HSD3B1 and resistance to androgen deprivation therapy in prostate cancer.

Authors

null

Jason W.D. Hearn

Cleveland Clinic, Cleveland, OH

Jason W.D. Hearn , Ghada AbuAli , Cristina Magi-Galluzzi , Chandana A. Reddy , Kai-Hsiung Chang , Eric A. Klein , Nima Sharifi

Organizations

Cleveland Clinic, Cleveland, OH, Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, Departments of Hematology and Oncology, Cancer Biology, and Urology, Taussig Cancer Institute and Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH

Research Funding

No funding sources reported

Background: The somatic mutation HSD3B1(1245A > C) has been mechanistically linked to castration-resistant prostate cancer by encoding a mutant enzyme that augments intratumoral dihydrotestosterone (DHT) synthesis. Given the HSD3B1(1245C) allele is also frequently found in the germline, we hypothesized men inheriting this variant allele would exhibit resistance to androgen deprivation therapy (ADT), as manifested by worse clinical outcomes. Methods: We used a large, prospectively maintained prostate cancer registry to identify men treated with ADT for biochemical failure in the post-prostatectomy setting who were without evidence of metastatic disease. We analyzed progression-free survival (PFS), distant metastasis-free survival (DMFS), and overall survival (OS) according to HSD3B1 genotype using Kaplan-Meier methods. Cox proportional hazards regression was performed to evaluate potential gene-dosage effects, with homozygous wild-type men serving as the reference group. Demographic and treatment characteristics were compared to assess for possible confounders using Fisher’s exact test and Kruskal-Wallis analysis of variance. Multivariable analysis (MVA) was performed to assess whether HSD3B1 genotype independently predicted clinical outcomes. Results: Of 118 men genotyped, 37% were homozygous wild-type, 53% were heterozygous, and 10% were homozygous variant. Demographic and treatment characteristics did not differ across groups. Median PFS diminished as a function of the number of variant alleles inherited (6.6 years in homozygous wild-type men, 4.1 years in heterozygotes, and 2.5 years in homozygous variant men; P = 0.01). Median DMFS likewise decreased according to the number of variant alleles inherited (9.1 years vs. 6.8 years vs. 3.6 years, respectively; P = 0.01). Finally, OS similarly diminished (5-year and 10-year OS: 82% and 55% vs. 74% and 35% vs. 58% and 0%, respectively; P = 0.006). On MVA, the associations between HSD3B1 genotype and metastasis (hazard ratio (HR) 2.76; P = 0.023) and death (HR 3.33; P = 0.016) were maintained. Conclusions: Inheritance of the variant HSD3B1(1245C) allele that enhances DHT synthesis may be a powerful predictor of resistance to ADT for prostate cancer.

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

2015 ASCO Annual Meeting

Session Type

Poster Discussion Session

Session Title

Genitourinary (Prostate) Cancer

Track

Genitourinary Cancer

Sub Track

Prostate Cancer

Citation

J Clin Oncol 33, 2015 (suppl; abstr 5020)

DOI

10.1200/jco.2015.33.15_suppl.5020

Abstract #

5020

Poster Bd #

12

Abstract Disclosures

Similar Abstracts

First Author: Edward Christopher Dee

Abstract

2024 ASCO Genitourinary Cancers Symposium

Impact of exposure to clinical trials and standard-of-care therapy in metastatic or recurrent prostate cancer: An update.

First Author: Grace G Kim

Abstract

2024 ASCO Gastrointestinal Cancers Symposium

Associations between BMI, morphomics and survival in 528 patients with metastatic pancreatic cancer.

First Author: Valerie Gunchick