Abstract

Simple SummaryOver the last two decades, our improved understanding of the pathobiology of androgen-addicted prostate cancer (PCa), and documented therapeutic advances/breakthroughs have not translated into any substantial or sustained curative benefit for patients treated with traditional ADT or novel immune checkpoint blockade therapeutics. This is invariably connected with the peculiar biology and intratumoral heterogeneity of PCa. Castration-resistant PCa, constituting ~30% of all PCa, remains a clinically enigmatic and therapeutically challenging disease sub-type, that is therapy-refractory and characterized by high risk for recurrence after initial response. Our findings highlight the role and exploitability of testosterone metabolic reprogramming in prostate TME for patient stratification and personalized/precision medicine based on the differential but concerted expression of molecular components of the proposed testosterone tetrad in patients with therapy-refractory, locally advanced, or recurrent PCa. The therapeutic exploitability and clinical feasibility of our proposed approach is suggested by our preclinical findings.Background: Testosterone plays a critical role in prostate development and pathology. However, the impact of the molecular interplay between testosterone-associated genes on therapy response and susceptibility to disease relapse in PCa patients remains underexplored. Objective: This study investigated the role of dysregulated or aberrantly expressed testosterone-associated genes in the enhanced dissemination, phenoconversion, and therapy response of treatment-resistant advanced or recurrent PCa. Methods: Employing a combination of multi-omics big data analyses, in vitro, ex vivo, and in vivo assays, we assessed the probable roles of HSD17B2, HSD17B3, SHBG, and SRD5A1-mediated testosterone metabolism in the progression, therapy response, and prognosis of advanced or castration-resistant PCa (CRPC). Results: Our bioinformatics-aided gene expression profiling and immunohistochemical staining showed that the aberrant expression of the HSD17B2, HSD17B3, SHBG, and SRD5A1 testosterone metabolic tetrad characterize androgen-driven PCa and is associated with disease progression. Reanalysis of the TCGA PRAD cohort (n = 497) showed that patients with SRD5A1-dominant high expression of the tetrad exhibited worse mid-term to long-term (≥5 years) overall survival, with a profoundly shorter time to recurrence, compared to those with low expression. More so, we observed a strong association between enhanced HSD17B2/SRD5A1 signaling and metastasis to distant lymph nodes (M1a) and bones (M1b), while upregulated HSD17B3/SHBG signaling correlated more with negative metastasis (M0) status. Interestingly, increased SHBG/SRD5A1 ratio was associated with metastasis to distant organs (M1c), while elevated SRD5A1/SHBG ratio was associated with positive biochemical recurrence (BCR) status, and shorter time to BCR. Molecular enrichment and protein–protein connectivity network analyses showed that the androgenic tetrad regulates testosterone metabolism and cross-talks with modulators of drug response, effectors of cell cycle progression, proliferation or cell motility, and activators/mediators of cancer stemness. Moreover, of clinical relevance, SHBG ectopic expression (SHBG_OE) or SRD5A1 knockout (sgSRD5A1) induced the acquisition of spindle fibroblastoid morphology by the round/polygonal metastatic PC-3 and LNCaP cells, attenuated their migration and invasion capability, and significantly suppressed their ability to form primary or secondary tumorspheres, with concomitant downregulation of stemness KLF4, OCT3/4, and drug resistance ABCC1, ABCB1 proteins expression levels. We also showed that metronomic dutasteride synergistically enhanced the anticancer effect of low-dose docetaxel, in vitro, and in vivo. Conclusion: These data provide proof of concept that re-reprogramming of testosterone metabolism through “SRD5A1 withdrawal” or “SHBG induction” is a workable therapeutic strategy for shutting down androgen-driven oncogenic signals, reversing treatment resistance, and repressing the metastatic/recurrent phenotypes of patients with PCa.

Highlights

  • Prostate cancer (PCa) is among the most diagnosed male malignancies globally

  • Bioinformatics-aided gene expression profiling showed that, compared to HSD17B2 (0.31-fold, p = 0.0092) and sex-hormone-binding globulin (SHBG) (0.93-fold, p = 0.78) mRNA expression levels, which are downregulated, HSD17B3 (2.96-fold, p = 1.11 × 10−16 ) and SRD5A1 (1.3-fold, p = 4.02 × 10−5 ) transcript levels are upregulated in patients with PCa from the TCGA PRAD cohort (n = 497) (Figure 1B)

  • Using samples from our PCa cohort (n = 56), compared to the moderate-strong immunoreactivity of HSD17B3 and SRD5A1 in high-grade PCa (Gleason score (GS) ≥ 8), normal prostate tissue (GS ≤ 5), and low-grade PCa (GS = 6–7) were characterized by null-mild protein expression, moderate-strong HSD17B2 and SHBG protein expression levels were observed in the normal prostate and low grade PCa tissues compared to null–mild protein expression in the high-grade samples (Figure 1C)

Read more

Summary

Introduction

Prostate cancer (PCa) is among the most diagnosed male malignancies globally. Considering that ~1/3 of patients who undergo radical prostatectomy for clinically localizedPCa suffer postoperative recurrence, androgen deprivation therapy (ADT) remains the treatment of choice for advanced and clinically localized PCa. The last decade has been characterized by increased exploration of selected genes and proteins (so called proteogenomics) in the oncogenicity, immunogenicity, disease progression, and therapy response, with piqued interest in how this gene(s) dictates treatment success or failure in patients (otherwise called precision medicine). This seamless integration of proteogenomics and precision medicine ( termed “precision proteogenomics”) is touted to provide comprehensive elucidation of disease mechanisms through discovery and validation of novel diagnostic/prognostic biomarkers, while concomitantly facilitating patient stratification into responders or non-responders to specific targeted therapies.

Objectives
Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call