Next Article in Journal
Effects of Green Tea Catechins on Prostate Cancer Chemoprevention: The Role of the Gut Microbiome
Previous Article in Journal
Functional Outcome in Spinal Meningioma Surgery and Use of Intraoperative Neurophysiological Monitoring
Previous Article in Special Issue
Prognostic Gene Expression-Based Signature in Clear-Cell Renal Cell Carcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Updating Clear Cell Renal Cell Carcinoma (a Tribute to Prof. Ondrej Hes)

1
Department of Pathology, San Giovanni Bosco Hospital, 10154 Turin, Italy
2
Department of Sciences of Public Health and Pediatrics, University of Turin, 10124 Turin, Italy
3
Unit of Biomarkers, Biocruces-Bizkaia Health Research Institute, 48903 Barakaldo, Spain
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(16), 3990; https://doi.org/10.3390/cancers14163990
Submission received: 3 August 2022 / Accepted: 17 August 2022 / Published: 18 August 2022
(This article belongs to the Special Issue Clear Cell Renal Cell Carcinoma 20212022)
This Special Issue provides an insight into critical issues concerning clear cell renal cell carcinomas (CCRCCs), reflecting the recent level of intricacy reached by renal oncology. The collection includes nineteen papers (nine articles, eight reviews, one perspective, and one commentary) which deal with contemporary diagnostic, prognostic, and therapeutic aspects of this tumor. Moreover, this Special Issue aims to provide a humble and sincere homage to the memory of Prof. Ondrej Hes, a worldwide referential Czech pathologist in renal cancer, who passed away unexpectedly on 2 July 2022 at the age of 54. We are honored to have two contributions co-authored by him (refs. [1,2]) in this collection.
Manini et al. [3] focus on tumor sampling as a cornerstone to scrutinize the complexity of intratumor heterogeneity (ITH) in CCRCC. Based on the recent molecular findings of tumor regionalization [4], the authors propose focalizing tumor sampling on peripheral zones, where ITH is expected to be the highest. Conversely, the tumor interior, where metastasizing subclones develop, is more homogeneous.
Sequeira et al. [5] show that a specific pattern of miRNA expression characterizes CCRCC with a sensitivity of 74.78%. This pattern includes hsa-miR-126-3p and hsa-miR-200b-3p levels. The authors conclude that this minimally invasive test may be useful to detect CCRCC in the early stages of tumor development.
Gopal et al. [6] review the current advances and future directions of the use of radiogenomics in the management of CCRCC. The authors update the issue and stress the promising correlation found between imaging features and gene expression patterns in several neoplasms, particularly in CCRCC.
Vano et al. [7] review the first-line treatment options in metastatic CCRCC. They state that a strategy based on the International Metastatic Database Consortium is currently recommended with either pembrolizumab and axitinib, cabozantinib and nivolumab, or levantinib and pembrolizumab given as the first-line treatment for all patients. Additionally, patients with an intermediate or poor risk should be treated with nivolumab and ipilimumab. They indicate that several issues, such as PD-L1 status, are unresolved and deserve further analyses. Thus, making therapeutic decisions based on a reliable immunohistochemical detection of the PD-L1 is still a matter of controversy [8].
Larrinaga et al. [9] compare the plasma and tissue expression of PD-1 and PD-L1 in a series of 89 CCRCCs. This unprecedented analysis yielded some significant results, for example, the plasmatic levels of both proteins were lower in CCRCC patients than in the controls. The study also confirms that the high expression of PD-1 and PD-L1 in tumor tissue was associated with tumor grade, size, and tumor necrosis. While PD-1 was associated with tumor stage (pT), PD-L1 was associated with metastases. The combination of plasmatic and tissue positivity increased the level of significance to predict the prognosis of these patients.
Several contributions deal with the ever-changing landscape of therapies and resistances to therapies occurring in these tumors. Three clinical reviews [10,11,12] and one article analyzing sunitinib resistance in CCRCC cell lines [13] revisit this particularly important issue.
Ballesteros et al. [10] focus on the molecular mechanisms of resistance to immunotherapy and antiangiogenic drugs. Resistance associated with tyrosine-kinase inhibitors include molecular mechanisms related to hypoxia, the angiogenic switch, epithelial-to-mesenchymal transition, the activation of bypass pathways, the lysosomal sequestration of tyrosine kinase inhibitors, non-coding RNAs and single-nucleotide polymorphism, and the tumor microenvironment. Among the pathways associated with resistance to immune checkpoint inhibitors, the authors analyze interferon gamma signaling, Wnt/β-catenin, MAPK, PI3K/AKT/mTOR, cell cycle checkpoint, the loss of major histocompatibility complexes I and II, and the tumor microenvironment.
Angulo et al. [11] analyze the epigenetic landscape of CCRCCs. Thus, abnormal DNA methylation, methyl-binding proteins, post-translational histone modifications, miRNAs, long non-coding RNAs, and RNA methylation are thoroughly reviewed. Furthermore, the authors revise the epigenetic-based therapeutic opportunities for CCRCCs and the caveats and limitations of these treatments.
Kim et al. [12] update the immune landscape and the immunotherapy opportunities of CCRCCs, i.e., cytokine-based immunotherapy, tyrosine kinase and mTOR inhibitors, and immune checkpoint inhibitors. The authors also focus on single-cell genomics to analyze the tumor microenvironment.
Sunitinib is a standard first-line treatment for metastatic CCRCCs [14]. Armesto et al. [13] have identified miRNA:target interactions involved in sunitinib resistance using three CCRCC cell lines (786-O, A498, and Caki-1). They have demonstrated that the use of in vitro models of sunitinib resistance, combined with an integrated approach of miRNA and gene expression, can identify divergent mechanisms of resistance with potential benefit for patients.
Paderi et al. [15] retrospectively evaluate the immune-related adverse effect of nivolumab and ipilimumab in 43 patients with metastatic renal cell carcinomas, 36 of them being CCRCCs. They conclude that adverse effects, such as thyroid dysfunction and cutaneous reactions, were associated with longer progression-free survivals and that patients that experienced more than one adverse effect presented a better response to treatment. Endocrine disorders, notably thyroid toxicities, must be taken into account since they present clinically with vague symptoms and unclear clinical pictures. The effect of nivolumab on the PD-1 expression in a culture model of CCRCC has been analyzed by Stenzel et al. [16]. They conclude that data obtained from ex vivo tissue slice culture may predict patient response to nivolumab. The influence of molecular subtypes based on genomic and transcriptomic features in the responsiveness of metastatic CCRCCs to immune checkpoint inhibitors has been reviewed by Jee et al. [17].
Mattila et al. [18] analyze the existing prognostic features and prediction models for localized CCRCCs, a growing group of tumors with an unpredictable clinical course. They conclude that prognostic factors and prediction models may help evaluate the risk of recurrence after surgical resection in localized CCRCCs, which would reduce follow-up imaging in low-risk cases. Additionally, better prediction models would help select patients for adjuvant trial therapies.
Lipidomic analysis adds interesting information in normal and neoplastic kidneys. Molecular histology has recently been profiled in non-tumor kidney tissue using the mass spectrometry of lipids [19]. Data obtained in this study demonstrate that up to seven lipidic patterns correlate with different parts of the nephron, allowing one to distinguish characteristic lipidic fingerprints in different individuals. The lipidomic analysis performed in samples from 12 CCRCCs has demonstrated the overexpression of stearoyl-CoA desaturase-1 (SCD-1) induced by the hypoxic microenvironment [20] which is characteristic of this neoplasm. The authors have detected a particular lipidomic composition involving SCD-1 in the center of CCRCC which in turns depends on the high hypoxic status found at this level. They conclude that SCD-1 may be a potential target in future treatments of these tumors. Other authors have detected that metastasizing clones of CCRCCs are located in the tumor’s center [4], where hypoxia is high and the struggle for survival is fierce.
The molecular heterogeneity in paired primary and metastatic samples of CCRCCs has previously been analyzed [21,22]. Prochazkova et al. [1] have studied the mutational variability between primary CCRCCs (four cases) and their multiple pulmonary metastases (nine metastases in total). The authors conclude that all the cases studied displayed high mutational variability not only when comparing the primary tumors, but also among the metastases themselves. These findings confirm the previous analyses which stress the high inter- and intratumor variability in most CCRCCs, a feature of critical importance when making therapeutic decisions for patients.
Recent studies have shown that the angiogenic type of CCRCC is linked to PBRM1 gene loss [23]. In this Special Issue, Saiga et al. [24] correlate the immunohistochemical expression of PBRM1 with specific architectural and vascular patterns in CCRCCs. The authors found that endothelial expression tends to be lost in cases with low PBRM1 expression. Previous studies of the same research group have demonstrated that a vascularity-based architectural classification of CCRCC has prognostic implications [25].
Khaleel et al. [26] analyze the translation between the radiologic phenotype and the underlying genotype available in the current radiogenomics literature of CCRCCs, reviewing PubMed, Medline, Cochrane Library, Google Scholar, and the Web of Science databases. Most studies use computed tomography images and the most common genomic mutations of CCRCC (VHL, PBRM1, BAP1, SETD2, and KDM5C) for such translation. They conclude that the field is promising but further studies are needed to implement this approach in clinical practice.
Roldán et al. [27] define a gene-expression-based signature in CCRCCs with prognostic implications based on a whole-transcriptome profiling of 26 cases. They found a total of 132 genes related to prognosis; however, following a Cox analysis, a nomogram including CERCAM, MIA2, HS6ST2, ONE-CUT2, SOX12, and TMEM132A genes, together with pT stage, tumor size, and ISUP grade, has been generated. The authors conclude that this nomogram discriminates between two different groups of CCRCCs with different probabilities of recurrence and predicts cancer-specific survival.
A commentary in this CCRCC Special Issue refers to the urologist’s perspective of the so-called multilocular cystic renal neoplasm of low malignant potential [2]. The clinical, radiological, and pathological findings as well as the therapeutic management are reviewed. They conclude that this entity is a lesion with excellent prognosis in which a conservative nephron-sparing treatment, if technically possible from the surgeon’s perspective, should be performed.

Author Contributions

C.M. and J.I.L. designed and wrote the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Prochazkova, K.; Ptakova, N.; Alaghehbandan, R.; Williamson, S.R.; Vaněček, T.; Vodicka, J.; Treska, V.; Rogala, J.; Pivovarcikova, K.; Michalova, K.; et al. Mutation Profile Variability in the Primary Tumor and Multiple Pulmonary Metastases of Clear Cell Renal Cell Carcinoma. A Review of the Literature and Analysis of Four Metastatic Cases. Cancers 2021, 13, 5906. [Google Scholar] [CrossRef] [PubMed]
  2. Pitra, T.; Pivovarcikova, K.; Alaghehbandan, R.; Vesela, A.B.; Tupy, R.; Hora, M.; Hes, O. A Comprehensive Commentary on the Multilocular Cystic Renal Neoplasm of Low Malignant Potential: A Urologist’s Perspective. Cancers 2022, 14, 831. [Google Scholar] [CrossRef] [PubMed]
  3. Manini, C.; López-Fernández, E.; López, J.I. Towards Personalized Sampling in Clear Cell Renal Cell Carcinomas. Cancers 2022, 14, 3381. [Google Scholar] [CrossRef]
  4. Zhao, Y.; Fu, X.; López, J.I.; Rowan, A.; Au, L.; Fendler, A.; Hazell, S.; Xu, H.; Horswell, S.; Shepherd, S.T.C.; et al. Selection of metastasis competent subclones in the tumour interior. Nat. Ecol. Evol. 2021, 5, 1033–1045. [Google Scholar] [CrossRef]
  5. Sequeira, J.P.; Constâncio, V.; Salta, S.; Lobo, J.; Barros-Silva, D.; Carvalho-Maia, C.; Rodrigues, J.; Braga, I.; Henrique, R.; Jerónimo, C. LiKidMiRs: A ddPCR-Based Panel of 4 Circulating miRNAs for Detection of Renal Cell Carcinoma. Cancers 2022, 14, 858. [Google Scholar] [CrossRef]
  6. Gopal, N.; Anari, P.Y.; Turkbey, E.; Jones, E.C.; Malayeri, A.A. The Next Paradigm Shift in the Management of Clear Cell Renal Cancer: Radiogenomics—Definition, Current Advances, and Future Directions. Cancers 2022, 14, 793. [Google Scholar] [CrossRef] [PubMed]
  7. Vano, Y.-A.; Ladoire, S.; Elaidi, R.; Dermeche, S.; Eymard, J.-C.; Falkowski, S.; Gross-Goupil, M.; Malouf, G.; Narciso, B.; Sajous, C.; et al. First-Line Treatment of Metastatic Clear Cell Renal Cell Carcinoma: What Are the Most Appropriate Combination Therapies? Cancers 2021, 13, 5548. [Google Scholar] [CrossRef]
  8. Nunes-Xavier, C.E.; Angulo, J.C.; Pulido, R.; López, J.I. A Critical Insight into the Clinical Translation of PD-1/PD-L1 Blockade Therapy in Clear Cell Renal Cell Carcinoma. Curr. Urol. Rep. 2019, 20, 1. [Google Scholar] [CrossRef]
  9. Larrinaga, G.; Solano-Iturri, J.D.; Errarte, P.; Unda, M.; Loizaga-Iriarte, A.; Pérez-Fernández, A.; Echevarría, E.; Asumendi, A.; Manini, C.; Angulo, J.C.; et al. Soluble PD-L1 Is an Independent Prognostic Factor in Clear Cell Renal Cell Carcinoma. Cancers 2021, 13, 667. [Google Scholar] [CrossRef]
  10. Alvarez Ballesteros, P.; Chamorro, J.; San Román-Gil, M.; Pozas, J.; Gómez Dos Santos, V.; Ruiz Granados, A.; Grande, E.; Alonso-Gordoa, T.; Molina-Cerrillo, J. Molecular mechanisms of resistance to immunotherapy and antiangiogenic treatments in clear cell renal cell carcinoma. Cancers 2021, 13, 5981. [Google Scholar] [CrossRef]
  11. Angulo, J.; Manini, C.; López, J.; Pueyo, A.; Colás, B.; Ropero, S. The Role of Epigenetics in the Progression of Clear Cell Renal Cell Carcinoma and the Basis for Future Epigenetic Treatments. Cancers 2021, 13, 2071. [Google Scholar] [CrossRef] [PubMed]
  12. Kim, M.-C.; Jin, Z.; Kolb, R.; Borcherding, N.; Chatzkel, J.A.; Falzarano, S.M.; Zhang, W. Updates on Immunotherapy and Immune Landscape in Renal Clear Cell Carcinoma. Cancers 2021, 13, 5856. [Google Scholar] [CrossRef] [PubMed]
  13. Armesto, M.; Marquez, M.; Arestín, M.; Errarte, P.; Rubio, A.; Manterola, L.; López, J.I.; Lawrie, C.H. Integrated mRNA and miRNA transcriptomic analyses reveals divergent mechanisms of sunitinib resistance in clear cell renal cell carcinoma (ccRCC). Cancers 2021, 13, 4401. [Google Scholar] [CrossRef] [PubMed]
  14. Motzer, R.J.; Hutson, T.E.; Tomczak, P.; Michaelson, M.D.; Bukowski, R.M.; Rixe, O.; Oudard, S.; Negrier, S.; Szczylik, C.; Kim, S.T.; et al. Sunitinib versus Interferon Alfa in Metastatic Renal-Cell Carcinoma. N. Engl. J. Med. 2007, 356, 115–124. [Google Scholar] [CrossRef] [PubMed]
  15. Paderi, A.; Giorgione, R.; Giommoni, E.; Micol Mela, M.; Rossi, V.; Doni, L.; Minervini, A.; Carini, M.; Pillozzi, S.; Antonuzzo, L. Association between Immune Related Adverse Events and Outcome in Patients with Metastatic Renal Cell Carcinoma Treated with Immune Checkpoint Inhibitors. Cancers 2021, 13, 860. [Google Scholar] [CrossRef] [PubMed]
  16. Stenzel, P.J.; Hörner, N.; Foersch, S.; Wagner, D.-C.; Tsaur, I.; Thomas, A.; Haferkamp, A.; Macher-Goeppinger, S.; Roth, W.; Porubsky, S.; et al. Nivolumab Reduces PD1 Expression and Alters Density and Proliferation of Tumor Infiltrating Immune Cells in a Tissue Slice Culture Model of Renal Cell Carcinoma. Cancers 2021, 13, 4511. [Google Scholar] [CrossRef]
  17. Jee, B.; Seo, E.; Park, K.; Kim, Y.R.; Byeon, S.-J.; Lee, S.M.; Chung, J.H.; Song, W.; Sung, H.H.; Jeon, H.G.; et al. Molecular Subtypes Based on Genomic and Transcriptomic Features Correlate with the Responsiveness to Immune Checkpoint Inhibitors in Metastatic Clear Cell Renal Cell Carcinoma. Cancers 2022, 14, 2354. [Google Scholar] [CrossRef]
  18. Mattila, K.E.; Vainio, P.; Jaakkola, P.M. Prognostic Factors for Localized Clear Cell Renal Cell Carcinoma and Their Application in Adjuvant Therapy. Cancers 2022, 14, 239. [Google Scholar] [CrossRef]
  19. Martín-Saiz, L.; Mosteiro, L.; Solano-Iturri, J.D.; Rueda, Y.; Martín-Allende, J.; Imaz, I.; Olano, I.; Ochoa, B.; Fresnedo, O.; Fernández, J.A.; et al. High-Resolution Human Kidney Molecular Histology by Imaging Mass Spectrometry of Lipids. Anal. Chem. 2021, 93, 9364–9372. [Google Scholar] [CrossRef]
  20. Melana, J.; Mignolli, F.; Stoyanoff, T.; Aguirre, M.; Balboa, M.; Balsinde, J.; Rodríguez, J. The Hypoxic Microenvironment Induces Stearoyl-CoA Desaturase-1 Overexpression and Lipidomic Profile Changes in Clear Cell Renal Cell Carcinoma. Cancers 2021, 13, 2962. [Google Scholar] [CrossRef]
  21. Eckel-Passow, J.E.; Serie, D.J.; Cheville, J.C.; Ho, T.H.; Kapur, P.; Brugarolas, J.; Thompson, R.H.; Leibovich, B.C.; Kwon, E.D.; Joseph, R.W.; et al. BAP1 and PBRM1 in metastatic clear cell renal cell carcinoma: Tumor heterogeneity and concordance with paired primary tumor. BMC Urol. 2017, 17, 19. [Google Scholar] [CrossRef] [PubMed]
  22. Turajlic, S.; Xu, H.; Litchfield, K.; Rowan, A.; Chambers, T.; Lopez, J.I.; Nicol, D.; O’Brien, T.; Larkin, J.; Horswell, S.; et al. Tracking Cancer Evolution Reveals Constrained Routes to Metastases: TRACERx Renal. Cell 2018, 173, 581–594. [Google Scholar] [CrossRef] [PubMed]
  23. Brugarolas, J.; Rajaram, S.; Christie, A.; Kapur, P. The Evolution of Angiogenic and Inflamed Tumors: The Renal Cancer Paradigm. Cancer Cell 2020, 38, 771–773. [Google Scholar] [CrossRef] [PubMed]
  24. Saiga, K.; Ohe, C.; Yoshida, T.; Ohsugi, H.; Ikeda, J.; Atsumi, N.; Noda, Y.; Yasukochi, Y.; Higasa, K.; Taniguchi, H.; et al. PBRM1 Immunohistochemical Expression Profile Correlates with Histomorphological Features and Endothelial Expression of Tumor Vasculature for Clear Cell Renal Cell Carcinoma. Cancers 2022, 14, 1062. [Google Scholar] [CrossRef]
  25. Ohe, C.; Yoshida, T.; Amin, M.B.; Atsumi, N.; Ikeda, J.; Saiga, K.; Noda, Y.; Yasukochi, Y.; Ohashi, R.; Ohsugi, H.; et al. Development and validation of a vascularity-based architectural classification for clear cell renal cell carcinoma: Correlation with conventional pathological prognostic factors, gene expression patterns, and clinical outcomes. Mod. Pathol. 2022, 35, 816–824. [Google Scholar] [CrossRef]
  26. Khaleel, S.; Katims, A.; Cumarasamy, S.; Rosenzweig, S.; Attalla, K.; Hakimi, A.A.; Mehrazin, R. Radiogenomics in Clear Cell Renal Cell Carcinoma: A Review of the Current Status and Future Directions. Cancers 2022, 14, 2085. [Google Scholar] [CrossRef]
  27. Roldán, F.L.; Izquierdo, L.; Ingelmo-Torres, M.; Lozano, J.J.; Carrasco, R.; Cuñado, A.; Reig, O.; Mengual, L.; Alcaraz, A. Prognostic Gene Expression-Based Signature in Clear-Cell Renal Cell Carcinoma. Cancers 2022, 14, 3754. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Manini, C.; López, J.I. Updating Clear Cell Renal Cell Carcinoma (a Tribute to Prof. Ondrej Hes). Cancers 2022, 14, 3990. https://doi.org/10.3390/cancers14163990

AMA Style

Manini C, López JI. Updating Clear Cell Renal Cell Carcinoma (a Tribute to Prof. Ondrej Hes). Cancers. 2022; 14(16):3990. https://doi.org/10.3390/cancers14163990

Chicago/Turabian Style

Manini, Claudia, and José I. López. 2022. "Updating Clear Cell Renal Cell Carcinoma (a Tribute to Prof. Ondrej Hes)" Cancers 14, no. 16: 3990. https://doi.org/10.3390/cancers14163990

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop