1
|
Tang H, Li YX, Lian JJ, Ng HY, Wang SSY. Personalized treatment using predictive biomarkers in solid organ malignancies: A review. TUMORI JOURNAL 2024; 110:386-404. [PMID: 39091157 DOI: 10.1177/03008916241261484] [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] [Indexed: 08/04/2024]
Abstract
In recent years, the influence of specific biomarkers in the diagnosis and prognosis of solid organ malignancies has been increasingly prominent. The relevance of the use of predictive biomarkers, which predict cancer response to specific forms of treatment provided, is playing a more significant role than ever before, as it affects diagnosis and initiation of treatment, monitoring for efficacy and side effects of treatment, and adjustment in treatment regimen in the long term. In the current review, we explored the use of predictive biomarkers in the treatment of solid organ malignancies, including common cancers such as colorectal cancer, breast cancer, lung cancer, prostate cancer, and cancers associated with high mortalities, such as pancreatic cancer, liver cancer, kidney cancer and cancers of the central nervous system. We additionally analyzed the goals and types of personalized treatment using predictive biomarkers, and the management of various types of solid organ malignancies using predictive biomarkers and their relative efficacies so far in the clinical settings.
Collapse
|
2
|
Wang L, Pattnaik A, Sahoo SS, Stone EG, Zhuang Y, Benton A, Tajmul M, Chakravorty S, Dhawan D, Nguyen MA, Sirit I, Mundy K, Ricketts CJ, Hadisurya M, Baral G, Tinsley SL, Anderson NL, Hoda S, Briggs SD, Kaimakliotis HZ, Allen-Petersen BL, Tao WA, Linehan WM, Knapp DW, Hanna JA, Olson MR, Afzali B, Kazemian M. Unbiased discovery of cancer pathways and therapeutics using Pathway Ensemble Tool and Benchmark. Nat Commun 2024; 15:7288. [PMID: 39179644 PMCID: PMC11343859 DOI: 10.1038/s41467-024-51859-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 08/19/2024] [Indexed: 08/26/2024] Open
Abstract
Correctly identifying perturbed biological pathways is a critical step in uncovering basic disease mechanisms and developing much-needed therapeutic strategies. However, whether current tools are optimal for unbiased discovery of relevant pathways remains unclear. Here, we create "Benchmark" to critically evaluate existing tools and find that most function sub-optimally. We thus develop the "Pathway Ensemble Tool" (PET), which outperforms existing methods. Deploying PET, we identify prognostic pathways across 12 cancer types. PET-identified prognostic pathways offer additional insights, with genes within these pathways serving as reliable biomarkers for clinical outcomes. Additionally, normalizing these pathways using drug repurposing strategies represents therapeutic opportunities. For example, the top predicted repurposed drug for bladder cancer, a CDK2/9 inhibitor, represses cell growth in vitro and in vivo. We anticipate that using Benchmark and PET for unbiased pathway discovery will offer additional insights into disease mechanisms across a spectrum of diseases, enabling biomarker discovery and therapeutic strategies.
Collapse
Affiliation(s)
- Luopin Wang
- Department of Computer Science, Purdue University, West Lafayette, IN, USA
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
| | - Aryamav Pattnaik
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
| | - Subhransu Sekhar Sahoo
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
| | - Ella G Stone
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Yuxin Zhuang
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
| | - Annaleigh Benton
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Md Tajmul
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
- Immunoregulation Section, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, USA
| | - Srishti Chakravorty
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
| | - Deepika Dhawan
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - My An Nguyen
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
- Department of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Isabella Sirit
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Kyle Mundy
- Department of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Christopher J Ricketts
- Urologic Oncology Branch of Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, MD, USA
| | - Marco Hadisurya
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
| | - Garima Baral
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Samantha L Tinsley
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Nicole L Anderson
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Smriti Hoda
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
| | - Scott D Briggs
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
| | | | - Brittany L Allen-Petersen
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - W Andy Tao
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
- Department of Chemistry, Purdue University, West Lafayette, IN, USA
| | - W Marston Linehan
- Urologic Oncology Branch of Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, MD, USA
| | - Deborah W Knapp
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - Jason A Hanna
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Matthew R Olson
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Behdad Afzali
- Immunoregulation Section, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, MD, USA.
| | - Majid Kazemian
- Department of Computer Science, Purdue University, West Lafayette, IN, USA.
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, USA.
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
3
|
Rajagopal A. Surveillance strategies for renal cell carcinoma in the renal allograft: balancing early detection and resource utilization. Proc AMIA Symp 2024; 37:839-840. [PMID: 39165816 PMCID: PMC11332631 DOI: 10.1080/08998280.2024.2384343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 07/22/2024] [Indexed: 08/22/2024] Open
Affiliation(s)
- Arthi Rajagopal
- Division of Nephrology, Baylor University Medical Center, Dallas, Texas, USA
| |
Collapse
|
4
|
Xu W, Gaborieau V, Niman SM, Mukeria A, Liu X, Maremanda KP, Takakura A, Zaridze D, Freedman ML, Xie W, McDermott DF, Choueiri TK, Catalano PJ, Sabbisetti V, Bonventre J, Pierorazio PM, Singla N, Brennan P, Bhatt RS. Plasma Kidney Injury Molecule-1 for Preoperative Prediction of Renal Cell Carcinoma Versus Benign Renal Masses, and Association With Clinical Outcomes. J Clin Oncol 2024; 42:2691-2701. [PMID: 38701382 PMCID: PMC11539753 DOI: 10.1200/jco.23.00699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 11/17/2023] [Accepted: 02/20/2024] [Indexed: 05/05/2024] Open
Abstract
PURPOSE Both clear cell and papillary renal cell carcinomas (RCCs) overexpress kidney injury molecule-1 (KIM-1). We investigated whether plasma KIM-1 (pKIM-1) may be a useful risk stratification tool among patients with suspicious renal masses. METHODS Prenephrectomy pKIM-1 was measured in two independent cohorts of patients with renal masses. Cohort 1, from the prospective K2 trial, included 162 patients found to have clear cell RCC (cases) and 162 patients with benign renal masses (controls). Cohort 2 included 247 patients with small (cT1a) renal masses from an academic biorepository, of whom 184 had RCC. We assessed the relationship between pKIM-1, surgical pathology, and clinical outcomes. RESULTS In Cohort 1, pKIM-1 distinguished RCC versus benign masses with area under the receiver operating curve (AUC-ROC, 0.81 [95% CI, 0.76 to 0.86]). In Cohort 2 (cT1a only), pKIM-1 distinguished RCC versus benign masses (AUC-ROC, 0.74 [95% CI, 0.67 to 0.80]) and the addition of pKIM-1 to an established nomogram for predicting malignancy improved the model AUC-ROC (0.65 [95% CI, 0.57 to 0.74] v 0.78 [95% CI, 0.72 to 0.85]). A pKIM-1 cutpoint identified using Cohort 2 demonstrated sensitivity of 92.5% and specificity of 60% for identifying RCC in Cohort 1. In long-term follow-up of RCC cases (Cohort 1), higher prenephrectomy pKIM-1 was associated with worse metastasis-free survival (multivariable MFS hazard ratio [HR] 1.29 per unit increase in log pKIM-1, 95% CI, 1.10 to 1.53) and overall survival (multivariable OS HR 1.31 per unit increase in log pKIM-1, 95% CI, 1.10 to 1.54). In long-term follow-up of Cohort 2, no metastatic events occurred, consistent with the favorable prognosis of resected cT1a RCC. CONCLUSION Among patients with renal masses, pKIM-1 is associated with malignant pathology, worse MFS, and risk of death. pKIM-1 may be useful for selecting patients with renal masses for intervention versus surveillance.
Collapse
Affiliation(s)
- Wenxin Xu
- Dana-Farber Cancer Institute, Boston, United States
| | | | | | - Anush Mukeria
- N.N. Blokhin National Medical Research Centre of Oncology, Moscow, Russia
| | - Xiaowen Liu
- Beth Israel Deaconess Medical Center, Boston, United States
| | | | | | - David Zaridze
- N.N. Blokhin National Medical Research Centre of Oncology, Moscow, Russia
| | | | - Wanling Xie
- Dana-Farber Cancer Institute, Boston, United States
| | | | | | | | | | | | | | - Nirmish Singla
- Brady Urological Institute, Johns Hopkins University, United States
| | - Paul Brennan
- International Agency for Research on Cancer, Lyon, France
| | - Rupal S. Bhatt
- Beth Israel Deaconess Medical Center, Boston, United States
| |
Collapse
|
5
|
Sanguedolce F, Mazzucchelli R, Falagario UG, Cormio A, Zanelli M, Palicelli A, Zizzo M, Eccher A, Brunelli M, Galosi AB, Carrieri G, Cormio L. Diagnostic Biomarkers in Renal Cell Tumors According to the Latest WHO Classification: A Focus on Selected New Entities. Cancers (Basel) 2024; 16:1856. [PMID: 38791935 PMCID: PMC11120103 DOI: 10.3390/cancers16101856] [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: 04/14/2024] [Revised: 05/02/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
The fifth edition of the World Health Organization (WHO) classification for urogenital tumors, released in 2022, introduces some novelties in the chapter on renal epithelial tumors compared to the previous 2016 classification. Significant changes include the recognition of new disease entities and adjustments in the nomenclature for certain pathologies. Notably, each tumor entity now includes minimum essential and desirable criteria for reliable diagnosis. This classification highlights the importance of biological and molecular characterization alongside traditional cytological and architectural features. In this view, immunophenotyping through immunohistochemistry (IHC) plays a crucial role in bridging morphology and genetics. This article aims to present and discuss the role of key immunohistochemical markers that support the diagnosis of new entities recognized in the WHO classification, focusing on critical topics associated with single markers, in the context of specific tumors, such as the clear cell capillary renal cell tumor (CCPRCT), eosinophilic solid and cystic renal cell carcinoma (ESC-RCC), and so-called "other oncocytic tumors", namely the eosinophilic vacuolated tumor (EVT) and low-grade oncocytic tumor (LOT). Their distinctive characteristics and immunophenotypic profiles, along with insights regarding diagnostic challenges and the differential diagnosis of these tumors, are provided. This state-of-the-art review offers valuable insights in biomarkers associated with novel renal tumors, as well as a tool to implement diagnostic strategies in routine practice.
Collapse
Affiliation(s)
| | - Roberta Mazzucchelli
- Section of Pathological Anatomy, Department of Biomedical Sciences and Public Health, United Hospitals, Università Politecnica delle Marche, 60126 Ancona, Italy;
| | - Ugo Giovanni Falagario
- Department of Urology and Renal Transplantation, Policlinico Foggia, University of Foggia, 71122 Foggia, Italy (G.C.); (L.C.)
- Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Angelo Cormio
- Department of Urology, Azienda Ospedaliero-Universitaria Ospedali Riuniti Di Ancona, Università Politecnica Delle Marche, Via Conca 71, 60126 Ancona, Italy
| | - Magda Zanelli
- Pathology Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (M.Z.); (A.P.)
| | - Andrea Palicelli
- Pathology Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy; (M.Z.); (A.P.)
| | - Maurizio Zizzo
- Surgical Oncology Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy;
| | - Albino Eccher
- Department of Pathology and Diagnostics, University and Hospital Trust of Verona, 37126 Verona, Italy;
| | - Matteo Brunelli
- Department of Pathology and Diagnostics and Public Health, Section of Pathology, University Hospital of Verona, 37126 Verona, Italy;
| | - Andrea Benedetto Galosi
- Department of Urology, Azienda Ospedaliero-Universitaria Ospedali Riuniti Di Ancona, Università Politecnica Delle Marche, Via Conca 71, 60126 Ancona, Italy
| | - Giuseppe Carrieri
- Department of Urology and Renal Transplantation, Policlinico Foggia, University of Foggia, 71122 Foggia, Italy (G.C.); (L.C.)
| | - Luigi Cormio
- Department of Urology and Renal Transplantation, Policlinico Foggia, University of Foggia, 71122 Foggia, Italy (G.C.); (L.C.)
- Department of Urology, Bonomo Teaching Hospital, 76123 Andria, Italy
| |
Collapse
|
6
|
Gong X, Gong Y, Wu G, Ke H. Bioinformatics analysis highlights CCNB1 as a potential prognostic biomarker and an anti-kidney renal papillary cell carcinoma drug target. Medicine (Baltimore) 2024; 103:e37609. [PMID: 38518000 PMCID: PMC10956941 DOI: 10.1097/md.0000000000037609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/23/2024] [Indexed: 03/24/2024] Open
Abstract
Kidney renal papillary cell carcinoma (KIRP) is a common urinary tumor that causes lymph node invasion. Once metastatic, the prognosis is poor and there is a lack of effective early diagnostic markers for this tumor. The expression of CCNB1 in KIRP tumor tissues was significantly higher than that in normal tissues in The Cancer Genome Atlas database with or without the genotype-tissue expression database, and a consistent result was obtained in 32 paired tissues. In addition, CCNB1 expression increased remarkably with the progression of the T and M stages. Moreover, using the online HPA database, we verified that the immunohistochemical scores of CCNB1 in KIRP were higher than those in the normal kidney tissues. The higher expression group of CCNB1 showed a worse prognosis in KIRP. Moreover, the receiver operating characteristic curve, univariate and multivariate analyses, and construction of the column diagram further illustrated that CCNB1 was an independent prognostic factor for KIRP. Meanwhile, CCNB1 could better predict the 1- and 3-year survival rates of KIRP. Six genes were significantly and positively co-expressed with CCNB1. We also found that the CCNB1 high-expression group was enriched in the ECM_RECEPTOR_INTERACTION and FOCAL_ADHESION pathways. Finally, drug sensitivity analysis combined with molecular docking identified 5 targeting drugs with the strongest binding activity to CCNB1. CCNB1 is a potential and reliable biomarker for KIRP diagnosis and can be used to predict the survival of patients with KIRP. The 5 selected drugs targeting CCNB1 may provide new hopes for patients with KIRP metastasis.
Collapse
Affiliation(s)
- Xiaoming Gong
- Department of Infectious Disease, Hubei AIDS Clinical Training Center, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Radiology, Xianning Central Hospital, The First Affiliate Hospital of Hubei University of Science and Technology, Xianning, China
| | - Yahong Gong
- Department of Radiology, Xianning Central Hospital, The First Affiliate Hospital of Hubei University of Science and Technology, Xianning, China
| | - GuiFang Wu
- Department of Infectious Disease, Hubei AIDS Clinical Training Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hengning Ke
- Department of Infectious Disease, Hubei AIDS Clinical Training Center, Zhongnan Hospital of Wuhan University, Wuhan, China
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China
- Cancer Research Institute, General Hospital, Ningxia Medical University, Yinchuan, China
| |
Collapse
|
7
|
Elkady N, Abdelaziz RA, Abdelmoneum RA, Ghonaimy AS, Allam DM. Regulation of Tumor Vascular Microenvironment by Nestin and Fms-related Tyrosine Kinase 1 (FLT1) and Their Prognostic Significance in Renal Cell Carcinoma. IRANIAN JOURNAL OF PATHOLOGY 2024; 19:332-341. [PMID: 39687450 PMCID: PMC11646198 DOI: 10.30699/ijp.2024.2024190.3266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 06/11/2024] [Indexed: 12/18/2024]
Abstract
Background & Objective Hypervascularity is a characteristic feature of renal cell carcinoma (RCC) and microvessel density (MVD) predicts tumor metastasis. Nestin is a stem cell marker that is expressed in proliferating endothelial cells and newly formed vessels and Fms-related tyrosine kinase 1 (FLT1) is a proangiogenic factor. This study aimed to evaluate the expression of Nestin and FLT1 in RCC and their prognostic impact. Methods This retrospective study included sixty cases of RCC after obtaining ethical approval. Sections were immunohistochemically stained by Nestin and FLT1 then their expressions were compared to different clinicopathological parameters. MVD was evaluated using Nestin and CD34 and compared to the different parameters. Results Nestin was expressed mainly in endothelial cells of small vessels in 65% of cases while FLT1 was expressed in tumor and endothelial cells in 73.3% of cases. Their expressions were significantly associated with aggressive tumor parameters including larger tumors, high-grade tumors, wider tumor extension, and advanced stage. Moreover, Nestin expression was significantly associated with metastasis. MVD evaluated by Nestin showed more associations with larger tumors, high-grade tumors, wider tumor extension, advanced stage, and metastasis than MVD measured by CD34. Nestin and FLT1 positivity and high MVD measured by Nestin were significantly associated with short overall survival. Conclusion Nestin and FLT1 expressions in RCC may be associated with aggressive tumor features and short patients' overall survival. MVD evaluated by Nestin may be correlated with tumor progression and metastasis. Nestin and FLT1 may be used as prognostic biomarkers in RCC.
Collapse
Affiliation(s)
- Noha Elkady
- Department of Pathology, Faculty of Medicine, Menoufia University, Shibin Elkom, Egypt
| | - Reham Ahmed Abdelaziz
- Department of Clinical Oncology and Nuclear Medicine, Faculty of Medicine, Menoufia University, Egypt
| | - Rasha Adel Abdelmoneum
- Department of Clinical Oncology and Nuclear Medicine, Faculty of Medicine, Menoufia University, Egypt
| | - Ahmed S Ghonaimy
- Department of Urology, Faculty of Medicine, Menoufia University, Shibin Elkom, Egypt
| | - Dina Mohamed Allam
- Department of Pathology, Faculty of Medicine, Menoufia University, Shibin Elkom, Egypt
| |
Collapse
|
8
|
Zieren RC, Zondervan PJ, Pienta KJ, Bex A, de Reijke TM, Bins AD. Diagnostic liquid biopsy biomarkers in renal cell cancer. Nat Rev Urol 2024; 21:133-157. [PMID: 37758847 DOI: 10.1038/s41585-023-00818-y] [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] [Accepted: 08/25/2023] [Indexed: 09/29/2023]
Abstract
The clinical presentation of renal cell cancer (RCC) is shifting towards incidental and early detection, creating new challenges in RCC diagnosis. Overtreatment might be reduced with the development of new diagnostic biomarkers to distinguish benign from malignant small renal masses (SRMs). Differently from tissue biopsies, liquid biopsies are obtained from a patient's blood or urine and, therefore, are minimally invasive and suitable for longitudinal monitoring. The most promising types of liquid biopsy biomarkers for RCC diagnosis are circulating tumour cells, extracellular vesicles (EVs) and cell-free DNA. Circulating tumour cell assays have the highest specificity, with low processing time and costs. However, the biological characteristics and low sensitivity limit the use of these markers in SRM diagnostics. Cell-free DNA might complement the diagnosis of high-volume RCC, but the potential for clinical application in SRMs is limited. EVs have the highest biological abundance and the highest sensitivity in identifying low-volume disease; moreover, the molecular characteristics of these markers make EVs suitable for multiple analytical applications. Thus, currently, EV assays have the greatest potential for diagnostic application in RCC (including identification of SRMs). All these liquid biomarkers have potential in clinical practice, pending validation studies. Biomarker implementation will be needed to also improve characterization of RCC subtypes. Last, diagnostic biomarkers might be extended to prognostic or predictive applications.
Collapse
Affiliation(s)
- Richard C Zieren
- Department of Urology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands.
- The Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Patricia J Zondervan
- Department of Urology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Kenneth J Pienta
- The Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Axel Bex
- Specialist Centre for Kidney Cancer, Royal Free Hospital, London, United Kingdom
- Division of Surgery and Interventional Science, University College London, London, United Kingdom
- The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Theo M de Reijke
- Department of Urology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Adriaan D Bins
- Department of Medical Oncology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
9
|
Kothapalli KSD, Park HG, Kothapalli NSL, Brenna JT. FADS2 function at the major cancer hotspot 11q13 locus alters fatty acid metabolism in cancer. Prog Lipid Res 2023; 92:101242. [PMID: 37597812 DOI: 10.1016/j.plipres.2023.101242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/31/2023] [Accepted: 08/15/2023] [Indexed: 08/21/2023]
Abstract
Dysregulation of fatty acid metabolism and de novo lipogenesis is a key driver of several cancer types through highly unsaturated fatty acid (HUFA) signaling precursors such as arachidonic acid. The human chromosome 11q13 locus has long been established as the most frequently amplified in a variety of human cancers. The fatty acid desaturase genes (FADS1, FADS2 and FADS3) responsible for HUFA biosynthesis localize to the 11q12-13.1 region. FADS2 activity is promiscuous, catalyzing biosynthesis of several unsaturated fatty acids by Δ6, Δ8, and Δ4 desaturation. Our main aim here is to review known and putative consequences of FADS2 dysregulation due to effects on the 11q13 locus potentially driving various cancer types. FADS2 silencing causes synthesis of sciadonic acid (5Z,11Z,14Z-20:3) in MCF7 cells and breast cancer in vivo. 5Z,11Z,14Z-20:3 is structurally identical to arachidonic acid (5Z,8Z,11Z,14Z-20:4) except it lacks the internal Δ8 double bond required for prostaglandin and leukotriene synthesis, among other eicosanoids. Palmitic acid has substrate specificity for both SCD and FADS2. Melanoma, prostate, liver and lung cancer cells insensitive to SCD inhibition show increased FADS2 activity and sapienic acid biosynthesis. Elevated serum mead acid levels found in hepatocellular carcinoma patients suggest an unsatisfied demand for arachidonic acid. FADS2 circular RNAs are at high levels in colorectal and lung cancer tissues. FADS2 circular RNAs are associated with shorter overall survival in colorectal cancer patients. The evidence thusfar supports an effort for future research on the role of FADS2 as a tumor suppressor in a range of neoplastic disorders.
Collapse
Affiliation(s)
- Kumar S D Kothapalli
- Dell Pediatric Research Institute, Dell Medical School and Department of Nutritional Sciences, The University of Texas at Austin, 1400 Barbara Jordan Blvd, Austin, TX 78723, USA.
| | - Hui Gyu Park
- Dell Pediatric Research Institute, Dell Medical School and Department of Nutritional Sciences, The University of Texas at Austin, 1400 Barbara Jordan Blvd, Austin, TX 78723, USA
| | | | - J Thomas Brenna
- Dell Pediatric Research Institute, Dell Medical School and Department of Nutritional Sciences, The University of Texas at Austin, 1400 Barbara Jordan Blvd, Austin, TX 78723, USA.
| |
Collapse
|
10
|
Dani KA, Rich JM, Kumar SS, Cen H, Duddalwar VA, D’Souza A. Comprehensive Systematic Review of Biomarkers in Metastatic Renal Cell Carcinoma: Predictors, Prognostics, and Therapeutic Monitoring. Cancers (Basel) 2023; 15:4934. [PMID: 37894301 PMCID: PMC10605584 DOI: 10.3390/cancers15204934] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 09/30/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Challenges remain in determining the most effective treatment strategies and identifying patients who would benefit from adjuvant or neoadjuvant therapy in renal cell carcinoma. The objective of this review is to provide a comprehensive overview of biomarkers in metastatic renal cell carcinoma (mRCC) and their utility in prediction of treatment response, prognosis, and therapeutic monitoring in patients receiving systemic therapy for metastatic disease. METHODS A systematic literature search was conducted using the PubMed database for relevant studies published between January 2017 and December 2022. The search focused on biomarkers associated with mRCC and their relationship to immune checkpoint inhibitors, targeted therapy, and VEGF inhibitors in the adjuvant, neoadjuvant, and metastatic settings. RESULTS The review identified various biomarkers with predictive, prognostic, and therapeutic monitoring potential in mRCC. The review also discussed the challenges associated with anti-angiogenic and immune-checkpoint monotherapy trials and highlighted the need for personalized therapy based on molecular signatures. CONCLUSION This comprehensive review provides valuable insights into the landscape of biomarkers in mRCC and their potential applications in prediction of treatment response, prognosis, and therapeutic monitoring. The findings underscore the importance of incorporating biomarker assessment into clinical practice to guide treatment decisions and improve patient outcomes in mRCC.
Collapse
Affiliation(s)
- Komal A. Dani
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA;
| | - Joseph M. Rich
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA;
| | - Sean S. Kumar
- Eastern Virginia Medical School, Norfolk, VA 23507, USA;
- Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| | - Harmony Cen
- University of Southern California, Los Angeles, CA 90033, USA;
| | - Vinay A. Duddalwar
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA;
- Institute of Urology, University of Southern California, Los Angeles, CA 90033, USA
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Anishka D’Souza
- Department of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
11
|
Nolazco JI, Soerensen SJC, Chung BI. Biomarkers for the Detection and Surveillance of Renal Cancer. Urol Clin North Am 2023; 50:191-204. [PMID: 36948666 DOI: 10.1016/j.ucl.2023.01.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
Renal cell carcinoma (RCC) is a heterogeneous disease characterized by a broad spectrum of disorders in terms of genetics, molecular and clinical characteristics. There is an urgent need for noninvasive tools to stratify and select patients for treatment accurately. In this review, we analyze serum, urinary, and imaging biomarkers that have the potential to detect malignant tumors in patients with RCC. We discuss the characteristics of these numerous biomarkers and their ability to be used routinely in clinical practice. The development of biomarkers continues to evolve with promising prospects.
Collapse
Affiliation(s)
- José Ignacio Nolazco
- Division of Urological Surgery, Brigham and Women's Hospital, Harvard Medical School, 45 Francis Street, Boston, MA 02115, USA; Servicio de Urología, Hospital Universitario Austral, Universidad Austral, Av Juan Domingo Perón 1500, B1629AHJ Pilar, Argentina.
| | - Simon John Christoph Soerensen
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA; Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, USA
| | - Benjamin I Chung
- Department of Urology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
12
|
Ali SN, Tano Z, Landman J. The Changing Role of Renal Mass Biopsy. Urol Clin North Am 2023; 50:217-225. [PMID: 36948668 DOI: 10.1016/j.ucl.2023.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
The incidence and prevalence of small renal masses (SRMs) continues to rise and with increased detection comes increases in surgical management, although the probability of an SRM being benign is upward of 30%. An extirpative treatment first diagnose-later strategy persists and clinical tools for risk stratification such as renal mass biopsy remain severely underutilized. The overtreatment of SRMs has multiple detrimental effects including surgical complications, psychosocial stress, financial loss, and reduced renal function leading to downstream effects such as the need for dialysis and cardiovascular disease.
Collapse
Affiliation(s)
| | - Zachary Tano
- Department of Urology, University of California, Irvine, CA, USA
| | - Jaime Landman
- Department of Urology, University of California, Irvine, CA, USA.
| |
Collapse
|
13
|
Posada Calderon L, Eismann L, Reese SW, Reznik E, Hakimi AA. Advances in Imaging-Based Biomarkers in Renal Cell Carcinoma: A Critical Analysis of the Current Literature. Cancers (Basel) 2023; 15:cancers15020354. [PMID: 36672304 PMCID: PMC9856305 DOI: 10.3390/cancers15020354] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/31/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
Cross-sectional imaging is the standard diagnostic tool to determine underlying biology in renal masses, which is crucial for subsequent treatment. Currently, standard CT imaging is limited in its ability to differentiate benign from malignant disease. Therefore, various modalities have been investigated to identify imaging-based parameters to improve the noninvasive diagnosis of renal masses and renal cell carcinoma (RCC) subtypes. MRI was reported to predict grading of RCC and to identify RCC subtypes, and has been shown in a small cohort to predict the response to targeted therapy. Dynamic imaging is promising for the staging and diagnosis of RCC. PET/CT radiotracers, such as 18F-fluorodeoxyglucose (FDG), 124I-cG250, radiolabeled prostate-specific membrane antigen (PSMA), and 11C-acetate, have been reported to improve the identification of histology, grading, detection of metastasis, and assessment of response to systemic therapy, and to predict oncological outcomes. Moreover, 99Tc-sestamibi and SPECT scans have shown promising results in distinguishing low-grade RCC from benign lesions. Radiomics has been used to further characterize renal masses based on semantic and textural analyses. In preliminary studies, integrated machine learning algorithms using radiomics proved to be more accurate in distinguishing benign from malignant renal masses compared to radiologists' interpretations. Radiomics and radiogenomics are used to complement risk classification models to predict oncological outcomes. Imaging-based biomarkers hold strong potential in RCC, but require standardization and external validation before integration into clinical routines.
Collapse
Affiliation(s)
- Lina Posada Calderon
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Lennert Eismann
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Stephen W. Reese
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ed Reznik
- Computational Oncology, Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Abraham Ari Hakimi
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Correspondence:
| |
Collapse
|
14
|
Are Renal Cell Carcinoma with Fibromyomatous Stroma (RCC-FMS) and Thyroid-like Follicular Carcinoma of the Kidney (TLFCK) Really Independent Variants? Diagnostics (Basel) 2022; 13:diagnostics13010086. [PMID: 36611378 PMCID: PMC9818596 DOI: 10.3390/diagnostics13010086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/16/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Renal cell carcinoma with fibromyomatous stroma (RCC-FMS) is a recent provisional entity already recognised in the 2016 WHO Classification of Cancer of the Urinary Tract and Male Genital Organs 4th Edition as renal cell carcinoma with (angio)leiomyomatous stroma, histologically defined as a tumour characterised by clear cells intertwined in a conspicuous vascular stroma. In the casuistry taken into consideration, another proposed variant, thyroid-like follicular carcinoma of the kidney (TLFCK), endowed with a morphology mimicking thyroid parenchyma, was examined. The aim of this work was to parse the theoretical system, experimental data and diagnostic impact of these new entities proposed in the field of renal neoplasms. MATERIALS AND METHODS An analysis of 120 cases of kidney tumours from the Department of Surgical, Medical, Molecular and Critical Area at the University of Pisa was run. Subsequently, all samples were reassessed by two pathologists with expertise in uropathology, whose revaluation provided a histomorphological study combined with subsequent and coherent immunohistochemical analyses of CK7, CD10, CAIX, CK34betaE12, CD117, vimentin, TTF-1 and thyroglobulin. These analyses were performed using the Ventana Benchmark Automated Staining System (Ventana Medical Systems, Tucson, AZ, USA) and Ventana reagents. RESULTS On the one hand, the data, thus brought to light, did not show an immunohistochemical profile consistent with that proposed for RCC-FMS. However, it should be emphasised that the morphological background also unearthed a poor specificity for RCC-FMS. This was specifically due to a stromal component which was, in any case, evident, although characterised by a wide range of presentation, in clear cell renal cell carcinoma (ccRCC). This latter is, indeed, the reference background for this theorised variant. On the other hand, a thyroid-like pattern was highlighted in 11 cases, more specifically in 10 ccRCCs and in one oncocytoma, presenting itself as a type of neoplastic appearance rather than as the peculiar morphological pattern of a standalone cancer. CONCLUSIONS In the light of these results, RCC-FMS and TLFCK appear to be more appropriately variants of already categorised neoplastic entities rather than new independent neoplasias.
Collapse
|
15
|
Del Re M, Crucitta S, Paolieri F, Cucchiara F, Verzoni E, Bloise F, Ciampi R, Mercinelli C, Capuano A, Sportiello L, Martinetti A, Procopio G, Galli L, Porta C, Bracarda S, Danesi R. The amount of DNA combined with TP53 mutations in liquid biopsy is associated with clinical outcome of renal cancer patients treated with immunotherapy and VEGFR-TKIs. Lab Invest 2022; 20:371. [PMID: 35974365 PMCID: PMC9382729 DOI: 10.1186/s12967-022-03557-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/25/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND Despite the increasing number of treatment options, reliable prognostic/predictive biomarkers are still missing for patients affected by metastatic clear cell renal cell carcinoma (mccRCC). METHODS Patients with mccRCC undergoing standard first line treatment were enrolled. Blood (12 ml) was drawn at treatment baseline and circulating free DNA (cfDNA) was extracted from plasma. Next-generation sequencing (NGS) was performed on cfDNA using the Oncomine Pan-Cancer Cell-Free Assay and clinical outcomes were correlated with liquid biopsy findings. RESULTS A total of 48 patients were enrolled, 12 received immunotherapy and 36 received a vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitor (TKI). A cfDNA cut-off of 0.883 ng/μl stratified patients based on progression-free survival (PFS) and overall survival (OS) (p = 0.001 and p = 0.008, respectively). cfDNA amount was also correlated with best response (p = 0.006). Additional cfDNA cut-points divided patients into short, intermediate and long responders, with PFS of 4.87 vs 9.13 vs 23.1 months, respectively (p < 0.001). PFS resulted to be significantly shorter in carriers of mutant TP53 compared to not carriers (p = 0.04). Patients with high cfDNA levels and mutant TP53 have the worst PFS, while patients with low cfDNA amounts and no mutations in TP53 displayed the longest PFS (p = 0.004). CONCLUSIONS The present study demonstrates that cfDNA and TP53 are potential predictive biomarkers of response in mccRCC to be further explored in larger and/or prospective studies.
Collapse
Affiliation(s)
- Marzia Del Re
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Stefania Crucitta
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Federico Paolieri
- Unit of Medical Oncology, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Federico Cucchiara
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Elena Verzoni
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Francesco Bloise
- Unit of Medical Oncology, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Raffaele Ciampi
- Endocrine Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Chiara Mercinelli
- Unit of Medical Oncology, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Annalisa Capuano
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Liberata Sportiello
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Antonia Martinetti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giuseppe Procopio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Luca Galli
- Unit of Medical Oncology, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Camillo Porta
- Division of Oncology, Department of Biomedical Sciences and Human Oncology, University of Bari, Bari, Italy
| | - Sergio Bracarda
- Unit of Medical and Translational Oncology, Department of Oncology, Civil Hospital of Terni, Terni, Italy
| | - Romano Danesi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.
| |
Collapse
|
16
|
Köditz B, Heidenreich A, von Brandenstein M. The Current Status of Kidney Cancer Urine Markers – A Systematic Review. KIDNEY CANCER 2022. [DOI: 10.3233/kca-220005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND: Renal cell carcinoma is the 9th most common malignant disease in the Western World. Typically, patients develop symptoms in a late stage of the disease and most of them are diagnosed by chance. Up to 30% of the patients at the time of diagnosis had metastatic disease. Therefore, highly specific and sensitive biomarkers for the detection and progression of kidney cancer are of great importance. Here, urine markers can be a major advantage and can have a huge clinical impact on the diagnosis, differentiation and prognosis of kidney cancer. At the moment there are several approaches to improve these conditions.. METHODS: A systematic literature research was performed according to the PRISMA guidelines to identify studies reporting urine markers for kidney cancer between 2012 and 2021. A two-step process for the selection of the studies was initiated. In total 287 studies were considering for the final analysis. In total, 6 studies, which presented potential urinary biomarker were analyzed in depth. RESULTS: The major focus was on urinary markers for the detection, progression and differentiation of renal cell carcinoma. In total, a study population of 1099 patients were investigated in the different studies that were analyzed in depth. The median patient sample size of the different studies was 157 patients. The focus was based on the investigation of different microRNAs and proteins as urinary marker for kidney cancer detection. CONCLUSION: Overall, there are different approaches present for the detection, prognosis and differentiation of kidney cancer in urine but most of the studies are based on a small sample size and need to be validated in a greater collective. Furthermore, the standard should be improved to bring these biomarkers into routine clinical practice.
Collapse
Affiliation(s)
- Barbara Köditz
- Department of Urology, Facility of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Axel Heidenreich
- Department of Urology, Facility of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Melanie von Brandenstein
- Department of Urology, Facility of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
17
|
Ördög N, Borsos BN, Majoros H, Ujfaludi Z, Pankotai-Bodó G, Bankó S, Sükösd F, Kuthi L, Pankotai T. The clinical significance of epigenetic and RNAPII variabilities occurring in clear cell renal cell carcinoma as a potential prognostic marker. Transl Oncol 2022; 20:101420. [PMID: 35417813 PMCID: PMC9018449 DOI: 10.1016/j.tranon.2022.101420] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 03/02/2022] [Accepted: 04/05/2022] [Indexed: 12/14/2022] Open
Abstract
30 ccRCC patients were used, to follow the epigenetic changes (γH2A.X, H3K4me3 and H3K9me3) and the alterations in the level of RNA polymerase II (RNAPII). The variabilities between the tumorous and non-tumorous parts of the tissue were measured by image analysis in which we monitored 30 cells from different positions of either the tumorous or the non-tumorous part of the tissue sections. These markers were classified to predict patient outcomes based on their individual cellular background. These results also support that detection of any alteration in the level of H3K4me3, H3K9me3, and γH2AX can account valuable information for presuming the progression of ccRCC and the clinical benefits to select the most efficient personalized therapy.
Patients diagnosed with clear cell renal cell carcinoma (ccRCC) have poor prognosis for recurrence and approximately 30–40% of them will later develop metastases. For this reason, the appropriate diagnosis and the more detailed molecular characterisation of the primary tumour, including its susceptibility to metastasis, are crucial to select the proper adjuvant therapy by which the most prosperous outcome can be achieved. Nowadays, clinicopathological variables are used for classification of the tumours. Apart from these, molecular biomarkers are also necessary to improve risk classification, which would be the most beneficial amongst modern adjuvant therapies. As a potential molecular biomarker, to follow the transcriptional kinetics in ccRCC patients (n=30), we analysed epigenetic changes (γH2A.X, H3K4me3, and H3K9me3) and the alterations in the level of RNA polymerase II (RNAPII) by immunohistochemical staining on dissected tissue sections. The variabilities between the tumorous and non-tumorous parts of the tissue were detected using quantitative image analysis by monitoring 30 cells from different positions of either the tumorous or the non-tumorous part of the tissue sections. Data obtained from the analyses were used to identify potential prognostic features and to associate them with the progression. These markers might have a value to predict patient outcomes based on their individual cellular background. These results also support that detection of any alteration in the level of H3K4me3, H3K9me3, and γH2A.X can account for valuable information for presuming the progression of ccRCC and the clinical benefits to select the most efficient personalised therapy.
Collapse
|
18
|
Renal oncocytoma: a challenging diagnosis. Curr Opin Oncol 2022; 34:243-252. [DOI: 10.1097/cco.0000000000000829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
19
|
LiKidMiRs: A ddPCR-Based Panel of 4 Circulating miRNAs for Detection of Renal Cell Carcinoma. Cancers (Basel) 2022; 14:cancers14040858. [PMID: 35205607 PMCID: PMC8869982 DOI: 10.3390/cancers14040858] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/06/2022] [Accepted: 02/07/2022] [Indexed: 01/26/2023] Open
Abstract
Simple Summary Early detection of renal cell carcinoma (RCC) significantly increases the likelihood of curative treatment, avoiding the need of adjuvant therapies, associated side effects and comorbidities. Thus, we aimed to discover circulating microRNAs that might aid in early, minimally invasive, RCC detection/diagnosis. Abstract Background: Decreased renal cell cancer-related mortality is an important societal goal, embodied by efforts to develop effective biomarkers enabling early detection and increasing the likelihood of curative treatment. Herein, we sought to develop a new biomarker for early and minimally invasive detection of renal cell carcinoma (RCC) based on a microRNA panel assessed by ddPCR. Methods: Plasma samples from patients with RCC (n = 124) or oncocytomas (n = 15), and 64 healthy donors, were selected. Hsa-miR-21-5p, hsa-miR-126-3p, hsa-miR-155-5p and hsa-miR-200b-3p levels were evaluated using a ddPCR protocol. Results: RCC patients disclosed significantly higher circulating levels of hsa-miR-155-5p compared to healthy donors, whereas the opposite was observed for hsa-miR-21-5p levels. Furthermore, hsa-miR-21-5p and hsa-miR-155-5p panels detected RCC with high sensitivity (82.66%) and accuracy (71.89%). The hsa-miR-126-3p/hsa-miR-200b-3p panel identified the most common RCC subtype (clear cell, ccRCC) with 74.78% sensitivity. Conclusion: Variable combinations of plasma miR levels assessed by ddPCR enable accurate detection of RCC in general, and of ccRCC. These findings, if confirmed in larger studies, provide evidence for a novel ancillary tool which might aid in early detection of RCC.
Collapse
|
20
|
Peter MR, Zhao F, Jeyapala R, Kamdar S, Xu W, Hawkins C, Evans AJ, Fleshner NE, Finelli A, Bapat B. Investigating Urinary Circular RNA Biomarkers for Improved Detection of Renal Cell Carcinoma. Front Oncol 2022; 11:814228. [PMID: 35174071 PMCID: PMC8841801 DOI: 10.3389/fonc.2021.814228] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/27/2021] [Indexed: 12/22/2022] Open
Abstract
Renal cell carcinomas (RCC) are usually asymptomatic until late stages, posing several challenges for early detection of malignant disease. Non-invasive liquid biopsy biomarkers are emerging as an important diagnostic tool which could aid with routine screening of RCCs. Circular RNAs (circRNAs) are novel non-coding RNAs that play diverse roles in carcinogenesis. They are promising biomarkers due to their stability and ease of detection in small quantities from non-invasive sources such as urine. In this study, we analyzed the expression of various circRNAs that were previously identified in RCC tumors (circEGLN3, circABCB10, circSOD2 and circACAD11) in urinary sediment samples from non-neoplastic controls, patients with benign renal tumors, and clear cell RCC (ccRCC) patients. We observed significantly reduced levels of circEGLN3 and circSOD2 in urine from ccRCC patients compared to healthy controls. We also assessed the linear variant of EGLN3 and found differential expression between patients with benign tumors compared to ccRCC patients. These findings highlight the potential of circRNA markers as non-invasive diagnostic tools to detect malignant RCC.
Collapse
Affiliation(s)
- Madonna R. Peter
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Fang Zhao
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Renu Jeyapala
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Shivani Kamdar
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Wei Xu
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Cynthia Hawkins
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumor Research Centre, Hospital for Sick Children, Toronto, ON, Canada
| | - Andrew J. Evans
- Department of Laboratory Medicine, Mackenzie Health, Richmond Hill, ON, Canada
| | - Neil E. Fleshner
- Division of Urology, Department of Surgery, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Antonio Finelli
- Division of Urology, Department of Surgery, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Bharati Bapat
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON, Canada
- *Correspondence: Bharati Bapat,
| |
Collapse
|
21
|
Arance E, Ramírez V, Rubio-Roldan A, Ocaña-Peinado FM, Romero-Cachinero C, Jódar-Reyes AB, Vazquez-Alonso F, Martinez-Gonzalez LJ, Alvarez-Cubero MJ. Determination of Exosome Mitochondrial DNA as a Biomarker of Renal Cancer Aggressiveness. Cancers (Basel) 2021; 14:cancers14010199. [PMID: 35008363 PMCID: PMC8750318 DOI: 10.3390/cancers14010199] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 12/23/2021] [Accepted: 12/28/2021] [Indexed: 01/12/2023] Open
Abstract
Simple Summary Components of liquid biopsy are potential non-invasive biomarkers for monitoring renal cell carcinoma (RCC) status. The aim of our study was to examine mitochondrial genes (such as HV1 and CYB) included in exosomal fractions as promising and innovative biomarkers in RCC. We found that phase C containing different types of vesicles and phase F rich in exosomes with a high mitochondrial DNA (mtDNA) content could be considered as powerful biomarkers for susceptibility to RCC. Interestingly, mtDNA was a good genetic marker when aggressiveness was evaluated. Abstract Here, the role of non-invasive biomarkers in liquid biopsy was evaluated, mainly in exosomes and mitochondrial DNA (mtDNA) as promising, novel, and stable biomarkers for renal cell carcinoma (RCC). A total of 140 fractions (named from B to F) obtained by ultracentrifugations of whole blood samples from 28 individuals (13 patients and 15 controls) were included. Nanoparticle Tracking Analysis (NTA) was conducted to characterized exosomal fraction. Subsequently, an analysis of digital PCR (dPCR) using the QuantStudio™ 3D Digital PCR platform was performed and the quantification of mtDNA copy number by QuantStudioTM 12K Flex Real-Time PCR System (qPCR) was developed. Moreover, Next Generation Sequencing (NGS) analyses were included using MiSeq system (Illumina, San Diego, CA, USA). An F fraction, which contains all exosome data and all mitochondrial markers, was identified in dPCR and qPCR with statistically significant power (adjusted p values ≤ 0.03) when comparing cases and controls. Moreover, present analysis in mtDNA showed a relevant significance in RCC aggressiveness. To sum up, this is the first time a relation between exosomal mtDNA markers and clinical management of RCC is analyzed. We suggest a promising strategy for future liquid biopsy RCC analysis, although more analysis should be performed prior to application in routine clinical practice.
Collapse
Affiliation(s)
- Elena Arance
- GENYO. Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada-Avenida de la Ilustración, 114-18016 Granada, Spain; (E.A.); (V.R.); (A.R.-R.)
| | - Viviana Ramírez
- GENYO. Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada-Avenida de la Ilustración, 114-18016 Granada, Spain; (E.A.); (V.R.); (A.R.-R.)
| | - Alejandro Rubio-Roldan
- GENYO. Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada-Avenida de la Ilustración, 114-18016 Granada, Spain; (E.A.); (V.R.); (A.R.-R.)
| | | | - Catalina Romero-Cachinero
- Nursery Department. University Hospital Virgen de las Nieves, Av. de las Fuerzas Armadas 2, 18014 Granada, Spain;
| | - Ana Belén Jódar-Reyes
- Biocolloid and Fluid Physics Group, Excellence Research Unit Modeling Nature (MNat), Department of Applied Physics, School of Sciences, University of Granada, 18071 Granada, Spain;
| | - Fernando Vazquez-Alonso
- Urology Department, University Hospital Virgen de las Nieves, Av. de las Fuerzas Armadas 2, 18014 Granada, Spain;
| | - Luis Javier Martinez-Gonzalez
- GENYO. Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada-Avenida de la Ilustración, 114-18016 Granada, Spain; (E.A.); (V.R.); (A.R.-R.)
- Correspondence: ; Tel.: +34-958-715-500; Fax: +34-958-637-071
| | - Maria Jesus Alvarez-Cubero
- Department of Biochemistry and Molecular Biology III, Faculty of Medicine, PTS Granada, University of Granada, 18016 Granada, Spain;
- Instituto de Investigación Biosanitaria ibs. GRANADA, 18014 Granada, Spain
| |
Collapse
|
22
|
Srivastava A, Shinder B, Singer EA. EDITORIAL COMMENT. Urology 2021; 158:115. [PMID: 34895625 DOI: 10.1016/j.urology.2021.04.076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 04/28/2021] [Indexed: 11/26/2022]
Affiliation(s)
- Arnav Srivastava
- Rutgers Cancer Institute of New Jersey, Section of Urologic Oncology
| | - Brian Shinder
- Rutgers Cancer Institute of New Jersey, Section of Urologic Oncology
| | - Eric A Singer
- Rutgers Cancer Institute of New Jersey, Section of Urologic Oncology
| |
Collapse
|
23
|
Ged Y, Voss MH. Novel emerging biomarkers to immunotherapy in kidney cancer. Ther Adv Med Oncol 2021; 13:17588359211059367. [PMID: 34868351 PMCID: PMC8640284 DOI: 10.1177/17588359211059367] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 10/25/2021] [Indexed: 12/23/2022] Open
Abstract
The treatment of metastatic renal cell carcinoma has significantly evolved in recent years, particularly with the advent of novel immune checkpoint inhibitors (ICI). Despite the striking benefits observed on a population level, outcomes vary and some patients do not respond to ICI-based regimens, ultimately require salvage therapies. An ever deeper understanding of the disease biology mediated by the development of multiple high-throughput molecular omics has led to significant progress in biomarkers discovery. But despite growing insights into the molecular underpinnings of the tumor microenvironment, biomarkers have not been integrated successfully into clinical practice. In this review, we discuss some of the novel emerging predictive biomarkers to ICIs in metastatic renal cell carcinoma.
Collapse
Affiliation(s)
- Yasser Ged
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Martin H Voss
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 300 East 66th Street, New York, NY 10065, USA
| |
Collapse
|
24
|
Schieda N, Krishna S, Pedrosa I, Kaffenberger SD, Davenport MS, Silverman SG. Active Surveillance of Renal Masses: The Role of Radiology. Radiology 2021; 302:11-24. [PMID: 34812670 DOI: 10.1148/radiol.2021204227] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Active surveillance of renal masses, which includes serial imaging with the possibility of delayed treatment, has emerged as a viable alternative to immediate therapeutic intervention in selected patients. Active surveillance is supported by evidence that many benign masses are resected unnecessarily, and treatment of small cancers has not substantially reduced cancer-specific mortality. These data are a call to radiologists to improve the diagnosis of benign renal masses and differentiate cancers that are biologically aggressive (prompting treatment) from those that are indolent (allowing treatment deferral). Current evidence suggests that active surveillance results in comparable cancer-specific survival with a low risk of developing metastasis. Radiology is central in this. Imaging is used at the outset to estimate the probability of malignancy and degree of aggressiveness in malignant masses and to follow up masses for growth and morphologic change. Percutaneous biopsy is used to provide a more definitive histologic diagnosis and to guide treatment decisions, including whether active surveillance is appropriate. Emerging applications that may improve imaging assessment of renal masses include standardized assessment of cystic and solid masses and radiomic analysis. This article reviews the current and future role of radiology in the care of patients with renal masses undergoing active surveillance.
Collapse
Affiliation(s)
- Nicola Schieda
- From the Department of Medical Imaging, The Ottawa Hospital, University of Ottawa, 1053 Carling Ave, Ottawa, ON, Canada K1H 1H6 (N.S.); Joint Department of Medical Imaging, University Health Network, Mount Sinai Hospital and Women's College Hospital, University of Toronto, Toronto, Canada (S.K.); Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Tex (I.P.); Departments of Urology (S.D.K., M.S.D.) and Radiology (M.S.D.), Michigan Medicine, University of Michigan, Ann Arbor, Mich; and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (S.G.S.)
| | - Satheesh Krishna
- From the Department of Medical Imaging, The Ottawa Hospital, University of Ottawa, 1053 Carling Ave, Ottawa, ON, Canada K1H 1H6 (N.S.); Joint Department of Medical Imaging, University Health Network, Mount Sinai Hospital and Women's College Hospital, University of Toronto, Toronto, Canada (S.K.); Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Tex (I.P.); Departments of Urology (S.D.K., M.S.D.) and Radiology (M.S.D.), Michigan Medicine, University of Michigan, Ann Arbor, Mich; and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (S.G.S.)
| | - Ivan Pedrosa
- From the Department of Medical Imaging, The Ottawa Hospital, University of Ottawa, 1053 Carling Ave, Ottawa, ON, Canada K1H 1H6 (N.S.); Joint Department of Medical Imaging, University Health Network, Mount Sinai Hospital and Women's College Hospital, University of Toronto, Toronto, Canada (S.K.); Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Tex (I.P.); Departments of Urology (S.D.K., M.S.D.) and Radiology (M.S.D.), Michigan Medicine, University of Michigan, Ann Arbor, Mich; and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (S.G.S.)
| | - Samuel D Kaffenberger
- From the Department of Medical Imaging, The Ottawa Hospital, University of Ottawa, 1053 Carling Ave, Ottawa, ON, Canada K1H 1H6 (N.S.); Joint Department of Medical Imaging, University Health Network, Mount Sinai Hospital and Women's College Hospital, University of Toronto, Toronto, Canada (S.K.); Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Tex (I.P.); Departments of Urology (S.D.K., M.S.D.) and Radiology (M.S.D.), Michigan Medicine, University of Michigan, Ann Arbor, Mich; and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (S.G.S.)
| | - Matthew S Davenport
- From the Department of Medical Imaging, The Ottawa Hospital, University of Ottawa, 1053 Carling Ave, Ottawa, ON, Canada K1H 1H6 (N.S.); Joint Department of Medical Imaging, University Health Network, Mount Sinai Hospital and Women's College Hospital, University of Toronto, Toronto, Canada (S.K.); Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Tex (I.P.); Departments of Urology (S.D.K., M.S.D.) and Radiology (M.S.D.), Michigan Medicine, University of Michigan, Ann Arbor, Mich; and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (S.G.S.)
| | - Stuart G Silverman
- From the Department of Medical Imaging, The Ottawa Hospital, University of Ottawa, 1053 Carling Ave, Ottawa, ON, Canada K1H 1H6 (N.S.); Joint Department of Medical Imaging, University Health Network, Mount Sinai Hospital and Women's College Hospital, University of Toronto, Toronto, Canada (S.K.); Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Tex (I.P.); Departments of Urology (S.D.K., M.S.D.) and Radiology (M.S.D.), Michigan Medicine, University of Michigan, Ann Arbor, Mich; and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (S.G.S.)
| |
Collapse
|
25
|
A Reliable Prediction Model for Renal Cell Carcinoma Subtype Based on Radiomic Features from 3D Multiphase Enhanced CT Images. JOURNAL OF ONCOLOGY 2021; 2021:6595212. [PMID: 34594377 PMCID: PMC8478553 DOI: 10.1155/2021/6595212] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 09/06/2021] [Indexed: 12/04/2022]
Abstract
Background This study aimed to develop a prediction model to distinguish renal cell carcinoma (RCC) subtypes. Methods The radiomic features (RFs) from 5 different computed tomography (CT) phases were used in the prediction models: noncontrast phase (NCP), corticomedullary phase (CMP), nephrographic phase (NP), excretory phase (EP), and all-phase (ALL-P). Results For the ALL-P model, all of the RFs obtained from the 4 single-phase images were combined to 420 RFs. The ALL-P model performed the best of all models, with an accuracy of 0.80; the sensitivity and specificity for clear cell RCC (ccRCC) were 0.85 and 0.83; those for papillary RCC (pRCC) were 0.60 and 0.91; those for chromophobe RCC (cRCC) were 0.66 and 0.91, respectively. Binary classification experiments showed for distinguishing ccRCC vs. not-ccRCC that the area under the receiver operating characteristic curve (AUC) of the ALL-P and CMP models was 0.89, but the overall sensitivity/specificity/accuracy of the ALL-P model was better. For cRCC vs. non-cRCC, the ALL-P model had the best performance. Conclusions A reliable prediction model for RCC subtypes was constructed. The performance of the ALL-P prediction model was the best as compared to individual single-phase models and the traditional prediction model.
Collapse
|
26
|
Flitcroft JG, Verheyen J, Vemulkar T, Welbourne EN, Rossi SH, Welsh SJ, Cowburn RP, Stewart GD. Early detection of kidney cancer using urinary proteins: a truly non-invasive strategy. BJU Int 2021; 129:290-303. [PMID: 34570419 DOI: 10.1111/bju.15601] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
OBJECTIVES To review urinary protein biomarkers as potential non-invasive, easily obtainable, early diagnostic tools in renal cell carcinoma (RCC). METHODS A PubMed database search was performed up to the year 2020 to identify primary studies reporting potential urinary protein biomarkers for RCC. Separate searches were conducted to identify studies describing appropriate methods of developing cancer screening programmes and detection of cancer biomarkers. RESULTS Several urinary protein biomarkers are under validation for RCC diagnostics, e.g. aquaporin-1, perilipin-2, carbonic anhydrase-9, Raf-kinase inhibitory protein, nuclear matrix protein-22, 14-3-3 Protein β/α and neutrophil gelatinase-associated lipocalin. However, none has yet been validated or approved for clinical use due to low sensitivity or specificity, inconsistencies in appropriate study design, or lack of external validation. CONCLUSIONS Evaluation of biomarkers' feasibility, sample preparation and storage, biomarker validation, and the application of novel technologies may provide a solution that maximises the potential for a truly non-invasive biomarker in early RCC diagnostics.
Collapse
Affiliation(s)
- Jordan G Flitcroft
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, UK
| | - Jeroen Verheyen
- Department of Surgery, Addenbrookes Hospital, University of Cambridge, Cambridge, UK
| | - Tarun Vemulkar
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, UK
| | - Emma N Welbourne
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, UK
| | - Sabrina H Rossi
- Department of Surgery, Addenbrookes Hospital, University of Cambridge, Cambridge, UK
| | - Sarah J Welsh
- Department of Surgery, Addenbrookes Hospital, University of Cambridge, Cambridge, UK
| | - Russell P Cowburn
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, UK
| | - Grant D Stewart
- Department of Surgery, Addenbrookes Hospital, University of Cambridge, Cambridge, UK
| |
Collapse
|
27
|
Clearing up Clear Cell: Clarifying the Immuno-Oncology Treatment Landscape for Metastatic Clear Cell RCC. Cancers (Basel) 2021; 13:cancers13164140. [PMID: 34439293 PMCID: PMC8391664 DOI: 10.3390/cancers13164140] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 07/28/2021] [Accepted: 08/13/2021] [Indexed: 02/07/2023] Open
Abstract
Patients with advanced or malignant renal cell carcinoma at the time of diagnosis have historically had a poor prognosis. Immunonologic agents have significantly altered the therapeutic landscape and clinical outcomes of these patients. In this review, we highlight recent and upcoming clinical trials investigating the role of immunotherapies in clear cell RCC. In particular, we emphasize immunotherapy-based combinations, including immune checkpoint inhibitor (ICI) combinations, neoadjuvant, and adjuvant ICI, and ICI agents combined with anti-VEGF therapy.
Collapse
|
28
|
Zimpfer A, Glass Ä, Bastian M, Schuff-Werner P, Hakenberg OW, Maruschke M. Ceruloplasmin expression in renal cell carcinoma correlates with higher-grade and shortened survival. Biomark Med 2021; 15:841-850. [PMID: 34284640 DOI: 10.2217/bmm-2020-0514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 02/26/2021] [Indexed: 12/24/2022] Open
Abstract
Aim: We aimed to explore ceruloplasmin (CP) expression in clear cell renal cell carcinoma (ccRCC). Materials & methods: CP was analyzed in biofluid samples of 63 ccRCC patients, divided into three grading groups, and immunohistochemically, in 308 ccRCC. Results: Significant differences of mean plasma and urine CP levels in different grading groups were found. CP immunoreactivity was significantly linked to high-grade disease. Log rank tests showed a significant shorter overall survival rate in CP-positive cases (all p < 0.05). Conclusion: CP protein levels in biofluid samples confirmed differential CP expressions, depending on nuclear grade in ccRCC as previously seen in RNA expression analysis. CP expression was linked to high-grade disease and reduced survival rate in RCC.
Collapse
Affiliation(s)
- Annette Zimpfer
- Institute of Pathology, University Medicine Rostock, Strempelstr 14, Rostock, 18055, Germany
| | - Änne Glass
- Institute for Biostatistics & Informatics in Medicine, University Medicine Rostock, Ernst-Heydemann-Str 8, Rostock, 18057, Germany
| | - Manuela Bastian
- Institute of Clinical Chemistry & Laboratory Medicine, University Medicine Rostock, Ernst-Heydemann-Straße 6, Rostock,18057, Germany
| | - Peter Schuff-Werner
- Institute of Clinical Chemistry & Laboratory Medicine, University Medicine Rostock, Ernst-Heydemann-Straße 6, Rostock,18057, Germany
| | - Oliver W Hakenberg
- Department of Urology, University Medicine Rostock, Ernst-Heydemann-Str 8, Rostock, 18057, Germany
| | - Matthias Maruschke
- Department of Urology, University Medicine Rostock, Ernst-Heydemann-Str 8, Rostock, 18057, Germany
- Department of Urology, HELIOS Hanseklinikum Stralsund, Große Parower Str 47-53, Stralsund, 18435, Germany
| |
Collapse
|
29
|
Oh E, Kim JH, Um J, Jung DW, Williams DR, Lee H. Genome-Wide Transcriptomic Analysis of Non-Tumorigenic Tissues Reveals Aging-Related Prognostic Markers and Drug Targets in Renal Cell Carcinoma. Cancers (Basel) 2021; 13:3045. [PMID: 34207247 PMCID: PMC8234889 DOI: 10.3390/cancers13123045] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 06/10/2021] [Accepted: 06/12/2021] [Indexed: 01/03/2023] Open
Abstract
The relationship between expression of aging-related genes in normal tissues and cancer patient survival has not been assessed. We developed a genome-wide transcriptomic analysis approach for normal tissues adjacent to the tumor to identify aging-related transcripts associated with survival outcome, and applied it to 12 cancer types. As a result, five aging-related genes (DUSP22, MAPK14, MAPKAPK3, STAT1, and VCP) in normal tissues were found to be significantly associated with a worse survival outcome in patients with renal cell carcinoma (RCC). This computational approach was investigated using nontumorigenic immune cells purified from young and aged mice. Aged immune cells showed upregulated expression of all five aging-related genes and promoted RCC invasion compared to young immune cells. Further studies revealed DUSP22 as a regulator and druggable target of metastasis. DUSP22 gene knockdown reduced RCC invasion and the small molecule inhibitor BML-260 prevented RCC dissemination in a tumor/immune cell xenograft model. Overall, these results demonstrate that deciphering the relationship between aging-related gene expression in normal tissues and cancer patient survival can provide new prognostic markers, regulators of tumorigenesis and novel targets for drug development.
Collapse
Affiliation(s)
- Euiyoung Oh
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-Gu, Gwangju 61005, Korea;
| | - Jun-Hyeong Kim
- New Drug Targets Laboratory, School of Life Sciences, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-Gu, Gwangju 61005, Korea; (J.-H.K.); (J.U.); (D.R.W.)
| | - JungIn Um
- New Drug Targets Laboratory, School of Life Sciences, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-Gu, Gwangju 61005, Korea; (J.-H.K.); (J.U.); (D.R.W.)
| | - Da-Woon Jung
- New Drug Targets Laboratory, School of Life Sciences, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-Gu, Gwangju 61005, Korea; (J.-H.K.); (J.U.); (D.R.W.)
| | - Darren R. Williams
- New Drug Targets Laboratory, School of Life Sciences, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-Gu, Gwangju 61005, Korea; (J.-H.K.); (J.U.); (D.R.W.)
| | - Hyunju Lee
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology, 123 Cheomdangwagi-ro, Buk-Gu, Gwangju 61005, Korea;
| |
Collapse
|
30
|
Rausch M, Rutz A, Allard PM, Delucinge-Vivier C, Docquier M, Dormond O, Wolfender JL, Nowak-Sliwinska P. Molecular and Functional Analysis of Sunitinib-Resistance Induction in Human Renal Cell Carcinoma Cells. Int J Mol Sci 2021; 22:6467. [PMID: 34208775 PMCID: PMC8235637 DOI: 10.3390/ijms22126467] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 05/24/2021] [Accepted: 06/11/2021] [Indexed: 02/06/2023] Open
Abstract
Resistance in clear cell renal cell carcinoma (ccRCC) against sunitinib is a multifaceted process encompassing numerous molecular aberrations. This induces clinical complications, reducing the treatment success. Understanding these aberrations helps us to select an adapted treatment strategy that surpasses resistance mechanisms, reverting the treatment insensitivity. In this regard, we investigated the dominant mechanisms of resistance to sunitinib and validated an optimized multidrug combination to overcome this resistance. Human ccRCC cells were exposed to single or chronic treatment with sunitinib to obtain three resistant clones. Upon manifestation of sunitinib resistance, morphometric changes in the cells were observed. At the molecular level, the production of cell membrane and extracellular matrix components, chemotaxis, and cell cycle progression were dysregulated. Molecules enforcing the cell cycle progression, i.e., cyclin A, B1, and E, were upregulated. Mass spectrometry analysis revealed the intra- and extracellular presence of N-desethyl sunitinib, the active metabolite. Lysosomal sequestration of sunitinib was confirmed. After treatment with a synergistic optimized drug combination, the cell metabolic activity in Caki-1-sunitinib-resistant cells and 3D heterotypic co-cultures was reduced by >80%, remaining inactive in non-cancerous cells. These results demonstrate geno- and phenotypic changes in response to sunitinib treatment upon resistance induction. Mimicking resistance in the laboratory served as a platform to study drug responses.
Collapse
Affiliation(s)
- Magdalena Rausch
- School of Pharmaceutical Sciences, University of Geneva, CMU-Rue Michel-Servet 1, CH-1211 Geneva, Switzerland; (M.R.); (A.R.); (P.-M.A.); (J.-L.W.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU-Rue Michel-Servet 1, CH-1211 Geneva, Switzerland
- Translational Research Center in Oncohaematology, 1205 Geneva, Switzerland
| | - Adriano Rutz
- School of Pharmaceutical Sciences, University of Geneva, CMU-Rue Michel-Servet 1, CH-1211 Geneva, Switzerland; (M.R.); (A.R.); (P.-M.A.); (J.-L.W.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU-Rue Michel-Servet 1, CH-1211 Geneva, Switzerland
| | - Pierre-Marie Allard
- School of Pharmaceutical Sciences, University of Geneva, CMU-Rue Michel-Servet 1, CH-1211 Geneva, Switzerland; (M.R.); (A.R.); (P.-M.A.); (J.-L.W.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU-Rue Michel-Servet 1, CH-1211 Geneva, Switzerland
| | | | - Mylène Docquier
- iGE3 Genomics Platform, University of Geneva, 1206 Geneva, Switzerland; (C.D.-V.); (M.D.)
- Department of Genetics and Evolution, University of Geneva, 1205 Geneva, Switzerland
| | - Olivier Dormond
- Department of Visceral Surgery, Lausanne University Hospital and University of Lausanne, 1015 Lausanne, Switzerland;
| | - Jean-Luc Wolfender
- School of Pharmaceutical Sciences, University of Geneva, CMU-Rue Michel-Servet 1, CH-1211 Geneva, Switzerland; (M.R.); (A.R.); (P.-M.A.); (J.-L.W.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU-Rue Michel-Servet 1, CH-1211 Geneva, Switzerland
| | - Patrycja Nowak-Sliwinska
- School of Pharmaceutical Sciences, University of Geneva, CMU-Rue Michel-Servet 1, CH-1211 Geneva, Switzerland; (M.R.); (A.R.); (P.-M.A.); (J.-L.W.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU-Rue Michel-Servet 1, CH-1211 Geneva, Switzerland
- Translational Research Center in Oncohaematology, 1205 Geneva, Switzerland
| |
Collapse
|
31
|
A 25 year perspective on the evolution and advances in an understanding of the biology, evaluation and treatment of kidney cancer. Urol Oncol 2021; 39:548-560. [PMID: 34092483 DOI: 10.1016/j.urolonc.2021.04.038] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/20/2021] [Accepted: 04/25/2021] [Indexed: 01/20/2023]
Abstract
The diagnosis, evaluation and management of patients with renal cell carcinoma has transformed in the 21st century. Utilizing biological discoveries and technological advances, the field has moved from blunt surgical and largely ineffective medical treatments, to nuanced and fine-tuned approaches based on biology, extent of disease and patient preferences. In this review we will summarize the last 25 years of progress in kidney cancer.
Collapse
|
32
|
Obradovic A, Chowdhury N, Haake SM, Ager C, Wang V, Vlahos L, Guo XV, Aggen DH, Rathmell WK, Jonasch E, Johnson JE, Roth M, Beckermann KE, Rini BI, McKiernan J, Califano A, Drake CG. Single-cell protein activity analysis identifies recurrence-associated renal tumor macrophages. Cell 2021; 184:2988-3005.e16. [PMID: 34019793 PMCID: PMC8479759 DOI: 10.1016/j.cell.2021.04.038] [Citation(s) in RCA: 181] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 02/10/2021] [Accepted: 04/23/2021] [Indexed: 12/11/2022]
Abstract
Clear cell renal carcinoma (ccRCC) is a heterogeneous disease with a variable post-surgical course. To assemble a comprehensive ccRCC tumor microenvironment (TME) atlas, we performed single-cell RNA sequencing (scRNA-seq) of hematopoietic and non-hematopoietic subpopulations from tumor and tumor-adjacent tissue of treatment-naive ccRCC resections. We leveraged the VIPER algorithm to quantitate single-cell protein activity and validated this approach by comparison to flow cytometry. The analysis identified key TME subpopulations, as well as their master regulators and candidate cell-cell interactions, revealing clinically relevant populations, undetectable by gene-expression analysis. Specifically, we uncovered a tumor-specific macrophage subpopulation characterized by upregulation of TREM2/APOE/C1Q, validated by spatially resolved, quantitative multispectral immunofluorescence. In a large clinical validation cohort, these markers were significantly enriched in tumors from patients who recurred following surgery. The study thus identifies TREM2/APOE/C1Q-positive macrophage infiltration as a potential prognostic biomarker for ccRCC recurrence, as well as a candidate therapeutic target.
Collapse
MESH Headings
- Adult
- Apolipoproteins E/genetics
- Apolipoproteins E/metabolism
- Biomarkers, Tumor/genetics
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/pathology
- Cohort Studies
- Female
- Gene Expression/genetics
- Gene Expression Regulation, Neoplastic/genetics
- Humans
- Kidney/metabolism
- Kidney Neoplasms/pathology
- Lymphocytes, Tumor-Infiltrating/pathology
- Macrophages/metabolism
- Male
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
- Middle Aged
- Neoplasm Recurrence, Local/genetics
- Neoplasm Recurrence, Local/metabolism
- Prognosis
- Receptors, Complement/genetics
- Receptors, Complement/metabolism
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Sequence Analysis, RNA/methods
- Single-Cell Analysis/methods
- Tumor Microenvironment
- Tumor-Associated Macrophages/metabolism
- Tumor-Associated Macrophages/physiology
Collapse
Affiliation(s)
- Aleksandar Obradovic
- Columbia Center for Translational Immunology (CCTI), Columbia University Irving Medical Center (CUMC), New York, NY 10032, USA; Department of Systems Biology, HICC, New York, NY 10032, USA
| | - Nivedita Chowdhury
- Columbia Center for Translational Immunology (CCTI), Columbia University Irving Medical Center (CUMC), New York, NY 10032, USA; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | | - Casey Ager
- Columbia Center for Translational Immunology (CCTI), Columbia University Irving Medical Center (CUMC), New York, NY 10032, USA
| | - Vinson Wang
- Department of Urology, Herbert Irving Comprehensive Cancer Center (HICC), New York, NY 10032, USA
| | - Lukas Vlahos
- Department of Systems Biology, HICC, New York, NY 10032, USA
| | - Xinzheng V Guo
- Columbia Center for Translational Immunology (CCTI), Columbia University Irving Medical Center (CUMC), New York, NY 10032, USA
| | - David H Aggen
- Columbia Center for Translational Immunology (CCTI), Columbia University Irving Medical Center (CUMC), New York, NY 10032, USA
| | | | - Eric Jonasch
- Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Marc Roth
- Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Brian I Rini
- Vanderbilt University Medical Center, Nashville, TN, USA
| | - James McKiernan
- Department of Urology, Herbert Irving Comprehensive Cancer Center (HICC), New York, NY 10032, USA; HICC, Columbia University, New York, NY, USA
| | - Andrea Califano
- Department of Systems Biology, HICC, New York, NY 10032, USA; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; HICC, Columbia University, New York, NY, USA; Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY; Department of Biomedical Informatics, Columbia University, New York, NY, USA; Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA; J.P. Sulzberger Columbia Genome Center, New York, NY, USA.
| | - Charles G Drake
- Columbia Center for Translational Immunology (CCTI), Columbia University Irving Medical Center (CUMC), New York, NY 10032, USA; Department of Urology, Herbert Irving Comprehensive Cancer Center (HICC), New York, NY 10032, USA; HICC, Columbia University, New York, NY, USA.
| |
Collapse
|
33
|
Greenberg JW, Kim H, Moustafa AA, Datta A, Barata PC, Boulares AH, Abdel-Mageed AB, Krane LS. Repurposing ketoconazole as an exosome directed adjunct to sunitinib in treating renal cell carcinoma. Sci Rep 2021; 11:10200. [PMID: 33986386 PMCID: PMC8119955 DOI: 10.1038/s41598-021-89655-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 04/16/2021] [Indexed: 12/19/2022] Open
Abstract
Renal Cell Carcinoma (RCC) is the most common form of kidney cancer, with clear cell RCC (ccRCC) representing about 85% of all RCC tumors. There are limited curable treatments available for metastatic ccRCC because this disease is unresponsive to conventional targeted systemic pharmacotherapy. Exosomes (Exo) are small extracellular vesicles (EVs) secreted from cancer cells with marked roles in tumoral signaling and pharmacological resistance. Ketoconazole (KTZ) is an FDA approved anti-fungal medication which has been shown to suppress exosome biogenesis and secretion, yet its role in ccRCC has not been identified. A time-course, dose-dependent analysis revealed that KTZ selectively decreased secreted Exo in tumoral cell lines. Augmented Exo secretion was further evident by decreased expression of Exo biogenesis (Alix and nSMase) and secretion (Rab27a) markers. Interestingly, KTZ-mediated inhibition of Exo biogenesis was coupled with inhibition of ERK1/2 activation. Next, selective inhibitors were employed and showed ERK signaling had a direct role in mediating KTZ's inhibition of exosomes. In sunitinib resistant 786-O cells lines, the addition of KTZ potentiates the efficacy of sunitinib by causing Exo inhibition, decreased tumor proliferation, and diminished clonogenic ability of RCC cells. Our findings suggest that KTZ should be explored as an adjunct to current RCC therapies.
Collapse
Affiliation(s)
- Jacob W Greenberg
- Departments of Urology, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, 70112, USA
| | - Hogyoung Kim
- Departments of Urology, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, 70112, USA.
| | - Ahmed A Moustafa
- Departments of Urology, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, 70112, USA.,Zoology and Entomology Department, Faculty of Science, Helwan University, Cairo, 11790, Egypt
| | - Amrita Datta
- Departments of Urology, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, 70112, USA.,College of Nursing and Health, Loyola University New Orleans, New Orleans, LA, 70118, USA
| | - Pedro C Barata
- Department of Internal Medicine, Section of Hematology/Oncology, Tulane University School of Medicine, New Orleans, LA, 70012, USA
| | - A Hamid Boulares
- The Stanley Scott Cancer Center/Louisiana Cancer Research Center, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Asim B Abdel-Mageed
- Departments of Urology, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, 70112, USA.,Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, 70012, USA
| | - Louis S Krane
- Departments of Urology, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, 70112, USA.
| |
Collapse
|
34
|
Xu W, Puligandla M, Halbert B, Haas NB, Flaherty KT, Uzzo RG, Dutcher JP, DiPaola RS, Sabbisetti V, Bhatt RS. Plasma KIM-1 Is Associated with Recurrence Risk after Nephrectomy for Localized Renal Cell Carcinoma: A Trial of the ECOG-ACRIN Research Group (E2805). Clin Cancer Res 2021; 27:3397-3403. [PMID: 33832947 DOI: 10.1158/1078-0432.ccr-21-0025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/26/2021] [Accepted: 04/06/2021] [Indexed: 12/24/2022]
Abstract
PURPOSE No circulating biomarkers are currently available to identify patients at highest risk of recurrence after nephrectomy for renal cell carcinoma (RCC). Kidney injury molecule-1 (KIM-1) is overexpressed in RCC and its ectodomain circulates in plasma. We investigated whether plasma KIM-1 is a prognostic biomarker in patients with localized RCC after nephrectomy. EXPERIMENTAL DESIGN The ECOG-ACRIN E2805 (ASSURE) trial evaluated adjuvant sunitinib, sorafenib, or placebo in resected high-risk RCC. KIM-1 levels were measured from banked plasma at trial enrollment 4-12 weeks after nephrectomy. Lognormal accelerated failure time models were used to test for association between KIM-1 and disease-free survival (DFS) as well as overall survival (OS). RESULTS Plasma from 418 patients was analyzed. Higher post-nephrectomy KIM-1 was associated with worse DFS across all study arms after adjustment for Fuhrman grade, T stage, N stage, and tumor histology [survival time ratio 0.56 for 75th vs. 25th percentile of KIM-1; 95% confidence interval (CI), 0.42-0.73; P < 0.001]. The association between KIM-1 and DFS was stronger among patients with pathologic nodal involvement (P interaction = 0.0086). The addition of post-nephrectomy KIM-1 improved the concordance of clinical prognostic models [Stage, Size, Grade, and Necrosis (SSIGN) concordance 0.57 vs. 0.43, P = 0.05; UCLA International Staging System (UISS) concordance 0.60 vs. 0.40, P = 0.0005]. Higher post-nephrectomy KIM-1 was also associated with worse OS after multivariable adjustment (survival time ratio 0.71 for 75th vs. 25th percentile of KIM-1; 95% CI, 0.56-0.91; P < 0.001). CONCLUSIONS Post-nephrectomy plasma KIM-1 is associated with DFS and OS in RCC, and may be a biomarker for microscopic residual disease.
Collapse
Affiliation(s)
- Wenxin Xu
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Brian Halbert
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Naomi B Haas
- University of Pennsylvania/Abramson Cancer Center, Philadelphia, Philadelphia
| | | | | | | | | | | | - Rupal S Bhatt
- Beth Israel Deaconess Medical Center, Boston, Massachusetts.
| |
Collapse
|
35
|
Xia S, Lin Y, Lin J, Li X, Tan X, Huang Z. Increased Expression of TICRR Predicts Poor Clinical Outcomes: A Potential Therapeutic Target for Papillary Renal Cell Carcinoma. Front Genet 2021; 11:605378. [PMID: 33505430 PMCID: PMC7831611 DOI: 10.3389/fgene.2020.605378] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023] Open
Abstract
Background: Papillary renal cell carcinoma (PRCC), although the second-most common type of renal cell carcinoma, still lacks specific biomarkers for diagnosis, treatment, and prognosis. TopBP1-interacting checkpoint and replication regulator (TICRR) is a DNA replication initiation regulator upregulated in various cancers. We aimed to evaluate the role of TICRR in PRCC tumorigenesis and prognosis. Methods: Based on the Kidney Renal Papillary cell carcinoma Project (KIRP) on The Cancer Genome Atlas (TCGA) database, we determined the expression of TICRR using the Wilcoxon rank sum test. The biological functions of TICRR were evaluated using the Metascape database and Gene Set Enrichment Analysis (GSEA). The association between TICRR and immune cell infiltration was investigated by single sample GSEA. Logistic analysis was applied to study the correlation between TICRR expression and clinicopathological characteristics. Finally, Cox regression analysis, Kaplan–Meier analysis, and nomograms were used to determine the predictive value of TICRR on clinical outcomes in PRCC patients. Results:TICRR expression was significantly elevated in PRCC tumors (P < 0.001). Functional annotation indicated enrichment with negative regulation of cell division, cell cycle, and corresponding pathways in the high TICRR expression phenotype. High TICRR expression in PRCC was associated with female sex, younger age, and worse clinical stages. Cox regression analysis revealed that TICRR was a risk factor for overall survival [hazard ratio (HR): 2.80, P = 0.002], progression-free interval (HR: 2.86, P < 0.001), and disease-specific survival (HR: 7.03, P < 0.001), especially in patients with male sex, age below 60 years, clinical stages II–IV and clinical T stage T1–T2. Conclusion: Increased TICRR expression in PRCC might play a role in tumorigenesis by regulating the cell cycle and has prognostic value for clinical outcomes.
Collapse
Affiliation(s)
- Shuang Xia
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yan Lin
- Department of Nephrology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiaqiong Lin
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiaoyong Li
- Department of Surgery, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xuexian Tan
- Department of Pathology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zena Huang
- Department of General Medicine, Guangdong Provincial Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
36
|
Zong J, Fan Z, Zhang Y. Serum Tumor Markers for Early Diagnosis of Primary Hepatocellular Carcinoma. J Hepatocell Carcinoma 2020; 7:413-422. [PMID: 33376710 PMCID: PMC7755348 DOI: 10.2147/jhc.s272762] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 12/05/2020] [Indexed: 12/11/2022] Open
Abstract
Primary hepatocellular carcinoma (HCC) is one of the most frequently occurring pernicious tumors in the world. It is typically very insidious in the early stages with no obvious symptoms. Its development and metastasis are very rapid. Upon diagnosis, most patients have already reached a local advanced stage or have established distant metastases. The treatment of HCC is limited, with poor prognosis and short natural survival time. In order to improve the efficiency of early diagnosis, it is particularly significant to choose economic and effective diagnosis methods. Ultrasound, magnetic resonance imaging, and computed tomography are usually used in the clinic, but these methods are extremely limited in the diagnosis of HCC. Tumor markers have become the main effective early clinical diagnosis method. Potential serum tumor markers include alpha fetoprotein heterogeneity, Golgi protein 73, phosphatidylinositol proteoglycan (GPC-3), osteopontin, abnormal prothrombin, and heat shock protein. These tumor markers provide new ideas and methods for the diagnosis of HCC. A combination of multiple markers can make up for the deficiency of single marker detection and provide a new strategy for the prognosis and auxiliary diagnosis of HCC. This review introduces protein tumor markers utilized over the past five years.
Collapse
Affiliation(s)
- Jingjing Zong
- Department of General Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing210009, People’s Republic of China
| | - Zhe Fan
- Department of General Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing210009, People’s Republic of China
- Department of General Surgery, The Third People’s Hospital of Dalian, Dalian Medical University, Dalian116033, People’s Republic of China
| | - Yewei Zhang
- Department of General Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing210009, People’s Republic of China
| |
Collapse
|
37
|
Nayak B, Panaiyadiyan S, Singh P, Karmakar S, Kaushal S, Seth A. Role of circulating tumor cells in patients with metastatic clear-cell renal cell carcinoma. Urol Oncol 2020; 39:135.e9-135.e15. [PMID: 33250345 DOI: 10.1016/j.urolonc.2020.10.077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/07/2020] [Accepted: 10/30/2020] [Indexed: 11/26/2022]
Abstract
PURPOSE Circulating tumor cells (CTC) have been demonstrated to have prognostic and predictive role in certain human cancers. However, studies exploring their role in metastatic renal cell carcinoma (mRCC) are scarce. We aimed to evaluate the prognostic and predictive role of CTC in mRCC. MATERIALS AND METHODS In this prospective study, 35 patients with mRCC were analyzed for the presence of CTC before starting tyrosine kinase inhibitors (TKI). Progression-free and overall survival rates were estimated using the Kaplan-Meier curves and log-rank test. The prediction to TKI therapy was calculated with the response to treatment determined by standard imaging techniques. RESULTS Outcomes were assessed according to the CTC positivity at baseline, before the patients started TKI for mRCC. At a mean follow-up of 12.4 ± 4.1 months, disease progression was noted in 17 patients (48.6%) including 8 deaths (22.9%). CTC positive patients had a significantly lower progression-free survival rate (12.5% vs. 64.1%, respectively; P = 0.009) but not in the overall survival rate (75% vs. 76.3%, respectively; P = 0.88) in the Kaplan-Meier estimation curves. CTC positivity at baseline significantly predicted a poorer response to TKI (87.5% vs. 37.1%, P = 0.01). The multivariate Cox proportional hazards analysis showed that CTC at baseline was the most significant predictor of progression-free survival (hazard ratio 4.17, 95% confidence interval 1.41-11.99, P = 0.01). CONCLUSIONS Baseline CTC detection can be an important prognostic factor of progression-free survival and significant predictor of poor response to TKI in patients with metastatic RCC.
Collapse
Affiliation(s)
- Brusabhanu Nayak
- Department of Urology, All India Institute of Medical Sciences, New Delhi, India
| | - Sridhar Panaiyadiyan
- Department of Urology, All India Institute of Medical Sciences, New Delhi, India.
| | - Prabhjot Singh
- Department of Urology, All India Institute of Medical Sciences, New Delhi, India
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Seema Kaushal
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Amlesh Seth
- Department of Urology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
38
|
The Role of Chemokine Receptor CXCR3 and Its Ligands in Renal Cell Carcinoma. Int J Mol Sci 2020; 21:ijms21228582. [PMID: 33202536 PMCID: PMC7696621 DOI: 10.3390/ijms21228582] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 11/08/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023] Open
Abstract
The major invasive subtype of kidney cancer is renal cell carcinoma (RCC). The essential components of cancer development are chronic inflammation and neoangiogenesis. It has been suggested that the chemokine ligand 9, -10, –11 (CXCL9–11) and chemokine receptor 3 (CXCR3) chemokines receptor expressed on monocytes, T and NK cells may be involved in the inhibition of angiogenesis. However, to date, little is known about the potential clinical significance of these chemokines and their receptor in renal cell carcinoma. Therefore, in this review, we described the role of CXCR3 and its ligands in pathogenesis of RCC. We performed an extensive search of the current literature in our investigation, using the MEDLINE/PubMed database. The changes of chemokines and their specific receptor in renal cell carcinoma were observed. Published studies revealed an increased expression of CXCR3 and elevated concentration of its ligands in RCC. The association between treatment of RCC and CXCL9–11/CXCR3 concentration and expression was also observed. Moreover, CXCR3 and its ligands levels were related to patient’s prognosis, risk of metastasis and tumor growth. This review describes the potential role of CXCR3 and its ligands in pathogenesis of RCC, as well as their potential immune-therapeutic significance. However, future studies should aim to confirm the clinical and prognostic role of CXCL9–11/CXCR3 in renal cell carcinoma.
Collapse
|
39
|
Tabakin AL, Stein MN, Anderson CB, Drake CG, Singer EA. Cytoreductive nephrectomy for metastatic renal cell carcinoma, the ultimate urologic 'Choosing Wisely' campaign: a narrative review. Transl Cancer Res 2020; 9:7337-7349. [PMID: 33354523 PMCID: PMC7751973 DOI: 10.21037/tcr-20-2343] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 10/21/2020] [Indexed: 01/04/2023]
Abstract
In the early 2000s, cytoreductive nephrectomy in addition to systemic cytokines became standard of care for treating metastatic renal cell carcinoma. Since that time, the development of novel systemic targeted therapies and immuno-oncologic agents have challenged the utility of cytoreductive nephrectomy in clinical practice. In 2019, the controversial CARMENA study was published, providing the first level one evidence suggesting that cytoreductive nephrectomy combined with targeted therapy yielded no survival advantage over targeted therapy alone in intermediate and poor risk metastatic renal cell carcinoma patients. Later that year, the SURTIME trial demonstrated that patients undergoing targeted therapy with delayed nephrectomy maintained a survival advantage over those that underwent upfront cytoreductive nephrectomy followed by targeted therapy. Both of these studies underscored the importance of patient selection and timing of cytoreductive nephrectomy and systemic therapy. As new immuno-oncologic agents are trialed, particularly in combination, the role of cytoreductive nephrectomy will continue to be questioned. In this narrative review, we discuss the evolution of the role of cytoreductive nephrectomy in treating metastatic renal cell carcinoma through the context of the ever-changing landscape of targeted therapies and immuno-oncologic agents. We assess the evidence for cytoreductive nephrectomy with respect to patient factors, timing of surgery, and combination with other therapies.
Collapse
Affiliation(s)
- Alexandra L. Tabakin
- Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08903, USA
| | - Mark N. Stein
- Division of Medical Oncology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Christopher B. Anderson
- Department of Urology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Charles G. Drake
- Division of Medical Oncology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Eric A. Singer
- Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08903, USA
| |
Collapse
|
40
|
Akkoc RF, Aydin S, Goksu M, Ozcan Yildirim S, Eroksuz Y, Ogeturk M, Ugur K, Dagli AF, Yakar B, Sahin I, Aydin S. Can renalase be a novel candidate biomarker for distinguishing renal tumors? Biotech Histochem 2020; 96:520-525. [PMID: 33956551 DOI: 10.1080/10520295.2020.1825805] [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: 01/05/2023] Open
Abstract
Renalase (RNLS) is synthesized mainly in renal tissues. The function of RNLS in cancerous renal tissues has not been investigated. We investigated the synthesis of RNLS in chromophobe renal cell carcinoma, papillary renal cell carcinoma and clear cell renal cell carcinoma with Fuhrman grades (FG): FG1, nucleoli are absent or inconspicuous and basophilic; FG2, nucleoli are conspicuous and eosinophilic and visible but not prominent; FG3, nucleoli are conspicuous and eosinophilic; FG4, extreme nuclear pleomorphism, multinucleate giant cells, and/or rhabdoid and/or sarcomatoid differentiation. We used 90 tissue samples including 15 healthy controls, 15 chromophobe renal cell carcinoma tissues and 10 papillary renal cell carcinoma renal tissues: 12 FG1, 14 FG 2, 14 FG 3 and 10 FG4. RNLS in the tissue samples was measured using enzyme linked immunosorbent assay and immunostaining of RNLS in these tissues. RNLS was significantly greater in the chromophobe renal cell carcinoma and papillary renal cell carcinoma tissues than the control. The least amount of RNLS was found in the renal tissues of clear cell renal cell carcinoma FG1; the amount of RNLS increased as the FG grades increased. Because RNLS increased significantly in renal tissues due to cancer, except for clear cell renal cell carcinoma FG1, RNLS may be useful biomarker for distinguishing grades of renal cancer. Because RNLS increases cell survival, anti-RNLS preparations may be useful for treating cancer in the future.
Collapse
Affiliation(s)
- R F Akkoc
- Department of Anatomy, School of Medicine, Firat University, Elazig, Turkey
| | - S Aydin
- Department of Cardiovascular Surgery, Elazig Fethi Sekin City Hospital, Elazig, Turkey
| | - M Goksu
- Department of Pediatric Surgery, School of Medicine, Adiyaman University, Adiyaman, Turkey
| | - S Ozcan Yildirim
- Department of Histology and Embryology, School of Medicine, Firat University, Elazig, Turkey
| | - Y Eroksuz
- Department of Pathology, School of Veterinary Medicine, Firat University, Elazig, Turkey
| | - M Ogeturk
- Department of Anatomy, School of Medicine, Firat University, Elazig, Turkey
| | - K Ugur
- Department of Endocrinology and Metabolism Disease, School of Medicine, Firat University, Elazig, Turkey
| | - A F Dagli
- Department of Pathology, School of Medicine, Firat University, Elazig, Turkey
| | - B Yakar
- Department of Family Medicine, School of Medicine, Firat University, Elazig, Turkey
| | - I Sahin
- Department of Medical Biochemistry and Clinical Biochemistry, Firat Hormones Research Group, School of Medicine, Firat University Elazig, Elazig, Turkey.,Department of Medical Biology, School of Medicine, Erzincan Binali Yildirim University, Erzincan, Turkey
| | - S Aydin
- Department of Medical Biochemistry and Clinical Biochemistry, Firat Hormones Research Group, School of Medicine, Firat University Elazig, Elazig, Turkey
| |
Collapse
|
41
|
Barth DA, Drula R, Ott L, Fabris L, Slaby O, Calin GA, Pichler M. Circulating Non-coding RNAs in Renal Cell Carcinoma-Pathogenesis and Potential Implications as Clinical Biomarkers. Front Cell Dev Biol 2020; 8:828. [PMID: 33042985 PMCID: PMC7523432 DOI: 10.3389/fcell.2020.00828] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 08/04/2020] [Indexed: 12/13/2022] Open
Abstract
Liquid biopsy-the determination of circulating cells, proteins, DNA or RNA from biofluids through a "less invasive" approach-has emerged as a novel approach in all cancer entities. Circulating non-(protein) coding RNAs including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and YRNAs can be passively released by tissue or cell damage or actively secreted as cell-free circulating RNAs, bound to lipoproteins or carried by exosomes. In renal cell carcinoma (RCC), a growing body of evidence suggests circulating non-coding RNAs (ncRNAs) such as miRNAs, lncRNAs, and YRNAs as promising and easily accessible blood-based biomarkers for the early diagnosis of RCC as well as for the prediction of prognosis and treatment response. In addition, circulating ncRNAs could also play a role in RCC pathogenesis and progression. This review gives an overview over the current study landscape of circulating ncRNAs and their involvement in RCC pathogenesis as well as their potential utility as future biomarkers in RCC diagnosis and treatment.
Collapse
Affiliation(s)
- Dominik A Barth
- Research Unit of Non-Coding RNAs and Genome Editing, Division of Clinical Oncology, Department of Internal Medicine, Comprehensive Cancer Center Graz, Medical University of Graz, Graz, Austria.,Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Rares Drula
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Research Centre for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Leonie Ott
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Linda Fabris
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ondrej Slaby
- Central European Institute of Technology, Masaryk University, Brno, Czechia.,Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - George A Calin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Martin Pichler
- Research Unit of Non-Coding RNAs and Genome Editing, Division of Clinical Oncology, Department of Internal Medicine, Comprehensive Cancer Center Graz, Medical University of Graz, Graz, Austria.,Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
42
|
Nizioł J, Ossoliński K, Tripet BP, Copié V, Arendowski A, Ruman T. Nuclear magnetic resonance and surface-assisted laser desorption/ionization mass spectrometry-based serum metabolomics of kidney cancer. Anal Bioanal Chem 2020; 412:5827-5841. [PMID: 32661677 PMCID: PMC7413895 DOI: 10.1007/s00216-020-02807-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/19/2020] [Accepted: 07/06/2020] [Indexed: 12/24/2022]
Abstract
Kidney cancer is one of the most frequently diagnosed and the most lethal urinary cancer. Despite all the efforts made, no serum-specific biomarker is currently used in the clinical management of patients with this tumor. In this study, comprehensive high-resolution proton nuclear magnetic resonance spectroscopy (1H NMR) and silver-109 nanoparticle-enhanced steel target laser desorption/ionization mass spectrometry (109AgNPET LDI MS) approaches were conducted, in conjunction with multivariate data analysis, to discriminate the global serum metabolic profiles of kidney cancer (n = 50) and healthy volunteers (n = 49). Eight potential biomarkers have been identified using 1H NMR metabolomics and nine mass spectral features which differed significantly (p < 0.05) between kidney cancer patients and healthy volunteers, as observed by LDI MS. A partial least squares discriminant analysis (OPLS-DA) model generated from metabolic profiles obtained by both analytical approaches could robustly discriminate normal from cancerous samples (Q2 > 0.7), area under the receiver operative characteristic curve (ROC) AUC > 0.96. Compared with healthy human serum, kidney cancer serum had higher levels of glucose and lower levels of choline, glycerol, glycine, lactate, leucine, myo-inositol, and 1-methylhistidine. Analysis of differences between these metabolite levels in patients with different types and grades of kidney cancer was undertaken. Our results, derived from the combination of LDI MS and 1H NMR methods, suggest that serum biomarkers identified herein appeared to have great potential for use in clinical prognosis and/or diagnosis of kidney cancer. Graphical abstract.
Collapse
Affiliation(s)
- Joanna Nizioł
- Faculty of Chemistry, Rzeszów University of Technology, 6 Powstańców Warszawy Ave, 35-959, Rzeszów, Poland.
| | - Krzysztof Ossoliński
- Department of Urology, John Paul II Hospital, Grunwaldzka 4 St, 36-100, Kolbuszowa, Poland
| | - Brian P Tripet
- The Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT, 59717, USA
| | - Valérie Copié
- The Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT, 59717, USA
| | - Adrian Arendowski
- Faculty of Chemistry, Rzeszów University of Technology, 6 Powstańców Warszawy Ave, 35-959, Rzeszów, Poland
| | - Tomasz Ruman
- Faculty of Chemistry, Rzeszów University of Technology, 6 Powstańców Warszawy Ave, 35-959, Rzeszów, Poland
| |
Collapse
|
43
|
Soleimani M, Nappi L, Kollmannsberger C. Avelumab and axitinib combination therapy for the treatment of advanced renal cell carcinoma. Future Oncol 2020; 16:3021-3034. [PMID: 32856478 DOI: 10.2217/fon-2020-0586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Owing to an improved understanding of the immunobiological profile of renal cell carcinoma (RCC), the past few years have ushered in significant changes in systemic therapies for advanced stage RCC. First-line treatment with single-agent tyrosine kinase inhibitors (TKI) has been virtually replaced for most patients by immunotherapy combinations. The first of such treatments was the dual immune checkpoint inhibitor combination of ipilimumab and nivolumab. More recently, the combination of an immune checkpoint inhibitor and a TKI has also moved into the first-line setting. This review summarizes the pharmacologic properties, evidence for use and safety of avelumab, a PD-L1 inhibitor and axitinib a small-molecule TKI, each as monotherapy, and in combination for the management of metastatic RCC.
Collapse
Affiliation(s)
- Maryam Soleimani
- Department of Medical Oncology, BC Cancer Vancouver Centre, Vancouver, BC, V5Z 4E6, Canada
| | - Lucia Nappi
- Department of Medical Oncology, BC Cancer Vancouver Centre, Vancouver, BC, V5Z 4E6, Canada.,Department of Urological Sciences, The Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H 3Z6, Canada
| | | |
Collapse
|
44
|
Khetani VV, Portal DE, Shah MR, Mayer T, Singer EA. Combination drug regimens for metastatic clear cell renal cell carcinoma. World J Clin Oncol 2020; 11:541-562. [PMID: 32879843 PMCID: PMC7443831 DOI: 10.5306/wjco.v11.i8.541] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 05/11/2020] [Accepted: 07/18/2020] [Indexed: 02/06/2023] Open
Abstract
Renal cell carcinomas (RCC) make up about 90% of kidney cancers, of which 80% are of the clear cell subtype. About 20% of patients are already metastatic at the time of diagnosis. Initial treatment is often cytoreductive nephrectomy, but systemic therapy is required for advanced RCC. Single agent targeted therapies are moderately toxic and only somewhat effective, leading to development of immunotherapies and combination therapies. This review identifies limitations of monotherapies for metastatic renal cell carcinoma, discusses recent advances in combination therapies, and highlights therapeutic options under development. The goal behind combining various modalities of systemic therapy is to potentiate a synergistic antitumor effect. However, combining targeted therapies may cause increased toxicity. The initial attempts to create therapeutic combinations based on inhibition of the vascular endothelial growth factor or mammalian target of rapamycin pathways were largely unsuccessful in achieving a profile of increased synergy without increased toxicity. To date, five combination therapies have been approved by the U.S. Food and Drug Administration, with the most recently approved therapies being a combination of checkpoint inhibition plus targeted therapy. Several other combination therapies are under development, including some in the phase 3 stage. The new wave of combination therapies for metastatic RCC has the potential to increase response rates and improve survival outcomes while maintaining tolerable side effect profiles.
Collapse
Affiliation(s)
- Viraj V Khetani
- Rutgers Cancer Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ 08903, United States
| | - Daniella E Portal
- Rutgers Cancer Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ 08903, United States
| | - Mansi R Shah
- Rutgers Cancer Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ 08903, United States
| | - Tina Mayer
- Rutgers Cancer Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ 08903, United States
| | - Eric A Singer
- Rutgers Cancer Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ 08903, United States
| |
Collapse
|
45
|
Loughlin KR. Commentary: Contemporary trends in percutaneous renal mass biopsy in the United States by Ozambela et al. Urol Oncol 2020; 38:844-845. [PMID: 32826163 DOI: 10.1016/j.urolonc.2020.07.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Kevin R Loughlin
- Vascular Biology Research Laboratory, Harvard Medical School, Boston, MA.
| |
Collapse
|
46
|
Pajdzik K, Wilamowski M, Żurawek D, Stopa KB, Nodzyński M, Kalita A, Jura J. Anterior gradient 2 promotes tumorigenesis through upregulation of CCAAT-enhancer binding protein beta and hypoxia-inducible factor-2α and subsequent secretion of interleukin-6, interleukin-8, and vascular endothelial growth factor in the Caki-1 clear cell renal cell carcinoma cell line. IUBMB Life 2020; 72:1807-1818. [PMID: 32593213 DOI: 10.1002/iub.2331] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 05/08/2020] [Accepted: 05/24/2020] [Indexed: 12/27/2022]
Abstract
It has been previously established that hypoxia leads to tumor development, treatment resistance, and a poor prognosis. Under oxygen deprivation, hypoxia-inducible factors (HIFs) are stimulated to activate the genes necessary for tumor development in a low-oxygen environment. These genes encode regulators of angiogenesis, epithelial-mesenchymal transition, and cellular metabolism. A disulfide isomerase, anterior gradient 2 (AGR2), has been shown to increase hypoxia-inducible factor 1, alpha subunit (HIF-1α) stability in breast cancer. Our goal was to determine if AGR2 affects the level of transcription factor hypoxia-inducible factor 2, alpha subunit (HIF-2α). As a model, we used the clear cell renal cell carcinoma (ccRCC) cell line Caki-1. The cells were transduced with lentiviral vector (Tet-On) encoding AGR2. After induction of AGR2 expression, cells were grown under either hypoxic (0.5% O2 ) or normoxic (21% O2 ) conditions. Our data showed that AGR2 upregulated both HIF-1α and HIF-2α expression in Caki-1 cells increasing the expression of HIF-activated genes (glucose transporter 1, phosphoglycerate kinase 1, vascular endothelial growth factor A, and transforming growth factor-alpha) under the hypoxic conditions. Under the normoxic conditions, AGR2 strongly activated CCAAT-enhancer binding protein beta (C/EBPβ). Upregulation of C/EBPβ correlated with increased expression and secretion of the interleukin-6 and interleukin-8, inducing angiogenesis and inflammation in Caki-1 cells. In summary, our studies revealed that AGR2 has essential functions in ccRCC progression through upregulation of C/EBPβ and HIF-2α expressions, which affects cell signaling and metabolism.
Collapse
Affiliation(s)
- Kinga Pajdzik
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Mateusz Wilamowski
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Dariusz Żurawek
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Kinga B Stopa
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Michał Nodzyński
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Agata Kalita
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Jolanta Jura
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| |
Collapse
|
47
|
Patel HV, Doppalapudi SK, Singer EA. Taking a SPOP at renal cell carcinoma - unraveling a novel pathway for Tumor progression in clear cell RCC. EBioMedicine 2020; 56:102823. [PMID: 32512506 PMCID: PMC7276556 DOI: 10.1016/j.ebiom.2020.102823] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 05/19/2020] [Indexed: 12/03/2022] Open
Affiliation(s)
- Hiren V Patel
- Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08902, United States
| | - Sai K Doppalapudi
- Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08902, United States
| | - Eric A Singer
- Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08902, United States.
| |
Collapse
|
48
|
Light A, Ahmed A, Dasgupta P, Elhage O. The genetic landscapes of urological cancers and their clinical implications in the era of high-throughput genome analysis. BJU Int 2020; 126:26-54. [PMID: 32306543 DOI: 10.1111/bju.15084] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVE With the advent of high-throughput genome analysis, we are increasingly able to sequence and hence understand the pathogenic processes underlying individual cancers. Recently, consortiums such as The Cancer Genome Atlas (TCGA) have performed large-scale projects to this end, providing significant amounts of information regarding the genetic landscapes of several cancers. PATIENTS AND METHODS We performed a narrative review of studies from the TCGA and other major studies. We aimed to summarise data exploring the clinical implications of specific genetic alterations, both prognostically and therapeutically, in four major urological cancers. These were renal cell carcinoma, muscle-invasive bladder cancer/carcinoma, prostate cancer, and testicular germ cell tumours. RESULTS With these four urological cancers, great strides have been made in the molecular characterisation of tumours. In particular, recent studies have focussed on identifying molecular subtypes of tumours with characteristic genetic alterations and differing prognoses. Other prognostic alterations have also recently been identified, including those pertaining to epigenetics and microRNAs. In regard to treatment, numerous options are emerging for patients with these cancers such as including immune checkpoint inhibition, epigenetic-based treatments, and agents targeting MAPK, PI3K, and DNA repair pathways. There are a multitude of trials underway investigating the effects of these novel agents, the results of which are eagerly awaited. CONCLUSIONS As medicine chases the era of personalised care, it is becoming increasingly important to provide individualised prognoses for patients. Understanding how specific genetic alterations affects prognosis is key for this. It will also be crucial to provide highly targeted treatments against the specific genetics of a patient's tumour. With work performed by the TCGA and other large consortiums, these aims are gradually being achieved. Our review provides a succinct overview of this exciting field that may underpin personalised medicine in urological oncology.
Collapse
Affiliation(s)
- Alexander Light
- Department of Surgery, Cambridge University Hospitals NHS Foundation Trust, University of Cambridge, Cambridge, UK.,Bedford Hospital NHS Trust, Bedford Hospital, Bedford, UK
| | - Aamir Ahmed
- Centre for Stem Cell and Regenerative Medicine, King's College London, London, UK
| | - Prokar Dasgupta
- Department of Urology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Oussama Elhage
- Department of Urology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| |
Collapse
|
49
|
Amaro F, Pinto J, Rocha S, Araújo AM, Miranda-Gonçalves V, Jerónimo C, Henrique R, Bastos MDL, Carvalho M, Guedes de Pinho P. Volatilomics Reveals Potential Biomarkers for Identification of Renal Cell Carcinoma: An In Vitro Approach. Metabolites 2020; 10:metabo10050174. [PMID: 32349455 PMCID: PMC7281256 DOI: 10.3390/metabo10050174] [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: 04/14/2020] [Revised: 04/22/2020] [Accepted: 04/24/2020] [Indexed: 12/11/2022] Open
Abstract
The identification of noninvasive biomarkers able to detect renal cell carcinoma (RCC) at an early stage remains an unmet clinical need. The recognition that altered metabolism is a core hallmark of cancer boosted metabolomic studies focused in the search for cancer biomarkers. The present work aims to evaluate the performance of the volatile metabolites present in the extracellular medium to discriminate RCC cell lines with distinct histological subtypes (clear cell and papillary) and metastatic potential from non-tumorigenic renal cells. Hence, volatile organic compounds (VOCs) and volatile carbonyl compounds (VCCs) were extracted by headspace solid-phase microextraction (HS-SPME) and analyzed by gas chromatography-mass spectrometry (GC-MS). Multivariate and univariate analysis unveiled a panel of metabolites responsible for the separation between groups, mostly belonging to ketones, alcohols, alkanes and aldehydes classes. Some metabolites were found similarly altered for all RCC cell lines compared to non-tumorigenic cells, namely 2-ethylhexanol, tetradecane, formaldehyde, acetone (increased) and cyclohexanone and acetaldehyde (decreased). Furthermore, significantly altered levels of cyclohexanol, decanal, decane, dodecane and 4-methylbenzaldehyde were observed in all metastatic RCC cell lines when compared with the non-metastatic ones. Moreover, some alterations in the volatile composition were also observed between RCC histological subtypes. Overall, our results demonstrate the potential of volatile profiling for identification of noninvasive candidate biomarkers for early RCC diagnosis.
Collapse
Affiliation(s)
- Filipa Amaro
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.R.); (A.M.A.); (M.d.L.B.); (P.G.d.P.)
- Correspondence: (F.A.); (J.P.); (M.C.); Tel.: +351-220-428-500 (F.A. & J.P.); +351-225-071-300 (M.C.)
| | - Joana Pinto
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.R.); (A.M.A.); (M.d.L.B.); (P.G.d.P.)
- Correspondence: (F.A.); (J.P.); (M.C.); Tel.: +351-220-428-500 (F.A. & J.P.); +351-225-071-300 (M.C.)
| | - Sílvia Rocha
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.R.); (A.M.A.); (M.d.L.B.); (P.G.d.P.)
- Master in Oncology, Institute of Biomedical Sciences Abel Salazar–University of Porto (ICBAS-UP), 4050-313 Porto, Portugal
| | - Ana Margarida Araújo
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.R.); (A.M.A.); (M.d.L.B.); (P.G.d.P.)
| | - Vera Miranda-Gonçalves
- Cancer Biology & Epigenetics Group, Research Centre (CI-IPOP) Portuguese Oncology Institute of Porto (IPO Porto), 4200-072 Porto, Portugal; (V.M.-G.); (C.J.); (R.H.)
| | - Carmen Jerónimo
- Cancer Biology & Epigenetics Group, Research Centre (CI-IPOP) Portuguese Oncology Institute of Porto (IPO Porto), 4200-072 Porto, Portugal; (V.M.-G.); (C.J.); (R.H.)
- Department of Pathology and Molecular Immunology-Biomedical Sciences Institute (ICBAS), University of Porto, 4050-313 Porto, Portugal
| | - Rui Henrique
- Cancer Biology & Epigenetics Group, Research Centre (CI-IPOP) Portuguese Oncology Institute of Porto (IPO Porto), 4200-072 Porto, Portugal; (V.M.-G.); (C.J.); (R.H.)
- Department of Pathology and Molecular Immunology-Biomedical Sciences Institute (ICBAS), University of Porto, 4050-313 Porto, Portugal
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto), 4200-072 Porto, Portugal
| | - Maria de Lourdes Bastos
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.R.); (A.M.A.); (M.d.L.B.); (P.G.d.P.)
| | - Márcia Carvalho
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.R.); (A.M.A.); (M.d.L.B.); (P.G.d.P.)
- UFP Energy, Environment and Health Research Unit (FP-ENAS), University Fernando Pessoa, 349, 4249-004 Porto, Portugal
- Correspondence: (F.A.); (J.P.); (M.C.); Tel.: +351-220-428-500 (F.A. & J.P.); +351-225-071-300 (M.C.)
| | - Paula Guedes de Pinho
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.R.); (A.M.A.); (M.d.L.B.); (P.G.d.P.)
| |
Collapse
|
50
|
Wei X, Ren X, Ding Y, Wang H, Li Y, Li X, Gao Y. Comparative outcomes of radio frequency ablation versus partial nephrectomy for T1 renal tumors: a systematic review. Transl Androl Urol 2020; 8:601-608. [PMID: 32038956 DOI: 10.21037/tau.2019.10.13] [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: 01/20/2023] Open
Abstract
Background The role of radio frequency ablation (RFA) in small renal tumors remains controversial. This systematic review was performed to compare clinical outcomes of RFA versus partial nephrectomy (PN) for the treatment of T1 renal tumors. Methods A total of 11 studies including 2,397 patients were analyzed in this systematic review after searching the databases of PubMed, EMBASE and Web of Science. P value and odds ratio (OR)/hazard ratio (HR) with 95% confidence interval (CI) were used to evaluate the strength of the association. Results A total of six studies (2,056 patients) provided either survival curves or HR and its 95% CI, demonstrating that the majority of the patients with RFA treatment tended to exhibit a similar long-term survival rate to those with PN treatment. In addition, according to four studies, no differences were found in the overall rate of complications between the two groups. Furthermore, there were significant differences in glomerular filtration rate (GFR) change between the two methods in four studies but no differences were observed in other two. Conclusions Our systematic review indicated that RFA is an effective treatment option which could provide comparable oncologic outcomes to PN. Moreover, it may present obvious advantages in renal function preservation.
Collapse
Affiliation(s)
- Xiyi Wei
- Department of Urology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & Affiliated Cancer Hospital of Nanjing Medical University, Nanjing 210009, China.,First Clinical Medical College of Nanjing Medical University, Nanjing 210029, China
| | - Xiaohan Ren
- First Clinical Medical College of Nanjing Medical University, Nanjing 210029, China
| | - Yichao Ding
- School of Nursing of Nanjing Medical University, Nanjing 210029, China
| | - Hongye Wang
- First Clinical Medical College of Nanjing Medical University, Nanjing 210029, China
| | - Yunxin Li
- First Clinical Medical College of Nanjing Medical University, Nanjing 210029, China
| | - Xiao Li
- Department of Urology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & Affiliated Cancer Hospital of Nanjing Medical University, Nanjing 210009, China
| | - Yang Gao
- Department of Radiology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & Affiliated Cancer Hospital of Nanjing Medical University, Nanjing 210009, China
| |
Collapse
|