Your browser doesn't support javascript.
loading
CDKN2C-Null Leiomyosarcoma: A Novel, Genomically Distinct Class of TP53/RB1-Wild-Type Tumor With Frequent CIC Genomic Alterations and 1p/19q-Codeletion.
Williams, Erik A; Sharaf, Radwa; Decker, Brennan; Werth, Adrienne J; Toma, Helen; Montesion, Meagan; Sokol, Ethan S; Pavlick, Dean C; Shah, Nikunj; Williams, Kevin Jon; Venstrom, Jeffrey M; Alexander, Brian M; Ross, Jeffrey S; Albacker, Lee A; Lin, Douglas I; Ramkissoon, Shakti H; Elvin, Julia A.
Afiliação
  • Williams EA; Foundation Medicine, Cambridge, MA.
  • Sharaf R; Foundation Medicine, Cambridge, MA.
  • Decker B; Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA.
  • Werth AJ; Christiana Hospital, Department of Obstetrics and Gynecology, Newark, DE.
  • Toma H; Christiana Hospital, Department of Obstetrics and Gynecology, Newark, DE.
  • Montesion M; Foundation Medicine, Cambridge, MA.
  • Sokol ES; Foundation Medicine, Cambridge, MA.
  • Pavlick DC; Foundation Medicine, Cambridge, MA.
  • Shah N; Foundation Medicine, Cambridge, MA.
  • Williams KJ; Department of Physiology, Department of Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA.
  • Venstrom JM; Foundation Medicine, Cambridge, MA.
  • Alexander BM; Foundation Medicine, Cambridge, MA.
  • Ross JS; Foundation Medicine, Cambridge, MA.
  • Albacker LA; Department of Pathology, State University of New York Upstate Medical University, Syracuse, NY.
  • Lin DI; Foundation Medicine, Cambridge, MA.
  • Ramkissoon SH; Foundation Medicine, Cambridge, MA.
  • Elvin JA; Foundation Medicine, Cambridge, MA.
Article em En | MEDLINE | ID: mdl-33015533
ABSTRACT

PURPOSE:

Leiomyosarcoma (LMS) harbors frequent mutations in TP53 and RB1 but few actionable genomic alterations. Here, we searched for recurrent actionable genomic alterations in LMS that occur in the absence of common untreatable oncogenic drivers.

METHODS:

Tissues from 276,645 unique advanced cancers, including 2,570 uterine and soft tissue LMS, were sequenced by hybrid-capture-based next-generation DNA and RNA sequencing/comprehensive genomic profiling of up to 406 genes. We characterized clinicopathologic features of relevant patient cases.

RESULTS:

Overall, 77 LMS exhibited homozygous copy loss of CDKN2C at chromosome 1p32.3 (3.0% of LMS). Genomic alterations (GAs) in TP53, RB1, and ATRX were rare compared with the remainder of the LMS cohort (11.7% v 73.4%, 0% v 54.5%, 2.6% v 24.5%, respectively; all P < .0001). CDKN2C-null LMS patient cases were significantly enriched for GAs in CIC (40.3% v 1.4%) at 19q13.2, CDKN2A (46.8% v 7.0%), and RAD51B (16.9% v 1.7%; all P < .0001). Chromosome arm-level aneuploidy analysis of available LMS patient cases (n = 1,284) found that 81% (58 of 72) of CDKN2C-null LMS exhibited 1p/19q-codeletion, a significant enrichment compared with 5.1% in the remainder of the LMS cohort (P < .0001). In total, 99% of CDKN2C-null LMS were in women; the median age was 61 years at surgery (range, 36-81 years). Fifty-five patient cases were uterine primary, four were nonuterine, and the remaining 18 were of uncertain primary site. Sixty percent of cases showed at least focal epithelioid variant histology. Most patients had advanced-stage disease, with 62% of confirmed uterine primary LMS at International Federation of Gynecology and Obstetrics stage IVB. We further validated our findings in two publicly available datasets The Cancer Genome Atlas and the Project GENIE initiative.

CONCLUSION:

CDKN2C-null LMS defines a genomically distinct tumor that may have prognostic and/or therapeutic clinical implications, including possible use of specific cyclin-dependent kinase inhibitors.

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Tipo de estudo: Prognostic_studies Idioma: En Ano de publicação: 2020 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Tipo de estudo: Prognostic_studies Idioma: En Ano de publicação: 2020 Tipo de documento: Article