Englander Institute for Precision Medicine

Detecting Neuroendocrine Prostate Cancer Through Tissue-Informed Cell-Free DNA Methylation Analysis.

TitleDetecting Neuroendocrine Prostate Cancer Through Tissue-Informed Cell-Free DNA Methylation Analysis.
Publication TypeJournal Article
Year of Publication2022
AuthorsBerchuck JE, Baca SC, McClure HM, Korthauer K, Tsai HK, Nuzzo PVitale, Kelleher KM, He M, Steinharter JA, Zacharia S, Spisak S, Seo J-H, Conteduca V, Elemento O, Auh J, Sigouros M, Corey E, Hirsch MS, Taplin M-E, Choueiri TK, Pomerantz MM, Beltran H, Freedman ML
JournalClin Cancer Res
Volume28
Issue5
Pagination928-938
Date Published2022 Mar 01
ISSN1557-3265
KeywordsCarcinoma, Neuroendocrine, Cell-Free Nucleic Acids, DNA Methylation, Humans, Male, Neuroendocrine Tumors, Prostate, Prostatic Neoplasms
Abstract

PURPOSE: Neuroendocrine prostate cancer (NEPC) is a resistance phenotype that emerges in men with metastatic castration-resistant prostate adenocarcinoma (CR-PRAD) and has important clinical implications, but is challenging to detect in practice. Herein, we report a novel tissue-informed epigenetic approach to noninvasively detect NEPC.

EXPERIMENTAL DESIGN: We first performed methylated immunoprecipitation and high-throughput sequencing (MeDIP-seq) on a training set of tumors, identified differentially methylated regions between NEPC and CR-PRAD, and built a model to predict the presence of NEPC (termed NEPC Risk Score). We then performed MeDIP-seq on cell-free DNA (cfDNA) from two independent cohorts of men with NEPC or CR-PRAD and assessed the accuracy of the model to predict the presence NEPC.

RESULTS: The test cohort comprised cfDNA samples from 48 men, 9 with NEPC and 39 with CR-PRAD. NEPC Risk Scores were significantly higher in men with NEPC than CR-PRAD (P = 4.3 × 10-7) and discriminated between NEPC and CR-PRAD with high accuracy (AUROC 0.96). The optimal NEPC Risk Score cutoff demonstrated 100% sensitivity and 90% specificity for detecting NEPC. The independent, multi-institutional validation cohort included cfDNA from 53 men, including 12 with NEPC and 41 with CR-PRAD. NEPC Risk Scores were significantly higher in men with NEPC than CR-PRAD (P = 7.5×10-12) and perfectly discriminated NEPC from CR-PRAD (AUROC 1.0). Applying the predefined NEPC Risk Score cutoff to the validation cohort resulted in 100% sensitivity and 95% specificity for detecting NEPC.

CONCLUSIONS: Tissue-informed cfDNA methylation analysis is a promising approach for noninvasive detection of NEPC in men with advanced prostate cancer.

DOI10.1158/1078-0432.CCR-21-3762
Alternate JournalClin Cancer Res
PubMed ID34907080
PubMed Central IDPMC8898270
Grant ListP50 CA097186 / CA / NCI NIH HHS / United States
T32 CA009172 / CA / NCI NIH HHS / United States
R37 CA241486 / CA / NCI NIH HHS / United States
P01 CA163227 / CA / NCI NIH HHS / United States

Weill Cornell Medicine Englander Institute for Precision Medicine 413 E 69th Street
Belfer Research Building
New York, NY 10021