This is an open access article under the terms of a license that permits non-commercial use, provided the original work is properly cited. © 2023 The Authors. Société Internationale d'Urologie Journal, published by the Société Internationale d'Urologie, Canada. SIUJ.ORG SIUJ • Volume 4, Number 4 • July 2023 Key Words Competing Interests Article Information Liquid biopsy, circulating tumor DNA, prostate cancer, personalized medicine, next-generation sequencing None declared. Received on December 3, 2022 Accepted on June 10, 2023 This article has been peer reviewed. Soc Int Urol J. 2023;4(4):273–286 DOI: 10.48083/RFSH8912 273 REVIEW — LIQUID BIOPSY Utility and Clinical Application of Circulating Tumor DNA (ctDNA) in Advanced Prostate Cancer Louise Kostos,1,2 Heidi Fettke,2,3 Edmond M. Kwan,4,5 Arun A. Azad1,2 1Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia 2Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia 3Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Australia 4Vancouver Prostate Centre, Department of Urologic Sciences, The University of British Columbia, Vancouver, Canada 5BC Cancer, Vancouver Centre, Vancouver, Canada Abstract The treatment landscape for metastatic prostate cancer has undergone significant changes in recent years. The availability of next-generation imaging techniques and the emergence of novel therapies have led to earlier and more aggressive treatment approaches for patients. However, despite these advancements, drug resistance and progression to castration-resistant disease remain inevitable. Understanding the molecular landscape of advanced prostate cancer lies at the forefront of being able to deliver personalized therapies and more robustly risk-stratify patients, when combined with clinical factors. Advanced prostate cancer is characterized by inter- and intratumoral heterogeneity, posing challenges in comprehensively analyzing the genomic tumor profile using a solitary tissue sample. Additionally, the disease often manifests as bone-predominant metastatic tumors, making biopsies impractical in many cases. Moreover, archival tissue samples from a prostatectomy specimen may not accurately represent the current state of the tumor. To overcome these limitations, liquid biopsies using plasma samples have emerged as a minimally invasive surrogate approach to obtain real-time information on the genomic tumor profile. Growing evidence confirms the excellent concordance of liquid biopsies with tissue samples, making them an attractive alternative to traditional tissue biopsies. These assays can provide predictive and prognostic information that may enhance patient discussions and influence treatment decisions. This review focuses on the evolution and utility of circulating tumor-derived DNA (ctDNA) liquid biopsy assays in metastatic prostate cancer. Background Despite recent treatment advances, metastatic prostate cancer (mPC) continues to be a leading global cause of cancer- related death in men worldwide, with a 5-year survival rate below 30%[1–3]. The treatment landscape for advanced disease has become increasingly complex over the past decade, with the availability of multiple systemic therapies such as taxanes, androgen receptor pathway inhibitors (ARPIs), poly (ADP-ribose) polymerase inhibitors (PARPi), and targeted radioligand therapy. Each of these therapies is administered alongside androgen deprivation therapy (ADT). There is an emphasis on early treatment intensification with the introduction of these therapies as doublet and even triplet regimens for metastatic hormone-sensitive prostate cancer (mHSPC)[4,5]. While some clinical subgroups achieve a clear survival benefit from these approaches, not all patients benefit from treatment intensification. Lingering questions remain regarding the optimal timing of treatment intensification or de-intensification, the ideal duration of treatment, and the optimal sequencing of available therapies. Therefore, there is an urgent need for novel predictive and prognostic biomarkers to assist with risk stratification and inform treatment decisions. To address this critical unmet need, it is crucial to prioritize the elucidation of the molecular landscape of advanced prostate cancer and apply it at an individual patient level. In this context, there is the continuous development of http://SIUJ.org tools for comprehensive molecular tumor profiling to guide treatment selection and sequencing. Currently, the gold standard approach for molecular biomarker assessment is analysis of tumor tissue[6]. However, collecting adequate tumor tissue in mPC, which often develops with bone lesions and deep abdominal lymph nodes, is not always feasible, with invasive biopsies often associated with significant procedural morbidities and low-quality samples that preclude serial, multisite biopsies[7–9]. Moreover, characterizing molecular changes during therapy and upon disease progression is challenging, potentially leading to the oversight of resistance-conferring or novel clinically actionable clones[10]. This is especially important considering that the lethal clone involved in metastatic dissemination may not originate from the dominant foci of the primary prostate tumor[11]. Liquid biopsy approaches to molecular tumor charac- terization have gained attention as attractive surrogates for tumor biopsy in advanced prostate cancer over the past decade. Liquid biopsies commonly detect biomark- ers such as circulating tumor cells (CTCs), cell-free DNA (cf DNA) or RNA (cf RNA), proteins, and extracellular vesicles[12]. Among these, plasma cfDNA has garnered the most interest because of its ease of sampling and established isolation and preparation protocols. The proportion of tumor-derived cf DNA is referred to as circulating tumor DNA (ctDNA), and although it is found in all fluid compartments of the body, it is best characterized from plasma. Furthermore, its short half- life (minutes to hours) and the ability to simultaneously profile both local and distant sites make it an ideal substrate for providing a comprehensive “snapshot” of the tumor[13–15]. The Current Landscape of ctDNA in Prostate Cancer Since the initial discovery of the connection between cancer and cfDNA in 1994, the field of ctDNA analysis in oncology has rapidly expanded, with FDA-approved commercia l assays a nd compa nion diagnost ics becoming standard-of-practice for genomic profiling in many cancer types[13,16,17]. In 2015, Azad et al. published the earliest clinical research involving genomic analysis of plasma ctDNA in advanced prostate cancer. The authors successfully identified somatic androgen receptor (AR) point mutations and focal copy number gains using targeted next-generation sequencing (NGS) and array comparative genomic hybridization, respectively[18]. Furthermore, they reported an association between plasma-detectable AR alterations and primary resistance to the ARPI enzalutamide, providing evidence that ctDNA can be exploited to identify and understand contemporary biomarkers. Subsequent studies have shown that in mPC, ctDNA is a high-fidelity substitute for solid tumor tissue-derived DNA and is capable of not only recapitulating the somatic landscape of a tumor but also identifying clinically relevant driver alterations missed by a single metastatic biopsy[10,19–21]. Additionally, through serial sampling before and during treatment, ctDNA has the potential to monitor tumor progression, provide prognostic information, and thus dictate tailored treatment plans[22,23]. The investigation of ctDNA biomarkers to prognosticate mPC and predict response to targeted therapies has become widespread, with liquid biopsy collection often incorporated into clinical trial design[24–27]. Technical Considerations for ctDNA Analysis As ctDNA gains significance in guiding precision-based care for men with mPC, a myriad of approaches and technological platforms is being employed (Table  1). Understanding which approach will provide the most robust data for a particular research question is crucial to translating ctDNA assays into the clinic. Advanced prostate cancer can be detected in 60% to 90% of patient plasma samples, with the ctDNA fraction varying widely among patients[10,21,28]. Consequently, high assay sensitivity is essential to avoid excluding patients from data analysis and minimize false-negative results t hat may compromise biomarker identif ication. Currently, ctDNA analysis techniques can be broadly categorized as candidate gene approaches (for < 10 loci) and high-throughput approaches[13]. Low-throughput candidate gene approaches such as digital droplet polymerase chain reaction (PCR) assays, have the highest sensitivity, enabling the detection of somatic mutations below 0.002% allelic frequency[29,30]. These approaches are valuable for monitoring treatment resistance and minimal residual disease when the targets are already known. High-throughput techniques such as NGS provide an unbiased approach to genomic analysis and are the preferred method for identifying mechanisms of treatment resistance and novel genomic biomarkers[31]. However, they are typically less sensitive and more expensive than candidate gene approaches. Recent advances in NGS technology, however, such as the inclusion of molecular barcoding, patient-specific custom panels, and significant cost reductions for short- read sequencing have enabled the detection of somatic alterations below 0.5% allelic frequency[32]. These improvements also allow for the detection of focal copy number abnormalities, which are crucial for examining the landscape of mPC. Previously, the prostate cancer genome was thought to be associated with few focal chromosomal gains or losses, but it is now clear that focal copy number alterations, such as focal deletions in PTEN or focal AR amplifications, play an integral role in tumor evolution and disease progression[33]. In addition to these pre-analytical assay decisions, the selection and design of the bioinformatics workflow used to profile ctDNA are crucial. mPC is typically char- acterized by high levels of copy number abnormalities, structural rearrangements, and genomic heterogene- ity among lesions[34,35]. Therefore, a comprehensive approach capable of detecting point mutations, struc- tural variants, copy number variants, and low-frequency subclonal somatic mutations is necessary for robust profiling of the prostate cancer genome. Application of ctDNA in Metastatic Castration-Resistant Prostate Cancer Most genomic studies have been conducted in patients with metastatic castration-resistant prostate cancer (mCRPC), initially using tissue samples and more recently incorporating plasma cf DNA analysis. Liquid biopsies exhibit excellent concordance with tissue samples and represent an attractive alternative to molecular profiling of the tumor[10,15]. As a peripheral blood sample contains ctDNA from multiple sites, this liquid biopsy approach has the added benefit Abbreviations ADT androgen deprivation therapy AR androgen receptor ARPI androgen receptor pathway inhibitor cfDNA cell-free DNA CNVs copy number variants ctDNA circulating tumor DNA DDR DNA damage response and repair HRR homologous recombination repair ICI immune checkpoint inhibitor IHC Immunohistochemistry mCRPC metastatic castration-resistant prostate cancer mHSPC metastatic hormone-sensitive prostate cancer mPC metastatic prostate cancer MSI microsatellite instability NGS next-generation sequencing OS overall survival PARPi poly (ADP-ribose) polymerase inhibitors PCR polymerase chain reaction PFS progression-free survival PSA prostate-specific antigen SNVs single nucleotide variants SVs structural variants TMB tumor mutational burden of capturing inter- and intratumoral heterogeneity, thereby offering valuable insights to inform treatment decisions that would otherwise be missed in a single-site metastatic biopsy. The potential clinical applications of ctDNA in mCRPC are outlined below (Figure 1). Pretreatment ctDNA fraction and profile for prognostication The prognostic value of pretreatment ctDNA levels has been firmly established in mCRPC, showing that higher ctDNA fraction is associated with shorter progression- free survival (PFS) and overall survival (OS) regardless of treatment received[10,18,47]. In a study evaluating 202 patients with mCRPC receiving first-line treatment with the ARPIs enzalutamide or abiraterone acetate, a high ctDNA fraction (> 30%) was associated not only with increased tumor burden (as indicated by elevated plasma levels of prostate-specific antigen [PSA], lactate dehydrogenase [LDH], and alkaline phosphatase [ALP]) but also with poor response to treatment even after adjusting for established clinical prognostic factors[48]. Similarly, a high baseline ctDNA fraction prior to taxane chemotherapy was associated with shorter radiographic PFS and OS, independent of other prognostic variables[49]. Furthermore, specific genomic abnormalities detected in ctDNA have prognostic i mpl icat ions for t reat ment outcomes. Pat ients treated with abiraterone acetate or enzalutamide who had baseline aberrations in tumor suppressor genes (TP53, RB1, or PTEN) exhibited worse survival outcomes compared to those who tested negative at baseline or showed undetectable levels by cycle 2 of treatment[47,48,50,51]. Therefore, a high pretreatment ctDNA fraction and the presence of tumor suppressor aberrations can facilitate informed discussions with patients about their treatment options and expected outcomes and potentially support a more aggressive approach to systemic therapy. Longitudinal monitoring of treatment response Trad it iona l ly, seria l ser u m PSA measu rements have been used to monitor response to treatment in mCRPC. However, PSA has limitations, as radiographic progression can occur in the absence of a PSA rise, and heavily pretreated patients with AR-independent disease may have no or low levels of PSA, making interpretation of potential response challenging[52,53]. Serial ctDNA assays offer an alternative method for treatment monitoring. An early reduction in cf DNA concentration or fraction (within the first 9 weeks) has been associated with longer PFS and OS in patients with mCRPC patients treated with taxanes, ARPIs, and PARP inhibitors[54–57]. This finding was maintained even after adjusting for known clinical risk factors. Similarly, a lack of response or persistent rise in ctDNA fraction has been associated with shorter PFS[57]. 275274 SIUJ.ORG SIUJ • Volume 4, Number 4 • July 2023SIUJ • Volume 4, Number 4 • July 2023 SIUJ.ORG REVIEW — LIQUID BIOPSY Utility and Clinical Application of Circulating Tumor DNA (ctDNA) in Advanced Prostate Cancer http://SIUJ.org http://SIUJ.org sample. Germline alterations can usually be detected through simultaneous analysis of leucocyte samples extracted from the buffy coat of peripheral blood after centrifugation. Determination of HRR status not only informs whether the patient can benefit from a PARP inhibitor but also predicts a favorable response to plat- inum chemotherapy[71]. It is important to note that clonal hematopoiesis of indeterminate potential involv- ing DNA repair genes may lead to false-positive results, and therefore ctDNA samples should be accompanied by a whole blood control to exclude such variants[72]. Prostate cancers with PTEN loss are more sensi- tive to AKT inhibition, as demonstrated by the radiographic PFS benefit when combining the AKT inhibitor ipatasertib with abiraterone acetate for patients with mCRPC and PTEN loss identified through tumor immunohistochemistry[73]. PTEN loss is also predictive of a poor response to abiraterone acetate while retaining sensitivity to docetaxel[74,75]. The prevalence of PTEN loss through cf DNA assay is comparable to that found in tissue, potentially eliminating the need for archival tissue or a fresh biopsy[76]. Prostate cancer is typically considered immunogen- ically “cold” due to minimal T-cell infiltrates failing to generate a significant peripheral antitumor response, with limited benefit from immune checkpoint inhibitor (ICI) therapy in unselected cohorts[77–79]. However, a subset of prostate cancer exhibits an immuno- genic phenotype that may benefit from such therapy. Early detection of treatment resistance Analysis of genomic alterations in patients with mCRPC has identified both primary and acquired mutations associated with treatment resistance. With the increasing integration of ARPIs earlier on in the mPC disease course, resistance and the development of aggressive neuroendocrine prostate cancer may become more prevalent[52,58]. Therefore, it is crucial to use ctDNA biopsies to investigate markers of ARPI resistance. The presence and magnitude of AR gene amplification have been associated with shorter PFS and OS[48,59–61]. Some AR short variants are more frequently detected in liquid biopsy samples than in tissue biopsies, making ctDNA an ideal tool for early detection of treatment-resistant clones[21]. This discordance between plasma and tissue is likely due to intratumoral heterogeneity in AR gene expression[62] and the ability of liquid biopsies to integrate genomic information from multiple metastatic sites. ctDNA may also be used to predict resistance to PARPi by detecting acquired BRCA reversion mutations, which are also more frequently detected in liquid biopsy samples compared to tissue and are thought to predict a poor response to PARPi[21,63]. However, a recent analysis of patients with BRCA-mutant mCRPC enrolled in the TRITON2 trial suggests this may not be the case, as patients who developed a BRCA reversion mutation while receiving rucaparib experienced better treatment outcomes[64]. In addition to detecting specific genomic aberrations, dynamic changes in ctDNA levels during TABLE 1. General comparison of ctDNA analysis platforms used in advanced prostate cancer Approach Detection method Genomic elements tested Limit of detection (VAF) Variant types detected Estimated turnaround time Cost Volume of plasma required Strengths Limitations Clinical uses References High-throughput WGS Entire genome 5%–10% SNVs, indels, CNVs, SVs, fusions, rearrangements 4–6 weeks High 10–20 mL Provides comprehensive genomic information, no prior knowledge of loci required High cost, time-consuming, requires complex bioinformatic analysis Comprehensive genomic profiling, identification of rare or novel alterations [36–39] WES Exons 5%–10% 3–4 weeks Moderate 10–20 mL Focuses on protein-coding regions of the genome, no prior knowledge of gene required High cost, time-consuming, misses noncoding regions, requires bioinformatic analysis Identification of novel targets and mechanisms of treatment resistance [39–42] Targeted NGS Panel of genes/regions 1%–5% 2–3 weeks Moderate 5–10 mL Cost-effective and allows focused analysis, high sensitivity High-cost, requires bioinformatic analysis, limited to selected targets, some methods cannot detect CNVs or SVs Targeted profiling, and monitoring of known alterations, i.e., MRD detection [39,42–44] Candidate gene Digital PCR Specific mutations ≤ 0.01% SNVs, indels, known mutations 1–2 weeks Low 1–5 mL Cost-effective, highly sensitive and precise detection of mutations Limited to known mutations and loci Detection of specific mutations for treatment allocation, monitoring treatment response [39,45,46] Conventional PCR Specific genes/regions < 1%–5% 1–2 weeks Low 1–5 mL Cost-effective and allows targeted analysis, no bioinformatic analysis required Limited to known targets, most methods only detect SNVs Targeted mutation analysis [39] CNVs: copy number variants; ctDNA: circulating tumor DNA; indel: insertion/deletion; MRD: minimal residual disease; NGS: next-generation sequencing; PCR: polymerase chain reaction; SNVs: single nucleotide variants; SVs: structural variants; VAF: variant allele frequency; WES: whole exome sequencing; WGS: whole genome sequencing. therapy are also valuable for early detection of a lack of treatment response or the development of progressive disease. Early reductions in plasma ctDNA levels from baseline have been observed in patients prior to clinical determinants of treatment response[65], while persistent detectable ctDNA have been associated with worse outcomes[47,49,66]. Facilitating selection of personalized treatment One of the most important advantages of ctDNA analysis is the ability to identify potentially actionable genomic aberrat ions, enabl i ng t he del iver y of personalized treatment plans (Table 2). Detecting AR gene amplifications from the outset may assist clinicians in providing tailored treatment plans, potentially favoring taxane chemotherapy due to the known resistance to ARPIs[67]. With the introduction of PARPi such as olaparib and rucaparib for patients with homologous recombi- nation repair (HRR) gene mutations[68,69], guidelines now recommend testing all patients with mCRPC for somatic and germline pathogenic HRR aberrations, including BRCA1 and BRCA2[70]. Typically, this testing is conducted on tissue samples, which often suffer from compromised DNA quality as they are archival pretreat- ment samples. However, ctDNA analysis provides an easily accessible alternative for HRR status testing, showing excellent concordance with tissue samples for HRR-related gene mutations[21,56], although this depends on tumor content (ie, ctDNA fraction) in the Biomarkers detectable in cfDNA assays can help iden- tify these patients and provide a rationale for treatment. A recent analysis found that patients with mCRPC and a tumor mutational burden (TMB) of greater than 10 mutations per megabase respond better to ICI therapy than chemotherapy[80]. Similarly, patients with mCRPC whose tumors harbor CDK12 mutations[81,82] and high microsatellite instability (MSI)[83] have shown vulner- ability to ICI therapy. Both CDK12 mutations and MSI can be detected using plasma ctDNA platforms, showing high concordance with matched tissue samples[84,85]. Somatic mutations in genes responsible for regulat- ing the Wnt signaling pathway are found in up to 20% of patients with mCRPC[43,86]. Activating mutations in the Wnt pathway, such as CTNNB1, are associated with resistance to ARPI, and CTNNB1 mutations occur more frequently in mCRPC cfDNA samples that have progressed on enzalutamide[50,87]. Consequently, the Wnt pathway has become an attractive target for therapeutic intervention, leading to extensive preclinical research into Wnt pathway inhibitors[88]. Despite the interest and development of several novel agents, Wnt-pathway–directed therapies are yet to be approved for clinical use. Finally, the transition to AR-independent mPC is driven by lineage plasticity and can result in neuroen- docrine differentiation. Confirming neuroendocrine features requires a repeat biopsy, which can be chal- lenging due to tumor heterogeneity and the associ- ated morbidity of metastatic biopsies. Neuroendocrine 277276 SIUJ.ORG SIUJ • Volume 4, Number 4 • July 2023SIUJ • Volume 4, Number 4 • July 2023 SIUJ.ORG REVIEW — LIQUID BIOPSY Utility and Clinical Application of Circulating Tumor DNA (ctDNA) in Advanced Prostate Cancer http://SIUJ.org http://SIUJ.org of tumor suppressor gene alterations in tissue samples is associated with early relapse and worse outcomes[95,96]. In plasma samples, baseline alterations in DNA damage response and repair (DDR) genes and loss-of-function alterations in TP53 are likewise associated with poorer PFS and OS[35]. Untreated mHSPC patients with somatic DDR mutations had significantly shorter OS and a shorter time to ADT failure[35], while the pres- ence of germline DDR alterations predicted shorter time to developing castration-resistant disease[98,99]. Such findings can assist clinicians with risk stratifica- tion and deciding when to intensify upfront treatment for patients with mHSPC. Patients with poor prognos- tic factors present at baseline, such as a high ctDNA fraction and/or DDR or tumor suppressor alterations, may be considered for a more aggressive treatment regimen or enrolment in clinical trials. Conversely, the absence of detectable ctDNA at baseline or the absence prostate cancer is enriched with tumor suppressor gene alterations (such as TP53, PTEN, RB1), heralding an aggressive disease phenotype resistant to standard therapeutic approaches[89,90]. cf DNA methylation assays matched with tissue samples have shown high concordance for identifying neuroendocrine features, potentially serving as a future surrogate for tissue biop- sies in cases where neuroendocrine transformation is suspected. ctDNA analysis is now being integrated into clinical trials, both as a supplementary test conducted alongside treatment and, more recently, as a means of determining treatment. There are two ongoing biomarker-directed clinical trials (ProBio and PC-BETS) using ctDNA anal- ysis to guide treatment allocation in mCRPC[91–93]. Metastatic Hormone-Sensitive Prostate Cancer The benefits of ctDNA in mHSPC are less established compared with mCRPC, primarily because of the TABLE 2. Examples of the clinical significance of specific ctDNA findings in advanced prostate cancer Disease setting ctDNA finding Clinical significance mHSPC Baseline ctDNA fraction • Higher pretreatment ctDNA fraction is predictive of ADT failure, shorter metastasis-free survival and OS[35,94]. Baseline tumor suppressor gene alterations • Associated with early relapse and worse survival outcomes[95,96]. • Abiraterone acetate + ADT less effective compared to patients without tumor suppressor gene alterations[97]. Baseline DDR alterations • Somatic DDR mutation associated with shorter PFS and OS[35]. • Germline DDR alterations predictive of a shorter time to developing castration-resistant disease[98,99]. Baseline AR aberrations • Any AR aberration was associated with poor OS compared to patients without detectable AR alterations[100]. SPOP mutation • Predictive of a favorable response to ARPIs and improved survival outcomes[101,102]. mCRPC ctDNA fraction • Higher ctDNA fraction correlates with shorter PFS as well as OS regardless of treatment received[10,13,47–49]. • An early reduction in cfDNA concentration or ctDNA fraction associated with longer PFS and OS[54–57,103]. • A lack of response or continued rise in ctDNA fraction has been associated with shorter PFS[47,49,57,66]. Baseline tumor suppressor gene alterations • Patients treated with ARPIs had worse survival outcomes compared to those without tumor suppressor gene alterations at baseline, or who reverted to undetectable by cycle 2 of treatment[47,48,50,51]. AR amplification • The presence, as well as magnitude, of AR gene amplification, has been associated with shorter PFS as well as OS[48,59–61]. HRR alterations • Presence of HRR mutation/s predicts sensitivity to PARPi as well as platinum chemotherapy[71]. CTNNB1 mutation • Wnt pathway activating mutations (such as CTNNB1) are associated with resistance to ARPI[50,87]. PTEN loss • Prostate cancers with PTEN loss on IHC are more sensitive to AKT inhibition[73]. PTEN loss is also predictive for poor response to abiraterone acetate, while sensitivity to docetaxel is retained[74,75]. CDK12 mutation, high-MSI, high TMB • A TMB of > 10 mutations per megabase[80], CDK12 mutations[81,82] and high MSI[83] predict sensitivity to ICI therapy. AR: androgen receptor; ARPI: androgen receptor pathway inhibitor; ctDNA: circulating tumor DNA; DDR: DNA damage response and repair; HRR: homologous recombination repair; IHC: Immunohistochemistry; mCRPC: metastatic castration-resistant prostate cancer; mHSPC: metastatic hormone-sensitive prostate cancer; MSI: microsatellite instability; OS: overall survival; PFS: progression-free survival; TMB: tumor mutational burden. AR: androgen receptor; ctDNA: circulating tumor DNA; NGS: next-generation sequencing. “Created with BioRender.com”. Reproduced with permission. FIGURE 1. Advantages, limitations, and clinical applications of ctDNA in advanced prostate cancer lower cf DNA yield and ctDNA fraction observed in lower-volume, less heavily pretreated disease and due to decreases in the abundance of ctDNA in plasma following administration of ADT[35]. ctDNA as a prognostic tool to guide upfront treatment intensification Kohli et al. demonstrated that baseline ctDNA fraction also holds prognostic value in mHSPC, with higher pretreatment ctDNA fractions predicting shorter OS. The combination of ctDNA fraction, volume of disease, and serum ALP levels was also more prognostic of survival than clinical factors alone, with low-volume metastatic disease and low ctDNA fraction associated with the longest OS[35]. A higher ctDNA fraction was also predictive of ADT failure and shorter metastasis- free survival[35,94]. Additionally, several prognostic genomic aberra- tions exist in mHSPC, and ctDNA analysis is a useful method for identifying them (see Table 2). The presence 279278 SIUJ.ORG SIUJ • Volume 4, Number 4 • July 2023SIUJ • Volume 4, Number 4 • July 2023 SIUJ.ORG REVIEW — LIQUID BIOPSY Utility and Clinical Application of Circulating Tumor DNA (ctDNA) in Advanced Prostate Cancer http://SIUJ.org http://SIUJ.org References 1. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer Statistics, 2021. 2021;71(1):7–33. doi: 10.3322/caac.21654. PMID: 33433946. 2. H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin.2021;71(3):209–249. doi: 10.3322/caac.21660. PMID: 33538338. 3. Rebello RJ, Oing C, Knudsen KE, Loeb S, Johnson DC, Reiter RE, et al. Prostate cancer. Nat Rev Dis Primers.2021;7(1):9. doi: 10.1038/ s41572-020-00243-0. PMID: 33542230. 4. Smith MR, Hussain M, Saad F, Fizazi K, Sternberg CN, Crawford ED, et al.; ARASENS Trial Investigators. Darolutamide and survival in metastatic, hormone-sensitive prostate cancer. N Engl J Med.2022;386(12):1132–1142. doi: 10.1056/NEJMoa2119115. PMID: 35179323; PMCID: PMC9844551. 5. Fizazi K, Foulon S, Carles J, Roubaud G, McDermott R, Fléchon A, et al.; PEACE-1 investigators. Abiraterone plus prednisone added to androgen deprivation therapy and docetaxel in de novo metastatic castration-sensitive prostate cancer (PE ACE-1): a multicentre, open-label, randomised, phase 3 study with a 2 × 2 factorial design. Lancet.2022;399(10336):1695–1707. doi: 10.1016/S0140- 6736(22)00367-1. PMID: 35405085. 6. Finzel A, Sadik H, Ghitti G, Laes JF. The combined analysis of solid and liquid biopsies provides additional clinical information to improve patient care. J Cancer Metastasis Treat.2018;4:21. doi: 10.20517/2394-4722.2018.10. 7. Hussain M, Corcoran C, Sibilla C, Fizazi K, Saad F, Shore N, et al. Tumor genomic testing for >4,000 men with metastatic castration-resistant prostate cancer in the phase III trial PROfound (Olaparib). Clin Cancer Res.2022;28(8):1518–1530. doi: 10.1158/1078-0432.CCR-21-3940. PMID: 35091440. 8. Holmes MG, Foss E, Joseph G, Foye A, Beckett B, Motamedi D, et al. CT-guided bone biopsies in metastatic castration-resistant prostate cancer: factors predictive of maximum tumor yield. J Vasc Interv Radiol.2017;28(8):1073–81.e1. doi: 10.1016/j.jvir.2017.04.019. PMID: 28549709. 9. Lorente D, Omlin A, Zafeiriou Z, Nava-Rodrigues D, Pérez-López R, Pezaro C, et al. Castration-resistant prostate cancer tissue acquisition from bone metastases for molecular analyses. Clin Genitourin Cancer.2016;14(6):485–493. doi: 10.1016/j.clgc.2016.04.016. PMID: 27246360; PMCID: PMC5132155. 10. Wyatt AW, Annala M, Aggarwal R, Beja K, Feng F, Youngren J, et al. Concordance of circulating tumor DNA and matched metastatic tissue biopsy in prostate cancer. J Natl Cancer Inst.2017;109(12):djx118. doi: 10.1093/jnci/djx118. PMID: 29206995; PMCID: PMC6440274. 11. Haffner MC, Mosbruger T, Esopi DM, Fedor H, Heaphy CM, Walker DA, et al. Tracking the clonal origin of lethal prostate cancer. J Clin Invest.2013;123(11):4918 – 4922. doi: 10.1172/JCI70354. PMID: 24135135; PMCID: PMC3809798. 12. Soda N, Rehm BHA, Sonar P, Nguyen NT, Shiddiky MJA. Advanced liquid biopsy technologies for circulating biomarker detection. J Mater Chem B.2019;7(43):6670 – 6704. doi: 10.1039/c9tb01490j. PMID: 31646316. 13. Fettke H, Kwan EM, Azad AA. Cell-free DNA in cancer: current insights. Cell Oncol (Dordr).2019;42(1):13–28. doi: 10.1007/s13402-018-0413-5. PMID: 30367445. 14. Kustanovich A, Schwartz R, Peretz T, Grinshpun A. Life and death of circulating cell-free DNA. Cancer Biol Ther.2019;20(8):1057–1067. doi: 10.1080/15384047.2019.1598759. PMID: 30990132; PMCID: PMC6606043. 15. Herberts C, Annala M, Sipola J, Ng SWS, Chen XE, Nurminen A, et al. Deep whole-genome ctDNA chronology of treatment-resistant prostate cancer. Nature.2022;608(7921):199 –208. doi: 10.1038/ s41586-022-04975-9. PMID: 35859180. 16. Sorenson GD, Pribish DM, Valone FH, Memoli VA, Bzik DJ, Yao SL. Soluble normal and mutated DNA sequences from single-copy genes in human blood. Cancer Epidemiol Biomarkers Prev.1994;3(1):67–71. 17. Bando H, Nakamura Y, Taniguchi H, Shiozawa M, Yasui H, Esaki T, et al. Effects of metastatic sites on circulating tumor DNA in patients with metastatic colorectal cancer. JCO Precis Oncol.2022(6):e2100535. doi: 10.1200/PO.21.00535. PMID: 35544728. 18. Azad AA, Volik SV, Wyatt AW, Haegert A, Le Bihan S, Bell RH, et al. Androgen receptor gene aberrations in circulating cell-free DNA: biomarkers of therapeutic resistance in castration-resistant prostate cancer. Clin Cancer Res.2015;21(10):2315–2324. doi: 10.1158/1078- 0432.CCR-14-2666. PMID: 25712683. 19. Chi KN, Barnicle A, Sibilla C, Lai Z, Corcoran C, Williams JA, et al. Concordance of BRCA1, BRCA2 (BRCA), and ATM mutations identified in matched tumor tissue and circulating tumor DNA (ctDNA) in men with metastatic castration-resistant prostate cancer (mCRPC) screened in the PROfound study. J Clin Oncol.2021;39(6_suppl):26. doi: 10.1200/JCO.2021.39.6_suppl.26. 20. Dong X, Zheng T, Zhang M, Dai C, Wang L, Wang L, et al. Circulating cell-free DNA-based detection of tumor suppressor gene copy number loss and its clinical implication in metastatic prostate cancer. Front Oncol.2021;11:720727. doi: 10.3389/fonc.2021.720727. PMID: 34504797; PMCID: PMC8422845. 21. Tukachinsky H, Madison RW, Chung JH, Gjoerup OV, Severson EA, Dennis L, et al. Genomic analysis of circulating tumor DNA in 3,334 patients with advanced prostate cancer identifies targetable BRCA alterations and AR resistance mechanisms. Clin Cancer Res.2021;27(11):3094–3105. doi: 10.1158/1078-0432.CCR-20-4805. PMID: 33558422; PMCID: PMC9295199. 22. Geeurickx E, Hendrix A. Targets, pitfalls and reference materials for liquid biopsy tests in cancer diagnostics. Mol Aspects Med.2020;72:100828. doi: 10.1016/j.mam.2019.10.005. PMID: 31711714. of poor prognostic aberrations may potentially spare the patient from unnecessary treatment toxicity. However, prospective data evaluating ctDNA as a prognostic tool to guide treatment decisions in mHSPC (alongside clin- ical parameters) is needed before ctDNA can be adopted into mainstream practice. Genomic aberrations to guide the choice of systemic therapy in metastatic hormone- sensitive prostate cancer Baseline tumor suppressor gene a lterations are associated with worse outcomes with AR PIs in mHSPC[97]. Furthermore, the phase 3 TITAN trial, where patients received apalutamide or placebo in combination with ADT for mHSPC, found that any AR aberration combined with detectable ctDNA at baseline is associated with poor OS[100]. In such cases, the addition of docetaxel as part of triplet therapy may be particularly important. Conversely, an SPOP mutation, which occurs in approximately 5% of patients with mHSPC, predicts a favourable response to ARPIs and improved survival outcomes[101]. Challenges and Limitations of ctDNA Profiling One significant limitation of ctDNA profiling in prostate cancer is the variability in ctDNA shed into the plasma, potentially resulting in undetectable plasma tumor content. Unfortunately, up to half of mPC patients have low plasma tumor fraction (< 20%), and the dynamics of ctDNA release mechanisms and relative contributions from different lesions are still not fully understood[59,76]. These samples pose challenges, as the high background signal can hinder the sensitive and specific detection of copy number variants, the identification of loss of heterozygosity (either by copy-loss or copy-neutral mechanisms), and the filtering of non-neoplastic somatic mutations arising from hematopoietic stem cells[72]. It is unlikely that ctDNA biopsies will completely replace genomic analysis of solid tissue, especially in earlier- disease stages where tumor burden is lower. Additional approaches such as methylation or tumor-informed sequencing are required to overcome the limitations of low ctDNA fraction. Incorporating ctDNA with current conventional methods will significantly advance our understanding of the biological processes underlying treatment resistance and response. Indeed, recent studies on PARP inhibitors in mCRPC have combined ctDNA analysis with tumor tissue testing to detect HRR alterations[104,105]. Continued advancements in DNA processing, sequencing technologies, and downstream bioinformatics ana lysis w ill enable the increasing integration of ctDNA into precision- oncology initiatives for mPC. However, due to the substantial infrastructural, technological, and financial requirements of ctDNA analysis, particularly for high-throughput assays, global access to ctDNA platforms at both the research and clinical levels is still limited. Furthermore, there is considerable lack of harmonization in the post-analytical stage, further complicating the implementation of ctDNA assays into the clinic (Figure 1). Significant efforts are still required to establish best practices for variant interpretation and reporting[106,107]. These considerations must be addressed before widespread clinical implementation of ctDNA profiling in mPC can occur. Conclusion The increasing complexity of optimal treatment selection and sequencing in mPC is compounded by the integration of multiple novel therapies. Clinicians urgently need the ability to molecularly profile patients to gain predictive and prognostic insights that will guide treatment decisions. The high concordance between ctDNA and tumor tissue samples, combined with its minimally invasive and easily accessible nature, makes ctDNA a highly attractive alternative to tissue biopsy for assessing a tumor’s molecular profile. By employing serial sampling, ctDNA can capture clonal heterogeneity across metastatic sites and track lineage plasticity as it develops, enabling early detection of resistant clones before they manifest clinically. However, before widespread adoption of ctDNA can be realized, several limitations must be addressed. These include improving the sensitivity of analysis techniques to detect aberrations at low allele frequencies and streamlining variant interpretation pipelines. Furthermore, extensive clinical validation with large sample sizes and eventual cost subsidization are prerequisites for the broad use of ctDNA in clinical practice. 281280 SIUJ.ORG SIUJ • Volume 4, Number 4 • July 2023SIUJ • Volume 4, Number 4 • July 2023 SIUJ.ORG REVIEW — LIQUID BIOPSY Utility and Clinical Application of Circulating Tumor DNA (ctDNA) in Advanced Prostate Cancer http://SIUJ.org http://SIUJ.org 44. Newman AM, Bratman SV, To J, Wynne JF, Eclov NC, Modlin LA, et al. An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage. Nat Med.2014;20(5):548–554. doi: 10.1038/ nm.3519. PMID: 24705333; PMCID: PMC4016134. 45. Du M, Huang CC, Tan W, Kohli M, Wang L. Multiplex digital PCR to detect amplifications of specific androgen receptor loci in cell-free DNA for prognosis of metastatic castration-resistant prostate cancer. Cancers (Basel).2020;12(8):2139. doi: 10.3390/cancers12082139. PMID: 32752286; PMCID: PMC7465398. 46. Ma Y, Luk A, Young FP, Lynch D, Chua W, Balakrishnar B, et al. Droplet digital PCR based androgen receptor variant 7 (AR-V7) detection from prostate cancer patient blood biopsies. Int J Mol Sci.2016;17(8). doi: 10.3390/ijms17081264. PMID: 27527157; PMCID: PMC5000662. 47. Jayaram A, Wingate A, Wetterskog D, Wheeler G, Sternberg CN, Jones R, et al. Plasma tumor gene conversions after one cycle abiraterone acetate for metastatic castration-resistant prostate cancer: a biomarker analysis of a multicenter international trial. Ann Oncol.2021;32(6):726–735. doi: 10.1016/j.annonc.2021.03.196. PMID: 33794293. 48. Annala M, Vandekerkhove G, Khalaf D, Taavitsainen S, Beja K, Warner EW, et al. Circulating tumor DNA genomics correlate with resistance to abiraterone and enzalutamide in prostate cancer. Cancer Discov.2018;8(4):444–457. doi: 10.1158/2159-8290.CD-17-0937. PMID: 29367197. 49. Mehra N, Dolling D, Sumanasuriya S, Christova R, Pope L, Carreira S, et al. Plasma Cell-free DNA concentration and outcomes from taxane therapy in metastatic castration-resistant prostate cancer from two phase III trials (FIRSTANA and PROSELICA). Eur Urol.2018;74(3):283– 291. doi: 10.1016/j.eururo.2018.02.013. PMID: 29500065; PMCID: PMC6090941. 50. Wyatt AW, Azad AA, Volik SV, Annala M, Beja K, McConeghy B, et al. Genomic alterations in cell-free DNA and enzalutamide resistance in castration-resistant prostate cancer. JAMA Oncol.2016;2(12):1598– 1606. doi: 10.1001/jamaoncol.2016.0494. PMID: 27148695; PMCID: PMC5097690. 51. Torquato S, Pallavajjala A, Goldstein A, Toro PV, Silberstein JL, Lee J, et al. Genetic alterations detected in cell-free DNA are associated with enzalutamide and abiraterone resistance in castration-resistant prostate cancer. JCO Precis Oncol.2019;3:PO.18.00227. doi: 10.1200/ PO.18.00227. PMID: 31131348; PMCID: PMC6532665. 52. Beltran H, Hruszkewycz A, Scher HI, Hildesheim J, Isaacs J, Yu EY, et al. The role of lineage plasticity in prostate cancer therapy resistance. Clin Cancer Res.2019;25(23):6916–6924. doi: 10.1158/1078-0432. CCR-19-1423. PMID: 31363002; PMCID: PMC6891154. 53. Bryce AH, Chen YH, Liu G, Carducci MA, Jarrard DM, Garcia JA, et al. Patterns of cancer progression of metastatic hormone-sensitive prostate cancer in the ECOG3805 CHA ARTED trial. Eur Urol Oncol.2020;3(6):717–724. doi: 10.1016/j.euo.2020.07.001. PMID: 32807727; PMCID: PMC7738423. 54. Sumanasuriya S, Seed G, Parr H, Christova R, Pope L, Bertan C, et al. Elucidating prostate cancer behaviour during treatment via low-pass whole-genome sequencing of circulating tumour DNA. Eur Urol.2021;80(2):243–253. doi: 10.1016/j.eururo.2021.05.030. PMID: 34103179; PMCID: PMC8329366. 55. Goodall J, Assaf ZJ, Shi Z, Seed G, Zhang L, Lauffer B, et al. Circulating tumor DNA (ctDNA) dynamics associate with treatment response and radiological progression-free survival (rPFS): analyses from a randomized phase II trial in metastatic castration-resistant prostate cancer (mCRPC). J Clin Oncol.2020;38(15_suppl):5508. doi: 10.1200/ JCO.2020.38.15_suppl.5508. 56. Goodall J, Mateo J, Yuan W, Mossop H, Porta N, Miranda S, et al.; TOPARP-A investigators. Circulating cell-free DNA to guide prostate cancer treatment with PARP inhibition. Cancer Discov.2017;7(9):1006–1017. doi: 10.1158/2159-8290.CD-17-0261. PMID: 28450425; PMCID: PMC6143169. 57. Annala M, Fu S, Bacon JV W, Sipola J, Iqbal N, Ferrario C, et al. Cabazitaxel versus abiraterone or enzalutamide in poor prognosis metastatic castration-resistant prostate cancer: a multicentre, randomised, open-label, phase II trial. Ann Oncol.2021;32(7):896–905. doi: 10.1016/j.annonc.2021.03.205. PMID: 33836265. 58. Bluemn EG, Coleman IM, Lucas JM, Coleman RT, Hernandez-Lopez S, Tharakan R, et al. Androgen receptor pathway-independent prostate cancer is sustained through FGF signaling. Cancer Cell.2017;32(4):474– 489.e6. doi: 10.1016/j.ccell.2017.09.003. PMID: 29017058; PMCID: PMC5750052. 59. Fettke H, Kwan EM, Docanto MM, Bukczynska P, Ng N, Graham LK, et al. Combined cell-free DNA and RNA profiling of the androgen receptor: clinical utility of a novel multianalyte liquid biopsy assay for metastatic prostate cancer. Eur Urol.2020;78(2):173 –180. doi: 10.1016/j. eururo.2020.03.044. PMID: 32487321; PMCID: PMC8216705. 60. Jayaram A, Wingate A, Wetterskog D, Conteduca V, Khalaf D, Sharabiani MTA, et al. Plasma androgen receptor copy number status at emergence of metastatic castration-resistant prostate cancer: a pooled multicohort analysis. JCO Precis Oncol.2019;3:PO.19.00123. doi: 10.1200/PO.19.00123. PMID: 32923850; PMCID: PMC7446348. 61. Tolmeijer SH, Boerrigter E, Schalken JA, Geerlings MJ, van Oort IM, van Erp NP, et al. A systematic review and meta-analysis on the predictive value of cell-free DNA-based androgen receptor copy number gain in patients with castration-resistant prostate cancer. JCO Precis Oncol.2020;4:714–729. doi: 10.1200/PO.20.00084. PMID: 35050750. 62. Kumar A, Coleman I, Morrissey C, Zhang X, True LD, Gulati R, et al. Substantial interindividual and limited intraindividual genomic diversity among tumors from men with metastatic prostate cancer. Nat Med.2016;22(4):369–378. doi: 10.1038/nm.4053. PMID: 26928463; PMCID: PMC5045679. 23. Crocetto F, Russo G, Di Zazzo E, Pisapia P, Mirto BF, Palmieri A, et al. Liquid biopsy in prostate cancer management-current challenges and future perspectives. Cancers (Basel).2022;14(13):3272. doi: 10.3390/ cancers14133272. PMID: 35805043; PMCID: PMC9265840. 24. Dhiantravan N, Emmett L, Joshua AM, Pattison DA, Francis RJ, Williams S, et al. UpFrontPSMA: a randomized phase 2 study of sequential (177) Lu-PSMA-617 and docetaxel vs docetaxel in metastatic hormone-naïve prostate cancer (clinical trial protocol). BJU Int.2021;128(3):331–342. doi: 10.1111/bju.15384. PMID: 33682320. 25. Maughan BL, Nussenzveig R, Swami U, Gupta S, Agarwal N. Prospective trial of nivolumab (Nivo) plus radium-223 (RA) in metastatic castration- resistant prostate cancer (mCRPC) evaluating circulating tumor DNA (ctDNA) levels as a biomarker of response. J Clin Oncol.2020;38(6_ suppl):TPS267. doi: 10.1200/JCO.2020.38.6_suppl.TPS267. 26. Agarwal N, Azad A, Fizazi K, Mateo J, Matsubara N, Shore ND, et al.; for the TALAPRO-3 investigational group. Talapro-3: a phase 3, double- blind, randomized study of enzalutamide (ENZA) plus talazoparib (TALA) versus placebo plus enza in patients with DDR gene mutated metastatic castration-sensitive prostate cancer (mCSPC). J Clin Oncol.2022;40(6_ suppl):TPS221. doi: 10.1200/JCO.2022.40.6_suppl.TPS221. 27. Clarke NW, Armstrong AJ, Thier y-Vuillemin A, Oya M, Shore N, Loredo E, et al.; for the PROpel Investigators. Abiraterone and olaparib for metastatic castration-resistant prostate cancer. NEJM Evidence.2022;1(9):EVIDoa2200043. doi: 10.1056/EVIDoa2200043. 28. Fettke H, Kwan EM, Bukczynska P, Steen JA, Docanto M, Ng N, et al. Independent prognostic impact of plasma NCOA 2 alterations in metastatic castration-resistant prostate cancer. Prostate.2021;81(13):992–1001. doi: 10.1002/pros.24194. PMID: 34254334. 29. Hussung S, Follo M, Klar RFU, Michalczyk S, Fritsch K, Nollmann F, et al. Development and clinical validation of discriminatory multitarget digital droplet PCR assays for the detection of hot spot KRAS and NRAS mutations in cell-free DNA. J Mol Diagn.2020;22(7):943–956. doi: 10.1016/j.jmoldx.2020.04.206. PMID: 32376474. 30. Odegaard JI, Vincent JJ, Mortimer S, Vowles JV, Ulrich BC, Banks KC, et al. Validation of a plasma-based comprehensive cancer genotyping assay utilizing orthogonal tissue- and plasma-based methodologies. Clin Cancer Res.2018;24(15):3539–3549. doi: 10.1158/1078-0432. CCR-17-3831. PMID: 29691297. 31. Chan HT, Chin Y M, Low SK. Circulating tumor DNA-based genomic profiling assays in adult solid tumors for precision oncology: recent advancements and future challenges. Cancers (Basel).2022;14(13):3275. doi: 10.3390/cancers14133275. PMID: 35805046; PMCID: PMC9265547. 32. Fet tke H, Steen JA, Kwan EM, Bukcz ynska P, Keer thikumar S, Goode D, et al. Analy tical validation of an error-corrected ultr a-sensitive c tDN A nex t- gener ation sequencing assay. BioTechniques.2020;69(2):133–140. doi: 10.2144/btn-2020-0045. PMID: 32654508. 33. Ulz P, Belic J, Graf R, Auer M, Lafer I, Fischereder K, et al. Whole- genome plasma sequencing reveals focal amplifications as a driving force in metastatic prostate cancer. Nat Comm.2016;7(1):12008. doi: 10.1038/ncomms12008. PMID: 27328849; PMCID: PMC4917969. 34. Baca SC, Garraway LA. The genomic landscape of prostate cancer. Front Endocrinol (Lausanne).2012;3:69. doi: 10.3389/fendo.2012.00069. PMID: 22649426; PMCID: PMC3355898. 35. Kohli M, Tan W, Zheng T, Wang A, Montesinos C, Wong C, et al. Clinical and genomic insights into circulating tumor DNA-based alterations across the spectrum of metastatic hormone-sensitive and castrate-resistant prostate cancer. EBioMedicine.2020;54:102728. doi: 10.1016/j.ebiom.2020.102728. PMID: 32268276; PMCID: PMC7186589. 36. van Dessel LF, van Riet J, Smits M, Zhu Y, Hamberg P, van der Heijden MS, et al. The genomic landscape of metastatic castration-resistant prostate cancers reveals multiple distinct genotypes with potential clinical impact. Nat Commun.2019;10(1):5251. doi: 10.1038/s41467- 019-13084-7. PMID: 31748536; PMCID: PMC6868175. 37. Crumbaker M, Chan EKF, Gong T, Corcoran N, Jaratlerdsiri W, Lyons RJ, et al. The impact of whole genome data on therapeutic decision- making in metastatic prostate cancer: a retrospective analysis. Cancers (Basel).2020;12(5):1178. doi: 10.3390/cancers12051178. PMID: 32392735; PMCID: PMC7280976. 38. Quigley DA, Dang HX, Zhao SG, Lloyd P, Aggarwal R, Alumkal JJ, et al. Genomic hallmarks and structural variation in metastatic prostate cancer. Cell.2018;174(3):758–769.e9. doi: 10.1016/j.cell.2018.06.039. PMID: 30033370; PMCID: PMC6425931. 39. Vnencak-Jones C, Berger M, Pao W. 2016. Types of molecular tumor testing. My Cancer Genome. Accessed June 25, 2023. https://www.mycancergenome.org/content/molecular-medicine/ types-of-molecular-tumor-testing/ 40. Beltran H, Eng K, Mosquera JM, Sigaras A, Romanel A, Rennert H, et al. Whole-exome sequencing of metastatic cancer and biomarkers of treatment response. JAMA Oncol.2015;1(4):466–474. doi: 10.1001/ jamaoncol.2015.1313. PMID: 26181256; PMCID: PMC4505739. 41. Ramesh N, Sei E, Tsai PC, Bai S, Zhao Y, Troncoso P, et al. Decoding the evolutionary response to prostate cancer therapy by plasma genome sequencing. Genome Biol.2020;21(1):162. doi: 10.1186/s13059-020- 02045-9. PMID: 32631448; PMCID: PMC7336456. 42. Han X, Wang J, Sun Y. Circulating tumor DNA as biomarkers for cancer detection. Genomics Proteomics Bioinformatics.2017;15(2):59–72. doi: 10.1016/j.gpb.2016.12.004. PMID: 28392479; PMCID: PMC5414889. 43. Beltran H, Yelensky R, Frampton GM, Park K, Downing SR, MacDonald T Y, et al. Targeted nex t-generation sequencing of advanced prostate cancer identifies potential therapeutic targets and disease heterogeneity. Eur Urol.2013;63(5):920–926. doi: 10.1016/j. eururo.2012.08.053. PMID: 22981675; PMCID: PMC3615043. 283282 SIUJ.ORG SIUJ • Volume 4, Number 4 • July 2023SIUJ • Volume 4, Number 4 • July 2023 SIUJ.ORG REVIEW — LIQUID BIOPSY Utility and Clinical Application of Circulating Tumor DNA (ctDNA) in Advanced Prostate Cancer https://www.mycancergenome.org/content/molecular-medicine/types-of-molecular-tumor-testing/ https://www.mycancergenome.org/content/molecular-medicine/types-of-molecular-tumor-testing/ http://SIUJ.org http://SIUJ.org 82. Wu YM, Cieślik M, Lonigro RJ, Vats P, Reimers MA, Cao X, et al.; PCF/SU2C International Prostate Cancer Dream Team. Inactivation of CDK12 delineates a distinct immunogenic class of advanced prostate cancer. Cell.2018;173(7):1770–1782.e14. doi: 10.1016/j. cell.2018.04.034. PMID: 29906450; PMCID: PMC6084431. 83. Abida W, Cheng ML, Armenia J, Middha S, Autio KA, Vargas HA, et al. Analysis of the prevalence of microsatellite instability in prostate cancer and response to immune checkpoint blockade. JAMA Oncol.2019;5(4):471–478. doi: 10.1001/jamaoncol.2018.5801. PMID: 30589920; PMCID: PMC6459218. 84. Willis J, Lefterova MI, Artyomenko A, Kasi PM, Nakamura Y, Mody K, et al. Validation of microsatellite instability detection using a comprehensive plasma-based genotyping panel. Clin Cancer Res.2019;25(23):7035–7045. doi: 10.1158/1078-0432.CCR-19-1324. PMID: 31383735. 85. Warner E, Herberts C, Fu S, Yip S, Wong A, Wang G, et al. BRCA2, ATM, and CDK12 defects differentially shape prostate tumor driver genomics and clinical aggression. Clin Cancer Res.2021;27(6):1650–1662. doi: 10.1158/1078-0432.CCR-20-3708. PMID: 33414135. 86. Robinson D, Van Allen EM, Wu YM, Schultz N, Lonigro RJ, Mosquera JM, et al. Integrative clinical genomics of advanced prostate cancer. Cell.2015;161(5):1215–1228. doi: 10.1016/j.cell.2015.05.001. 87. Isaacsson Velho P, Fu W, Wang H, Mirkheshti N, Qazi F, Lima FAS, et al. Wnt-pathway activating mutations are associated with resistance to first-line abiraterone and enzalutamide in castration-resistant prostate cancer. Eur Urol.2020;77(1):14–21. doi: 10.1016/j.eururo.2019.05.032. PMID: 31176623; PMCID: PMC6893106. 88. Zhang Z, Cheng L, Li J, Farah E, Atallah NM, Pascuzzi PE, et al. Inhibition of the Wnt/β-catenin pathway overcomes resistance to enzalutamide in castration-resistant prostate cancer. Cancer Res.2018;78(12):3147–3162. doi: 10.1158/0008-5472.CAN-17-3006. PMID: 29700003; PMCID: PMC6004251. 89. Beltran H, Prandi D, Mosquera JM, Benelli M, Puca L, Cyrta J, et al. Divergent clonal evolution of castration-resistant neuroendocrine prostate cancer. Nat Med.2016;22(3):298–305. doi: 10.1038/nm.4045. PMID: 26855148; PMCID: PMC4777652. 90. Conteduca V, Ku SY, Fernandez L, Dago-Rodriquez A, Lee J, Jendrisak A, et al. Circulating tumor cell heterogeneity in neuroendocrine prostate cancer by single cell copy number analysis. NPJ Precis Oncol. 2021;5 (1):76. doi: 10.1038/s41698- 021- 00211-1. PMID: 34385567; PMCID: PMC8361159. 91. Chi KN, Mukherjee S, Saad F, Winquist E, Ong M, Kolinsky MP, et al. Prostate cancer biomarker enrichment and treatment selection (PC-BETS) study: a Canadian cancer trials group phase II umbrella trial for metastatic castration-resistant prostate cancer (mCRPC). J Clin Oncol.2020;38(15_suppl):5551. doi: 10.1200/ JCO.2020.38.15_suppl.5551. 92. De Laere B, Crippa A, Discacciati A, Larsson B, Oldenburg J, Mortezavi A, et al.; ProBio Investigators. Clinical trial protocol for ProBio: an outcome-adaptive and randomised multiarm biomarker- driven study in patients with metastatic prostate cancer. Eur Urol Focus.2022;8(6):1617–1621. doi: 10.1016/j.euf.2022.03.005. PMID: 35317973. 93. Crippa A, De Laere B, Discacciati A, Larsson B, Connor JT, Gabriel EE, et al. The ProBio trial: molecular biomarkers for advancing personalized treatment decision in patients with metastatic castration-resistant prostate cancer. Trials.2020;21(1):579. doi: 10.1186/s13063-020- 04515-8. PMID: 32586393; PMCID: PMC7318749. 94. Vandekerkhove G, Struss WJ, Annala M, Kallio HML, Khalaf D, Warner EW, et al. Circulating tumor DNA abundance and potential utility in de novo metastatic prostate cancer. Eur Urol.2019;75(4):667–675. doi: 10.1016/j.eururo.2018.12.042. PMID: 30638634. 95. Hamid AA, Gray KP, Shaw G, MacConaill LE, Evan C, Bernard B, et al. Compound genomic alterations of TP53, PTEN, and RB1 tumor suppressors in localized and metastatic prostate cancer. Eur Urol.2019;76(1):89 –97. doi: 10.1016/j.eururo.2018.11.045. PMID: 30553611. 96. Schweizer MT, Ha G, Gulati R, Brown LC, McKay RR, Dorff T, et al. CDK12-mutated prostate cancer: clinical outcomes with standard therapies and immune checkpoint blockade. JCO Precis Oncol.2020;4:382–392. doi: 10.1200/po.19.00383. PMID: 32671317; PMCID: PMC7363399. 97. Velez MG, Kosiorek HE, Egan JB, McNatty AL, Riaz IB, Hwang SR, et al. Differential impact of tumor suppressor gene (TP53, PTEN, RB1) alterations and treatment outcomes in metastatic, hormone-sensitive prostate cancer. Prostate Cancer Prostatic Dis.2022 Sep;25(3):479- 483. doi: 10.1038/s41391-021-00430-4. PMID: 34294873; PMCID: PMC9385473. 98. Wei Y, Wu J, Gu W, Wang J, Lin G, Qin X, et al. Prognostic value of germline DNA repair gene mutations in de novo metastatic and castration-sensitive prostate cancer. Oncologist.2020;25(7):e1042– e1050. doi: 10.1634/theoncologist.2019-0495. PMID: 32190957; PMCID: PMC7356812. 99. Annala M, Struss WJ, Warner EW, Beja K, Vandekerkhove G, Wong A, et al. Treatment outcomes and tumor loss of heterozygosity in germline DNA repair-deficient prostate cancer. Eur Urol.2017;72(1):34–42. doi: 10.1016/j.eururo.2017.02.023. PMID: 28259476. 100. Agarwal N, Lucas J, Aguilar-Bonavides C, Thomas S, Gormley M, Chowdhury S, et al. Genomic aberrations associated with overall survival (OS) in metastatic castration-sensitive prostate cancer (mCSPC) treated with apalutamide (APA) or placebo (PBO) plus androgen deprivation therapy (ADT) in TITAN. J Clin Oncol.2022;40(16_ suppl):5066. doi: 0.1200/JCO.2022.40.16_suppl.5066. 63. Quigley D, Alumkal JJ, Wyatt AW, Kothari V, Foye A, Lloyd P, et al. Analysis of circulating cell-free DNA identifies multiclonal heterogeneit y of BRCA 2 reversion mutations associated with resistance to PARP inhibitors. Cancer Discov.2017;7(9):999–1005. doi: 10.1158/2159-8290.CD-17-0146. PMID: 28450426; PMCID: PMC5581695. 64. Loehr A, Hussain A, Patnaik A, Bryce AH, Castellano D, Font A, et al. Emergence of BRCA reversion mutations in patients with metastatic castration-resistant prostate cancer after treatment with rucaparib. Eur Urol.2022;83(3):200–209. doi: 10.1016/j.eururo.2022.09.010. PMID: 36243543. 65. Tolmeijer SH, Boerrigter E, Sumiyoshi T, Ng S, Kwan EM, Annala M, et al. On-treatment plasma ctDNA fraction and treatment outcomes in metastatic castration-resistant prostate cancer. J Clin Oncol. 2022;4 0 (16 _ suppl):5 051. doi: 10.120 0/ JCO.2022.40.16_suppl.5051. 66. Conteduca V, Casadei C, Scarpi E, Brighi N, Schepisi G, Lolli C, et al. Baseline plasma tumor DNA (ctDNA) correlates with PSA kinetics in metastatic castration-resistant prostate cancer (mCRPC) treated with abiraterone or enzalutamide. Cancers (Basel).2022;14(9):2219. doi: 10.3390/cancers14092219. PMID: 35565349; PMCID: PMC9102454. 67. Conteduca V, Jayaram A, Romero-Laorden N, Wetterskog D, Salvi S, Gurioli G, et al. Plasma androgen receptor and docetaxel for metastatic castration-resistant prostate cancer. Eur Urol.2019;75(3):368–373. doi: 10.1016/j.eururo.2018.09.049. PMID: 30773204; PMCID: PMC6377278. 68. Abida W, Patnaik A, Campbell D, Shapiro J, Bryce AH, McDermott R, et al.; TRITON2 investigators. Rucaparib in men with metastatic castration-resistant prostate cancer harboring a BRCA1 or BRCA2 gene alteration. J Clin Oncol.2020;38(32):3763–3772. doi: 10.1200/ JCO.20.01035. PMID: 32795228; PMCID: PMC7655021. 69. de Bono J, Mateo J, Fizazi K, Saad F, Shore N, Sandhu S, et al. Olaparib for metastatic castration-resistant prostate cancer. N Engl J Med.2020;382(22):2091–2102. doi: 10.1056/NEJMoa1911440. PMID: 32343890. 70. National Comprehensive Cancer Network (NCCN). Prostate Cancer V1.2023 2022. Available at: https://www.nccn.org/professionals/ physician_gls/pdf/prostate.pdf. Accessed June 25, 2023. 71. Mota JM, Barnett E, Nauseef JT, Nguyen B, Stopsack KH, Wibmer A, et al. Platinum-based chemotherapy in metastatic prostate cancer with DNA repair gene alterations. JCO Precis Oncol.2020;4:355–366. doi: 10.1200/po.19.00346. PMID: 32856010; PMCID: PMC7446522. 72. Jensen K, Konnick EQ, Schweizer MT, Sokolova AO, Grivas P, Cheng HH, et al. Association of clonal hematopoiesis in DNA repair genes with prostate cancer plasma cell-free DNA testing interference. JAMA Oncol.2021;7(1):107–110. doi: 10.1001/jamaoncol.2020.5161. PMID: 33151258; PMCID: PMC7645740. 73. Sweeney C, Bracarda S, Sternberg CN, Chi KN, Olmos D, Sandhu S, et al. Ipatasertib plus abiraterone and prednisolone in metastatic castration- resistant prostate cancer (IPATential150): a multicentre, randomised, double-blind, phase 3 trial. Lancet.2021;398(10295):131–142. doi: 10.1016/S0140-6736(21)00580-8. PMID: 34246347. 74. Ferraldeschi R, Nava Rodrigues D, Riisnaes R, Miranda S, Figueiredo I, Rescigno P, et al. PTEN protein loss and clinical outcome from castration-resistant prostate cancer treated with abiraterone acetate. Eur Urol.2015;67(4):795–802. doi: 10.1016/j.eururo.2014.10.027. PMID: 25454616; PMCID: PMC4410287. 75. Rescigno P, Lorente D, Dolling D, Ferraldeschi R, Rodrigues DN, Riisnaes R, et al. Docetaxel treatment in PTEN- and ERG-aberrant metastatic prostate cancers. Eur Urol Oncol.2018;1(1):71–77. doi: 10.1016/j.euo.2018.02.006. PMID: 29911685; PMCID: PMC5995869. 76. Kwan EM, Dai C, Fettke H, Hauser C, Docanto MM, Bukczynska P, et al. Plasma cell–free DNA profiling of PTEN-PI3K-AKT pathway aberrations in metastatic castration-resistant prostate cancer. JCO Precis Oncol.2021:(5):PO.20.00424. doi: 10.1200/PO.20.00424. PMID: 34250422; PMCID: PMC8232889. 77. May KF Jr, Gulley JL, Drake CG, Dranoff G, Kantoff PW. Prostate cancer immunotherapy. Clin Cancer Res.2011;17(16):5233–5238. doi: 10.1158/1078-0432.CCR-10-3402. PMID: 21700764; PMCID: PMC3263933. 78. Sharma P, Pachynski RK, Narayan V, Fléchon A, Gravis G, Galsky MD, et al. Nivolumab plus ipilimumab for metastatic castration-resistant prostate cancer: preliminary analysis of patients in the CheckMate 650 trial. Cancer Cell.2020;38(4):4 89 – 499.e3. doi: 10.1016/j. ccell.2020.08.007. PMID: 32916128. 79. Beer TM, Kwon ED, Drake CG, Fizazi K, Logothetis C, Gravis G, et al. Randomized, double-blind, phase III trial of ipilimumab versus placebo in asymptomatic or minimally symptomatic patients with metastatic chemotherapy-naive castration-resistant prostate cancer. J Clin Oncol.2017;35(1):40–47. doi: 10.1200/JCO.2016.69.1584. PMID: 28034081. 80. Graf RP, Fisher V, Weberpals J, Gjoerup O, Tierno MB, Huang RSP, et al. Comparative effectiveness of immune checkpoint inhibitors vs chemotherapy by tumor mutational burden in metastatic castration- resistant prostate cancer. JAMA Netw Open.2022;5(3):e225394. doi: 10.1001/jamanetworkopen.2022.5394. PMID: 35357449; PMCID: PMC8972027. 81. Gongora ABL, Marshall CH, Velho PI, Lopes CDH, Marin JF, Camargo AA, et al. Extreme responses to a combination of DNA-damaging therapy and immunotherapy in CDK12-altered metastatic castration- resistant prostate cancer: a potential therapeutic vulnerability. Clin Genitourin Cancer.2022;20(2):183–188. doi: 10.1016/j.clgc.2021.11.015. PMID: 35027313. 285284 SIUJ.ORG SIUJ • Volume 4, Number 4 • July 2023SIUJ • Volume 4, Number 4 • July 2023 SIUJ.ORG REVIEW — LIQUID BIOPSY Utility and Clinical Application of Circulating Tumor DNA (ctDNA) in Advanced Prostate Cancer https://www.nccn.org/professionals/physician_gls/pdf/prostate.pdf https://www.nccn.org/professionals/physician_gls/pdf/prostate.pdf http://SIUJ.org http://SIUJ.org 101. Nizialek E, Lim SJ, Wang H, Isaacsson Velho P, Yegnasubramanian S, Antonarakis ES. Genomic profiles and clinical outcomes in primary versus secondary metastatic hormone-sensitive prostate cancer. Prostate.2021;81(9):572– 579. doi: 10.1002/pros.24135. PMID: 33955569. 102. Swami U, Graf RP, Nussenzveig RH, Fisher V, Tukachinsky H, Schrock AB, et al. SPOP mutations as a predictive biomarker for androgen receptor axis–targeted therapy in de novo metastatic castration- sensitive prostate cancer. Clin Cancer Res.2022;28(22):4917–4925. doi: 10.1158/1078-0432.CCR-22-2228. PMID: 36088616. 103. Tolmeijer SH, Boerrigter E, Sumiyoshi T, Kwan EM, Ng S, Annala M, et al. Early on-treatment changes in circulating tumor DNA fraction and response to enzalutamide or abiraterone in metastatic castration- resistant prostate cancer. Clin Cancer Res.2023;CCR-22-2998. doi: 10.1158/1078-0432.CCR-22-2998. PMID: 36996325. 104. Loehr A, Patnaik A, Campbell D, Shapiro J, Bryce AH, McDermott R, et al. Response to rucaparib in BRCA-mutant metastatic castration- resistant prostate cancer identified by genomic testing in the TRITON2 study. Clin Cancer Res.2021;27(24):6677–6686. doi: 10.1158/1078- 0432.CCR-21-2199. PMID: 34598946; PMCID: PMC8678310. 105. Chi KN, Barnicle A, Sibilla C, Lai Z, Corcoran C, Barrett JC, et al. Detection of BRCA1, BRCA 2, and ATM alterations in matched tumor tissue and circulating tumor DNA in patients with prostate cancer screened in PROfound. Clin Cancer Res.2023;29(1):81–91. doi: 10.1158/1078-0432.CCR-22-0931. PMID: 36043882; PMCID: PMC9811161. 106. Taavitsainen S, Annala M, Ledet E, Beja K, Miller PJ, Moses M, et al. Evaluation of commercial circulating tumor DNA test in metastatic prostate cancer. JCO Precis Oncol.2019;3:PO.19.00014. doi: 10.1200/ PO.19.00014. PMID: 32914020; PMCID: PMC7446428. 107. Kwan EM, Wyatt AW, Chi KN. Towards clinical implementation of circulating tumor DNA in metastatic prostate cancer: opportunities for integration and pitfalls to interpretation. Front Oncol.2022;12:1054497. doi: 10.3 389/fonc.2022.105 4 4 97. PMID: 36 4 39 4 51; PMCID: PMC9685669. 286 SIUJ • Volume 4, Number 4 • July 2023 SIUJ.ORG REVIEW — LIQUID BIOPSY http://SIUJ.org