1
|
Liu G, Xiong D, Che Z, Chen H, Jin W. A novel inflammation‑associated prognostic signature for clear cell renal cell carcinoma. Oncol Lett 2022; 24:307. [PMID: 35949606 PMCID: PMC9353224 DOI: 10.3892/ol.2022.13427] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 05/20/2022] [Indexed: 12/05/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) are typically situated in a complex inflammatory and immune microenvironment, which has been reported to contribute to the unfavorable prognosis of patients with ccRCC. There would be beneficial clinical implications for elucidating the roles of its molecular characteristics in the inflammatory microenvironment. This is because it would facilitate the development of reliable biomarkers for pre-stratification prior to the designation of individualized treatment strategies. In the present study, RNA-sequencing data from 607 patients were retrospectively analyzed to elucidate the profile of inflammatory molecules. Based on this, an inflammatory prognostic signature (IPS) was developed and further validated using clinical ccRCC samples. Subsequently, the associated mechanisms in terms of the immune microenvironment and molecular pathways were then investigated. This proposed IPS was found to exhibit superior accuracy compared with the criterion of a good prognostic model for the prediction of patient prognosis from ccRCC [area under the receiver operating characteristic curve (AUC)=0.811] in addition to being an independent factor for prognostic risk stratification [hazard ratio: 11.73 (95% CI, 26.98-5.10); log-rank test, P<0.001]. Pathologically, ccRCC cells identified as high-risk according to their IPS presented with a more malignant tumor structure, including voluminous eosinophilic cytoplasm, acinar/lamellar/tubular growth patterns and atypic nuclei. High-risk ccRCC also exhibited higher infiltration levels by four types of immune cells, including T regulatory cells, but lower infiltration levels by mast cells. Pathways associated with immune-inflammation interaction, including the IL-17 pathway, were found to be upregulated in IPS-identified high-risk ccRCC. Furthermore, by combining the IPS with clinical factors, an integrated prognostic index was developed and validated for increasing the accuracy of patient risk-stratification for ccRCC (AUC=0.911). In conclusion, the complex regulatory mechanisms and molecular characteristics involved in ccRCC-inflammation interaction, coupled with their prognostic potential, were systematically elucidated in the present study. This may have important implications in furthering the understanding into the molecular mechanisms underlying this ccRCC-inflammation interaction, which can in turn be exploited for identifying high-risk patients with ccRCC prior to designing their clinical treatment strategy.
Collapse
Affiliation(s)
- Gangcheng Liu
- Department of Urology Surgery, Affiliated Renhe Hospital of China Three Gorges University Second Clinical Medical College of China Three Gorges University, Yichang, Hubei 443000, P.R. China
| | - Donglan Xiong
- Department of Respiratory Medicine, Affiliated Renhe Hospital of China Three Gorges University Second Clinical Medical College of China Three Gorges University, Yichang, Hubei 443000, P.R. China
| | - Zhifei Che
- Department of Urology, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570100, P.R. China
| | - Hualei Chen
- Department of Urology Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570100, P.R. China
| | - Wenyi Jin
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
2
|
Kim SH, Park J, Park WS, Hong D, Chung J. A retrospective single-centered, comprehensive targeted genetic sequencing analysis of prognostic survival using tissues from Korean patients with metastatic renal cell carcinoma after targeted therapy. Investig Clin Urol 2022; 63:602-611. [PMID: 36347549 PMCID: PMC9643729 DOI: 10.4111/icu.20210341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/14/2022] [Accepted: 09/21/2022] [Indexed: 11/05/2022] Open
Abstract
Purpose To identify candidate gene mutations to significantly predict the risk of survival prognosis after treatment with systemic first-line targeted therapy (TT) in metastatic renal cell carcinoma (mRCC) patients. Materials and Methods Between 2005 and 2017, 168 triplet-tissue block samples from 56 mRCC patients were selected for targeted gene sequencing (TGS). Fifty-six patients’ medical records including overall survival (OS) and progression-free survival (PFS) at the time of mRCC diagnosis were evaluated. The patients were grouped into favorable (>12 months/>3 years), intermediate (3–12/12–36 months), and poor groups according to their PFS/OS (<3 months/<12 months). We identified any significant therapeutic targeted genes relating to the survival with a significance at p<0.050. Results The first line therapeutic response showed 1.8% complete remission, 14.2% partial response, 42.9% stable disease, and 41.1% progressive disease. Among the overall TGS results, the cumulative effect of CDH1, and/or PTK2 genes significantly reflected the therapeutic responses in terms of PFS/OS; CDH1 and PTK2 mutations were associated with poor prognostic outcomes (p<0.050). Among only triplet-quality check passed tissues, the SGO2, BRAF, URB1, and NEDD1 mutated genes significantly correlated with OS. Regarding metastasis, patients with liver metastasis had the worst OS (p=0.050). The combinational mutation number from these two candidate genes in the liver metastatic samples with mutated EGFR2 and FABP7 also showed a significantly worse OS than those with other metastatic lesions (p<0.050). Conclusions This study reports several significant mutated genes related to the survival prognosis in mRCC patients treated with first-line TT.
Collapse
Affiliation(s)
- Sung Han Kim
- Department of Urology, Urologic Cancer Center, National Cancer Center, Goyang, Korea
| | - Jongkeun Park
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Weon Seo Park
- Department of Pathology, Urologic Cancer Center, National Cancer Center, Goyang, Korea
| | - Dongwan Hong
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jinsoo Chung
- Department of Urology, Urologic Cancer Center, National Cancer Center, Goyang, Korea
| |
Collapse
|
3
|
Park JS, Lee ME, Jang WS, Rha KH, Lee SH, Lee J, Ham WS. The DEAD/DEAH Box Helicase, DDX11, Is Essential for the Survival of Advanced Clear Cell Renal Cell Carcinoma and Is a Determinant of PARP Inhibitor Sensitivity. Cancers (Basel) 2021; 13:cancers13112574. [PMID: 34073906 PMCID: PMC8197413 DOI: 10.3390/cancers13112574] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 05/13/2021] [Accepted: 05/20/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary DDX11, a helicase involved in sister chromatid cohesion, was identified as a significant biomarker of aggressive renal cell carcinoma (RCC) in our previous studies. In this study, we evaluated the molecular pathways through which DDX11 is involved in RCC cell survival. Furthermore, we assessed the sensitivity of poly (ADP-ribose) polymerase (PARP) inhibitors, which have not been used in RCC treatment, in association with DDX11 expression. DDX11-deficient RCC inhibited RCC proliferation, caused defects in segregation, and increased apoptosis. DDX11-deficient RCC was associated with increased sensitivity to PARP inhibition. DDX11 could be a novel therapeutic and prognostic biomarker for RCC patients, and this study is the first to suggest the use of PARP inhibitors in DDX11-deficient RCC patients. Abstract Genes associated with the DEAD-box helicase DDX11 are significant biomarkers of aggressive renal cell carcinoma (RCC), but their molecular function is poorly understood. We analyzed the molecular pathways through which DDX11 is involved in RCC cell survival and poly (ADP-ribose) polymerase (PARP) inhibitor sensitivity. Immunohistochemistry and immunoblotting determined DDX11 expression in normal kidney tissues, benign renal tumors, and RCC tissues and cell lines. Quantitative polymerase chain reaction validated the downregulation of DDX11 in response to transfection with DDX11-specific small interfering RNA. Proliferation analysis and apoptosis assays were performed to determine the impact of DDX11 knockdown on RCC cells, and the relevant effects of sunitinib, olaparib, and sunitinib plus olaparib were evaluated. DDX11 was upregulated in high-grade, advanced RCC compared to low-grade, localized RCC, and DDX11 was not expressed in normal kidney tissues or benign renal tumors. DDX11 knockdown resulted in the inhibition of RCC cell proliferation, segregation defects, and rapid apoptosis. DDX11-deficient RCC cells exhibited significantly increased sensitivity to olaparib compared to sunitinib alone or sunitinib plus olaparib combination treatments. Moreover, DDX11 could determine PARP inhibitor sensitivity in RCC. DDX11 could serve as a novel therapeutic biomarker for RCC patients who are refractory to conventional targeted therapies and immunotherapies.
Collapse
Affiliation(s)
- Jee Soo Park
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul 03722, Korea; (J.S.P.); (M.E.L.); (W.S.J.); (K.H.R.); (S.H.L.); (J.L.)
- Department of Urology, Sorokdo National Hospital, Goheung 59562, Korea
| | - Myung Eun Lee
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul 03722, Korea; (J.S.P.); (M.E.L.); (W.S.J.); (K.H.R.); (S.H.L.); (J.L.)
| | - Won Sik Jang
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul 03722, Korea; (J.S.P.); (M.E.L.); (W.S.J.); (K.H.R.); (S.H.L.); (J.L.)
| | - Koon Ho Rha
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul 03722, Korea; (J.S.P.); (M.E.L.); (W.S.J.); (K.H.R.); (S.H.L.); (J.L.)
| | - Seung Hwan Lee
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul 03722, Korea; (J.S.P.); (M.E.L.); (W.S.J.); (K.H.R.); (S.H.L.); (J.L.)
| | - Jongsoo Lee
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul 03722, Korea; (J.S.P.); (M.E.L.); (W.S.J.); (K.H.R.); (S.H.L.); (J.L.)
| | - Won Sik Ham
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul 03722, Korea; (J.S.P.); (M.E.L.); (W.S.J.); (K.H.R.); (S.H.L.); (J.L.)
- Correspondence: ; Tel.: +82-10-6242-7938; Fax: +82-2-312-2538
| |
Collapse
|
4
|
Gundert L, Strick A, Hagen F, Schmidt D, Klümper N, Tolkach Y, Toma M, Kristiansen G, Ritter M, Ellinger J. Systematic expression analysis of m
6
A RNA methyltransferases in clear cell renal cell carcinoma. BJUI COMPASS 2021; 2:402-411. [PMID: 35474700 PMCID: PMC8988738 DOI: 10.1002/bco2.89] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 04/12/2021] [Accepted: 04/12/2021] [Indexed: 01/17/2023] Open
Abstract
Objectives To investigate the regulation of the N‐6‐methyladenosine (m6A) methyltransferases METTL3, METTL14, WTAP, KIAA1429, and METTL4, referred to as “m6A writers,” in clear cell renal cell carcinoma (ccRCC), and other RCC subtypes in respect of the potential prognostic value. Patients and methods Tissue samples were collected within the framework of the Biobank at the Center for Integrated Oncology Bonn. The expression of the methyltransferases was systematically determined in clear cell renal carcinoma (ccRCC) on the RNA (real‐time PCR) and protein level (immunohistochemistry). Additionally, protein expression of the m6A writers was further investigated in papillary RCC, chromophobe RCC, sarcomatoid RCC, oncocytoma, and normal renal tissue (immunohistochemistry). Results The expression of all m6A‐methyltransferases was significantly downregulated in ccRCC compared to benign renal tissue. Low m6A‐methyltransferase levels were correlated with higher histological grade, advanced pT‐stage, pN‐stage, and metastatic disease. Reduced m6A‐methyltransferase expression was associated with shorter overall survival. Conclusion In conclusion, m6A‐methyltransferases are dysregulated in ccRCC and might act as tumor suppressor genes, which could be of particular importance for future diagnostic and therapeutic options.
Collapse
Affiliation(s)
| | | | - Felix Hagen
- Department of Urology University Hospital Bonn Bonn Germany
| | - Doris Schmidt
- Department of Urology University Hospital Bonn Bonn Germany
| | - Niklas Klümper
- Department of Urology University Hospital Bonn Bonn Germany
| | - Yuri Tolkach
- Department of Pathology University Hospital Bonn Bonn Germany
- Department of Pathology University Hospital Cologne Cologne Germany
| | - Marieta Toma
- Department of Pathology University Hospital Bonn Bonn Germany
| | | | - Manuel Ritter
- Department of Urology University Hospital Bonn Bonn Germany
| | - Jörg Ellinger
- Department of Urology University Hospital Bonn Bonn Germany
| |
Collapse
|
5
|
Park JS, Lee ME, Kim SH, Jang WS, Ham WS. Development of a highly pulmonary metastatic orthotopic renal cell carcinoma murine model. Biol Open 2021; 10:256557. [PMID: 33913471 PMCID: PMC8084570 DOI: 10.1242/bio.058566] [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: 01/09/2021] [Accepted: 03/25/2021] [Indexed: 01/01/2023] Open
Abstract
The incidence of renal cell carcinoma (RCC) is high, and its outcomes remain poor. Mortality is attributable largely to metastatic disease and a dearth of effective therapeutic interventions. The lungs are the most common metastatic site. To elucidate the biological mechanisms underlying pulmonary metastasis and identify superior therapeutic strategies, we developed a novel and clinically relevant murine RCC model exhibiting enhanced pulmonary metastasis. Mice underwent intrarenal implantation using luciferase-expressing Renca, a murine renal adenocarcinoma cell line. Primary renal tumor progression and development of metastatic lung lesions were monitored in live mice using bioluminescent imaging, followed by post-mortem organ assessment. Cells were isolated from pulmonary metastases for reimplantation, followed by repeat monitoring and assessment. This process was repeated once more for a total of two in vivo passages to select for pulmonary metastatic Renca cell subpopulations. However, a single round of in vivo selection was sufficient to produce a near-maximally metastatic subpopulation. Relative to Renca cell-implanted mice, subpopulation-implanted mice exhibited shorter implantation-metastasis intervals (5 days), shorter implantation-moribundity intervals (sacrificed at 18.6±2.9 versus 22.3±1.1 days), a higher number of metastatic lung lesions at 23 days (183.9±39.0 versus 172.6±38.2) and poorer survival. Implantation of cells derived from the second round of in vivo selection produced no further significant differences in the above metrics. This model consistently and efficiently recapitulates RCC pulmonary metastasis while allowing in vivo monitoring of tumor progression, thereby facilitating elucidation of the molecular mechanisms underlying pulmonary metastasis and evaluation of therapeutic modalities.
Collapse
Affiliation(s)
- Jee Soo Park
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Myung Eun Lee
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Seung Hwan Kim
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Won Sik Jang
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Won Sik Ham
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
6
|
Lommen K, Vaes N, Aarts MJ, van Roermund JG, Schouten LJ, Oosterwijk E, Melotte V, Tjan-Heijnen VC, van Engeland M, Smits KM. Diagnostic DNA Methylation Biomarkers for Renal Cell Carcinoma: A Systematic Review. Eur Urol Oncol 2021; 4:215-226. [PMID: 31402218 DOI: 10.1016/j.euo.2019.07.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 06/23/2019] [Accepted: 07/16/2019] [Indexed: 01/22/2023]
Abstract
CONTEXT The 5-yr survival of early-stage renal cell carcinoma (RCC) is approximately 93%, but once metastasised, the 5-yr survival plummets to 12%, indicating that early RCC detection is crucial to improvement in survival. DNA methylation biomarkers have been suggested to be of potential diagnostic value; however, their current state of clinical translation is unclear and a comprehensive overview is lacking. OBJECTIVE To systematically review and summarise all literature regarding diagnostic DNA methylation biomarkers for RCC. EVIDENCE ACQUISITION We performed a systematic literature review of PubMed, EMBASE, Medline, and Google Scholar up to January 2019, according to the Preferred Reporting Items for Systematic Review and Meta-Analysis of Diagnostic Test Accuracy Studies (PRISMA-DTA) guidelines. Included studies were scored according to the Standards for Reporting of Diagnostic Accuracy Studies (STARD) criteria. Forest plots were generated to summarise diagnostic performance of all biomarkers. Level of evidence (LoE) and potential risk of bias were determined for all included studies. EVIDENCE SYNTHESIS After selection, 19 articles reporting on 44 diagnostic DNA methylation biomarkers and 11 multimarker panels were included; however, only 15 biomarkers were independently validated. STARD scores varied from 4 to 13 out of 23 points, with a median of 10 points. Large variation in subgroups, methods, and primer locations was observed. None of the reported biomarkers exceeded LoE III, and the majority of studies reported inadequately. CONCLUSIONS None of the reported biomarkers exceeded LoE III, indicating their limited clinical utility. Moreover, study reproducibility and further development of these RCC biomarkers are greatly hampered by inadequate reporting. PATIENT SUMMARY In this report, we reviewed whether specific biomarkers could be used to diagnose the most common form of kidney cancer. We conclude that due to limited evidence and reporting inconsistencies, none of these biomarkers can be used in clinical practice, and further development towards clinical use is hindered.
Collapse
Affiliation(s)
- Kim Lommen
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Nathalie Vaes
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Maureen J Aarts
- Department of Medical Oncology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Joep G van Roermund
- Department of Urology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Leo J Schouten
- Department of Epidemiology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | | | - Veerle Melotte
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Vivianne C Tjan-Heijnen
- Department of Medical Oncology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Manon van Engeland
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Kim M Smits
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands; Department of Medical Oncology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands.
| |
Collapse
|
7
|
PET/CT in Renal, Bladder, and Testicular Cancer. Clin Nucl Med 2020. [DOI: 10.1007/978-3-030-39457-8_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
8
|
Zahoor H, Duddalwar V, D’Souza A, Merseburger AS, Quinn DI. What Comes After Immuno-Oncology Therapy for Kidney Cancer? KIDNEY CANCER 2019. [DOI: 10.3233/kca-190053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Haris Zahoor
- Keck School of Medicine, University of Southern California, Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Vinay Duddalwar
- Keck School of Medicine, University of Southern California, Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Anishka D’Souza
- Keck School of Medicine, University of Southern California, Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Axel S. Merseburger
- Department of Urology, Campus Lübeck, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - David I. Quinn
- Keck School of Medicine, University of Southern California, Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| |
Collapse
|
9
|
Ergün S, Altay DU, Güneş S, Büyükalpelli R, Karahan SC, Tomak L, Abur Ü. Tr-KIT/c-KIT ratio in renal cell carcinoma. Mol Biol Rep 2019; 46:5287-5294. [DOI: 10.1007/s11033-019-04985-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 07/17/2019] [Indexed: 01/08/2023]
|
10
|
Evangelista L, Zattoni F, Alongi P. 68Ga-dotatoc vs. 18F-FDG vs. radiolabelled PSMA PET/CT in renal cancer patients. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:S150. [PMID: 31576357 DOI: 10.21037/atm.2019.06.28] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Laura Evangelista
- Nuclear Medicine Unit, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Fabio Zattoni
- Urology Unit, Academical Medical Centre Hospital, Udine, Italy
| | - Pierpaolo Alongi
- Department of Radiological Sciences, Nuclear Medicine Service, Fondazione Istituto G. Giglio, Cefalu', Italy
| |
Collapse
|
11
|
Circulating Tumor Cells for the Management of Renal Cell Carcinoma. Diagnostics (Basel) 2018; 8:diagnostics8030063. [PMID: 30177639 PMCID: PMC6164661 DOI: 10.3390/diagnostics8030063] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 08/30/2018] [Accepted: 08/31/2018] [Indexed: 12/17/2022] Open
Abstract
Renal cell carcinoma is a highly malignant cancer that would benefit from non-invasive innovative markers providing early diagnosis and recurrence detection. Circulating tumor cells are a particularly promising marker of tumor invasion that could be used to improve the management of patients with RCC. However, the extensive genetic and immunophenotypic heterogeneity of cells from RCC and their trend to transition to the mesenchymal phenotype when they circulate in blood constitute a challenge for their sensitive and specific detection. This review analyzes published studies targeting CTC in patients with RCC, in the context of the biological, pathological, and molecular complexity of this particular cancer. Although further analytical and clinical studies are needed to pinpoint the most suitable approach for highly sensitive CTC detection in RCC patients, it is clear that this field can bring a relevant guide to clinicians and help to RCC patients. Furthermore, as described, a particular subtype of RCC-the ccRCC-can be used as a model to study the relationship between cytomorphological and genetic cellular markers of malignancy, an important issue for the study of CTC from any type of solid cancer.
Collapse
|
12
|
Fiasconaro E, Caobelli F, Quartuccio N, Messina M, Spada M, Albano D, Alongi P. PET/CT for the diagnostic assessment of patients with renal cancer. Clin Transl Imaging 2018; 6:207-216. [DOI: 10.1007/s40336-018-0278-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 04/18/2018] [Indexed: 02/07/2023]
|
13
|
Broncy L, Njima BB, Méjean A, Béroud C, Romdhane KB, Ilie M, Hofman V, Muret J, Hofman P, Bouhamed HC, Paterlini-Bréchot AP. Single-cell genetic analysis validates cytopathological identification of circulating cancer cells in patients with clear cell renal cell carcinoma. Oncotarget 2018; 9:20058-20074. [PMID: 29732003 PMCID: PMC5929446 DOI: 10.18632/oncotarget.25102] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Accepted: 03/24/2018] [Indexed: 12/14/2022] Open
Abstract
CONTEXT Circulating Rare Cells (CRC) are non-haematological cells circulating in blood. They include Circulating Cancer Cells (CCC) and cells with uncertain malignant features (CRC-UMF) according to cytomorphology. Clear cell renal cell carcinomas frequently bear a mutated Von Hippel-Lindau (VHL) gene. AIM To match blind genetic analysis of CRC and tumor samples with CRC cytopathological diagnosis. RESULTS 29/30 patients harboured CRC (20 harboured CCC, 29 CRC-UMF) and 25/29 patients carried VHL mutations in their tumour. 205 single CRC (64 CCC, 141 CRC-UMF) provided genetic data. 57/57 CCC and 104/125 CRC-UMF from the 25 patients with VHL-mutated tumor carried the same VHL mutation detected in the tumor. Seven CCC and 16 CRC-UMF did not carry VHL mutations but were found in patients with wild-type VHL tumor tissue. CONCLUSIONS All the CCC and 83,2% (104/125) of the CRC-UMF were found to carry the same VHL mutation identified in the corresponding tumorous tissue, validating cytopathological identification of CCC in patients with clear cell renal cell carcinoma. METHODS The blood of 30 patients with clear cell renal cell carcinoma was treated by ISET® for CRC isolation, cytopathology and single-cell VHL mutations analysis, performed blindly and compared to VHL mutations of corresponding tumor tissues and leukocytes.
Collapse
Affiliation(s)
- Lucile Broncy
- INSERM Unit 1151, Faculté de Médecine Paris Descartes, Paris, France
| | - Basma Ben Njima
- Genetics and Pathology Departments, University of Tunis, Tunis, Tunisia
| | - Arnaud Méjean
- Service d'Urologie, Hôpital Européen Georges Pompidou, Paris, France
| | - Christophe Béroud
- Aix Marseille University, INSERM, MMG, Marseille, France
- APHM, Hôpital TIMONE Enfants, Laboratoire de Génétique Moléculaire, Marseille, France
| | | | - Marius Ilie
- Laboratoire de pathologie clinique et Biobank BB-0033-00025, Centre Hospitalo-Universitaire de Nice, Nice, France
| | - Veronique Hofman
- Laboratoire de pathologie clinique et Biobank BB-0033-00025, Centre Hospitalo-Universitaire de Nice, Nice, France
| | - Jane Muret
- Institut Curie, PSL Research University, Département d'Anesthésie Réanimation Douleur, Paris, France
| | - Paul Hofman
- Laboratoire de pathologie clinique et Biobank BB-0033-00025, Centre Hospitalo-Universitaire de Nice, Nice, France
| | | | - And Patrizia Paterlini-Bréchot
- INSERM Unit 1151, Faculté de Médecine Paris Descartes, Paris, France
- Laboratoire de Biochimie A, Hôpital Necker-Enfants Malades, Paris, France
| |
Collapse
|
14
|
Abstract
In 2011, Varela et al. reported that the PBRM1 gene is mutated in approximately 40% of clear cell renal cell carcinoma cases. Since then, the number of studies relating PBRM1 mutations to cancers has substantially increased. BAF180 has now been linked to more than 30 types of cancers, including ccRCC, cholangiocarcinomas, esophageal squamous cell carcinoma, bladder cancer, and breast cancer. The mutations associated with BAF180 are most often truncations, which result in a loss of protein expression. This loss has been shown to adversely affect the expression of genes, likely because BAF180 is the chromatin recognition subunit of PBAF. In addition, BAF180 functions in numerous DNA repair mechanisms. Its roles in mediating DNA repair are likely the mechanism by which BAF180 acts a tumor suppressor protein. As research on this protein gains more interest, scientists will begin to piece together the complicated puzzle of the BAF180 protein and why its loss often results in cancer.
Collapse
Affiliation(s)
- Sarah Hopson
- Department of Chemistry, Michigan Technological University, 1400 Townsend Drive, Houghton, Michigan 49931, United States
| | - Martin J. Thompson
- Department of Chemistry, Michigan Technological University, 1400 Townsend Drive, Houghton, Michigan 49931, United States
| |
Collapse
|
15
|
MicroRNA-497 suppresses renal cell carcinoma by targeting VEGFR-2 in ACHN cells. Biosci Rep 2017; 37:BSR20170270. [PMID: 28465356 PMCID: PMC5437937 DOI: 10.1042/bsr20170270] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 04/15/2017] [Accepted: 05/02/2017] [Indexed: 11/25/2022] Open
Abstract
Abnormal expression of miRNAs contributed to cancers through regulation of proliferation, apoptosis and drug resistance of cancer cells. The present study was designed to investigate the effect of miR-497 on renal cell carcinoma (RCC) and its possible mechanism. Forty paired clear cell RCC (ccRCC) tissues and adjacent normal kidney tissues were obtained from patients, who were not treated by chemotherapy or radiotherapy. RT-PCR was performed to detect expression of miR-497 in the ccRCC tissues. Effects of miR-497 on cell viability, apoptosis, migration and invasion were detected in ACHN cells. Western blotting (WB) was employed to detect the downstream targets of miR-497. We found that miR-497 in ccRCC tissues was decreased. We treated ACHN cells with miR-497 mimics and inhibitors in vitro and found that miR-497 inhibited viability, migration and invasion of ACHN cells. miR-497 promoted ACHN cells’ apoptosis. VEGFR-2 was predicted as a possible target of miR-497. Luciferase reporter assay proved that miR-497 suppressed VEGFR-2 directly by binding to its 3′-UTR. Further studies showed that miR-497 influenced the MEK/ERK and p38 MAPK signalling pathways. Our findings demonstrated that miR-497 could suppress RCC by targeting VEGFR-2.
Collapse
|
16
|
Brodaczewska KK, Szczylik C, Fiedorowicz M, Porta C, Czarnecka AM. Choosing the right cell line for renal cell cancer research. Mol Cancer 2016; 15:83. [PMID: 27993170 PMCID: PMC5168717 DOI: 10.1186/s12943-016-0565-8] [Citation(s) in RCA: 195] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Accepted: 11/30/2016] [Indexed: 01/08/2023] Open
Abstract
Cell lines are still a tool of choice for many fields of biomedical research, including oncology. Although cancer is a very complex disease, many discoveries have been made using monocultures of established cell lines. Therefore, the proper use of in vitro models is crucial to enhance our understanding of cancer. Therapeutics against renal cell cancer (RCC) are also screened with the use of cell lines. Multiple RCC in vitro cultures are available, allowing in vivo heterogeneity in the laboratory, but at the same time, these can be a source of errors. In this review, we tried to sum up the data on the RCC cell lines used currently. An increasing amount of data on RCC shed new light on the molecular background of the disease; however, it revealed how much still needs to be done. As new types of RCC are being distinguished, novel cell lines and the re-exploration of old ones seems to be indispensable to create effective in vitro tools for drug screening and more.
Collapse
Affiliation(s)
- Klaudia K Brodaczewska
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland
| | - Cezary Szczylik
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland
| | - Michal Fiedorowicz
- Department of Experimental Pharmacology, Polish Academy of Science Medical Research Centre, Warsaw, Poland
| | - Camillo Porta
- Department of Medical Oncology, IRCCS San Matteo University Hospital Foundation, Pavia, Italy
| | - Anna M Czarnecka
- Department of Oncology with Laboratory of Molecular Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland.
| |
Collapse
|
17
|
Ong PS, Wang LZ, Dai X, Tseng SH, Loo SJ, Sethi G. Judicious Toggling of mTOR Activity to Combat Insulin Resistance and Cancer: Current Evidence and Perspectives. Front Pharmacol 2016; 7:395. [PMID: 27826244 PMCID: PMC5079084 DOI: 10.3389/fphar.2016.00395] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 10/07/2016] [Indexed: 12/16/2022] Open
Abstract
The mechanistic target of rapamycin (mTOR), via its two distinct multiprotein complexes, mTORC1, and mTORC2, plays a central role in the regulation of cellular growth, metabolism, and migration. A dysregulation of the mTOR pathway has in turn been implicated in several pathological conditions including insulin resistance and cancer. Overactivation of mTORC1 and disruption of mTORC2 function have been reported to induce insulin resistance. On the other hand, aberrant mTORC1 and mTORC2 signaling via either genetic alterations or increased expression of proteins regulating mTOR and its downstream targets have contributed to cancer development. These underlined the attractiveness of mTOR as a therapeutic target to overcome both insulin resistance and cancer. This review summarizes the evidence supporting the notion of intermittent, low dose rapamycin for treating insulin resistance. It further highlights recent data on the continuous use of high dose rapamycin analogs and related second generation mTOR inhibitors for cancer eradication, for overcoming chemoresistance and for tumor stem cell suppression. Within these contexts, the potential challenges associated with the use of mTOR inhibitors are also discussed.
Collapse
Affiliation(s)
- Pei Shi Ong
- Department of Pharmacy, Faculty of Science, National University of Singapore Singapore, Singapore
| | - Louis Z Wang
- Department of Pharmacy, Faculty of Science, National University of SingaporeSingapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of SingaporeSingapore, Singapore
| | - Xiaoyun Dai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore Singapore, Singapore
| | - Sheng Hsuan Tseng
- Department of Pharmacy, Faculty of Science, National University of Singapore Singapore, Singapore
| | - Shang Jun Loo
- Department of Pharmacy, Faculty of Science, National University of Singapore Singapore, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore Singapore, Singapore
| |
Collapse
|
18
|
Zahoor H, Rini BI. Emerging growth factor receptor antagonists for the treatment of renal cell carcinoma. Expert Opin Emerg Drugs 2016; 21:431-440. [DOI: 10.1080/14728214.2016.1244263] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
19
|
Stukalin I, Alimohamed N, Heng DY. Contemporary Treatment of Metastatic Renal Cell Carcinoma. Oncol Rev 2016; 10:295. [PMID: 27471582 PMCID: PMC4943094 DOI: 10.4081/oncol.2016.295] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/30/2016] [Accepted: 06/29/2016] [Indexed: 12/23/2022] Open
Abstract
The introduction of targeted therapy has revolutionized the treatment of patients with metastatic renal cell carcinoma (mRCC). The current standard of care focuses on the inhibition of angiogenesis through the targeting of the vascular endothelial growth factor receptor (VEGFR) and the mammalian target of rapamycin (mTOR). Over the past few years, research exploring novel targeted agents has blossomed, leading to the approval of various targeted therapies. Furthermore, results from the CheckMate025 and the METEOR trials have brought about two additional novel options: the programmed cell death 1 (PD-1) checkpoint inhibitor nivolumab and the MET/VEGFR/AXL inhibitor cabozantinib, respectively. With the variety of therapeutic agents available for treatment of mRCC, research examining appropriate sequencing and combinations of the drugs is ongoing. This review discusses the role of prognostic criteria, such as those from the International Metastatic Renal Cell Carcinoma Database Consortium (IMDC) criteria. It also covers the current standard of treatment for mRCC with targeted therapy in first-, second-, and third-line setting. Additionally, the novel mechanism of action of nivolumab and cabozantinib, therapeutic sequencing and ongoing clinical trials are discussed.
Collapse
Affiliation(s)
| | | | - Daniel Y.C. Heng
- Department of Medical Oncology, Tom Baker Cancer Centre, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
20
|
Chinello C, Cazzaniga M, De Sio G, Smith AJ, Grasso A, Rocco B, Signorini S, Grasso M, Bosari S, Zoppis I, Mauri G, Magni F. Tumor size, stage and grade alterations of urinary peptidome in RCC. J Transl Med 2015; 13:332. [PMID: 26482227 PMCID: PMC4617827 DOI: 10.1186/s12967-015-0693-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 10/10/2015] [Indexed: 01/23/2023] Open
Abstract
Background Several promising biomarkers have been found for RCC, but none of them has been used in clinical practice for predicting tumour progression. The most widely used features for predicting tumour aggressiveness still remain the cancer stage, size and grade. Therefore, the aim of our study is to investigate the urinary peptidome to search and identify peptides whose concentrations in urine are linked to tumour growth measure and clinical data. Methods A proteomic approach applied to ccRCC urinary peptidome (n = 117) based on prefractionation with activated magnetic beads followed by MALDI-TOF profiling was used. A systematic correlation study was performed on urinary peptide profiles obtained from MS analysis. Peptide identity was obtained by LC–ESI–MS/MS. Results Fifteen, twenty-six and five peptides showed a statistically significant alteration of their urinary concentration according to tumour size, pT and grade, respectively. Furthermore, 15 and 9 signals were observed to have urinary levels statistically modified in patients at different pT or grade values, even at very early stages. Among them, C1RL, A1AGx, ZAG2G, PGBM, MMP23, GP162, ADA19, G3P, RSPH3, DREB, NOTC2 SAFB2 and CC168 were identified. Conclusions We identified several peptides whose urinary abundance varied according to tumour size, stage and grade. Among them, several play a possible role in tumorigenesis, progression and aggressiveness. These results could be a useful starting point for future studies aimed at verifying their possible use in the managements of RCC patients. Electronic supplementary material The online version of this article (doi:10.1186/s12967-015-0693-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Clizia Chinello
- Department of Health Science, School of Medicine, University of Milano-Bicocca (UNIMIB), Via Cadore, 48, 20900, Monza, Italy.
| | - Marta Cazzaniga
- Department of Health Science, School of Medicine, University of Milano-Bicocca (UNIMIB), Via Cadore, 48, 20900, Monza, Italy.
| | - Gabriele De Sio
- Department of Health Science, School of Medicine, University of Milano-Bicocca (UNIMIB), Via Cadore, 48, 20900, Monza, Italy.
| | - Andrew James Smith
- Department of Health Science, School of Medicine, University of Milano-Bicocca (UNIMIB), Via Cadore, 48, 20900, Monza, Italy.
| | - Angelica Grasso
- Urology Unit, Department of Specialistic Surgical Sciences, Ospedale Maggiore Policlinico Foundation, Milan, Italy.
| | - Bernardo Rocco
- Urology Unit, Department of Specialistic Surgical Sciences, Ospedale Maggiore Policlinico Foundation, Milan, Italy.
| | | | - Marco Grasso
- Department of Surgical Pathology, Cytology, Medical Genetics and Nephropathology, Azienda Ospedaliera San Gerardo, Monza, Italy.
| | - Silvano Bosari
- Department of Medicine, Surgery and Dental Sciences, Pathology Unit, IRCCS-Policlinico Foundation, Mangiagalli and Regina Elena, University of Milan, Milan, Italy.
| | - Italo Zoppis
- Department of Informatics, Systems and Communication, University of Milano-Bicocca, Milan, Italy.
| | - Giancarlo Mauri
- Department of Informatics, Systems and Communication, University of Milano-Bicocca, Milan, Italy.
| | - Fulvio Magni
- Department of Health Science, School of Medicine, University of Milano-Bicocca (UNIMIB), Via Cadore, 48, 20900, Monza, Italy.
| |
Collapse
|
21
|
Kamińska K, Czarnecka AM, Escudier B, Lian F, Szczylik C. Interleukin-6 as an emerging regulator of renal cell cancer. Urol Oncol 2015; 33:476-85. [PMID: 26296264 DOI: 10.1016/j.urolonc.2015.07.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 07/10/2015] [Accepted: 07/11/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Our knowledge on the molecular basis of kidney cancer metastasisis still relatively low. About 25-30% of patients suffering from clear cell renal cell carcinoma (ccRCC)present metastatic disease at the time of primary diagnosis. Only 10% of patients diagnosed with stage IV disease survive 5 years and 20-50% of patients diagnosed with localized tumor develop metastases within 3 years. High mortality of patients with this cancer is associated with a large potential for metastasis and resistance to oncologic treatments such as chemo- and radiotherapy. Literature data based on studies conducted on other types of cancers suggest that in metastatic ccRCC, the complex of interleukin-6 (IL-6) and its soluble receptor (sIL-6R; complex IL-6/sIL-6R) and the signal transduction pathway (gp130/STAT3) might play a key role in this process. PURPOSE Therefore, in this review we focus on the role of IL-6 and its signaling pathways as a factor for development and spread of RCC. Analyzing the molecular basis of cancer spreading will enable the development of prognostic tests, evaluate individual predisposition for metastasis, and produce drugs that target metastases. As the development of effective systemic treatments evolve from advancements in molecular biology, continued studies directed at understanding the genetic and molecular complexities of this disease are critical to improve RCC treatment options.
Collapse
Affiliation(s)
| | - Anna M Czarnecka
- Department of Oncology, Military Institute of Medicine, Warsaw, Poland
| | - Bernard Escudier
- Medical Oncology Department, Institut Gustave Roussy, Villejuif, France
| | - Fei Lian
- Emory University School of Medicine, Atlanta, GA
| | - Cezary Szczylik
- Department of Oncology, Military Institute of Medicine, Warsaw, Poland
| |
Collapse
|
22
|
Abstract
Imaging plays an important role in the clinical management of cancer patients. Hybrid imaging with PET/computed tomography (CT) is having a broad impact in oncology, and in recent years PET/CT is beginning to have an impact in urooncology. In both bladder and renal cancers, there is a need to study the efficacy of other tracers than F-18 fluorodeoxyglucose (FDG), particularly tracers with limited renal excretion. Thus, new tracers are being introduced. This review focuses on the clinical role of FDG and other PET agents in renal, bladder, and testicular cancers.
Collapse
|
23
|
Cheng S, Castillo V, Eliaz I, Sliva D. Honokiol suppresses metastasis of renal cell carcinoma by targeting KISS1/KISS1R signaling. Int J Oncol 2015; 46:2293-8. [PMID: 25846316 PMCID: PMC4441299 DOI: 10.3892/ijo.2015.2950] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 02/10/2015] [Indexed: 01/01/2023] Open
Abstract
Renal cell carcinoma (RCC) is a common urological cancer worldwide and is known to have a high risk of metastasis, which is considered responsible for more than 90% of cancer associated deaths. Honokiol is a small-molecule biphenol isolated from Magnolia spp. bark and has been shown to be a potential anticancer agent involved in multiple facets of signal transduction. In this study, we demonstrated that honokiol inhibited the invasion and colony formation of highly metastatic RCC cell line 786-0 in a dose-dependent manner. DNA-microarray data showed the significant upregulation of metastasis-suppressor gene KISS1 and its receptor, KISS1R. The upregulation was confirmed by qRT-PCR analysis. Overexpression of KISS1 and KISS1R was detected by western blotting at the translation level as well. Of note, the decreased invasive and colonized capacities were reversed by KISS1 knockdown. Taken together, the results first indicate that activation of KISS1/KISS1R signaling by honokiol suppresses multistep process of metastasis, including invasion and colony formation, in RCC cells 786-0. Honokiol may be considered as a natural agent against RCC metastasis.
Collapse
Affiliation(s)
- Shujie Cheng
- Cancer Research Laboratory, Methodist Research Institute, Indiana University Health, Indianapolis, IN, USA
| | - Victor Castillo
- Cancer Research Laboratory, Methodist Research Institute, Indiana University Health, Indianapolis, IN, USA
| | - Isaac Eliaz
- Amitabha Medical Clinic and Healing Center, Santa Rosa, CA, USA
| | - Daniel Sliva
- Cancer Research Laboratory, Methodist Research Institute, Indiana University Health, Indianapolis, IN, USA
| |
Collapse
|
24
|
Czarnecka AM, Szczylik C, Rini B. The use of sunitinib in renal cell carcinoma: where are we now? Expert Rev Anticancer Ther 2014; 14:983-99. [DOI: 10.1586/14737140.2014.941815] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|