Unique Transcriptomic Profile of Collecting Duct Carcinomas Relative to Upper Tract Urothelial Carcinomas and other Kidney Carcinomas

Gabriel G Malouf, Eva Compérat, Hui Yao, Roger Mouawad, Veronique Lindner, Nathalie Rioux-Leclercq, Virginie Verkarre, Xavier Leroy, Linda Dainese, Marion Classe, Jean-Luc Descotes, Philippe Barthelemy, Mokrane Yacoub, Morgan Rouprêt, Jean-Christophe Bernhard, Chad J Creighton, Jean-Philippe Spano, Xiaoping Su, David Khayat, Gabriel G Malouf, Eva Compérat, Hui Yao, Roger Mouawad, Veronique Lindner, Nathalie Rioux-Leclercq, Virginie Verkarre, Xavier Leroy, Linda Dainese, Marion Classe, Jean-Luc Descotes, Philippe Barthelemy, Mokrane Yacoub, Morgan Rouprêt, Jean-Christophe Bernhard, Chad J Creighton, Jean-Philippe Spano, Xiaoping Su, David Khayat

Abstract

Collecting duct carcinoma (CDC) is a kidney cancer subtype that is thought to arise from principal cells in distal parts of the collecting ducts. Some studies suggested an overlap of CDC with upper tract urothelial carcinoma (UTUC), making the pathological diagnosis challenging. Herein, we performed for the first time transcriptome sequencing of CDC and compared them to UTUC and renal cell carcinoma subtypes. We discovered that CDC displays a unique transcriptomic signature among kidney cancer subtypes, with a putative cell of origin in the distal convoluted tubules. Hierarchical unsupervised clustering reveals that the CDC signature is closer to that of other RCC subtypes than to UTUC, which is similar to that of bladder carcinoma. CDC is characterized by a metabolic shift, with impairment of oxidoreductase activity, pyruvate metabolism and the tricarboxlyic acid cycle, as well as an immunogenic response consistent with increased tumor infiltrating lymphocytes, particularly within metastatic cases. In addition, pathways differentially altered between CDC and UTUC point to a basal-like phenotype of CDC in contrast to the luminal-like signature of UTUC. We conclude that CDC harbors a pathognomonic transcriptomic signature characterized by immunogenic and a metabolic aberrations, indicating that targeting these processes might provide therapeutic options for patients.

Figures

Figure 1. Unsupervised clustering for the most…
Figure 1. Unsupervised clustering for the most differentially expressed mRNA.
(A) CDC forms a unique cluster compared to muscle-invasive UTUC and normal kidney tissue. (B) CDC compose a unique cluster compared to urothelial tumor (bladder carcinoma and UTUC) and other renal cell carcinoma subtypes, clear-cell RCC (KIRC), papillary RCC (KIRP), translocation RCC (Xp11RCC) and chromophobe RCC (KICH).
Figure 2. Association of CDC expression patterns…
Figure 2. Association of CDC expression patterns with those of specific regions of the nephron.
(A) The 8 kidney nephron regions evaluated: Glom, glomerulus; S1 and S3, the proximal tubule; mTAL, medullary thick ascending limb of Henle’s loop; cTAL, cortical thick ascending limb of Henle’s loop; DCT, distal convoluted tubule; CCD, cortical collecting duct; OMCD, outer medullary collecting duct. (B) Heat maps showing inter-sample correlations (red, positive) between mRNA profiles of RCC (columns) and mRNA profiles of nephron anatomical sites (rows). CDC, collecting duct carcinomas; KIRC, TCGA clear-cell RCC cases; KIRP, TCGA papillary RCC cases; Xp11RCC, TCGA translocation RCC cases; KICH, TCGA chromophobe RCC cases.
Figure 3
Figure 3
(A,B) Functional enrichment analysis of the genes differentially expressed between CDC and normal kidney tissue; (A) overexpressed in CDC; (B) underexpressed in CDC. (C) Gene set enrichment analysis showing impairment of pyruvate metabolism and tricarboxylic acid cycle in CDC. (D) Schematic of differential expression patterns of CDC versus clear-cell RCC in metabolism-related pathways, with a focus on gene expression patterns previously associated with Warburg-like effects in kidney cancer. P-values calculated by a t-test.
Figure 4. CDC and the immune system.
Figure 4. CDC and the immune system.
(A,B) Gene set enrichment analysis showing enrichment for positive regulation of T cell proliferation pathways, with the corresponding heatmap of differentially expressed genes. (C) Percentage of CD3 tumor infiltrating lymphocytes in all CDC cases and in metastatic versus non-metastatic cases. (D) Percentage of tumor infiltrating CD8 lymphocytes in all CDC cases and in metastatic versus non-metastatic cases. P-values calculated by a t-test.
Figure 5
Figure 5
(A,B) Functional enrichment analysis of the genes differentially expressed between CDC and UTUC; (A) overexpressed in CDC; (B) underexpressed in CDC. (C) Gene set enrichment analysis showing enrichment of luminal-like breast cancer signature versus mesenchymal signature in CDC compared to UTUC. (D) Box-plots of the expression of 10 genes previously reported as differentially expressed between luminal-like and basal-like breast cancer tumors.

References

    1. Kwon K. A. et al.. Clinical features and treatment of collecting duct carcinoma of the kidney from the korean cancer study group genitourinary and gynecology cancer committee. Cancer Res Treat 46, 141–147, 10.4143/crt.2014.46.2.141 (2014).
    1. Amin M. B. et al.. Collecting duct carcinoma versus renal medullary carcinoma: an appeal for nosologic and biological clarity. Am J Surg Pathol 38, 871–874, 10.1097/PAS.0000000000000222 (2014).
    1. Bose D., Das R. N., Chatterjee U. & Banerjee U. Collecting duct carcinoma: a rare malignancy. J Cancer Res Ther 9, 94–95, 10.4103/0973-1482.110387 (2013).
    1. Karakiewicz P. I. et al.. Collecting duct renal cell carcinoma: a matched analysis of 41 cases. Eur Urol 52, 1140–1145, 10.1016/j.eururo.2007.01.070 (2007).
    1. Gonzalez-Roibon N. et al.. The role of GATA binding protein 3 in the differential diagnosis of collecting duct and upper tract urothelial carcinomas. Hum Pathol 44, 2651–2657, 10.1016/j.humpath.2013.07.006 (2013).
    1. Oudard S. et al.. Prospective multicenter phase II study of gemcitabine plus platinum salt for metastatic collecting duct carcinoma: results of a GETUG (Groupe d’Etudes des Tumeurs Uro-Genitales) study. J Urol 177, 1698–1702, 10.1016/j.juro.2007.01.063 (2007).
    1. Becker F. et al.. Collecting duct carcinomas represent a unique tumor entity based on genetic alterations. PLoS One 8, e78137, 10.1371/journal.pone.0078137 (2013).
    1. Pal S. K. et al.. Characterization of Clinical Cases of Collecting Duct Carcinoma of the Kidney Assessed by Comprehensive Genomic Profiling. Eur Urol, 10.1016/j.eururo.2015.06.019 (2015).
    1. Albadine R. et al.. PAX8 (+)/p63 (−) immunostaining pattern in renal collecting duct carcinoma (CDC): a useful immunoprofile in the differential diagnosis of CDC versus urothelial carcinoma of upper urinary tract. Am J Surg Pathol 34, 965–969, 10.1097/PAS.0b013e3181dc5e8a (2010).
    1. Orsola A. et al.. Renal collecting (Bellini) duct carcinoma displays similar characteristics to upper tract urothelial cell carcinoma. Urology 65, 49–54, 10.1016/j.urology.2004.08.012 (2005).
    1. Linehan W. M., Srinivasan R. & Schmidt L. S. The genetic basis of kidney cancer: a metabolic disease. Nat Rev Urol 7, 277–285, 10.1038/nrurol.2010.47 (2010).
    1. Cheval L., Pierrat F., Rajerison R., Piquemal D. & Doucet A. Of mice and men: divergence of gene expression patterns in kidney. PLoS One 7, e46876, 10.1371/journal.pone.0046876 (2012).
    1. Charafe-Jauffret E. et al.. Gene expression profiling of breast cell lines identifies potential new basal markers. Oncogene 25, 2273–2284, 10.1038/sj.onc.1209254 (2006).
    1. Chen Z. et al.. Discovery of structure-based small molecular inhibitor of alphaB-crystallin against basal-like/triple-negative breast cancer development in vitro and in vivo. Breast Cancer Res Treat 145, 45–59, 10.1007/s10549-014-2940-8 (2014).
    1. Cheval L. et al.. Atlas of gene expression in the mouse kidney: new features of glomerular parietal cells. Physiol Genomics 43, 161–173, 10.1152/physiolgenomics.00093.2010 (2011).
    1. Linehan W. M. et al.. Comprehensive Molecular Characterization of Papillary Renal-Cell Carcinoma. N Engl J Med, 10.1056/NEJMoa1505917 (2015).
    1. Cancer Genome Atlas Research N. Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature 499, 43–49, 10.1038/nature12222 (2013).
    1. Srinivasan R., Ricketts C. J., Sourbier C. & Linehan W. M. New strategies in renal cell carcinoma: targeting the genetic and metabolic basis of disease. Clin Cancer Res 21, 10–17, 10.1158/1078-0432.CCR-13-2993 (2015).
    1. Mahmoud S. M. et al.. Tumor-infiltrating CD8+ lymphocytes predict clinical outcome in breast cancer. J Clin Oncol 29, 1949–1955, 10.1200/JCO.2010.30.5037 (2011).
    1. Giraldo N. A. et al.. Orchestration and Prognostic Significance of Immune Checkpoints in the Microenvironment of Primary and Metastatic Renal Cell Cancer. Clin Cancer Res 21, 3031–3040, 10.1158/1078-0432.CCR-14-2926 (2015).
    1. Srigley J. R. et al.. The International Society of Urological Pathology (ISUP) Vancouver Classification of Renal Neoplasia. Am J Surg Pathol 37, 1469–1489, 10.1097/PAS.0b013e318299f2d1 (2013).
    1. Davis C. F. et al.. The somatic genomic landscape of chromophobe renal cell carcinoma. Cancer Cell 26, 319–330, 10.1016/j.ccr.2014.07.014 (2014).

Source: PubMed

3
S'abonner