Comparative transcriptomic profiling of renal medullary carcinoma (RMC) to determine distinct signatures and pathways associated with response to chemotherapy.

Authors

null

Pavlos Msaouel

University of Texas MD Anderson Cancer Center, Houston, TX

Pavlos Msaouel , Gabriel G. Malouf , Xiaoping Su , Hui Yao , Durga N Tripathi , Jianjun Gao , Priya Rao , Rong He , Jose A. Karam , Christopher G. Wood , Cheryl Lyn Walker , Nizar M. Tannir

Organizations

University of Texas MD Anderson Cancer Center, Houston, TX, Pitié-Salpêtrière Hospital, Paris, France, Baylor College of Medicine, Houston, TX, The University of Texas MD Anderson Cancer Center, Houston, TX

Research Funding

Conquer Cancer Foundation of the American Society of Clinical Oncology

Background: RMC is a highly aggressive renal cell carcinoma (RCC) with poor prognosis and variable response to chemotherapy. It is often treated with cytotoxic regimens used for collecting duct carcinoma (CDC) and upper tract urothelial carcinoma (UTUC). We analyzed the transcriptomic profile of RMC samples and compared it with that of CDC, UTUC, clear-cell renal cell carcinoma (ccRCC), papillary RCC (pRCC), and chromophobe RCC (chRCC). Methods: RNA sequencing (RNA-seq) was performed in primary tumor tissues from treatment-naïve patients with RMC (n = 11), CDC (n = 9), and UTUC (n = 22). We compared these profiles to ccRCC, pRCC, and chRCC samples (n = 20 each) randomly selected from The Cancer Genome Atlas (TCGA). Gene ontology (GO) analysis was performed using DAVID. In vitro cell proliferation was assessed using the MTT assay. Results: Unsupervised hierarchical clustering and principal component analyses showed that 9/11 RMC samples formed a distinct cluster while 2 samples overlapped with CDC (p = 0.0003). There was no overlap between RMC and UTUC, ccRCC, pRCC, or chRCC. Five patients with RMC showed at least partial response to subsequent chemotherapy (4/5 received a paclitaxel-based regimen) and their pre-treatment samples demonstrated upregulation of 62 genes (FDR < 0.05) compared with the other patients. GO analysis revealed that these genes are associated with transmembrane transport pathways (enrichment score 3.87). The most upregulated gene ( > 3-fold) was guanine deaminase (GDA), a regulator of microtubule assembly normally expressed in human kidney collecting ducts. In vitro siRNA knockdown of GDA in the paclitaxel-sensitive RCC cell lines HCR-59 and UOK-146 conferred resistance to paclitaxel but not to drugs that do not target microtubules such as gemcitabine, doxorubicin, or platinum agents. Conclusions: RMC displays a unique transcriptomic signature that is closer to CDC than to UTUC or to other RCC subtypes. RMC tumors more likely to respond to chemotherapy show upregulation of genes such as GDA. Our results provide the first evidence that GDA may be a biomarker of response to microtubule-targeting agents in RMC.

Disclaimer

This material on this page is ©2024 American Society of Clinical Oncology, all rights reserved. Licensing available upon request. For more information, please contact licensing@asco.org

Abstract Details

Meeting

2018 ASCO Annual Meeting

Session Type

Poster Session

Session Title

Genitourinary (Nonprostate) Cancer

Track

Genitourinary Cancer—Kidney and Bladder

Sub Track

Kidney Cancer

Citation

J Clin Oncol 36, 2018 (suppl; abstr 4575)

DOI

10.1200/JCO.2018.36.15_suppl.4575

Abstract #

4575

Poster Bd #

401

Abstract Disclosures

Funded by Conquer Cancer

Similar Abstracts

Abstract

2024 ASCO Genitourinary Cancers Symposium

Spatial analysis of the tumor immune microenvironment in papillary renal cell carcinoma.

First Author: Mitch Hayes

Abstract

2016 ASCO Annual Meeting

Transcriptomic profiling of collecting duct carcinoma reveals metabolic and immune aberrations.

First Author: Gabriel G. Malouf

Abstract

2022 ASCO Genitourinary Cancers Symposium

CD70 is a promising CAR-T cell target in patients with advanced renal cell carcinoma.

First Author: Huihui Ye

Abstract

2020 Genitourinary Cancers Symposium

Transcriptomic profiling as proof of concept for investigation of collecting duct carcinoma (CDC).

First Author: Giuseppe Procopio