elements evaluated included gender, demographics, reported pathologic grading, and cancer staging. American Urological Association risk levels were used. Poisson regression was performed to estimate the population-based and sample-specific estimation for prevalence rate and corresponding 95% confidence interval. SAS 9.4 was used for data analysis. RESULTS Compared to PUF, KiTS19 and TCGA KIRC oversampled Caucasian by 9.5% (95% CI, -3.7 to 22.7%) and 15.1% (95% CI, 1.5 to 28.8%), undersampled African American by -6.7% (95% CI, -10% to -3.3%), and -5.5% (95% CI, -9.3% to -1.8%). Tertiary also undersampled African American by -6.6% (95% CI, -8.7% to -4.6%). The tertiary cohort largely undersampled aggressive cancers by -14.7% (95% CI, -20.9% to -8.4%). No statistically significant difference was found among PUF, TCGA, and KiTS19 in aggressive rate; however, heterogeneities in risk are notable. CONCLUSION Heterogeneities between cohorts need to be considered in future AI training and cross-validation for renal masses.
Collapse
Affiliation(s)
- Harmony Selena Cen
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA,
| | - Siddhartha Dandamudi
- College of Human Medicine, Michigan State University, East Lansing, Michigan, USA
| | - Xiaomeng Lei
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Chris Weight
- Urologic Oncology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Mihir Desai
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Inderbir Gill
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Vinay Duddalwar
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
11
|
Alam R, Rezaee ME, Pallauf M, Elias R, Yerrapragada A, Enikeev D, Fang D, Shariat SF, Woldu SL, Ged YMA, Singla N. Socioeconomic determinants of racial disparities in survival outcomes among patients with renal cell carcinoma. Urol Oncol 2023; 41:460.e1-460.e9. [PMID: 37709565 DOI: 10.1016/j.urolonc.2023.08.016] [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: 04/18/2023] [Revised: 07/25/2023] [Accepted: 08/22/2023] [Indexed: 09/16/2023]
Abstract
PURPOSE Racially driven outcomes in cancer are challenging to study. Studies evaluating the impact of race in renal cell carcinoma (RCC) outcomes are inconsistent and unable to disentangle socioeconomic disparities from inherent biological differences. We therefore seek to investigate socioeconomic determinants of racial disparities with respect to overall survival (OS) when comparing Black and White patients with RCC. METHODS We queried the National Cancer Database (NCDB) for patients diagnosed with RCC between 2004 and 2017 with complete clinicodemographic data. Patients were examined across various stages (all, cT1aN0M0, and cM1) and subtypes (all, clear cell, or papillary). We performed Cox proportional hazards regression with adjustment for socioeconomic and disease factors. RESULTS There were 386,589 patients with RCC, of whom 46,507 (12.0%) were Black. Black patients were generally younger, had more comorbid conditions, less likely to be insured, in a lower income quartile, had lower rates of high school completion, were more likely to have papillary RCC histology, and more likely to be diagnosed at a lower stage of RCC than their white counterparts. By stage, Black patients demonstrated a 16% (any stage), 22.5% (small renal mass [SRM]), and 15% (metastatic) higher risk of mortality than White patients. Survival differences were also evident in histology-specific subanalyses. Socioeconomic factors played a larger role in predicting OS among patients with SRMs than in patients with metastasis. CONCLUSIONS Black patients with RCC demonstrate worse survival outcomes compared to White patients across all stages. Socioeconomic disparities between races play a significant role in influencing survival in RCC.
Collapse
Affiliation(s)
- Ridwan Alam
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Michael E Rezaee
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Maximilian Pallauf
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, Paracelsus Medical University Salzburg, University Hospital Salzburg, Salzburg, Austria
| | - Roy Elias
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Anirudh Yerrapragada
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Dmitry Enikeev
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Institute for Urology and Reproductive Health, Sechenov University, Moscow, Russia
| | - Dong Fang
- Department of Urology, Peking University First Hospital, Institute of Urology, Peking University, National Urological Cancer Center, Beijing, China
| | - Shahrokh F Shariat
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Solomon L Woldu
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Yasser M A Ged
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Nirmish Singla
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD.
| |
Collapse
|
12
|
Asif A, Rajpoot K, Graham S, Snead D, Minhas F, Rajpoot N. Unleashing the potential of AI for pathology: challenges and recommendations. J Pathol 2023; 260:564-577. [PMID: 37550878 PMCID: PMC10952719 DOI: 10.1002/path.6168] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 08/09/2023]
Abstract
Computational pathology is currently witnessing a surge in the development of AI techniques, offering promise for achieving breakthroughs and significantly impacting the practices of pathology and oncology. These AI methods bring with them the potential to revolutionize diagnostic pipelines as well as treatment planning and overall patient care. Numerous peer-reviewed studies reporting remarkable performance across diverse tasks serve as a testimony to the potential of AI in the field. However, widespread adoption of these methods in clinical and pre-clinical settings still remains a challenge. In this review article, we present a detailed analysis of the major obstacles encountered during the development of effective models and their deployment in practice. We aim to provide readers with an overview of the latest developments, assist them with insights into identifying some specific challenges that may require resolution, and suggest recommendations and potential future research directions. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Amina Asif
- Tissue Image Analytics Centre, Department of Computer ScienceUniversity of WarwickCoventryUK
| | - Kashif Rajpoot
- School of Computer ScienceUniversity of BirminghamBirminghamUK
| | - Simon Graham
- Histofy Ltd, Birmingham Business ParkBirminghamUK
| | - David Snead
- Histofy Ltd, Birmingham Business ParkBirminghamUK
- Department of PathologyUniversity Hospitals Coventry & Warwickshire NHS TrustCoventryUK
| | - Fayyaz Minhas
- Tissue Image Analytics Centre, Department of Computer ScienceUniversity of WarwickCoventryUK
- Cancer Research CentreUniversity of WarwickCoventryUK
| | - Nasir Rajpoot
- Tissue Image Analytics Centre, Department of Computer ScienceUniversity of WarwickCoventryUK
- Histofy Ltd, Birmingham Business ParkBirminghamUK
- Cancer Research CentreUniversity of WarwickCoventryUK
- The Alan Turing InstituteLondonUK
| |
Collapse
|
13
|
Chow RD, Long JB, Hassan S, Wheeler SB, Spees LP, Leapman MS, Hurwitz ME, McManus HD, Gross CP, Dinan MA. Disparities in immune and targeted therapy utilization for older US patients with metastatic renal cell carcinoma. JNCI Cancer Spectr 2023; 7:pkad036. [PMID: 37202354 PMCID: PMC10276895 DOI: 10.1093/jncics/pkad036] [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: 04/24/2023] [Accepted: 05/07/2023] [Indexed: 05/20/2023] Open
Abstract
Disparities in metastatic renal cell carcinoma (mRCC) outcomes persist in the era of oral anticancer agents (OAAs) and immunotherapies (IOs). We examined variation in the utilization of mRCC systemic therapies among US Medicare beneficiaries from 2015 to 2019. Logistic regression models evaluated the association between therapy receipt and demographic covariates including patient race, ethnicity, and sex. In total, 15 407 patients met study criteria. After multivariable adjustment, non-Hispanic Black race and ethnicity was associated with reduced IO (adjusted relative risk ratio [aRRR] = 0.76, 95% confidence interval [CI] = 0.61 to 0.95; P = .015) and OAA receipt (aRRR = 0.76, 95% CI = 0.64 to 0.90; P = .002) compared with non-Hispanic White race and ethnicity. Female sex was associated with reduced IO (aRRR = 0.73, 95% CI = 0.66 to 0.81; P < .001) and OAA receipt (aRRR = 0.74, 95% CI = 0.68 to 0.81; P < .001) compared with male sex. Thus, disparities by race, ethnicity, and sex were observed in mRCC systemic therapy utilization for Medicare beneficiaries from 2015 to 2019.
Collapse
Affiliation(s)
| | - Jessica B Long
- Yale Cancer Outcomes, Public Policy, and Effectiveness Research Center, New Haven, CT, USA
| | - Sirad Hassan
- Yale Cancer Outcomes, Public Policy, and Effectiveness Research Center, New Haven, CT, USA
| | - Stephanie B Wheeler
- Department of Health Policy and Management, Gillings School of Global Public Health, University of North Carolina at Chapel Hill (UNC-CH), Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, UNC-CH, Chapel Hill, NC, USA
| | - Lisa P Spees
- Department of Health Policy and Management, Gillings School of Global Public Health, University of North Carolina at Chapel Hill (UNC-CH), Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, UNC-CH, Chapel Hill, NC, USA
| | - Michael S Leapman
- Yale Cancer Outcomes, Public Policy, and Effectiveness Research Center, New Haven, CT, USA
- Department of Urology, Yale School of Medicine, New Haven, CT, USA
| | - Michael E Hurwitz
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Hannah D McManus
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Cary P Gross
- Yale Cancer Outcomes, Public Policy, and Effectiveness Research Center, New Haven, CT, USA
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Michaela A Dinan
- Yale Cancer Outcomes, Public Policy, and Effectiveness Research Center, New Haven, CT, USA
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT, USA
| |
Collapse
|
14
|
Lichtensztajn DY, Hofer BM, Leppert JT, Brooks JD, Chung BI, Shah SA, DeRouen MC, Cheng I. Associations of Renal Cell Carcinoma Subtype with Patient Demographics, Comorbidities, and Neighborhood Socioeconomic Status in the California Population. Cancer Epidemiol Biomarkers Prev 2023; 32:202-207. [PMID: 36480301 PMCID: PMC9905278 DOI: 10.1158/1055-9965.epi-22-0784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/07/2022] [Accepted: 12/01/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Renal cell carcinoma (RCC) subtypes differ in molecular characteristics and prognosis. We investigated the associations of RCC subtype with patient demographics, comorbidity, and neighborhood socioeconomic status (nSES). METHODS Using linked California Cancer Registry and Office of Statewide Health Planning and Development data, we identified history of hypertension, diabetes, and kidney disease prior to RCC diagnosis in Asian/Pacific Islander, non-Latino Black, Latino, and non-Latino White adults diagnosed with their first pathologically confirmed RCC from 2005 through 2015. We used multinomial multivariable logistic regression to model the association of demographics, comorbidity, and nSES with clear-cell, papillary, and chromophobe RCC subtype. RESULTS Of the 40,016 RCC cases included, 62.6% were clear cell, 10.9% papillary, and 5.9% chromophobe. The distribution of subtypes differed strikingly by race and ethnicity, ranging from 40.4% clear cell and 30.4% papillary in non-Latino Black adults to 70.7% clear cell and 4.5% papillary in Latino adults. In multivariable analysis, non-Latino Black individuals had a higher likelihood of presenting with papillary (OR, 3.99; 95% confidence interval, 3.61-4.42) and chromophobe (OR, 1.81; 1.54-2.13) versus clear-cell subtype compared with non-Latino White individuals. Both hypertension (OR, 1.19; 1.10-1.29) and kidney disease (OR, 2.38; 2.04-2.77 end-stage disease; OR, 1.52; 1.33-1.72 non-end-stage disease) were associated with papillary subtype. Diabetes was inversely associated with both papillary (OR, 0.63; 0.58-0.69) and chromophobe (OR, 0.61; 0.54-0.70) subtypes. CONCLUSIONS RCC subtype is independently associated with patient demographics, and comorbidity. IMPACT Targeted RCC treatments or RCC prevention efforts may have differential impact across population subgroups.
Collapse
Affiliation(s)
| | - Brenda M Hofer
- California Cancer Reporting and Epidemiologic Surveillance (CalCARES) Program, University of California, Davis, Davis, California
| | - John T Leppert
- Stanford University School of Medicine, Stanford, California.,Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| | - James D Brooks
- Stanford University School of Medicine, Stanford, California
| | | | - Sumit A Shah
- Stanford University School of Medicine, Stanford, California
| | - Mindy C DeRouen
- University of California, San Francisco, San Francisco, California
| | - Iona Cheng
- University of California, San Francisco, San Francisco, California
| |
Collapse
|
15
|
McClain KM, Sampson JN, Petrick JL, Mazzilli KM, Gerszten RE, Clish CB, Purdue MP, Lipworth L, Moore SC. Metabolomic Analysis of Renal Cell Carcinoma in the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial. Metabolites 2022; 12:metabo12121189. [PMID: 36557227 PMCID: PMC9785244 DOI: 10.3390/metabo12121189] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 11/18/2022] [Accepted: 11/19/2022] [Indexed: 12/02/2022] Open
Abstract
Background: In the US in 2021, 76,080 kidney cancers are expected and >80% are renal cell carcinomas (RCCs). Along with excess fat, metabolic dysfunction is implicated in RCC etiology. To identify RCC-associated metabolites, we conducted a 1:1 matched case−control study nested within the Prostate, Lung, Colorectal, and Ovarian (PLCO) Cancer Screening Trial. Methods: We measured 522 serum metabolites in 267 cases/control pairs. Cases were followed for a median 7.1 years from blood draw to diagnosis. Using conditional logistic regression, we computed adjusted odds ratios (ORs) and 95% confidence intervals (CIs) comparing risk between 90th and 10th percentiles of log metabolite intensity, with the significance threshold at a false discovery rate <0.20. Results: Four metabolites were inversely associated with risk of RCC during follow-up—C38:4 PI, C34:0 PC, C14:0 SM, and C16:1 SM (ORs ranging from 0.33−0.44). Two were positively associated with RCC risk—C3-DC-CH3 carnitine and C5 carnitine (ORs = 2.84 and 2.83, respectively). These results were robust when further adjusted for metabolic risk factors (body mass index (BMI), physical activity, diabetes/hypertension history). Metabolites associated with RCC had weak correlations (|r| < 0.2) with risk factors of BMI, physical activity, smoking, alcohol, and diabetes/hypertension history. In mutually adjusted models, three metabolites (C38:4 PI, C14:0 SM, and C3-DC-CH3 carnitine) were independently associated with RCC risk. Conclusions: Serum concentrations of six metabolites were associated with RCC risk, and three of these had independent associations from the mutually adjusted model. These metabolites may point toward new biological pathways of relevance to this malignancy.
Collapse
Affiliation(s)
- Kathleen M. McClain
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
- Correspondence: ; Tel.: +240-276-6317
| | - Joshua N. Sampson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
| | | | - Kaitlyn M. Mazzilli
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
| | - Robert E. Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Clary B. Clish
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Mark P. Purdue
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
| | - Loren Lipworth
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Steven C. Moore
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
| |
Collapse
|
16
|
Xu W, Anwaier A, Liu W, Tian X, Su J, Shi G, Qu Y, Zhang H, Ye D. The unique genomic landscape and prognostic mutational signature of Chinese clear cell renal cell carcinoma. JOURNAL OF THE NATIONAL CANCER CENTER 2022; 2:162-170. [PMID: 39036450 PMCID: PMC11256728 DOI: 10.1016/j.jncc.2022.07.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 12/12/2022] Open
Abstract
Background The genomic background affects the occurrence and metastasis of cancers, including clear cell renal cell carcinoma (ccRCC). However, reports focusing on the prognostic mutational signature of Chinese ccRCC are lacking. Methods Overall, 929 patients, including a training cohort with Chinese patients (n = 201), a testing cohort with Caucasian patients (n = 274), and a validation cohort (n = 454) were analyzed for the genomic landscape of ccRCC. Then, machine-learning algorithms were used to identify and evaluate the genomic mutational signature (GMS) in ccRCC. Analyses for prognosis, immune microenvironment, association with independent clinicopathological features, and predictive responses for immune checkpoint therapies (ICTs) were performed. Results The DNA variation data of 929 patients with ccRCC suggested markedly differential genomic mutational frequency of the most frequent genes, such as VHL, PBRM1, BAP1, SETD2, and KDM5C between the Chinese and Caucasian populations. PBRM1 showed significant co-occurrence with VHL and SETD2. We then successfully identified a seven-gene mutational signature (GMSMut) that included mutations in FBN1, SHPRH, CELSR1, COL6A6, DST, ABCA13, and BAP1. The GMSMut significantly predicted progressive progression (P < 0.0001, HR = 2.81) and poor prognosis (P < 0.0001, HR = 3.89) in the Chinese training cohort. Moreover, ccRCC patients with the GMSMut had poor survival rates in the testing cohort (P = 0.020) and poor outcomes were predicted for those treated with ICTs in the validation cohort (P = 0.036). Interestingly, a favorable clinical response to ICTs, elevated expression of immune checkpoints, and increased abundance of tumor-infiltrated lymphocytes, specifically CD8+ T cells, Tregs, and macrophages, were observed in the GMSMut cluster. Conclusions This study described the pro-tumorigenic GMSMut cluster that improved the prognostic accuracy in Chinese patients with ccRCC. Our discovery of the novel independent prognostic signature highlights the relationship between tumor phenotype and genomic mutational characteristics of ccRCC.
Collapse
Affiliation(s)
- Wenhao Xu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Aihetaimujiang Anwaier
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wangrui Liu
- Department of Interventional Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xi Tian
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiaqi Su
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Guohai Shi
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuanyuan Qu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hailiang Zhang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
17
|
Li X, Xu Z, Xu T, Qi F, Song N. Basic Characteristics and Survival Outcomes of Asian-American Patients with Clear Cell Renal Cell Carcinoma and Comparisons with White Patients: A Population-Based Analysis. Int J Gen Med 2021; 14:7869-7883. [PMID: 34795508 PMCID: PMC8593352 DOI: 10.2147/ijgm.s340284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 10/21/2021] [Indexed: 12/16/2022] Open
Abstract
Background To explore the baseline characteristics, pathological and survival outcomes of Asian-American patients with clear cell renal cell carcinoma (ccRCC), and make comparisons with White patients. Materials and Methods In this study, patients diagnosed with ccRCC between 2010 and 2015 were extracted from the Surveillance, Epidemiology, and End Results (SEER) database. Basic characteristics of Asian-American patients were analysed and compared with White patients. Then, proportional mortality ratio (PMR) analyses were performed in Asian population to investigate the proportions of different cause of deaths (CODs), and make comparisons with White patients. Moreover, Kaplan-Meier (KM) analyses were developed to investigate the survival disparities of ccRCC patients between Asian-Americans and White patients. Finally, a competing risk regression model was constructed to identify potential prognostic factors for ccRCC patients in the whole population. Results A total of 1586 Asian-American patients were eventually identified, and the median age at diagnosis was 61 years old. In Asian patients, those from South Asian had the youngest age at diagnosis (P<0.001) and the earliest stage of diseases (localized: 76.83%, T1: 70.73%, all P<0.05) when compared with other ethnicities. No significant differences were detected in tumor characteristics between Asian-Americans and White patients. Older age (P<0.001), earlier stage (P<0.001) and the administration of surgery (P=0.050) were tightly associated with a lower risk of dying of RCC in Asian-American patients. Additionally, Asian-American patients had comparable survival outcomes when compared with White patients. Lastly, competing risk regression model revealed that age at diagnosis (P<0.001), tumor grade (P<0.001), histological stage (P<0.001), median household income (P<0.001) and the administration of surgery (P<0.001) were prognostic factors for cancer-specific survival (CSS) in ccRCC patients, while died of other causes was regarded as a competing event. Conclusion Asian-American patients had similar tumor characteristics and survival outcomes with White patients. In Asian patients, those from South Asian had the youngest age at diagnosis and the earliest stage of diseases. Age, grade, histological stage, household income and surgery were identified to be closely related to CSS in ccRCC patients. In the future, prospective and well-designed studies are needed to verify our findings.
Collapse
Affiliation(s)
- Xiao Li
- Department of Urology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, 210009, People's Republic of China
| | - Zicheng Xu
- Department of Urology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, 210009, People's Republic of China
| | - Ting Xu
- Department of Urology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, 210009, People's Republic of China
| | - Feng Qi
- Department of Urology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, 210009, People's Republic of China
| | - Ninghong Song
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People's Republic of China
| |
Collapse
|
18
|
Bossé D, Xie W, Lin X, Simantov R, Lalani AKA, Graham J, Wells JC, Donskov F, Rini B, Beuselinck B, Alva A, Hansen A, Wood L, Soulières D, Kollmannsberger C, Patenaude F, Heng DYC, Choueiri TK, McKay RR. Outcomes in Black and White Patients With Metastatic Renal Cell Carcinoma Treated With First-Line Tyrosine Kinase Inhibitors: Insights From Two Large Cohorts. JCO Glob Oncol 2021; 6:293-306. [PMID: 32109159 PMCID: PMC7055470 DOI: 10.1200/jgo.19.00380] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
PURPOSE To investigate whether black race is an independent predictor of overall survival (OS) in metastatic renal cell carcinoma (mRCC). METHODS We performed a retrospective 2-cohort (International Metastatic Renal Cell Carcinoma Database Consortium [IMDC] and trial-database) study of patients with mRCC treated with first-line tyrosine kinase inhibitors (TKIs). Unmatched (UM) and matched (M) analyses accounting for imbalances in region, year of treatment, age, and sex between races were performed. Cox models adjusting for histology, number of metastatic sites, nephrectomy, and IMDC risk compared time to treatment failure (TTF; IMDC cohort), progression-free survival (PFS; trial-database cohort), and OS. RESULTS The IMDC cohort included 73 black versus 3,381 (UM) and 1,236 (M) white patients. The trial-database cohort included 21 black versus 1,040 (UM) and 431 (M) white patients. Median OS for black versus white patients was 18.5 versus 25.8 months in the IMDC group and 21.0 versus 25.6 months in the trial-database group. Differences in OS were not significant in multivariable analysis in the IMDC group (hazard ratio [HR]M, 1.0; 95% CI, 0.7 to 1.5; HRUM, 1.1; 95% CI, 0.8 to 1.4) and trial-database (HRM, 1.5; 95% CI, 0.8 to 2.7; HRUM, 1.4; 95% CI, 0.8 to 2.6) cohorts. TTF for black patients was shorter in the UM IMDC cohort (HRUM, 1.4; 95% CI, 1.1 to 1.8; P = .003), but not in the M analysis. PFS was shorter for black patients in both analyses in the trial-database cohort (HRM, 2.3; 95% CI, 1.4 to 3.9; P = .002; HRUM, 2.3; 95% CI, 1.4 to 3.9; P = .002). CONCLUSION Black patients had more IMDC risk factors and worse outcomes with TKIs versus white patients. Race was not an independent predictor of OS. Strategies to understand biologic determinants of outcomes for minority patients are needed to optimize care.
Collapse
Affiliation(s)
- Dominick Bossé
- The Ottawa Hospital, Division of Medical Oncology, University of Ottawa, Ottawa, Ontario, Canada.,Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Wanling Xie
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Xun Lin
- Pfizer Oncology, La Jolla, CA
| | | | - Aly-Khan A Lalani
- Juravinski Cancer Centre, Department of Oncology, McMaster University, Hamilton, Ontario, Canada
| | | | - J Connor Wells
- Tom Baker Cancer Centre, University of Calgary, Calgary, Alberta, Canada
| | | | - Brian Rini
- Department of Hematology and Medical Oncology, Cleveland Clinic-Taussig Cancer Institute, Cleveland, OH
| | - Benoit Beuselinck
- University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | | | - Aaron Hansen
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Lori Wood
- Queen Elizabeth II Health Sciences Centre, Halifax, Nova Scotia, Canada
| | - Denis Soulières
- Centre Hospitalier de l'Université de Montréal, Division of Oncology, Montreal, Quebec, Canada
| | | | - Francois Patenaude
- Department of Oncology, McGill University Health Centre, McGill University, Montreal, Quebec, Canada
| | - Daniel Y C Heng
- University of California San Diego, Moores Cancer Center, San Diego, CA
| | - Toni K Choueiri
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Rana R McKay
- University of California San Diego, Moores Cancer Center, San Diego, CA
| |
Collapse
|
19
|
Olsen TA, Martini DJ, Goyal S, Liu Y, Evans ST, Magod B, Brown JT, Yantorni L, Russler GA, Caulfield S, Goldman JM, Harris WB, Kucuk O, Carthon BC, Master VA, Nazha B, Bilen MA. Racial Differences in Clinical Outcomes for Metastatic Renal Cell Carcinoma Patients Treated With Immune-Checkpoint Blockade. Front Oncol 2021; 11:701345. [PMID: 34222024 PMCID: PMC8242950 DOI: 10.3389/fonc.2021.701345] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 05/28/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Immune-checkpoint-inhibitors (ICIs) have become the cornerstone of metastatic renal-cell-carcinoma (mRCC) therapy. However, data are limited regarding clinical outcomes by race. In this study, we compared the real-world outcomes between African American (AA) and Caucasian mRCC patients treated with ICIs. METHODS We performed a retrospective study of 198 patients with mRCC who received ICI at the Emory Winship Cancer Institute from 2015-2020. Clinical outcomes were measured by overall survival (OS), progression-free survival (PFS), and overall response rate (ORR) defined as a complete or partial response maintained for at least 6 months per response evaluation criteria in solid tumors version 1.1. Univariate and multivariable analyses were carried out for OS and PFS by Cox proportional-hazard model and ORR by logistical-regression model. Descriptive statistics compared rates of immune-related adverse events (irAEs) and non-clear-cell-RCC (nccRCC) histology were assessed using Chi-square test. RESULTS Our cohort was comprised of 38 AA and 160 Caucasian patients. Most were diagnosed with clear-cell-RCC (ccRCC) (78%) and more than half received (57%) PD-1/PD-L1 monotherapy. Most patients were intermediate or poor-risk groups (83%). Comparing to Caucasians, our AA cohort contained more females and nccRCC cases. Kaplan-Meier method showed AAs had no statistically different median OS (17 vs 25 months, p=0.368) and PFS (3.1 vs 4.4 months, p=0.068) relative to Caucasian patients. On multivariable analysis, AA patients had significantly shorter PFS (HR=1.52, 95% CI: 1.01-2.3, p=0.045), similar ORR (OR=1.04, 95% CI: 0.42-2.57, p=0.936) and comparable OS (HR=1.09, 95% CI: 0.61-1.95, p=0.778) relative to Caucasians. CONCLUSIONS Our real-world analysis of ICI-treated mRCC patients showed that AAs experienced shorter PFS but similar OS relative to Caucasians. This similarity in survival outcomes is reassuring for the use of ICI amongst real-world patient populations, however, the difference in treatment response is poorly represented in early outcomes data from clinical trials. Thus, the literature requires larger prospective studies to validate these findings.
Collapse
Affiliation(s)
- T. Anders Olsen
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, United States
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | - Dylan J. Martini
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, United States
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | - Subir Goyal
- Departments of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, United States
| | - Yuan Liu
- Departments of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, United States
| | - Sean T. Evans
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, United States
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | - Benjamin Magod
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, United States
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | - Jacqueline T. Brown
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, United States
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | - Lauren Yantorni
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | - Greta Anne Russler
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | - Sarah Caulfield
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, United States
- Department of Pharmaceutical Services, Emory University School of Medicine, Atlanta, GA, United States
| | - Jamie M. Goldman
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, United States
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | - Wayne B. Harris
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, United States
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | - Omer Kucuk
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, United States
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | - Bradley C. Carthon
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, United States
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | - Viraj A. Master
- Department of Urology, Emory University School of Medicine, Atlanta, GA, United States
| | - Bassel Nazha
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, United States
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | - Mehmet Asim Bilen
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, United States
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
| |
Collapse
|
20
|
Batai K, Hooker S, Kittles RA. Leveraging genetic ancestry to study health disparities. AMERICAN JOURNAL OF PHYSICAL ANTHROPOLOGY 2021; 175:363-375. [PMID: 32935870 PMCID: PMC8246846 DOI: 10.1002/ajpa.24144] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 07/22/2020] [Accepted: 08/20/2020] [Indexed: 12/14/2022]
Abstract
Research to understand human genomic variation and its implications in health has great potential to contribute in the reduction of health disparities. Biological anthropology can play important roles in genomics and health disparities research using a biocultural approach. This paper argues that racial/ethnic categories should not be used as a surrogate for sociocultural factors or global genomic clusters in biomedical research or clinical settings, because of the high genetic heterogeneity that exists within traditional racial/ethnic groups. Genetic ancestry is used to show variation in ancestral genomic contributions to recently admixed populations in the United States, such as African Americans and Hispanic/Latino Americans. Genetic ancestry estimates are also used to examine the relationship between ancestry-related biological and sociocultural factors affecting health disparities. To localize areas of genomes that contribute to health disparities, admixture mapping and genome-wide association studies (GWAS) are often used. Recent GWAS have identified many genetic variants that are highly differentiated among human populations that are associated with disease risk. Some of these are population-specific variants. Many of these variants may impact disease risk and help explain a portion of the difference in disease burden among racial/ethnic groups. Genetic ancestry is also of particular interest in precision medicine and disparities in drug efficacy and outcomes. By using genetic ancestry, we can learn about potential biological differences that may contribute to the heterogeneity observed across self-reported racial groups.
Collapse
Affiliation(s)
- Ken Batai
- Department of UrologyUniversity of ArizonaTucsonArizonaUSA
| | - Stanley Hooker
- Division of Health Equities, Department of Population SciencesCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Rick A. Kittles
- Division of Health Equities, Department of Population SciencesCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| |
Collapse
|
21
|
Pinheiro PS, Medina HN, Callahan KE, Koru-Sengul T, Sharma J, Kobetz EN, Penedo FJ. Kidney cancer mortality disparities among Hispanics in the US. Cancer Epidemiol 2021; 72:101938. [PMID: 33862414 DOI: 10.1016/j.canep.2021.101938] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/01/2021] [Accepted: 04/02/2021] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Kidney cancer incidence is increasing among Hispanics but rate differences by distinct group, such as Cuban, Puerto Rican, and Mexican have not been studied. To fill this knowledge gap, we use mortality data, reflecting fatal kidney cancers, to examine patterns by race-ethnicity, including detailed Hispanic groups, and correlate the mortality rates with each group's prevalence of known kidney cancer risk factors: smoking, obesity, hypertension, diabetes, and chronic kidney disease. METHODS We used individual-level death data for California, Florida, and New York (2008-2018), and population prevalence data from the National Health Interview Surveys (2008-2018). Age-adjusted mortality rates (AAMRs) and regression-derived mortality rate ratios (MRRs) were computed. Pearson correlation analyses assessed the extent to which group-specific risk factor prevalence explained variability in observed AAMRs. RESULTS US-born Mexican Americans and American Indians had the highest rates and MRRs compared to Whites: 1.44 (95 %CI: 1.35-1.53) and 1.51 (1.38-1.64) for Mexican American men and women, respectively, and 1.54 (95 %CI: 1.25-1.89) and 1.53 (95 %CI: 1.15-2.04) for American Indians. In contrast, non-Mexican Hispanics had lower rates than Whites. Among males, positive correlations between AAMRs and smoking, obesity, and chronic kidney disease prevalence by race-ethnicity were found. CONCLUSION Mexican Americans and American Indians are high-risk for fatal kidney cancer. Disparities are only partially attributable to higher smoking and obesity prevalence, and more so among men than women. A shared risk factor profile, as well as possible genetic similarities, may explain their disproportionately higher kidney cancer mortality, but further research is warranted.
Collapse
Affiliation(s)
- Paulo S Pinheiro
- Sylvester Comprehensive Cancer Center, University of Miami School of Medicine, USA; Public Health Sciences, University of Miami School of Medicine, USA.
| | - Heidy N Medina
- Public Health Sciences, University of Miami School of Medicine, USA.
| | | | - Tulay Koru-Sengul
- Sylvester Comprehensive Cancer Center, University of Miami School of Medicine, USA; Public Health Sciences, University of Miami School of Medicine, USA.
| | - Janaki Sharma
- Sylvester Comprehensive Cancer Center, University of Miami School of Medicine, USA.
| | - Erin N Kobetz
- Sylvester Comprehensive Cancer Center, University of Miami School of Medicine, USA; Public Health Sciences, University of Miami School of Medicine, USA.
| | - Frank J Penedo
- Sylvester Comprehensive Cancer Center, University of Miami School of Medicine, USA; Department of Psychology, University of Miami, USA.
| |
Collapse
|
22
|
Suss NR, Bruha MJ, Monaghan TF, Robins D, Flores V, Agudelo CW, Smith M, Hyacinthe L, McNeil BK, Weiss J, Winer A. Assessing the role of race in pathological upstaging of renal cell carcinoma: Results from the National Cancer Database. Int J Clin Pract 2021; 75:e13818. [PMID: 33159366 DOI: 10.1111/ijcp.13818] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 11/02/2020] [Indexed: 12/26/2022] Open
Abstract
PURPOSE Pathologic upstaging in renal cell carcinoma (RCC) is common and confers a significant risk of poor surgical and survival outcomes. Preoperative predictors of upstaging are of great clinical relevance but empirical evidence specific to racial minorities remains scarce. METHODS National Cancer Database (NCDB) analysis of T3a-specific upstaging among White, African-American, Hispanic and Asian Pacific Islander (API) patients with AJCC cT1N0M0 RCC who underwent partial or radical nephrectomy between 2010 and 2015. Independent preoperative predictors of tumour upstaging were identified using multivariate logistic regression analyses. RESULTS A total of 81 002 patients met the criteria for inclusion (5.6% T3a-specific upstaging). Increased age, increased Charlson-Deyo comorbidity index, clinical stages cT1b and unspecified cT1, and increased Fuhrman nuclear grade were identified as independent risk factors for upstaging. Independent protective factors for upstaging were younger age, female sex, African-American race and papillary, chromophobe, and unspecified RCC histologic subtypes. Significant risk factors and protective factors within individual racial subgroups were highly consistent with those observed in the overall study sample. All independent factors identified on race-specific subgroup analyses were significant in the same direction relative to the overall study sample. Variables found to be non-significant in the overall study sample remained non-significant across all racial subgroup analyses. CONCLUSION The present study of nationally representative data found no clinically significant differences in upstaging risk across individual racial subgroups relative to the overall study sample. Preoperative factors that can be used to predict pT3a-specific tumour upstaging in CT1N0M0 RCC likely persist across different racial groups.
Collapse
Affiliation(s)
- Nicholas R Suss
- Department of Urology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Matthew J Bruha
- Department of Urology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Thomas F Monaghan
- Department of Urology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Dennis Robins
- Department of Urology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Viktor Flores
- Department of Urology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Christina W Agudelo
- Department of Urology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Matthew Smith
- Department of Urology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Llewellyn Hyacinthe
- Department of Urology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Brian K McNeil
- Department of Urology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Jeffrey Weiss
- Department of Urology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Andrew Winer
- Department of Urology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| |
Collapse
|
23
|
Abel MK, Liao CI, Chan C, Lee D, Rohatgi A, Darcy KM, Tian C, Mann AK, Maxwell GL, Kapp DS, Chan JK. Racial disparities in high-risk uterine cancer histologic subtypes: A United States Cancer Statistics study. Gynecol Oncol 2021; 161:470-476. [PMID: 33722415 DOI: 10.1016/j.ygyno.2021.02.037] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 02/27/2021] [Indexed: 10/21/2022]
Abstract
OBJECTIVE Black women with uterine cancer on average have worse survival outcomes compared to White women, in part due to higher rates of aggressive, non-endometrioid subtypes. However, analyses of incidence trends by specific high-risk subtypes are lacking, including those with hysterectomy and active pregnancy correction. The objective of our study was to evaluate racial disparities in age-adjusted incidence of non-endometrioid uterine cancer in 720,984 patients. METHODS Data were obtained from United States Cancer Statistics using SEER*Stat. We used the Behavioral Risk Factor Surveillance System to correct for hysterectomy and active pregnancy. Age-adjusted, corrected incidence of uterine cancer from 2001 to 2016 and annual percent change (APC) were calculated using Joinpoint regression. RESULTS Of 720,984 patients, 560,131 (77.7%) were White, 72,328 (10.0%) were Black, 56,239 (7.8%) were Hispanic, and 22,963 (3.2%) were Asian/Pacific Islander. Age-adjusted incidence of uterine cancer increased from 40.8 (per 100,000) in 2001 to 42.9 in 2016 (APC = 0.5, p < 0.001). Black women had the highest overall incidence at 49.5 (APC = 2.3, p < 0.001). The incidence of non-endometrioid subtypes was higher in Black compared to White women, with the most pronounced differences seen in serous carcinoma (9.1 vs. 3.0), carcinosarcoma (6.1 vs. 1.8), and leiomyosarcoma (1.3 vs. 0.6). In particular, Black women aged 70-74 with serous carcinoma had the highest incidence (61.3) and the highest APC (7.3, p < 0.001). CONCLUSIONS Black women have a two to four-fold higher incidence of high-risk uterine cancer subtypes, particularly serous carcinoma, carcinosarcoma, and leiomyosarcoma, compared to White women after correcting for hysterectomy and active pregnancy.
Collapse
Affiliation(s)
- Mary Kathryn Abel
- University of California, San Francisco School of Medicine, San Francisco, CA, USA
| | - Cheng-I Liao
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chloe Chan
- Division of Gynecologic Oncology, Palo Alto Medical Foundation, California Pacific Medical Center, Sutter Health, San Francisco, CA, USA
| | - Danny Lee
- Division of Gynecologic Oncology, Palo Alto Medical Foundation, California Pacific Medical Center, Sutter Health, San Francisco, CA, USA
| | - Atharva Rohatgi
- Division of Gynecologic Oncology, Palo Alto Medical Foundation, California Pacific Medical Center, Sutter Health, San Francisco, CA, USA
| | - Kathleen M Darcy
- Gynecologic Cancer Center of Excellence, Department of Obstetrics & Gynecology, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD, USA; The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Chunqiao Tian
- Gynecologic Cancer Center of Excellence, Department of Obstetrics & Gynecology, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD, USA; The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Amandeep K Mann
- Division of Gynecologic Oncology, Palo Alto Medical Foundation Research Institute, Palo Alto, CA, USA
| | - George L Maxwell
- Gynecologic Cancer Center of Excellence, Department of Obstetrics & Gynecology, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Daniel S Kapp
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - John K Chan
- Division of Gynecologic Oncology, Palo Alto Medical Foundation, California Pacific Medical Center, Sutter Health, San Francisco, CA, USA.
| |
Collapse
|
24
|
Kim H, Lee SJ, Park SJ, Choi IY, Hong SH. Machine Learning Approach to Predict the Probability of Recurrence of Renal Cell Carcinoma After Surgery: Prediction Model Development Study. JMIR Med Inform 2021; 9:e25635. [PMID: 33646127 PMCID: PMC7961397 DOI: 10.2196/25635] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/23/2021] [Accepted: 01/29/2021] [Indexed: 12/15/2022] Open
Abstract
Background Renal cell carcinoma (RCC) has a high recurrence rate of 20% to 30% after nephrectomy for clinically localized disease, and more than 40% of patients eventually die of the disease, making regular monitoring and constant management of utmost importance. Objective The objective of this study was to develop an algorithm that predicts the probability of recurrence of RCC within 5 and 10 years of surgery. Methods Data from 6849 Korean patients with RCC were collected from eight tertiary care hospitals listed in the KOrean Renal Cell Carcinoma (KORCC) web-based database. To predict RCC recurrence, analytical data from 2814 patients were extracted from the database. Eight machine learning algorithms were used to predict the probability of RCC recurrence, and the results were compared. Results Within 5 years of surgery, the highest area under the receiver operating characteristic curve (AUROC) was obtained from the naïve Bayes (NB) model, with a value of 0.836. Within 10 years of surgery, the highest AUROC was obtained from the NB model, with a value of 0.784. Conclusions An algorithm was developed that predicts the probability of RCC recurrence within 5 and 10 years using the KORCC database, a large-scale RCC cohort in Korea. It is expected that the developed algorithm will help clinicians manage prognosis and establish customized treatment strategies for patients with RCC after surgery.
Collapse
Affiliation(s)
- HyungMin Kim
- Department of Medical Informatics, College of Medicine, The Catholic University, Seoul, Republic of Korea.,Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University, Seoul, Republic of Korea
| | - Sun Jung Lee
- Department of Medical Informatics, College of Medicine, The Catholic University, Seoul, Republic of Korea.,Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University, Seoul, Republic of Korea
| | - So Jin Park
- Department of Medical Informatics, College of Medicine, The Catholic University, Seoul, Republic of Korea.,Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University, Seoul, Republic of Korea
| | - In Young Choi
- Department of Medical Informatics, College of Medicine, The Catholic University, Seoul, Republic of Korea.,Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University, Seoul, Republic of Korea
| | - Sung-Hoo Hong
- Department of Urology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University, Seoul, Republic of Korea
| |
Collapse
|
25
|
EDITORIAL COMMENT. Urology 2021; 148:190. [PMID: 33549214 DOI: 10.1016/j.urology.2020.10.060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 10/18/2020] [Indexed: 11/21/2022]
|
26
|
Wong KJ, Shusterman M, Goel S, Negassa A, Lin J, Gartrell BA. Racial Diversity Among Histology of Renal Cell Carcinoma at an Urban Medical Center. Clin Genitourin Cancer 2021; 19:e166-e170. [PMID: 33602582 DOI: 10.1016/j.clgc.2020.12.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 12/18/2020] [Accepted: 12/26/2020] [Indexed: 01/20/2023]
Abstract
BACKGROUND Non-Hispanic blacks (NHB) with renal cell carcinoma (RCC) are more likely to have papillary RCC (pRCC) than non-Hispanic whites (NHW). Data on histologic subtypes in RCC in Hispanics (H) are also sparse. Previous studies have shown that pRCC is more prevalent in NHB than in NHW, but they analyzed predominantly NHW populations. The Montefiore-Einstein Center for Cancer Care (MECC) serves a predominantly NHB and H population in the Bronx, NY. We investigated histologic subtype specific associations with established RCC risk factors in this population. PATIENTS AND METHODS The MECC tumor registry was used to identify patients ≥ 18 years of age treated with partial or radical nephrectomy between January 2000 and December 2015. An institutional software program and individual chart review were used to obtain demographic data (including self-reported race, age, and sex), pathology data, and RCC risk factors (hypertension, diabetes, renal function, weight). Data were modeled by multinomial logistic regression to estimate odds ratios (ORs) and 95% confidence intervals (CIs). RESULTS A total of 1010 RCC cases were identified. Of these, 232 (23.0%) occurred in NHW, 383 (37.9%) NHB, 181 (17.9%) H, and 214 (21.2%) other. A total of 530 cases (52.5%) were clear cell (ccRCC) histology, 257 (25.4%) pRCC, 100 (9.9%) chromophobe (cRCC), and 123 (12.2%) other. Individuals with pRCC compared to ccRCC were more likely to be NHB than NHW (OR, 4.41; 95% CI, 2.81-6.93) but were less likely to be female (OR, 0.50; 95% CI, 0.35-0.72). Individuals with pRCC were also less likely to be H than NHW (OR, 0.52; 95% CI, 0.27-0.99). Patients with cRCC were also more likely to be NHB than NHW (OR, 2.23; 95% CI, 1.06-4.67). CONCLUSION In the MECC data set, histology of RCC varies by race, confirming earlier reports that non-ccRCC is more common in NHB than NHW. We also report that pRCC is less common in H than NHW.
Collapse
Affiliation(s)
- Kevin Junmun Wong
- Department of Oncology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY
| | - Michael Shusterman
- Department of Oncology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY
| | - Sanjay Goel
- Department of Oncology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY
| | - Abdissa Negassa
- Department of Epidemiology & Population, Albert Einstein College of Medicine, Bronx, NY
| | - Juan Lin
- Department of Epidemiology & Population, Albert Einstein College of Medicine, Bronx, NY
| | - Benjamin Adam Gartrell
- Department of Oncology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY.
| |
Collapse
|
27
|
Chen C, Geng X, Liang R, Zhang D, Sun M, Zhang G, Hou J. Nomograms-based prediction of overall and cancer-specific survivals for patients with chromophobe renal cell carcinoma. Exp Biol Med (Maywood) 2020; 246:729-739. [PMID: 33302735 DOI: 10.1177/1535370220977107] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
This study built and tested two effective nomograms for the purpose of predicting cancer-specific survival and overall survival of chromophobe renal cell carcinoma (chRCC) patients. Multivariate Cox regression analysis was employed to filter independent prognostic factors predictive of cancer-specific survival and overall survival, and the nomograms were built based on a training set incorporating 2901 chRCC patients in a retrospective study (from 2004 to 2015) downloaded from the surveillance, epidemiology, and end results (SEER) database. The nomograms were verified on a validation cohort of 1934 patients, subsequently the performances of the nomograms were examined according to the receiver operating characteristic curve, calibration curves, the concordance (C-index), and decision curve analysis. The results showed that tumor grade, AJCC and N stages, race, marital status, age, histories of chemotherapy, radiotherapy and surgery were the individual prognostic factors for overall survival, and that AJCC, N and SEER stages, histories of surgery, radiotherapy and chemotherapy, age, tumor grade were individual prognostic factors for cancer-specific survival. According to C-indexes, receiver operating characteristic curves, and decision curve analysis outcomes, the nomograms showed a higher accuracy in predicting overall survival and OSS when compared with TNM stage and SEER stage. All the calibration curves were significantly consistent between predictive and validation sets. In this study, the nomograms, which were validated to be highly accurate and applicable, were built to facilitate individualized predictions of the cancer-specific survival and overall survival to patients diagnosed with chRCC between 2004 and 2015.
Collapse
Affiliation(s)
- Chunyang Chen
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou 215006, People's Republic of China
| | - Xinyu Geng
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou 215006, People's Republic of China
| | - Rui Liang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou 215006, People's Republic of China
| | - Dongze Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou 215006, People's Republic of China.,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou 215006, People's Republic of China
| | - Meiyun Sun
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou 215006, People's Republic of China.,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou 215006, People's Republic of China
| | - Guangbo Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou 215006, People's Republic of China.,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou 215006, People's Republic of China
| | - Jianquan Hou
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou 215006, People's Republic of China
| |
Collapse
|
28
|
Zhang B, Chu W, Wen F, Zhang L, Sun L, Hu B, Wang J, Su Q, Mei Y, Cao J, Zheng J, Mou X, Dong H, Lin X, Wang N, Ji H. Dysregulation of Long Non-coding RNAs and mRNAs in Plasma of Clear Cell Renal Cell Carcinoma Patients Using Microarray and Bioinformatic Analysis. Front Oncol 2020; 10:559730. [PMID: 33330027 PMCID: PMC7729199 DOI: 10.3389/fonc.2020.559730] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 08/17/2020] [Indexed: 11/13/2022] Open
Abstract
Objective: The roles of long non-coding RNAs (lncRNAs) in the diagnosis of clear cell renal cell carcinoma (ccRCC) are still not well-defined. We aimed to identify differentially expressed lncRNAs and mRNAs in plasma of ccRCC patients and health controls systematically. Methods: Expression profile of plasma lncRNAs and mRNAs in ccRCC patients and healthy controls was analyzed based on microarray assay. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway-based approaches were used to investigate biological function and signaling pathways mediated by the differentially expressed mRNAs. SOCS2-AS1 was selected for validation using Real-Time PCR. The differentially expressed lncRNAs and mRNAs were further compared with E-MTAB-1830 datasets using Venn and the NetworkAnalyst website. The GEPIA and ULCAN websites were utilized for the evaluation of the expression level of differentially expressed mRNA and their association with overall survival (OS). Results: A total of 3,664 differentially expressed lncRNAs were identified in the plasma of ccRCC patients, including 1,511 up-regulated and 2,153 down-regulated lncRNAs (fold change ≥2 and P < 0.05), respectively. There were 2,268 differentially expressed mRNAs, including 932 up-regulated mRNAs and 1,336 down-regulated mRNAs, respectively (fold change ≥2 and P < 0.05). Pathway analysis based on deregulated mRNAs was mainly involved in melanogenesis and Hippo signaling pathway (P < 0.05). In line with the lncRNA microarray findings, the SOCS2-AS1 was down-regulated in ccRCC plasma and tissues, as well as in cell lines. Compared with the E-MTAB-1830 gene expression profiles, we identified 18 lncRNAs and 87 mRNAs differently expressed in both plasma and neoplastic tissues of ccRCC. The expression of 10 mRNAs (EPB41L4B, CCND1, GGT1, CGNL1, CYSLTR1, PLAUR, UGT3A1, PROM2, MUC12, and PCK1) was correlated with the overall survival (OS) rate in ccRCC patients based on the GEPIA and ULCAN websites. Conclusions: We firstly reported differentially expressed lncRNAs in ccRCC patients and healthy controls systemically. Several differentially expressed lncRNAs and mRNAs were identified, which might serve as diagnostic or prognostic markers. The biological function of these lncRNAs and mRNAs should be further validated. Our study may contribute to the future treatment of ccRCC and provide novel insights into cancer biology.
Collapse
Affiliation(s)
- Bing Zhang
- Department of Urology, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou, China
| | - Wei Chu
- Department of Pathology, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou, China
| | - Feifei Wen
- Department of Pathology, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou, China
| | - Li Zhang
- Department of Anesthesiology, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou, China
| | - Lixia Sun
- Department of Pathology, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou, China
| | - Baoguang Hu
- Department of Gastrointestinal Surgery, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou, China
| | - Jingjing Wang
- Department of Pathology, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou, China
| | - Qingguo Su
- Department of Urology, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou, China
| | - Yanhui Mei
- Department of Urology, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou, China
| | - Jingyuan Cao
- Department of Urology, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou, China
| | - Jing Zheng
- Clinical Medicine Laboratory, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou, China
| | - Xiaodong Mou
- Department of Pathology, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou, China
| | - Hongliang Dong
- Clinical Medicine Laboratory, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou, China
| | - Xiaoyan Lin
- Department of Pathology, Shandong Province Hospital, Jinan, China
| | - Nan Wang
- Clinical Medicine Laboratory, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou, China
| | - Hong Ji
- Department of Pathology, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou, China
| |
Collapse
|
29
|
Dizman N, Salgia NJ, Bergerot PG, Hsu J, Ruel N, Pal SK. Race/Ethnicity and Survival in Metastatic Renal Cell Carcinoma: Outcomes for Patients Receiving First Line Targeted Therapies. KIDNEY CANCER 2020. [DOI: 10.3233/kca-200092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND: No study to date has assessed the relationship between treatment-specific therapeutic outcomes and race/ethnicity in metastatic renal cell carcinoma (mRCC). As targeted therapies have formed the backbone of first-line treatment options for mRCC until very recently, we assessed the relationship between race/ethnicity and targeted therapy-related outcomes in mRCC. OBJECTIVE: To retrospectively compare response rates and survival outcomes across ethnicities in patients who received first-line targeted therapies for mRCC. METHODS: Patients with mRCC receiving a first-line targeted therapy were identified from an institutional database encompassing consecutive patients treated between 2009 and 2019. Patient demographics, clinical characteristics and survival outcomes were recorded. The racial/ethnic groups included for analysis were Caucasian American, Hispanic American, and Asian American. Survival and response outcomes including progression-free survival (PFS), overall survival (OS), objective response rate (ORR) and disease control rate (DCR) were calculated and compared across ethnic groups using Kaplan-Meier method and Chi-square test, respectively. RESULTS: In total, 295 patients were included for analysis. There were 184 (62.4%) Caucasian American patients, 82 (27.8%) Hispanic American patients, and 29 (9.8%) Asian American patients. No statistically significant differences in PFS nor OS were found between groups (PFS: 5.6 vs. 4.7 vs. 4.7 months, respectively) (OS: 32 vs. 31.7 vs. 51.7 months, respectively). No significant difference was found in ORR nor DCR across groups. Univariate cox regression analyses demonstrated no independent effect of race/ethnicity on PFS or OS. CONCLUSIONS: The apparent lack of differences in treatment-related outcomes across racial/ethnic groups is encouraging. However, further validation is required in larger series.
Collapse
Affiliation(s)
- Nazli Dizman
- Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Nicholas J. Salgia
- Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Paulo G. Bergerot
- Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - JoAnn Hsu
- Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Nora Ruel
- Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Sumanta K. Pal
- Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| |
Collapse
|
30
|
Sun DE, Ye SY. Emerging Roles of Long Noncoding RNA Regulator of Reprogramming in Cancer Treatment. Cancer Manag Res 2020; 12:6103-6112. [PMID: 32765105 PMCID: PMC7382586 DOI: 10.2147/cmar.s253042] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 06/09/2020] [Indexed: 12/17/2022] Open
Abstract
Despite numerous advances in cancer treatment, the global prevalence and cancer-related mortality remain high. Understanding tumor initiation and progression mechanisms are critical as it will lead to the development of interventions for improving the prognosis of cancer patients. The roles of long noncoding RNAs (lncRNAs) in cancer have attracted immense research interest. Growing evidence indicates that lncRNA regulator of reprogramming (linc-ROR), a well-studied RNA, regulates the progression of various cancers, such as lung cancer (LC), hepatocellular carcinoma (HCC), breast cancer (BC), colorectal cancer (CRC), pancreatic cancer (PC), papillary thyroid carcinoma (PTC), or esophageal squamous cell carcinoma (ESCC). linc-ROR promotes the proliferation, invasion, migration and chemoresistance of cancer cells. Herein, we reviewed current literature on the modulatory functions and mechanisms of linc-ROR in cancer development. We highlight new linc-ROR-related therapeutic strategies in cancer treatment.
Collapse
Affiliation(s)
- Di-Er Sun
- Clinical Laboratory, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang, People's Republic of China.,Clinical Laboratory, Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, People's Republic of China
| | - Shu-Yuan Ye
- Clinical Laboratory, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang, People's Republic of China.,Clinical Laboratory, Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, People's Republic of China
| |
Collapse
|
31
|
Sekino Y, Han X, Babasaki T, Miyamoto S, Kitano H, Kobayashi G, Goto K, Inoue S, Hayashi T, Teishima J, Sakamoto N, Sentani K, Oue N, Yasui W, Matsubara A. TUBB3 Is Associated with High-Grade Histology, Poor Prognosis, p53 Expression, and Cancer Stem Cell Markers in Clear Cell Renal Cell Carcinoma. Oncology 2020; 98:689-698. [PMID: 32585672 DOI: 10.1159/000506775] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 02/21/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND βIII-Tubulin, encoded by the TUBB3 gene, is a microtubule protein. Several studies have shown that overexpression of TUBB3 is linked to poor prognosis and is involved in taxane resistance in some cancers. OBJECTIVE The aim of this study was to analyze the expression and function of TUBB3 in clear cell renal cell carcinoma (ccRCC). METHODS The expression of TUBB3 was determined using immuno-histochemistry in ccRCC specimens. The effects of TUBB3 knockdown on cell growth and invasion were evaluated in RCC cell lines. We analyzed the interaction between TUBB3, p53, cancer stem cell markers, and PD-L1. RESULTS In 137 cases of ccRCC, immunohistochemistry showed that 28 (20%) of the ccRCC cases were positive for TUBB3. High TUBB3 expression was significantly correlated with high nuclear grade, high T stage, and N stage. A Kaplan-Meier analysis showed that high expression of TUBB3 was associated with poor overall survival after nephrectomy. In silico analysis also showed that high TUBB3 expression was correlated with overall survival. Knockdown of TUBB3 suppressed cell growth and invasion in 786-O and Caki-1 cells. High TUBB3 expression was associated with CD44, CD133, PD-L1, and p53 in ccRCC. We generated p53 knockout cells using the CRISPR-Cas9 system. Western blotting revealed that p53 knockout upregulated the expression of TUBB3. CONCLUSION These results suggest that TUBB3 may play an oncogenic role and could be a potential therapeutic target in ccRCC.
Collapse
Affiliation(s)
- Yohei Sekino
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan,
| | - Xiangrui Han
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takashi Babasaki
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shunsuke Miyamoto
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hiroyuki Kitano
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Go Kobayashi
- Department of Molecular Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Keisuke Goto
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shogo Inoue
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Tetsutaro Hayashi
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Jun Teishima
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Naoya Sakamoto
- Department of Molecular Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kazuhiro Sentani
- Department of Molecular Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Naohide Oue
- Department of Molecular Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Wataru Yasui
- Department of Molecular Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Akio Matsubara
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
32
|
Padala SA, Barsouk A, Thandra KC, Saginala K, Mohammed A, Vakiti A, Rawla P, Barsouk A. Epidemiology of Renal Cell Carcinoma. World J Oncol 2020; 11:79-87. [PMID: 32494314 PMCID: PMC7239575 DOI: 10.14740/wjon1279] [Citation(s) in RCA: 590] [Impact Index Per Article: 118.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 05/04/2020] [Indexed: 01/11/2023] Open
Abstract
Though renal cell carcinoma (RCC) accounts for 2% of global cancer diagnoses and deaths, it has more than doubled in incidence in the developed world over the past half-century, and today is the ninth most common neoplasm in the United States (US). While North America and Western Europe have the highest disease burden (with the Belarus highest in incidence), Latin America, Asia and Africa are projected to see an increase in incidence as nation’s transition to a Western lifestyle. Most cases of RCC are discovered incidentally on imaging, and survival is highly dependent on the stage at diagnosis, with the metastatic disease having only a 12% 5-year survival rate. Two-thirds of RCC diagnoses are made in men, and the average age of diagnosis in the US is 64. Those with genetic predispositions, namely von Hippel-Lindau disease, tend to be diagnosed 20 years earlier. RCC has a greater incidence among Hispanics and Native Americans, and a lower survival rate among African Americans in the US. Modifiable risk factors for RCC include smoking, obesity, poorly-controlled hypertension, diet and alcohol, and occupational exposures. Prevention strategies aimed at improving survival and reducing disparities include addressing lifestyle factors and access to regular healthcare among underserved populations and in developing nations, as well as more rigorous imaging guidelines to detect RCC at an earlier stage. A stronger understanding of global RCC epidemiology can facilitate prevention efforts, especially in developing nations and underserved communities where disease burden is predicted to rise in the coming decades.
Collapse
Affiliation(s)
- Sandeep Anand Padala
- Department of Medicine, Nephrology, Augusta University, Medical College of Georgia, Augusta, GA 30912, USA
| | - Adam Barsouk
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA
| | - Krishna Chaitanya Thandra
- Department of Pulmonary and Critical Care Medicine, Sentara Virginia Beach General Hospital, Virginia Beach, VA, USA
| | - Kalyan Saginala
- Plains Regional Medical Group Internal Medicine, Clovis, NM 88101, USA
| | - Azeem Mohammed
- Department of Medicine, Nephrology, Augusta University, Medical College of Georgia, Augusta, GA 30912, USA
| | - Anusha Vakiti
- Department of Medicine, Hematology-Oncology, Augusta University, Medical College of Georgia, Augusta, GA 30912, USA
| | - Prashanth Rawla
- Department of Medicine, Sovah Health, Martinsville, VA 24112, USA
| | | |
Collapse
|
33
|
Zhou K, Mai H, Zheng S, Cai W, Yang X, Chen Z, Zhan B. OTUB1-mediated deubiquitination of FOXM1 up-regulates ECT-2 to promote tumor progression in renal cell carcinoma. Cell Biosci 2020; 10:50. [PMID: 32257108 PMCID: PMC7106863 DOI: 10.1186/s13578-020-00408-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/13/2020] [Indexed: 12/19/2022] Open
Abstract
Background OTUB1 (ovarian tumor domain protease domain-containing ubiquitin aldehyde-binding proteins)-mediated deubiquitination of FOXM1 (Forkhead box M1) participates in carcinogenesis of various tumors. We aim to investigate the effect and mechanism of OTUB1/FOXM1 on RCC (renal cell carcinoma) progression. Expression levels of OTUB1 in RCC tissues and cell lines were examined by qRT-PCR (quantitative real-time polymerase chain reaction) and immunohistochemistry. Cell proliferation was measured with CCK8 (Cell Counting Kit-8) and colony formation assays. Wound healing and transwell assays were used to determine cell migration and invasion, respectively. The effect of OTUB1 on FOXM1 ubiquitination was examined by Immunoprecipitation. Western blot was used to uncover the underlying mechanism. In vivo subcutaneous xenotransplanted tumor model combined with immunohistochemistry and western blot were used to examine the tumorigenic function of OTUB1. Results OTUB1 was up-regulated in RCC tissues and cell lines, and was associated with poor prognosis of RCC patients. Knockdown of OTUB1 inhibited cell viability and proliferation, as well as migration and invasion of RCC cells. Mechanistically, knockdown of OTUB1 down-regulated FOXM1 expression by promoting its ubiquitination. Down-regulation of FOXM1 inhibited ECT2 (epithelial cell transforming 2)-mediated Rho signaling. Moreover, the inhibition of RCC progression caused by OTUB1 knockdown was reversed by FOXM1 over-expression. In vivo subcutaneous xenotransplanted tumor model also revealed that knockdown of OTUB1 could suppress in vivo RCC growth via down-regulation of FOXM1-mediated ECT2 expression. Conclusions OTUB1-mediated deubiquitination of FOXM1 up-regulates ECT-2 to promote tumor progression in RCC, providing a new potential therapeutic target for RCC treatment.
Collapse
Affiliation(s)
- Kai Zhou
- 1Department of Urology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001 Fujian China
| | - Haixing Mai
- 2Department of Urology, Chinese PLA General Hospital, Beijing, 100853 China
| | - Song Zheng
- 1Department of Urology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001 Fujian China
| | - Weizhong Cai
- 1Department of Urology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001 Fujian China
| | - Xu Yang
- 1Department of Urology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001 Fujian China
| | - Zhenlin Chen
- 1Department of Urology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001 Fujian China
| | - Bin Zhan
- 1Department of Urology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001 Fujian China
| |
Collapse
|
34
|
Racial and ethnic differences in survival in contemporary metastatic renal cell carcinoma patients, according to alternative treatment modalities. Cancer Causes Control 2020; 31:263-272. [PMID: 31993859 DOI: 10.1007/s10552-020-01270-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 01/21/2020] [Indexed: 10/25/2022]
Abstract
PURPOSE To test the association between African-American race and overall mortality (OM) rates in patients with metastatic renal cell carcinoma (mRCC). METHODS Within the Surveillance, Epidemiology, and End Results registry (2006-2015), we identified patients with clear cell (ccmRCC) and non-clear cell mRCC (non-ccmRCC). African-Americans, Caucasians, and Hispanics were identified. Stratification was made according to histology and treatments: (1) no treatment, (2) systemic therapy (ST), (3) cytoreductive nephrectomy (CNT), (4) CNT + ST. Kaplan-Meier plots and multivariable Cox regression analyses were used. RESULTS Of ccmRCC patients, 410 (7%), 4353 (75%), and 1005 (17%) were African-American, Caucasian, and Hispanic, respectively. Of non-ccmRCC patients, 183 (25%), 479 (65%), and 77 (10%) were African-American, Caucasian, and Hispanic, respectively. In ccmRCC, African-Americans were associated with higher OM rates (HR 1.20; 95% CI 1.05-1.37). Conversely, in non-ccmRCC, African-Americans were associated with lower OM rates (HR 0.75; 95% CI 0.59-0.97). CONCLUSION African-American race is associated with prolonged survival in non-ccmRCC, but it is also associated with lower survival rates in ccmRCC. The exception to these observations consisted of patients treated with combination of CNT + ST for either ccmRCC or non-ccmRCC.
Collapse
|
35
|
Yeong J, Zhao Z, Lim JCT, Li H, Thike AA, Koh VCY, Teh BT, Kanesvaran R, Toh CK, Tan PH, Khor LY. PD-L1 expression is an unfavourable prognostic indicator in Asian renal cell carcinomas. J Clin Pathol 2020; 73:463-469. [PMID: 31980560 DOI: 10.1136/jclinpath-2019-206092] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/25/2019] [Accepted: 11/26/2019] [Indexed: 01/15/2023]
Abstract
BACKGROUND/AIMS The programmed cell death receptor 1 (PD-1) checkpoint inhibitor, nivolumab, has been approved for the treatment of metastatic renal cell carcinoma (RCC). However, the understanding of the expression and distribution of PD ligand 1 (PD-L1) in the tumour immune microenvironment and its prognostic role in an Asian cohort is limited. Our group investigated PD-L1 protein expression in a cohort of Asian patients with RCC of mixed ethnicity, using two commercially available antibody clones. METHODS E1L3N and SP263 anti-PD-L1 clones were used to categorise RCCs of various histological subtypes, diagnosed at our institution between 1995 and 2008, into PD-L1-positive or PD-L1-negative groups, based on a 1% Tumour Proportion Score (TPS) cut-off. RESULTS In total, 267 (83%) clear cell (cc)RCC and 55 (17%) non-ccRCC cases were studied. Overall PD-L1 protein expression rates for the entire cohort were 13% and 8% for the E1L3N and SP263 clones, respectively. Patients bearing PD-L1-positive tumours experienced significantly decreased disease-free survival (DFS; E1L3N: p=0.01; SP263: p=0.03) but not overall survival, compared with those with PD-L1-negative tumours. Multivariate survival analysis further confirmed the results of the E1L3N clone (HR 1.85, 95% CI 1.10 to 3.13, p=0.02), but not SP263, after adjusting for pathological stage, histological subtype and grade. The addition of PD-L1 (E1L3N) TPS to clinicopathological features significantly increased the prognostic value for DFS (∆LRχ2=5.25; p=0.022), compared with clinicopathological features alone. CONCLUSIONS PD-L1 protein expression was associated with an unfavourable prognosis in our study cohort. PD-L1 (E1L3N) expression was an independent prognostic indicator of clinical outcome in all RCCs when using a 1% cut-off.
Collapse
Affiliation(s)
- Joe Yeong
- Institute of Molecular Cell Biology (IMCB), Agency of Science, Technology and Research (A*STAR), Singapore.,Anatomical Pathology, Singapore General Hospital, Singapore
| | - Zitong Zhao
- Anatomical Pathology, Singapore General Hospital, Singapore
| | - Jeffrey Chun Tatt Lim
- Institute of Molecular Cell Biology (IMCB), Agency of Science, Technology and Research (A*STAR), Singapore.,Pathology, Singapore General Hospital, Singapore
| | - Huihua Li
- Center for Quantitative Medicine, Duke-NUS Medical School, Singapore.,Division of Medicine, Singapore General Hospital, Singapore
| | | | | | - Bin Tean Teh
- Laboratory of Cancer Epigenome, National Cancer Centre, Singapore.,Division of Cancer and Stem Cell Biology, Duke-NUS Graduate Medical School, Singapore
| | - Ravindran Kanesvaran
- Dean's Office, Duke-NUS Medical School, Singapore.,National Cancer Centre Singapore, Singapore
| | | | - Puay Hoon Tan
- Anatomical Pathology, Singapore General Hospital, Singapore.,Pathology, Singapore General Hospital, Singapore.,Dean's Office, Duke-NUS Medical School, Singapore
| | - Li Yan Khor
- Anatomical Pathology, Singapore General Hospital, Singapore .,Dean's Office, Duke-NUS Medical School, Singapore
| |
Collapse
|
36
|
Park KH, Yoon JA, Kim HS, Kim H, Park SK, Kim YH, Hong B, You D, Jeong IG, Baek CH. Clinical features and outcomes in kidney transplant recipients with renal cell carcinoma: a single-center study. Kidney Res Clin Pract 2019; 38:517-524. [PMID: 31826389 PMCID: PMC6913591 DOI: 10.23876/j.krcp.19.078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/24/2019] [Accepted: 10/27/2019] [Indexed: 01/20/2023] Open
Abstract
Background Previous studies have recommended a 2- to 5-year waiting time prior to kidney transplantation (KT) in patients with end-stage renal disease (ESRD) and symptomatic renal cell carcinoma (RCC) and no delay for incidental early-stage RCC. Data on Asian KT recipients are unavailable. Methods This is a Korean single-center retrospective study on 35 KT recipients with ESRD and RCC. Patients were classified into two groups: early KT (KT performed within 1 year after nephrectomy for RCC, including KT with simultaneous nephrectomy) and delayed KT (KT performed over than 1 year after nephrectomy for RCC). Patient survival, graft survival, and cancer recurrence were compared between both groups. Results There were no statistically significant differences in patient survival (P = 0.388), graft survival (P = 0.317), or graft rejection rate (P = 0.207) between the early and delayed KT groups. Additionally, there were no differences in pathological characteristics or RCC stage other than cancer histology: acquired cystic disease-associated RCC (47.4%) was the most common RCC type in the early KT group, whereas clear cell type (62.5%) was the most common RCC type in the delayed KT group. No RCC recurrence was observed. Conclusion Patients with early-stage and asymptomatic RCC do not require a mandatory observational period prior to KT after curative nephrectomy.
Collapse
Affiliation(s)
- Keun Hoi Park
- Division of Nephrology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Rebublic of Korea
| | - Jung A Yoon
- Division of Nephrology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Rebublic of Korea
| | - Hak Soo Kim
- Division of Nephrology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Rebublic of Korea
| | - Hyosang Kim
- Division of Nephrology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Rebublic of Korea
| | - Su-Kil Park
- Division of Nephrology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Rebublic of Korea
| | - Young Hoon Kim
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Rebublic of Korea
| | - Bumsik Hong
- Department of Urology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Rebublic of Korea
| | - Dalsan You
- Department of Urology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Rebublic of Korea
| | - In Gab Jeong
- Department of Urology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Rebublic of Korea
| | - Chung Hee Baek
- Division of Nephrology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Rebublic of Korea
| |
Collapse
|
37
|
Wang A, Chen M, Wang H, Huang J, Bao Y, Gan X, Liu B, Lu X, Wang L. Cell Adhesion-Related Molecules Play a Key Role in Renal Cancer Progression by Multinetwork Analysis. BIOMED RESEARCH INTERNATIONAL 2019; 2019:2325765. [PMID: 31950034 PMCID: PMC6948336 DOI: 10.1155/2019/2325765] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 08/23/2019] [Indexed: 12/24/2022]
Abstract
Renal cell carcinoma (RCC) is one of the most common malignancies in the urinary system. The study aimed to identify genetic characteristics and reveal the underlying mechanisms in RCC. GSE53757, GSE46699, and TCGA KIRC database (n = 897) were analyzed to screen differentially expressed genes (DEGs) in RCC. The gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed, followed by the analysis of the protein-protein interaction (PPI) network of the DEGs by Cytoscape software. In all, 834 DEGs were identified in RCC, including 416 upregulated genes and 418 downregulated genes. The top 10 hub genes, VEGFA, EGFR, EGF, CD44, CD86, FN1, ITGAM, ITGB2, TLR2, and PTPRC, were identified from the PPI network according to the core degree. The following subnetwork revealed that these significant modules were enriched in positive regulation of response to external stimulus, regulation of leukocyte-mediated immunity, and regulation of exocytosis. The expressions of these hub genes were also validated using qRT-PCR and IHC in Changzheng RCC database (n = 160). We especially found that half of the top ten hub genes were cell adhesion-related molecules, which were associated with RCC progression and poor prognosis. In conclusion, these hub genes, particularly cell adhesion-related molecules, could be used as prognostic biomarkers and potential therapeutic targets for RCC.
Collapse
Affiliation(s)
- Anbang Wang
- Department of Urology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Ming Chen
- Department of Urology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Hui Wang
- Department of Urology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Jinming Huang
- Department of Urology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Yi Bao
- Department of Urology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Xinxin Gan
- Department of Urology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Bing Liu
- Department of Urology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Xin Lu
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Linhui Wang
- Department of Urology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| |
Collapse
|
38
|
Batai K, Harb‐De la Rosa A, Zeng J, Chipollini JJ, Gachupin FC, Lee BR. Racial/ethnic disparities in renal cell carcinoma: Increased risk of early-onset and variation in histologic subtypes. Cancer Med 2019; 8:6780-6788. [PMID: 31509346 PMCID: PMC6826053 DOI: 10.1002/cam4.2552] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 08/23/2019] [Accepted: 08/28/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Racial/ethnic minority groups have a higher burden of renal cell carcinoma (RCC), but RCC among Hispanic Americans (HAs) and American Indians and Alaska Natives (AIs/ANs) are clinically not well characterized. We explored variations in age at diagnosis and frequencies of RCC histologic subtypes across racial/ethnic groups and Hispanic subgroups using National Cancer Database (NCDB) and Arizona Cancer Registry Data. METHODS Adult RCC cases with known race/ethnicity were included. Logistic regression analysis was performed to estimate odds and 95% confidence interval (CI) of early-onset (age at diagnosis <50 years) and diagnosis with clear cell RCC (ccRCC) or papillary RCC. RESULTS A total of 405 073 RCC cases from NCDB and 9751 cases from ACR were identified and included. In both datasets, patients from racial/ethnic minority groups had a younger age at diagnosis than non-Hispanic White (NHW) patients. In the NCDB, AIs/ANs had twofold increased odds (OR, 2.21; 95% CI, 1.88-2.59) of early-onset RCC compared with NHWs. HAs also had twofold increased odds of early-onset RCC (OR, 2.14; 95% CI, 1.79-2.55) in the ACR. In NCDB, ccRCC was more prevalent in AIs (86.3%) and Mexican Americans (83.5%) than NHWs (72.5%). AIs/ANs had twofold increased odds of diagnosis with ccRCC (OR, 2.18; 95% CI, 1.85-2.58) in the NCDB, but the association was stronger in the ACR (OR, 2.83; 95% CI, 2.08-3.85). Similarly, Mexican Americans had significantly increased odds of diagnosis with ccRCC (OR, 2.00; 95% CI, 1.78-2.23) in the NCDB. CONCLUSIONS This study reports younger age at diagnosis and higher frequencies of ccRCC histologic subtype in AIs/ANs and Hispanic subgroups. These variations across racial/ethnic groups and Hispanic subgroups may have potential clinical implications.
Collapse
Affiliation(s)
- Ken Batai
- Department of UrologyUniversity of ArizonaTucsonArizona
| | | | - Jiping Zeng
- Department of UrologyUniversity of ArizonaTucsonArizona
| | | | | | | |
Collapse
|
39
|
Gorczynski A, Czapiewski P, Korwat A, Budynko L, Prelowska M, Okon K, Biernat W. ALK-rearranged renal cell carcinomas in Polish population. Pathol Res Pract 2019; 215:152669. [PMID: 31677810 DOI: 10.1016/j.prp.2019.152669] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 09/22/2019] [Accepted: 09/24/2019] [Indexed: 12/24/2022]
Abstract
Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase, the activation of which is considered an important event in the pathogenesis of several neoplasms and a predictive factor for the targeted therapy with ALK inhibitors. Thus far, ALK rearrangements have been identified in 22 renal cell carcinomas in both pediatric and adult patients. We evaluated the incidence of ALK rearrangement-associated RCC in adult Central European population. An immunohistochemical evaluation of 1019 kidney tumors was performed with use of three different clones of anti-ALK antibodies. None of the tested samples showed positive staining, which suggests that the incidence of ALK rearrangement-associated renal cell carcinomas is significantly lower in the Polish population, and indicates a potential association between ethnicity and occurrence of these rare neoplasms.
Collapse
Affiliation(s)
- Adam Gorczynski
- Department of Pathomorphology, Medical University of Gdansk, Mariana Smoluchowskiego 17, 80-214 Gdansk, Poland.
| | - Piotr Czapiewski
- Department of Pathomorphology, Medical University of Gdansk, Mariana Smoluchowskiego 17, 80-214 Gdansk, Poland.
| | - Aleksandra Korwat
- Department of Pathomorphology, Medical University of Gdansk, Mariana Smoluchowskiego 17, 80-214 Gdansk, Poland.
| | - Lukasz Budynko
- Department of Pathomorphology, Medical University of Gdansk, Mariana Smoluchowskiego 17, 80-214 Gdansk, Poland.
| | - Monika Prelowska
- Department of Pathomorphology, Medical University of Gdansk, Mariana Smoluchowskiego 17, 80-214 Gdansk, Poland.
| | - Krzysztof Okon
- Department of Pathomorphology, Jagiellonian University, Grzegorzecka 16, 33-332, Krakow, Poland.
| | - Wojciech Biernat
- Department of Pathomorphology, Medical University of Gdansk, Mariana Smoluchowskiego 17, 80-214 Gdansk, Poland.
| |
Collapse
|
40
|
Race and renal cell carcinoma stage at diagnosis: an analysis of the Surveillance, Epidemiology, and End Results data. Eur J Cancer Prev 2019; 28:350-354. [DOI: 10.1097/cej.0000000000000484] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
41
|
Carlo MI, Khan N, Zehir A, Patil S, Ged Y, Redzematovic A, Coskey DT, Hyman DM, Ladanyi M, Chen YB, Robson M, Hakimi AA, Lee CH, Feldman DR, Gao J, Chakravarty D, Motzer RJ, Voss MH. Comprehensive Genomic Analysis of Metastatic Non-Clear-Cell Renal Cell Carcinoma to Identify Therapeutic Targets. JCO Precis Oncol 2019; 3:1800372. [PMID: 32914011 DOI: 10.1200/po.18.00372] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/27/2018] [Indexed: 12/27/2022] Open
Abstract
PURPOSE Non-clear-cell renal cell carcinoma (nccRCC) encompasses approximately 20% of renal cell carcinomas and includes subtypes that vary in clinical and molecular biology. Compared with clear cell renal cell carcinoma, nccRCC demonstrates limited sensitivity to conventional vascular endothelial growth factor- and mammalian target of rapamycin-directed agents, indicating a need for better therapies. Characterizing the genomic landscape of metastatic nccRCC variants may help define novel therapeutic strategies. PATIENTS AND METHODS We retrospectively analyzed tumor tissue from patients with metastatic nccRCC who consented to genomic analysis of their tumor and germline DNA. A hybridization capture-based assay was used to identify single nucleotide variants and small insertions and deletions across more than 340 cancer-associated genes with germline comparison. Clinical actionability of somatic mutations was assessed using OncoKB levels of evidence. Microsatellite instability (MSI) in the tumor was investigated. RESULTS Of 116 patients included in the analysis, 57 (49%) presented with de novo metastatic disease, and 59 (51%) presented with localized disease that later metastasized. Subtype classifications included unclassified (n = 41; 35%), papillary (n = 26; 22%), chromophobe (n = 17; 15%), translocation associated (n = 13; 11%), and other (n = 19; 16%). Of all tumors, 15 (13%) had putative driver somatic alterations amenable to targeted therapies, including alterations in MET, TSC1/2, and an ALK translocation. Of 45 patients who had germline testing, 11 (24%) harbored mutations, seven of which could potentially guide therapy. Of 115 available tumors for analysis, two (1.7%) had high and six (5%) had intermediate MSI status. CONCLUSION The mutation profiles of metastatic nccRCC vary by subtype. Comprehensive analysis of somatic mutations, germline mutations, and MSI, interpreted via an annotated precision oncology knowledge base, identified potentially targetable alterations in 22% of patients, which merits additional investigation.
Collapse
Affiliation(s)
- Maria I Carlo
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Nabeela Khan
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ahmet Zehir
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sujata Patil
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Yasser Ged
- Memorial Sloan Kettering Cancer Center, New York, NY
| | | | | | - David M Hyman
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Marc Ladanyi
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ying-Bei Chen
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Mark Robson
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - A Ari Hakimi
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Chung-Han Lee
- Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Jianjiong Gao
- Memorial Sloan Kettering Cancer Center, New York, NY
| | | | | | - Martin H Voss
- Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
42
|
Harris WB. Epidemiology of Renal Cell Carcinoma and Its Predisposing Risk Factors. Urol Oncol 2019. [DOI: 10.1007/978-3-319-42623-5_55] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
43
|
Callahan CL, Hofmann JN, Corley DA, Zhao WK, Shuch B, Chow WH, Purdue MP. Obesity and renal cell carcinoma risk by histologic subtype: A nested case-control study and meta-analysis. Cancer Epidemiol 2018; 56:31-37. [PMID: 30029068 DOI: 10.1016/j.canep.2018.07.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/28/2018] [Accepted: 07/06/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Although obesity is an established risk factor for renal cell carcinoma (RCC), it is unclear whether this relationship varies across histologic subtypes. METHODS We conducted a nested case-control study within the Kaiser Permanente Northern California (KPNC) health care network, and meta-analysis combining our results with those of previously published studies. Our KPNC study included 685 RCC cases [421 clear cell; 65 papillary; 24 chromophobe; 35 other; 141 not otherwise specified (NOS)] and 4266 controls. Subtype-specific odds ratios (ORs) and 95% confidence intervals (CIs) for categories of body mass index (BMI) and were computed from the case-control data using polytomous logistic regression. Findings from this and other relevant studies were combined by meta-analysis using a random effects model. RESULTS In the KPNC study, obesity (BMI ≥ 30 kg/m2) was associated with clear cell RCC (OR 1.5, 95% CI 1.1-2.1) and chromophobe RCC (OR 2.5, 95%CI 0.8-8.1), but not with papillary RCC (OR 1.0, 95% CI 0.5-1.9). In meta-analysis including three additional studies, a similar pattern of summary relative risks (SRR) for obesity was observed across subtypes (clear cell: SRR 1.8, 95% CI 1.5-2.2; chromophobe: SRR 2.2, 95% CI 1.3-3.7; papillary, SRR 1.2, 95% CI 0.8-1.6). CONCLUSIONS These findings support the hypothesis that histologic subtypes of RCC possess distinct etiologic pathways, with obesity important for the development of clear cell and, possibly, chromophobe RCC, but not papillary RCC.
Collapse
Affiliation(s)
- Catherine L Callahan
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, United States
| | - Jonathan N Hofmann
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, United States
| | - Douglas A Corley
- Division of Research, Kaiser Permanente Northern California, Oakland, California, United States
| | - Wei K Zhao
- Division of Research, Kaiser Permanente Northern California, Oakland, California, United States
| | - Brian Shuch
- Department of Urology, Yale School of Medicine, New Haven, Connecticut, United States
| | - Wong-Ho Chow
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | - Mark P Purdue
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, United States.
| |
Collapse
|
44
|
Marchioni M, Harmouch SS, Nazzani S, Bandini M, Preisser F, Tian Z, Kapoor A, Cindolo L, Briganti A, Shariat SF, Schips L, Karakiewicz PI. Effect of African-American race on cancer specific mortality differs according to clear cell vs. non-clear cell histologic subtype in metastatic renal cell carcinoma. Cancer Epidemiol 2018; 54:112-118. [DOI: 10.1016/j.canep.2018.04.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 03/21/2018] [Accepted: 04/13/2018] [Indexed: 11/16/2022]
|
45
|
Sims JN, Yedjou CG, Abugri D, Payton M, Turner T, Miele L, Tchounwou PB. Racial Disparities and Preventive Measures to Renal Cell Carcinoma. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2018; 15:E1089. [PMID: 29843394 PMCID: PMC6024978 DOI: 10.3390/ijerph15061089] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 04/27/2018] [Accepted: 05/01/2018] [Indexed: 02/06/2023]
Abstract
Kidney cancer ranks among the top 10 cancers in the United States. Although it affects both male and female populations, it is more common in males. The prevalence rate of renal cell carcinoma (RCC), which represents about 85% of kidney cancers, has been increasing gradually in many developed countries. Family history has been considered as one of the most relevant risk factors for kidney cancer, although most forms of an inherited predisposition for RCC only account for less than four percent. Lifestyle and other factors such as occupational exposure, high blood pressure, poor diet, and heavy cigarette smoking are highly associated with its incidence and mortality rates. In the United States, White populations have the lowest prevalence of RCC compared to other ethnic groups, while Black Americans suffer disproportionally from the adverse effects of RCC. Hence, this review article aims at identifying the major risk factors associated with RCC and highlighting the new therapeutic approaches for its control/prevention. To achieve this specific aim, articles in peer-reviewed journals with a primary focus on risk factors related to kidney cancer and on strategies to reduce RCC were identified. The review was systematically conducted by searching the databases of MEDLINE, PUBMED Central, and Google Scholar libraries for original articles. From the search, we found that the incidence and mortality rates of RCC are strongly associated with four main risk factors, including family history (genetics), lifestyle (poor diet, cigarette smoking, excess alcohol drinking), environment (community where people live), and occupation (place where people work). In addition, unequal access to improvement in RCC cancer treatment, limited access to screening and diagnosis, and limited access to kidney transplant significantly contribute to the difference observed in survival rate between African Americans and Caucasians. There is also scientific evidence suggesting that some physicians contribute to racial disparities when performing kidney transplant among minority populations. New therapeutic measures should be taken to prevent or reduce RCC, especially among African Americans, the most vulnerable population group.
Collapse
Affiliation(s)
- Jennifer N Sims
- Department of Behavioral and Environmental Health, School of Public Health, Jackson State University, 350 W. Woodrow Wilson Dr., P.O. Box 17038, Jackson, MS 39217, USA.
| | - Clement G Yedjou
- Department of Biology, College of Science, Engineering and Technology, Jackson State University, 1400 Lynch St., Jackson, MS 39217, USA.
- Natural Chemotherapeutics Research Laboratory, RCMI Center for Environmental Health, Jackson State University, 1400 Lynch St., Jackson, MS 39217, USA.
| | - Daniel Abugri
- Department of Chemistry and Department of Biology, Laboratory of Ethno-Medicine, Parasitology and Drug Discovery, College of Arts and Science, Tuskegee University, 1200 Old Montgomery Road, Tuskegee, AL 36088, USA.
| | - Marinelle Payton
- Department of Behavioral and Environmental Health, School of Public Health, Jackson State University, 350 W. Woodrow Wilson Dr., P.O. Box 17038, Jackson, MS 39217, USA.
| | - Timothy Turner
- Department of Biology, College of Science, Engineering and Technology, Jackson State University, 1400 Lynch St., Jackson, MS 39217, USA.
| | - Lucio Miele
- Department of Genetics, Louisiana State University, Health Sciences Center, School of Medicine, 533 Bolivar St., Room 657, New Orleans, LA 70112, USA.
| | - Paul B Tchounwou
- Natural Chemotherapeutics Research Laboratory, RCMI Center for Environmental Health, Jackson State University, 1400 Lynch St., Jackson, MS 39217, USA.
| |
Collapse
|
46
|
Rao A, Wiggins C, Lauer RC. Survival outcomes for advanced kidney cancer patients in the era of targeted therapies. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:165. [PMID: 29911113 DOI: 10.21037/atm.2018.04.44] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Background Advanced renal cell carcinoma (RCC) results in over 14,000 deaths each year in the United States alone. The therapeutic landscape for advanced RCC changed dramatically with the approval of tyrosine kinase inhibitors (TKI) between 2006 and 2012. A large-scale analysis of survival trends has not been performed in the TKI era to determine their impact on outcomes for advanced RCC patients. Methods The Surveillance, Epidemiology and End-Results (SEER) database was queried for adult patients with advanced RCC diagnosed between 2000 and 2013. Patients were divided into two cohorts based on the year of diagnosis-pre-TKI cohort [2000-2006] and TKI cohort [2007-2013]. Kaplan-Meier survival and multivariate Cox proportional hazards analyses were performed. Results A total of 14,976 patients were included in our study. Median age at diagnosis was 64 years (range, 18-89 years). Median (cancer-specific) overall survival was 10.0 months in the TKI cohort compared with 8.0 months in the pre-TKI cohort, corresponding to a 13% improvement in survival in the TKI area [hazard ratio (HR) for death 0.87; 95% confidence interval (CI), 0.84-0.91, P<0.0001]. Median survival was improved by 2 months for patients with clear-cell RCC histology [HR for death 0.86; 95% CI, 0.84-0.91, P<0.0001]. Patients with non-clear cell RCC had a 25% higher risk of mortality compared with those with clear-cell RCC. Additionally, median survival for non-clear cell RCC patients was not statistically different between the two cohorts (HR for death 0.98; 95% CI, 0.88-1.09, P=0.714). Subgroup analysis showed that elderly patients (age 71 years and above) had a 45% higher risk of death from advanced RCC compared with young patients (aged 18-50 years) [HR for death 1.45; 95% CI, 1.36-1.54, P<0.0001]. Gender and racial disparities in outcomes were also noted with women having a 10% higher risk of death compared with men (HR for death 1.10; 95% CI, 1.06-1.14, P<0.001) and Black patients having a 15% higher risk of death compared with White patients (HR for death 1.15; 95% CI, 1.08-1.23, P<0.0001). Conclusions Our study provides a largest registry-based analysis of survival outcomes in the TKI era. In majority of patients, the survival has improved significantly with the advent of TKIs as standard of care therapy. Survival for patients with non-clear cell RCC is clearly worse than clear-cell RCC and does not appear to have changed with TKIs. Elderly patients, women, and Black patients appear to have worse outcomes and these disparities merit further investigation.
Collapse
Affiliation(s)
- Arpit Rao
- Division of Hematology, Oncology & Transplantation, University of Minnesota, Minnesota, MN, USA
| | - Charles Wiggins
- Department of Internal Medicine, New Mexico Tumor Registry, University of New Mexico, Albuquerque, NM, USA
| | - Richard C Lauer
- Division of Hematology and Oncology, University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
| |
Collapse
|
47
|
Li G, Chong T, Yang J, Li H, Chen H. Kinesin Motor Protein KIFC1 Is a Target Protein of miR-338-3p and Is Associated With Poor Prognosis and Progression of Renal Cell Carcinoma. Oncol Res 2018; 27:125-137. [PMID: 29562961 PMCID: PMC7848269 DOI: 10.3727/096504018x15213115046567] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
KIFC1 (kinesin family member C1) plays a critical role in clustering of extra centrosomes in various cancer cells and thus could be considered as a promising therapeutic target. However, whether KIFC1 is involved in the procession of renal cell carcinoma (RCC) still remains unclear. In this study, we found that KIFC1 was upregulated in RCC tissues and is responsible for RCC tumorigenesis (p < 0.001). The high expression of KIFC1 correlates with aggressive clinicopathologic parameters. Kaplan–Meier analysis suggested that KIFC1 was associated with poor survival prognosis in RCC. Silencing KIFC1 dramatically resulted in inhibition of proliferation, delayed the cell cycle at G2/M phase, and suppressed cell invasion and migration in vitro. The antiproliferative effect of KIFC1 silencing was also observed in xenografted tumors in vivo. miR-338-3p could directly bind to the 3′-untranslated region (3′-UTR) of KIFC1, and ectopic miR-338-3p expression mimicked the inhibitory functions of KIFC1 silencing on RCC cells through inactivation of the PI3K/AKT signaling pathway. Therefore, these results revealed that KIFC1 may be a novel biomarker and an effective therapeutic target for the treatment of RCC.
Collapse
Affiliation(s)
- Gang Li
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| | - Tie Chong
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| | - Jie Yang
- Department of Nursing, Xi'an Beifang Chinese Medicine Skin Disease Hospital, Xi'an, Shaanxi Province, P.R. China
| | - Hongliang Li
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| | - Haiwen Chen
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| |
Collapse
|
48
|
Abdel-Rahman O. Impact of histological subtype on outcomes of renal cell carcinoma patients. J Drug Assess 2018. [DOI: 10.1080/21556660.2018.1439831] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Affiliation(s)
- Omar Abdel-Rahman
- Clinical Oncology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
49
|
Batai K, Bergersen A, Price E, Hynes K, Ellis NA, Lee BR. Clinical and Molecular Characteristics and Burden of Kidney Cancer Among Hispanics and Native Americans: Steps Toward Precision Medicine. Clin Genitourin Cancer 2018; 16:e535-e541. [PMID: 29449090 DOI: 10.1016/j.clgc.2018.01.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 01/16/2018] [Indexed: 01/20/2023]
Abstract
Cancer disparities in Native Americans (NAs) and Hispanic Americans (HAs) vary significantly in terms of cancer incidence and mortality rates across geographic regions. This review reports that kidney and renal pelvis cancers are unevenly affecting HAs and NAs compared to European Americans of non-Hispanic origin, and that currently there is significant need for improved data and reporting to be able to advance toward genomic-based precision medicine for the assessment of such cancers in these medically underserved populations. More specifically, in states along the US-Mexico border, HAs and NAs have higher kidney cancer incidence rates as well as a higher prevalence of kidney cancer risk factors, including obesity and chronic kidney disease. They are also more likely to receive suboptimal care compared to European Americans. Furthermore, they are underrepresented in epidemiologic, clinical, and molecular genomic studies of kidney cancer. Therefore, we maintain that progress in precision medicine for kidney cancer care requires an understanding of various factors among HAs and NAs, including the real kidney cancer burden, variations in clinical care, issues related to access to care, and specific clinical and molecular characteristics.
Collapse
Affiliation(s)
- Ken Batai
- Department of Surgery, Division of Urology, University of Arizona, Tucson, AZ.
| | - Andrew Bergersen
- Department of Surgery, Division of Urology, University of Arizona, Tucson, AZ
| | - Elinora Price
- Department of Surgery, Division of Urology, University of Arizona, Tucson, AZ
| | - Kieran Hynes
- Department of Surgery, Division of Urology, University of Arizona, Tucson, AZ
| | - Nathan A Ellis
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ
| | - Benjamin R Lee
- Department of Surgery, Division of Urology, University of Arizona, Tucson, AZ
| |
Collapse
|
50
|
Zou Y, Zhong Y, Wu J, Xiao H, Zhang X, Liao X, Li J, Mao X, Liu Y, Zhang F. Long non-coding PANDAR as a novel biomarker in human cancer: A systematic review. Cell Prolif 2018; 51:e12422. [PMID: 29226461 PMCID: PMC6528858 DOI: 10.1111/cpr.12422] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 11/02/2017] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES Long non-coding RNAs (lncRNAs) are characterized as a group of RNAs that more than 200 nucleotides in length and have no protein-coding function. More and more evidences provided that lncRNAs serve as key molecules in the development of cancer. Deregulation of lncRNAs functions as either oncogenes or tumour suppressor genes in various diseases. Recently, increasing studies about PANDAR in cancer progression were reported. In our review, we will focus on the current research on the character of PANDAR include the clinical management, tumour progression and molecular mechanisms in human cancers. MATERIALS AND METHODS We summarize and analyze current studies concerning the biological functions and mechanisms of lncRNA PANDA in tumour development. The related studies were obtained through a systematic search of Pubmed. RESULTS PANDAR was a well-characterized oncogenic lncRNA and widely overexpressed in many tumours. PANDAR is upregulated in many types of cancer, including colorectal cancer, lung cancer, renal cell carcinoma, cholangiocarcinoma, osteosarcoma, thyroid cancer and other cancers. Upregulation of PANDAR was significantly associated with advanced tumour weights, TNM stage and overall survival. Furthermore, repressed of PANDAR would restrain proliferation, migration and invasion. CONCLUSION PANDAR may act as a powerful tumour biomarker for cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Yifan Zou
- Key Laboratory of Medical Reprogramming TechnologyShenzhen Second People's HospitalGuangzhou Medical UniversityShenzhenChina
- Shantou University Medical CollegeShantouChina
| | - Yuantang Zhong
- Key Laboratory of Medical Reprogramming TechnologyShenzhen Second People's HospitalGuangzhou Medical UniversityShenzhenChina
| | - Junjie Wu
- Shantou University Medical CollegeShantouChina
| | - Huizhong Xiao
- Key Laboratory of Medical Reprogramming TechnologyShenzhen Second People's HospitalGuangzhou Medical UniversityShenzhenChina
| | - Xintao Zhang
- Key Laboratory of Medical Reprogramming TechnologyShenzhen Second People's HospitalGuangzhou Medical UniversityShenzhenChina
| | - Xinhui Liao
- Key Laboratory of Medical Reprogramming TechnologyShenzhen Second People's HospitalGuangzhou Medical UniversityShenzhenChina
| | - Jianfa Li
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and GeneticsInstitute of UrologyPeking University Shenzhen HospitalShenzhen PKU‐HKUST Medical CenterShenzhenChina
| | - Xuhua Mao
- Key Laboratory of Medical Reprogramming TechnologyShenzhen Second People's HospitalGuangzhou Medical UniversityShenzhenChina
| | - Yuchen Liu
- Key Laboratory of Medical Reprogramming TechnologyShenzhen Second People's HospitalGuangzhou Medical UniversityShenzhenChina
| | - Fuyou Zhang
- Key Laboratory of Medical Reprogramming TechnologyShenzhen Second People's HospitalGuangzhou Medical UniversityShenzhenChina
| |
Collapse
|