Evidence-Based Liquid Biopsy Knowledge
⚠️ IMPORTANT DISCLAIMER:

This content is AI-generated and has not yet been reviewed by medical professionals. The information presented here is for educational purposes only and should NOT be used to make medical decisions. Always consult with qualified healthcare providers for medical advice.

Prostate Cancer

Genotyping Identifies Actionable Targets; PSA Remains Superior for MRD Monitoring

Clinical Overview

Prostate cancer is the most common non-cutaneous malignancy in men, with metastatic castration-resistant prostate cancer (mCRPC) representing the lethal phase of disease progression. The clinical utility of ctDNA in prostate cancer differs substantially from other solid tumors: genotyping for treatment selection has established clinical value, while MRD monitoring remains investigational.

Prostate-specific antigen (PSA) remains the established biomarker for disease monitoring, with higher sensitivity and earlier detection of recurrence compared to ctDNA. However, molecular profiling through ctDNA testing identifies actionable genomic alterations in 12-20% of mCRPC patients, particularly DNA damage repair (DDR) mutations that predict response to PARP inhibitor therapy.

Clinical Utility Summary

  • Established Utility: Genotyping in mCRPC to identify BRCA1/2 and other DDR mutations for PARP inhibitor therapy
  • Emerging Utility: AR-V7 splice variant detection to select between androgen receptor-targeted therapy and taxane chemotherapy
  • Investigational: MRD monitoring (PSA remains more sensitive and cost-effective)
  • Limited Utility: MRD detection in localized disease (detection rates below 20%)

ctDNA Testing Methodology

Prostate cancer ctDNA testing employs targeted sequencing approaches to detect somatic mutations and structural variants in cell-free DNA isolated from plasma samples.

Technical Considerations:

  • Approach: Tumor-agnostic testing using fixed gene panels covering DDR genes, AR pathway, and common oncogenic drivers
  • Sample Type: Peripheral blood plasma (10-20 mL typical collection)
  • Detection Methodology: Next-generation sequencing or digital PCR for specific variants
  • Turnaround Time: 7-14 days for comprehensive genomic profiling

Key Technical Challenge: Clonal hematopoiesis of indeterminate potential (CHIP) is particularly problematic in prostate cancer given the older patient population (median age 68-72 years). CHIP mutations in DDR genes (especially ATM) can create false-positive results requiring careful bioinformatic filtering and variant interpretation.

MRD Detection: Limited Clinical Utility

Clinical Context: Unlike colorectal and breast cancer where ctDNA MRD monitoring has demonstrated clinical utility, prostate cancer MRD detection faces significant limitations due to low ctDNA shedding and the availability of PSA as a superior biomarker.

Detection Performance by Disease Stage

Disease Setting Detection Rate Clinical Utility
Localized Disease (Post-Prostatectomy) <20% Minimal - PSA is more sensitive
Biochemical Recurrence 20-40% Limited - PSA kinetics more informative
Metastatic Hormone-Sensitive 50-70% Investigational
Metastatic Castration-Resistant 50-77% Prognostic correlation demonstrated

Prognostic Value in Metastatic Disease

Published Data on ctDNA Prognostic Value:

  • Hazard Ratio for Progression: HR 2.7-5.4 for ctDNA-positive vs ctDNA-negative patients
  • Detection Sensitivity in mCRPC: 50-77% (varies by disease burden and testing platform)
  • Correlation with Outcomes: Baseline ctDNA levels correlate with tumor burden and overall survival
  • Dynamic Changes: ctDNA clearance on treatment associated with improved progression-free survival

Critical Limitation: PSA Superiority

PSA monitoring remains the established biomarker for prostate cancer surveillance with distinct advantages over ctDNA:

  • Earlier Detection: PSA typically rises before ctDNA becomes detectable, particularly in low-volume disease
  • Higher Sensitivity: PSA detectable in >95% of biochemical recurrence vs 20-40% ctDNA detection
  • Cost-Effectiveness: PSA testing costs significantly less than ctDNA sequencing
  • Established Thresholds: PSA kinetics (doubling time, velocity) have validated prognostic significance
  • Clinical Validation: Decades of data linking PSA changes to clinical outcomes

Clinical Implication: ctDNA does NOT replace PSA monitoring. The primary value of ctDNA in prostate cancer is genotyping, not MRD detection.

Genotyping Clinical Utility

Clinical Context: The established clinical utility of ctDNA in prostate cancer is molecular profiling to identify actionable therapeutic targets, particularly in mCRPC where tissue biopsy may be challenging due to bone-predominant metastases.

1. BRCA1/2 and DNA Damage Repair Mutations

Homologous recombination repair (HRR) deficiency creates synthetic lethality with PARP inhibition, forming the basis for precision therapy in DDR-mutant prostate cancer.

Prevalence and Clinical Significance:

  • BRCA1/2 Mutations: 12-20% of mCRPC patients
  • BRCA2: Most common (8-12%), associated with aggressive disease and earlier metastasis
  • BRCA1: 2-5% of mCRPC, similar PARP inhibitor sensitivity
  • ATM Mutations: 5-7% of mCRPC, predictive for rucaparib response
  • Other DDR Genes: PALB2, RAD51, CDK12 collectively account for additional 5-10%

PROFOUND Trial: Level 1 Evidence for PARP Inhibition

The randomized phase 3 PROFOUND trial established olaparib efficacy in DDR-mutant mCRPC:

  • Study Design: 387 patients with HRR gene alterations randomized to olaparib vs physician's choice of enzalutamide or abiraterone
  • Primary Endpoint (BRCA1/2 cohort): Radiographic PFS HR 0.34 (95% CI 0.25-0.47, p<0.001)
  • Median rPFS: 7.4 months (olaparib) vs 3.6 months (control) in BRCA1/2 patients
  • Objective Response Rate: 33% vs 2% in BRCA1/2 cohort
  • Overall Survival Benefit: HR 0.69 (95% CI 0.50-0.97) in BRCA1/2 cohort
  • Broader HRR Cohort: More modest benefit (rPFS HR 0.49), driven primarily by BRCA1/2 patients

Clinical Application: BRCA1/2 testing is now standard of care in mCRPC. ctDNA testing enables identification without invasive biopsy of bone metastases, which are technically challenging and often non-diagnostic.

Additional DDR Mutations with Therapeutic Implications:

  • ATM mutations: Rucaparib approved based on TRITON2 and TRITON3 trials showing activity in ATM-mutant disease
  • CDK12 mutations (5% of mCRPC): Associated with increased tumor mutational burden and potential immunotherapy responsiveness, though clinical validation ongoing
  • PTEN loss: Common alteration (20-40%) associated with aggressive phenotype but no specific targeted therapy currently available

2. AR-V7 Splice Variant: Treatment Selection

The androgen receptor splice variant 7 (AR-V7) lacks the ligand-binding domain, conferring constitutive activation and resistance to AR-targeted therapies (abiraterone, enzalutamide).

AR-V7 Clinical Significance:

  • Prevalence: Detected in 15-20% of mCRPC patients at baseline, increasing to 40% after AR-targeted therapy failure
  • Mechanism: Constitutively active AR variant driving transcription independent of androgen binding
  • Detection Method: Requires RNA-based assays (not standard DNA sequencing), typically from circulating tumor cells or ctDNA

Clinical Utility: Predicting Taxane Benefit

Multiple studies demonstrate AR-V7 predicts differential benefit from taxane chemotherapy vs AR-targeted therapy:

  • AR-V7 Positive Patients: Superior outcomes with taxane chemotherapy (cabazitaxel, docetaxel) compared to AR-targeted agents
  • AR-V7 Negative Patients: Both taxanes and AR-targeted therapy effective, choice based on toxicity profile and patient preference
  • Median PSA Response: AR-V7+ patients: 0% response to AR-targeted therapy vs 29-37% response to taxanes
  • Progression-Free Survival: HR 2-3 for AR-targeted therapy in AR-V7+ vs AR-V7- patients

Clinical Application: AR-V7 testing can guide treatment selection in mCRPC, identifying patients most likely to benefit from taxane chemotherapy over additional AR-pathway inhibition.

3. Microsatellite Instability-High (MSI-H)

Mismatch repair deficiency leads to hypermutated tumors with high immunogenicity and exceptional responses to immune checkpoint inhibition.

MSI-H in Prostate Cancer:

  • Prevalence: 3% of mCRPC (rare but clinically significant)
  • Enrichment: Higher in ductal and intraductal histologic variants (~10%)
  • Mechanism: Germline or somatic mutations in MMR genes (MLH1, MSH2, MSH6, PMS2)
  • Therapeutic Implication: Pembrolizumab approved for MSI-H/dMMR solid tumors (tissue-agnostic indication)
  • Response Rate: Approximately 50% objective response rate in MSI-H prostate cancer

Clinical Application: MSI-H detection through ctDNA testing enables identification of this small subset of patients with dramatic immunotherapy responsiveness, avoiding need for tissue re-biopsy.

Clinical Summary

Prostate cancer illustrates an important distinction in precision oncology: ctDNA genotyping for treatment selection has established clinical utility, while MRD monitoring remains investigational due to PSA's superior performance characteristics.

Evidence-Based Recommendations

Established Clinical Utility:

  • Genomic profiling in mCRPC to identify BRCA1/2 and other DDR mutations predictive of PARP inhibitor benefit (Level 1 evidence from PROFOUND trial, HR 0.34)
  • MSI-H detection to identify the 3% of patients with dramatic immunotherapy responsiveness
  • AR-V7 testing to select between taxane chemotherapy and AR-targeted therapy in appropriate clinical contexts

Not Recommended:

  • MRD monitoring in localized disease - detection rates too low (<20%), PSA is superior
  • Replacing PSA monitoring - PSA remains more sensitive, earlier, and more cost-effective across all disease settings
  • Treatment escalation based on ctDNA alone - no interventional trial evidence supporting improved outcomes

Under Investigation:

  • ctDNA-guided treatment intensification or de-escalation in mCRPC
  • Serial monitoring to detect emergence of resistance mutations (AR, BRCA reversion)
  • ctDNA as surrogate endpoint in clinical trials

Bottom Line: The clinical value of ctDNA in prostate cancer is genotyping to guide targeted therapy selection, particularly PARP inhibitors in BRCA-mutant disease. PSA monitoring remains superior for MRD surveillance across all disease settings. Clinicians should focus ctDNA testing on molecular profiling in mCRPC rather than MRD monitoring, where PSA provides earlier, more sensitive, and more cost-effective disease surveillance.

References

  1. Lozano R et al. Circulating tumour DNA as a biomarker in metastatic prostate cancer. Nat Rev Urol 2024;21:544-559
  2. de Bono J et al. Olaparib for metastatic castration-resistant prostate cancer. N Engl J Med 2020;382:2091-2102
  3. Antonarakis ES et al. AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer. N Engl J Med 2014;371:1028-1038
  4. Abida W et al. Analysis of the prevalence of microsatellite instability in prostate cancer. JAMA Oncol 2019;5:471-478

Evidence summary as of January 2026 | Document Version: 2.0