Elsevier

European Urology

Volume 69, Issue 6, June 2016, Pages 1055-1061
European Urology

Platinum Priority – Kidney Cancer
Editorial by Antonio Lopez-Beltran, Liang Cheng, Maria R. Raspollini and Rodolfo Montironi on pp. 1062–1064 of this issue
Balanced Translocations Disrupting SMARCB1 Are Hallmark Recurrent Genetic Alterations in Renal Medullary Carcinomas

https://doi.org/10.1016/j.eururo.2015.09.027Get rights and content

Abstract

Background

Renal medullary carcinoma (RMC) is a rare and highly aggressive neoplasm that most often occurs in the setting of sickle cell trait or sickle cell disease (SCD). Most patients present with metastatic disease resistant to conventional chemotherapy, and therefore there is an urgent need for molecular insight to propose new therapies.

Objective

To determine the molecular alterations and oncogenic pathways that drive RMC development.

Design, setting, and participants

A series of five frozen samples of patients with RMC was investigated by means of gene expression profiling, array comparative genomic hybridization, and RNA and whole exome sequencing (WES).

Outcome measurements and statistical analysis

RNA and DNA sequencing read data were analyzed to detect gene fusions and somatic mutations. Gene fusions mutations were validated by real-time polymerase chain reaction and fluorescence in situ hybridization. Gene expression profiling was analyzed by unsupervised hierarchical clustering and Gene Set Enrichment Analysis (Broad Institute, Cambridge, MA, USA).

Results and limitations

We observed inactivation of the tumor suppressor gene SMARCB1 in all tumors. In all four cases developed in patients with SCD, we identified an original mechanism of interchromosomal balanced translocations that disrupt the SMARCB1 sequence and thus contribute to its inactivation. Gene expression profiling revealed that RMC shares common oncogenic pathways with pediatric malignant rhabdoid tumors, another tumor subtype characterized by SMARCB1 deficiency.

Conclusions

RMCs are characterized by an original mechanism of interchromosomal balanced translocations that disrupt the SMARCB1 sequence. WES reveals that RMCs show no other recurrent genetic alteration and an overall stable genome, underscoring the oncogenic potency of SMARCB1 inactivation.

Patient summary

Our comprehensive molecular study supports a pivotal role of the tumor suppressor gene SMARCB1 in the development of renal medullary carcinoma. The use of therapeutic strategies based on the biologic effects of its inactivation should now open new perspectives for this typically lethal malignancy.

Introduction

Renal medullary carcinoma (RMC) is a rare and highly malignant neoplasm that most often occurs in teenagers or young adults with sickle cell trait or disease (hereafter, SCD) [1]. RMCs have a very poor prognosis. Most patients present with metastatic disease resistant to conventional chemotherapy, and a fatal outcome occurs within a few months [1]. There is therefore an urgent need for molecular insight to propose new therapies.

The only recurrent alteration reported so far in RMC is BAF47 loss of immunohistochemical expression [2], [3], [4]. BAF47, encoded by the SMARCB1 gene, is a core component of the ubiquitous SWI/SNF complex that regulates gene transcription by modifying positions of nucleosomes and occupancy along the chromatin [5], [6]. Inactivation of this tumor suppressor gene was first identified in malignant rhabdoid tumors (RTs) [7], a highly aggressive pediatric neoplasm originally described in the kidney.

RTs constantly show biallelic inactivation of SMARCB1 through truncating mutations and/or deletions that invariably results in BAF47 loss of expression [7]. The complete abrogation of SMARCB1 dramatically impairs SWI/SNF function and thus the transcription of genes involved in the cell cycle or differentiating programs [5], [8], [9]. In RMC, however, only hemizygous deletions of SMARCB1 have been reported so far [2], [3]. Hence whether BAF47 loss is a genuine driver for RMC oncogenesis remains elusive, and the molecular mechanisms that underlie RMC development are overall poorly understood.

By investigating five cases of this very rare malignancy by means of array comparative genomic hybridization (array CGH), whole exome sequencing (WES), and RNA sequencing (RNA-seq), our study aimed to determine the molecular alterations that drive RMC development.

Section snippets

Tumor samples

Tumors were snap frozen in liquid nitrogen after surgical resection. The diagnosis of RMC was mainly assessed by Y.A. and J.C. or by the French panel of experts from the nephroblastoma group. BAF47 immunohistochemistry was assessed as described in Calderaro et al [2], and all tumors showed negative staining. Genomic analyses were performed according to French ethical Huriet law regarding research on human biologic samples. The tumor cell content was visually estimated before nucleic acid

Molecular alterations driving renal medullary carcinoma development

We first analyzed all five RMC samples by array CGH and identified a few large-scale CNVs per tumor (Fig. 1a and 1b; Supplementary Fig. 1). The only recurrent CNV implicated the chromosome 22q11 region and encompassed the SMARCB1 locus. All four RMC cases associated with SCD showed hemizygous SMARCB1 deletion and seemingly retained one intact copy of the other allele (Supplementary Fig. 1). The only homozygous deletion was observed in the tumor developed in an 8-yr-old patient with normal

Discussion

Because only hemizygous 22q11.2 deletions were evidenced in all RMCs reported so far, the actual driver role of SMARCB1 in this malignancy remained speculative. We have now demonstrated that RMCs are true SMARCB1-deficient cancers, by two lines of evidence.

First, we showed that SMARCB1 is genetically inactivated in RMC and that, in SCD patients, the second hit is constantly provided by a balanced translocation. This is remarkable because acquired translocations disrupting a tumor suppressor

Conclusions

Our study, the first comprehensive analysis of RMC, supports a pivotal role for SMARCB1 in its development, and the use of therapeutic strategies based on the biologic effects of its abrogation should now open new perspectives for this typically lethal malignancy.

References (24)

  • S. Lee et al.

    Aurora A is a repressed effector target of the chromatin remodeling protein INI1/hSNF5 required for rhabdoid tumor cell survival

    Cancer Res

    (2011)
  • B. Langmead et al.

    Fast gapped-read alignment with Bowtie 2

    Nat Methods

    (2012)
  • Cited by (83)

    • Molecular testing in gynecologic cancer

      2023, Diagnostic Molecular Pathology: A Guide to Applied Molecular Testing, Second Edition
    • Medullary carcinoma of the ampulla has distinct clinicopathologic characteristics including common association with microsatellite instability and PD-L1 expression

      2023, Human Pathology
      Citation Excerpt :

      More studies are warranted to further clarify this potential association. Loss of SMARCB1/INI1 protein expression has been reported in renal and colonic ‘MCs’ [36,37]. However, in this study, all ampullary MCs (8/8 with tissue available) showed intact SMARCB1/INI1.

    View all citing articles on Scopus

    Contributed equally.

    Contributed equally.

    View full text