Abstract
Understanding the genomic profiling of prostate cancer is crucial, owing to the emergence of precision medicine to guide therapeutic approaches. Over the last decade, integrative genomic profiling of prostate tumors has provided insights that improve the understanding and treatment of the disease. Minimally invasive liquid biopsy procedures have emerged to investigate cancer-related molecules with the advantage of detecting heterogeneity as well as acquired resistance in cancer. The metastatic castration-resistant prostate cancer (mCRPC) tumors have a highly complex genomic landscape compared to primary prostate tumors; a number of mCRPC harbor clinically actionable molecular alterations, including DNA damage repair (e.g., BRCA1/2 and ATM) and PTEN/phosphoinositide 3-kinase signaling. Heterogeneity in the genomic landscape of prostate cancer has become apparent and genomic alterations of TP53, RB1, AR, and cell cycle pathway are associated with poor clinical outcomes in patients. Prostate cancer with mutant SPOP shows a distinct pattern of genomic alterations, associating with better clinical outcomes. Several genomic profiling tests, which can be used in the clinic, are approved by the U.S. Food and Drug Administration, including MSK-IMPACT, FoundationOne CDx, and FoundationOne Liquid CDx. Here, we review emerging evidence for genomic profiling of prostate cancer, especially focusing on associations between genomic alteration and clinical outcome, liquid biopsy, and actionable molecular alterations.
Keywords: Biomarkers, Decision making, Genomics, Liquid biopsy, Prostate cancer
INTRODUCTION
Prostate cancer is the second most frequent cancer among males and the cause of an estimated 385,000 deaths worldwide in 2018 [1]. Prostate carcinogenesis and progression are correlated with loss of specific chromosome regions and candidate tumor suppressor genes, such as loss of 8p21 and NKX3.1, loss of 10q and PTEN, loss of 13q and RB1, and loss of 17p and TP53 [2]. Recurrent gene fusions of TMPRSS2 and ETS transcription factor genes are frequently detected in prostate cancer, suggesting that the androgen-responsive promoter elements of TMPRSS2 mediate the overexpression of ETS family members [3]. Prostate cancer development and disease progression are driven by the androgen receptor (AR) signaling pathway, which has led to the use of androgen deprivation therapy (ADT) for patients with advanced prostate cancer. Sustained AR signaling is the primary driver of castration-resistant prostate cancer (CRPC), leading researchers to develop novel treatments targeting the AR pathway, such as abiraterone and enzalutamide [4]. Molecular mechanisms behind AR reactivation in CRPC include AR gene amplification, AR mutations (e.g., T878A, F876L, L702H, L701H, and T877A), AR splice variants (AR-Vs), changes of androgen biosynthesis, and changes in AR cofactor [5]. Recently, novel mechanisms of AR activation have been reported, such as amplification of an upstream enhancer of AR and AR gene rearrangements [6,7,8]. During disease progression, a subset of metastatic CRPC (mCRPC) tumors loses AR dependence and often have neuroendocrine features [9].
Recently, precision medicine has emerged to guide therapeutic approaches for patients with prostate cancer by understanding each altered gene or pathway in an individual, leading to the improvement of clinical outcomes [10]. A phase 3 clinical trial demonstrated that the alteration of BRCA1/2 or ATM was associated with response to poly (adenosine diphosphate–ribose) polymerase (PARP) inhibitor olaparib in patients with mCRPC [11]. An Akt inhibitor, ipatasertib, showed antitumor activity in patients with PTEN-loss tumors, in a phase 2 study [12]. Over the last decade, the integrative genomic profiling of human prostate tumors had provided the foundations for discoveries that can impact disease understanding and treatment [13,14,15]. Furthermore, minimally invasive liquid biopsy procedures have emerged to investigate cancer-related molecules with the advantage of detecting heterogeneity as well as acquired resistance in cancer [16,17]. Here, we review emerging evidence for genomic profiling of prostate cancer, especially focusing on association of genomic alteration and clinical outcome, liquid biopsy, and actionable molecular alterations (Fig. 1). In this review, we identified the relevant studies using electronic databases, including PubMed and Web of Science.
Fig. 1. Overview of genomic profiling of prostate cancer. The specific gene/pathway alterations are associated with clinical outcomes. Genomic profiling is useful to identify actionable molecular alterations. cfDNA: cell free DNA, ctDNA: circulating tumor DNA, CTC: circulating tumor cell, AR: androgen receptor, PARP: poly (adenosine diphosphate–ribose) polymerase, MSI-H: microsatellite instability-high, dMMR: deficiency in mismatch repair genes.
MAIN BODY
1. Genomic landscape of prostate cancer
Common genetic alterations in primary prostate cancer include losses of NKX3.1 and PTEN [2] and fusion of ETS family transcription factor genes with androgen-responsive promoters [3]. In addition, a significant proportion of primary prostate tumors harbor large-scale genomic rearrangements [18,19]. Recurrent somatic mutations were identified in multiple genes, including SPOP and FOXA1, in patients with primary prostate cancer [20]. In 2015, The Cancer Genome Atlas (TCGA) presented a comprehensive molecular analysis of 333 primary prostate cancers, in which the tumors fell into subtypes according to specific gene fusions or mutations (SPOP, FOXA1, and IDH1) [14]. AR activity varied widely in a subtype-specific manner, with SPOP and FOXA1 mutant tumors having the highest levels of AR-induced transcripts [14]. In 2015, Robinson et al [15] demonstrated that aberrations of AR, ETS genes, TP53, and PTEN were detected in 40% to 60% of cases in patients with mCRPC. The mCRPC tumors have a highly complex genomic landscape compared to primary prostate tumors (Fig. 2) [21,22]. Genomic alterations in AR, TP53, RB1, and PTEN are enriched during disease progression [23,24,25]. Approximately 90% of mCRPC harbor clinically actionable molecular alterations, including AR signaling, DNA damage repair and phosphoinositide 3-kinase (PI3K) signaling [15].
Fig. 2. Gene alterations in the different stages of prostate cancer. Localized PCa, TCGA (n=333) [14]; mCSPC, MSK (n=424) [38]; mCRPC, SU2C/PCF Dream Team (n=444) [36]. The frequency of each gene alteration was calculated based on clinical data provided by cBioPortal (https://www.cbioportal.org/) The Figures from the cBioportal are permitted to use in the publications (https://docs.cbioportal.org/1.-general/faq#can-i-use-figures-from-the-cbioportal-in-my-publications-or-presentations) [21,22]. PCa: prostate cancer, TCGA: The Cancer Genome Atlas, mCSPC: metastatic castration-sensitive prostate cancer, MSK: memorial sloan kettering, mCRPC: metastatic castration-resistant prostate cancer, SU2C/PCF: stand up to cancer/prostate cancer foundation.
In 2018, two studies, Quigley et al [6] and Viswanathan et al [7], demonstrated the structural alterations driving mCRPC using whole-genome sequencing. Tandem duplications affect an upstream enhancer of AR in 70% to 87% of cases, correlating with increased AR expression [6,7]. Progression on androgen pathway inhibitors, abiraterone and enzalutamide, was associated with gains in AR and AR enhancer [7]. Tandem duplication hotspots also occur near MYC, associated with post-translational MYC regulation [6]. Classes of structural variations were linked to distinct DNA repair deficiencies, including associations of CDK12 mutation with tandem duplications, TP53 inactivation with inverted rearrangements and chromothripsis, and BRCA2 inactivation with deletions [6,7,26].
The ethnic and racial background can influence the incidence and mortality of prostate cancer, partly due to the interplay of socioeconomic factors and environmental exposures [27]. To date, most prostate cancer genomics data have been derived from Western populations. Thus, precision oncologic studies have under-represented patients from Asia and Africa, limiting comprehensive understanding of disparities in the diagnosis and prognosis of prostate cancer among these populations [28]. The incidence and mortality rates of prostate cancer for Asians are lower than Western populations [29]. In 2020, Li et al [30] reported on the genomic landscape of primary prostate cancer in Asian populations, in which 41% of tumors contained mutations in FOXA1 and 18% had deletions in CHD1. Lower incidence of FOXA1/CHD1 alterations in Western populations and lower incidence of TMPRSS2:ERG fusion gene and PTEN loss in Asian populations compared with counterparts were reported [30,31,32,33]. Thus, the genomic alteration signatures in Asian patients were markedly different from those of Western cohorts.
2. Association of genomic alteration and clinical outcome
Heterogeneity in the genomic landscape of prostate cancer has become apparent through several comprehensive profiling studies. Growing evidence suggests that the genomic alterations correlate with clinical outcomes (Table 1). In 2014, Hieronymus et al [34] reported an association between biochemical recurrence and the pattern of DNA copy number alteration (CNA) in primary prostate cancer, raising the possibility of CNA as a prognostic biomarker. Since 2018, several studies have demonstrated the association of specific gene/pathway alterations and clinical outcomes based on the genome-wide study of prostate cancer [25,35,36,37,38,39]. Wang et al [35] reported that the gene-based pathway of cell cycle progression was associated with shorter time to treatment change (TTTC) in patients with mCRPC who were treated with abiraterone (hazard ratio [HR], 2.11; 95% confidence interval [CI], 1.17–3.80; p=0.01). Abida et al [36] demonstrated that RB1 alteration was associated with poor overall survival (OS), whereas alterations in RB1, AR, and TP53 were associated with shorter TTTC in patients with mCRPC treated with abiraterone or enzalutamide. Chen et al [37] reported that two DNA alterations in RB1 were predictive of poor OS (median 14.1 mo vs. 42.0 mo; p=0.007), and CTNNB1 mutations were exclusive to enzalutamide-resistant patients (p=0.01), associating with poor OS (median 13.6 mo vs. 41.7 mo; p=0.025) in patients with mCRPC treated with enzalutamide. Stopsack et al [38] reported that rates of castration resistance (HR, 1.84; 95% CI, 1.40–2.41) and death (HR, 3.71; 95% CI, 2.28–6.02) were higher in high-volume metastatic castration-sensitive prostate cancer (mCSPC), associating with genomic alterations. Rates of castration resistance differed 1.5-fold to 5-fold according to alterations in AR, cell cycle pathway, MYC pathway, TP53, WNT pathway (inverse), and SPOP (inverse), whereas OS rates differed 2-fold to 4-fold according to AR, cell cycle pathway, WNT pathway (inverse), and SPOP (inverse) [38]. Mateo et al [25] reported that patients with RB1 loss in the primary prostate cancer had a worse prognosis. Among men with matched hormone-naive and mCRPC biopsies, RB1/TP53/AR aberrations were enriched in later stages [25]. Deek et al [39] reported that the frequency of driver mutations in TP53 (p=0.01), WNT (p=0.08), and cell cycle (p=0.04) genes increased across the mCSPC spectrum. Mutations in TP53 were independently associated with shorter radiographic progression free survival (PFS) (HR, 1.59; p=0.03) and the development of CRPC (HR, 1.71; p=0.01) [39]. Hamid et al [40] reported that deleterious tumor suppressor genes, TP53, PTEN, and RB1, were associated with an increased risk of relapse and death in patients with CSPC.
Table 1. Genomic alterations in prostate cancer tissue samples associated with clinical outcome.
Author | Year | Patients | Number of patients | Therapy | Endpoint | Genomic alterations | Outcome |
---|---|---|---|---|---|---|---|
Hieronymus et al [34] | 2014 | Localized PCa | 168 | Px | Risk of BCR | CNA burden | HR, 1.99; 95% CI, 1.11–3.55; p=0.021 |
Wang et al [35] | 2018 | mCRPC | 77 | ABI | TTTC | Cell cycle progression scores (≥50) | HR, 2.11; 95% CI, 1.17–3.80; p=0.01 |
Boysen et al [41] | 2018 | mCRPC | 89 | ABI | TTTC | SPOP | HR, 0.37; 95% CI, 0.20–0.69; p=0.002 |
Abida et al [36] | 2019 | mCRPC | 128 | ABI or ENZ | TTTC | RB1 | CPE=0.818; p<0.001 |
AR | CPE=0.651; p=0.005 | ||||||
TP53 | CPE=0.609; p=0.046 | ||||||
OS | RB1 | CPE=0.768; p=0.002 | |||||
Chen et al [37] | 2019 | mCRPC | 101 | ENZ | OS | RB1 | Median 14.1 mo vs. 42.0 mo; p=0.007 |
CTNNB1 | Median 13.6 mo vs. 41.7 mo; p=0.025 | ||||||
Hamid et al [40] | 2019 | Localized PCa | 205 | Local therapy | PFS | TP53, PTEN, and RB1 | HR, 1.95; 95% CI, 1.22–3.13; p=0.005 |
Time to CRPC | TP53, PTEN, and RB1 | HR, 3.36; 95% CI, 1.01–11.16; p=0.04 | |||||
Stopsack et al [38] | 2020 | mCSPC | 424 | N/A | Time to CRPC | AR | HR, 5.30; 95% CI, 2.97–9.46 |
Cell cycle pathway | HR, 2.12; 95% CI, 1.50–3.00 | ||||||
MYC pathway | HR, 2.04; 95% CI, 1.35–3.10 | ||||||
TP53 | HR, 1.57; 95% CI, 1.17–2.12 | ||||||
WNT pathway | HR, 0.66; 95% CI, 0.47–0.95 | ||||||
SPOP | HR, 0.63; 95% CI, 0.39–1.00 | ||||||
OS | AR | HR, 4.06; 95% CI, 1.71–9.68 | |||||
Cell cycle pathway | HR, 2.03; 95% CI, 1.18–3.50 | ||||||
WNT pathway | HR, 0.45; 95% CI, 0.22–0.90 | ||||||
SPOP | HR, 0.33; 95% CI, 0.13–0.84 | ||||||
Mateo et al [25] | 2020 | Primary PCa | 203 | N/A | OS | RB1 | Median 2.32 y vs. 4.28 y; p=0.006 |
Swami et al [42] | 2020 | mCSPC | 121 | ADT | PFS | SPOP | Median 35 mo vs. 13 mo; HR, 0.47; 95% CI, 0.25–0.87; p=0.016 |
OS | SPOP | Median 97 mo vs. 69 mo; HR, 0.32; 95% CI, 0.12–0.88; p=0.027 | |||||
Deek et al [39] | 2021 | mCSPC | 294 | N/A | rPFS | TP53 | HR, 1.59; 95% CI, 1.04–2.41; p=0.03 |
Time to CRPC | TP53 | HR, 1.71; 95% CI, 1.16–2.52; p=0.01 |
PCa: prostate cance, BCR: biochemical recurrence, CNA: copy number alteration, HR: hazard ratio, CI: confidence interval, mCRPC: metastatic castration-resistant prostate cancer, ABI: abiraterone, TTTC: time to treatment change, ENZ: enzalutamide, CPE: concordance probability estimate, OS: overall survival, mCSPC: metastatic castration-sensitive prostate cancer, PFS: progression free survival, N/A: not applicable, ADT: androgen deprivation therapy, rPFS: radiographic PFS.
Prostate cancer with mutant SPOP shows a distinct pattern of genomic alterations, defining a new molecular subtype [20]. Boysen et al [41] reported that SPOP mutations were associated with a higher response rate to abiraterone (odds ratio, 14.50; 95% CI, 2.92–71.94; p=0.001) and a longer time on abiraterone (HR, 0.37; 95% CI, 0.20–0.69; p=0.002) in patients with mCRPC. Swami et al [42] reported that SPOP mutations were significantly associated with better PFS (median 35 mo vs. 13 mo; HR, 0.47; 95% CI, 0.25–0.87; p=0.016) and OS (97 mo vs. 69 mo; HR, 0.32; 95% CI, 0.12–0.88; p=0.027) in patients with mCSPC treated with ADT. Although AR is a ubiquitination degradation substrate of SPOP E3 ligase, prostate-cancer-associated SPOP mutants cannot bind to and promote AR degradation [43]. The SPOP mutant tumors have the highest AR transcriptional activity among prostate cancer subtypes [14]. Thus, the SPOP mutant tumors may primarily be driven by AR signaling and in turn will be responsive to AR targeted therapies [42].
Taken together, genomic alterations of TP53, RB1, AR, and cell cycle pathway are associated with poor clinical outcomes in patients with prostate cancer, whereas SPOP mutations are associated with better clinical outcomes (Table 1).
3. Liquid biopsy
A liquid biopsy is a minimally invasive procedure to investigate the cancer-related molecules in circulating tumor cells (CTCs) and cell-free tumor nucleic acids. There is a high consistency between metastatic tumor tissue and matched circulating tumor DNA (ctDNA) or CTCs [44,45,46,47]. Liquid biopsies have the advantage of detecting acquired resistance in prostate cancer [17,48]. In 2016, Ulz et al [16] performed whole-genome sequencing on plasma samples derived from patients with metastatic prostate cancer, and identified driver aberrations in cancer-related genes, including gene fusions (TMPRSS2:ERG), focal deletions (PTEN, RYBP, and SHQ1), and amplifications (AR and MYC). In serial plasma analyses, the focal amplifications were detected in 40% of cases, suggesting a high plasticity of prostate cancer genomes with newly occurring focal amplifications as a driving force in progression [16]. Although ADT rapidly reduces ctDNA availability [49], the emergence of AR amplification in ctDNA is detected during treatment with abiraterone and enzalutamide [50]. Tumor fraction in cell free DNA (cfDNA) correlates with metastatic burden, and the decline of ctDNA can be a promising biomarker for therapeutic response in patients with CRPC [51]. Decreases in cfDNA concentration independently associated with outcome in patients with metastatic prostate cancer who were treated with PARP inhibitor olaparib (HR for OS at week 8, 0.19; 95% CI, 0.06–0.56; p=0.003) [52].
Recently, a number of studies demonstrated the association between genomic alterations in liquid biopsy and clinical outcome in prostate cancer (Table 2). As sustained AR signaling pathway remains a key driver for CRPC progression [5], considerable efforts have been made to profile AR aberrations using circulating nucleic acids [53]. Resistance to AR pathway inhibitors, abiraterone and enzalutamide, has been observed in patients with CRPC harboring AR copy number gain/amplification [54,55,56,57,58,59], somatic AR mutations [54,55,56], and constitutively active AR-Vs, such as AR-V3, AR-V7, and AR-V9 [58,60]. AR copy number gain has also been associated with poor outcomes in patients receiving chemotherapy [58,61], likely reflecting aggressive intrinsic disease biology. Furthermore, genomic alterations of RB1, TP53, MYC, cell cycle pathway, and DNA repair pathway are detected in liquid biopsy, and are reported to be associated with poor clinical outcomes in patients with prostate cancer [55,62,63,64,65,66].
Table 2. Genomic alterations in liquid biopsy associated with clinical outcome.
Author | Year | Sample | Patients | Number of patients | Therapy | Endpoint | Genomic alterations | Outcome |
---|---|---|---|---|---|---|---|---|
Azad et al [54] | 2015 | Plasma cfDNA | mCRPC | 39 | ENZ | c/rPFS | AR gain/mut | Median 2.3 mo vs.7.0 mo; p<0.001 |
Wyatt et al [55] | 2016 | Plasma cfDNA | mCRPC | 65 | ENZ | PFS | AR gain/amp | HR, 2.92; 95% CI, 1.59–5.37; p=0.001 |
Multiple AR mut | HR, 3.94; 95% CI, 1.46–10.64; p=0.007 | |||||||
RB1 loss | HR, 4.46; 95% CI, 2.28–8.74; p<0.001 | |||||||
MET gain | HR, 4.53; 95% CI, 1.97–10.45; p<0.001 | |||||||
MYC gain | HR, 2.58; 95% CI, 1.39–4.77; p=0.003 | |||||||
Conteduca et al [56] | 2017 | Plasma cfDNA and CTC | CRPC | 171 | ABI or ENZ | PFS | AR gain | HR, 2.22; 95% CI, 1.48–3.34; p<0.001 |
AR mut | HR, 2.59; 95% CI, 1.24–5.44; p=0.012 | |||||||
OS | AR gain | HR, 4.26; 95% CI, 2.76–6.55; p<0.001 | ||||||
AR mut | HR, 3.80; 95% CI, 1.77–8.15; p=0.001 | |||||||
94 | ENZ | rPFS | AR gain | HR, 8.06; 95% CI, 3.26–19.93; p<0.001 | ||||
OS | AR gain | HR, 11.08; 95% CI, 2.16–56.95; p=0.004 | ||||||
De Laere et al [60] | 2017 | Plasma cfDNA and CTC | CRPC | 17 | ABI or ENZ | PFS | ARVs | HR, 4.53; 95% CI, 1.424–14.41; p=0.0105 |
Kohli et al [57] | 2018 | Plasma cfDNA | mCRPC | 70 | ABI | OS | AR amp | HR, 5.25; 95% CI, 2.21–12.46; p=0.0002 |
Annala et al [62] | 2018 | Plasma cfDNA | mCRPC | 202 | ABI or ENZ | PFS | BRCA2/ATM | HR, 6.14; 95% CI, 3.35–11.26; p<0.001 |
TP53 | HR, 2.70; 95% CI, 1.86–3.91; p<0.001 | |||||||
Conteduca et al [61] | 2019 | Plasma cfDNA | mCRPC | 163 | DTX | OS | AR gain | HR, 1.61; 95% CI, 1.08–2.39; p=0.018 |
De Laere et al [63] | 2019 | Plasma cfDNA and CTC | mCRPC | 168 | ABI or ENZ | PFS | TP53 | HR, 1.88; 95% CI, 1.18–3.00; p=0.008 |
Sonpavde et al [64] | 2019 | Plasma cfDNA | mCRPC | 163 | N/A | OS | MYC amp | HR, 5.85; 95% CI, 2.17–15.77; p<0.001 |
Fettke et al [58] | 2020 | Plasma cfDNA/cfRNA | mCRPC | 67 | ABI, ENZ, DTX, CBT | c/rPFS | AR gain | HR, 3.2; 95% CI, 1.3–8.0; p=0.01 |
OS | AR gain | HR, 2.8; 95% CI, 1.1–7.2; p=0.04 | ||||||
Du et al [59] | 2020 | Plasma cfDNA | mCRPC | 88 | ABI | TTTC | AR amp | HR, 3.27; 95% CI, 1.78–6.84; p=0.0003 |
OPHN1 amp | HR, 3.70; 95% CI, 1.08–7.00; p=0.0002 | |||||||
Ritch et al [65] | 2020 | Plasma cfDNA | mCSPC | 210 | ADT | Time to CRPC | dMMR | Median 9.1 mo vs.18.2 mo; p=0.00025 |
Kohli et al [66] | 2020 | Plasma cfDNA | mCRPC | 69 | N/A | OS | RB1 | HR, 4.2; 95% CI, 2.0–8.7; p=0.00015 |
mCSPC | 73 | N/A | OS | ATM, BRCA1, BRCA2, and CHEK2 | HR, 4.0; 95% CI, 1.4–11.8; p=0.0000475 |
cfDNA: cell free DNA, mCRPC: metastatic castration-resistant prostate cancer, ENZ: enzalutamide, c/rPFS: clinical/radiographic progression free survival, HR: hazard ratio, CI: confidence interval, CTC: circulating tumor cell, CRPC: castration-resistant prostate cancer, ABI: abiraterone, OS: overall survival, ARVs: androgen receptor splice variants, DTX: docetaxel, N/A: not applicable, cfRNA: cell free RNA, CBT: cabazitaxel, ADT: androgen deprivation therapy, dMMR: deficiency in mismatch repair genes.
4. Actionable molecular alterations
DNA repair alterations are observed in about one fourth of prostate cancer, in which most commonly mutated genes include BRCA2, BRCA1, and ATM [23]. These gene alterations can occur at either a somatic or a germline level [23]. Although the mutations in DNA-repair genes occurred more often in Black men than in White men [28], the germline alterations in DNA-repair genes were identified in 31% of the patients in Asian populations, including mutations in BRCA2 (5.3%) [67]. The germline mutations in BRCA1/2 and ATM are associated with prostate cancer risk [68], as well as aggressive prostate cancer phenotype [69,70,71,72,73,74]. Family history of cancer remains a foundation of genetic risk assessment, especially inquiring about prostate cancer as well as non-prostate cancers, including breast, ovary, pancreas, and melanoma, with their known association with mutations in BRCA1/2. [75]. BRCA1/2 and ATM are involved in homologous recombination repair. Tumors that lose the homologous recombination pathway are preferentially sensitive to PARP inhibition via the mechanism of synthetic lethality [76]. A randomized, phase 3 trial evaluated the PARP inhibitor olaparib in men with mCRPC who had disease progression while receiving a new hormonal agent (e.g., enzalutamide or abiraterone) [11]. Among patients who had at least one alteration in BRCA1, BRCA2, or ATM, radiological PFS was significantly longer in the olaparib group than in the control group (median 7.4 mo vs. 3.6 mo; HR, 0.34; 95% CI, 0.25–0.47; p<0.001) [11].
The solid tumors which harbor deficiency in mismatch repair genes (dMMR), such as MSH2, MSH6, PMS2, and MLH1, can be effectively treated by the anti–programmed cell death protein 1 (PD-1) antibody pembrolizumab, regardless of tissue of origin [77]. In 2019, Abida et al [78] reported that 32 of 1,033 patients with prostate cancer (3.1%) had microsatellite instability (MSI)–high or dMMR, of whom 7 (21.9%) carried a germline mutation in a Lynch syndrome–associated gene. The dMMR prostate cancers are associated with higher MSI scores, and enriched for higher T cell infiltration and PDL1 protein expression [79]. Screening for MSI-H/dMMR in advanced prostate cancer is beneficial for identifying patients who have potential for durable responses to anti–PD-1/PD-L1 therapy.
Approximately 40% to 60% of mCRPC tumors have a functional loss of PTEN, a tumor suppressor phosphatase, which causes hyperactivation of the PI3K–Akt–mTOR pathway [13,15]. Ipatasertib (GDC-0068) is a novel selective ATP-competitive small-molecule inhibitor of all three isoforms of Akt. Sensitivity to ipatasertib is associated with high tumoral levels of phosphorylated Akt, PTEN protein loss or genetic mutations, and PIK-3CA kinase domain mutations [80]. In a phase 2 study, combined treatment with abiraterone and ipatasertib showed superior antitumor activity to abiraterone alone in patients with mCRPC, especially in patients with PTEN-loss tumors [12]. A phase 3 trial is ongoing to test the efficiency of ipatasertib plus abiraterone in patients with mCRPC (IPATential150, NCT03072238).
5. Neuroendocrine prostate cancer
Neuroendocrine prostate cancer is an aggressive variant of prostate cancer, which may arise de novo or in patients who were previously treated with hormonal therapies [81]. A subset of mCRPC tumors show small-cell neuroendocrine features during disease progression on metastatic biopsy [82]. This phenomenon may reflect an epithelial plasticity that enables tumor adaptation in response to AR-targeted therapies [9]. Neuroendocrine prostate cancer is associated with worse OS, even when platinum-based chemotherapy is used [81,83]. In 2016, Beltran et al [9] demonstrated that CRPC with neuroendocrine features (CRPC-NE) is associated with low AR signaling and a paucity of somatic AR gene alterations, concurrent loss of RB1 and TP53 (in 53.3% of CRPC-NE vs 13.7% of CRPC-Adenocarcinoma; p<0.0004), changes in DNA methylation profile, and upregulation of mRNA encoding the histone methyltransferase EZH2. There was high concordance between ctDNA and biopsy tissue genomic alterations in patients with CRPC-NE, supporting the use of ctDNA profile to recognize transformation to CRPC-NE during the course of CRPC treatment [84].
6. Clinical utility of genomic profiling
Tumor genomic profiling is a fundamental component of precision medicine, enabling the identification of genomic alterations in genes and pathways that can be targeted therapeutically. In 2017, the U.S. Food and Drug Administration (FDA) approved two comprehensive next generation sequencing panel assays, MSK-IMPACT and FoundationOne CDx [85]. At Memorial Sloan Kettering Cancer Center, MSK-IMPACT was developed and implemented to detect protein-coding mutations, CNAs, and selected promoter mutations and structural rearrangements in 341 (and, more recently, 468) cancer-associated genes [85,86]. FoundationOne CDx, a similar 324 gene assay, was developed to identify actionable genomic aberrations in cancer [85]. For the effective analysis of genomic tests, the quality of tumor tissue samples is crucial. Although formalin-fixed paraffin-embedded blocks obtained from prostate tumor biopsies are widely used to identify clinically actionable molecular alterations, DNA degradation can occur during mid- to long-term storage of samples [87]. Genomic heterogeneity is commonly detected in primary prostate cancer [88,89,90]. Furthermore, genomic alterations can occur during CRPC progression [16,91]. Thus, a metastatic biopsy provides a reasonable assessment for genomic profiling in patients with mCRPC [92]. In 2020, FoundationOne Liquid CDx, a novel 324-Gene cfDNA-based comprehensive genomic profiling assay, was approved by the FDA [93]. This laboratory test can be used as a companion diagnostic tool that can identify if patients with mCRPC harbor BRCA1/2 alterations which may benefit from treatment with PARP inhibitors [93]. After eliminating clonal hematopoiesis variants, ctDNA was detected in 87.9% of patients with prostate cancer showing its high detectability [94]. Thus, cfDNA-based genomic tests provide a noninvasive approach to elucidate a patient's genomic landscape and actionable information.
CONCLUSIONS
The integrative genomic profiling of prostate tumors has provided comprehensive information and novel discoveries which improve our understanding of the disease. A number of mCRPC harbor clinically actionable molecular alterations, including changes to DNA damage repair pathway and PTEN/PI3K signaling. The genomic alterations of TP53, RB1, AR, and cell cycle pathway are associated with poor clinical outcomes, whereas SPOP mutation is associated with better clinical outcomes. Several genomic profiling tests are emerging to identify patients who could benefit from targeted therapy. Thus, the genomic profiling of prostate cancer provides useful information for diagnosis and treatment in this new era of precision medicine.
ACKNOWLEDGEMENTS
This research was supported by the Japan Society for the Promotion of Science KAKENHI (grant numbers: 20K18090 and T19K096890).
Footnotes
Conflict of Interest: The authors have nothing to disclose.
- Conceptualization: KH, NN.
- Data curation: KH.
- Formal analysis: KH.
- Funding acquisition: KH, NN.
- Investigation: KH.
- Methodology: KH.
- Project administration: KH, NN.
- Resources: KH.
- Software: KH.
- Supervision: NN.
- Validation: KH, NN.
- Visualization: KH.
- Writing — original draft: KH.
- Writing — review & editing: KH, NN.
References
- 1.Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424. doi: 10.3322/caac.21492. [DOI] [PubMed] [Google Scholar]
- 2.Abate-Shen C, Shen MM. Molecular genetics of prostate cancer. Genes Dev. 2000;14:2410–2434. doi: 10.1101/gad.819500. [DOI] [PubMed] [Google Scholar]
- 3.Tomlins SA, Rhodes DR, Perner S, Dhanasekaran SM, Mehra R, Sun XW, et al. Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science. 2005;310:644–648. doi: 10.1126/science.1117679. [DOI] [PubMed] [Google Scholar]
- 4.Watson PA, Arora VK, Sawyers CL. Emerging mechanisms of resistance to androgen receptor inhibitors in prostate cancer. Nat Rev Cancer. 2015;15:701–711. doi: 10.1038/nrc4016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Fujita K, Nonomura N. Role of androgen receptor in prostate cancer: a review. World J Mens Health. 2019;37:288–295. doi: 10.5534/wjmh.180040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.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;174:758–769.e9. doi: 10.1016/j.cell.2018.06.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Viswanathan SR, Ha G, Hoff AM, Wala JA, Carrot-Zhang J, Whelan CW, et al. Structural alterations driving castration-resistant prostate cancer revealed by linked-read genome sequencing. Cell. 2018;174:433–447.e19. doi: 10.1016/j.cell.2018.05.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Li Y, Yang R, Henzler CM, Ho Y, Passow C, Auch B, et al. Diverse AR gene rearrangements mediate resistance to androgen receptor inhibitors in metastatic prostate cancer. Clin Cancer Res. 2020;26:1965–1976. doi: 10.1158/1078-0432.CCR-19-3023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.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;22:298–305. doi: 10.1038/nm.4045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Ku SY, Gleave ME, Beltran H. Towards precision oncology in advanced prostate cancer. Nat Rev Urol. 2019;16:645–654. doi: 10.1038/s41585-019-0237-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.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;382:2091–2102. doi: 10.1056/NEJMoa1911440. [DOI] [PubMed] [Google Scholar]
- 12.de Bono JS, De Giorgi U, Rodrigues DN, Massard C, Bracarda S, Font A, et al. Randomized phase II study evaluating Akt blockade with ipatasertib, in combination with abiraterone, in patients with metastatic prostate cancer with and without PTEN loss. Clin Cancer Res. 2019;25:928–936. doi: 10.1158/1078-0432.CCR-18-0981. [DOI] [PubMed] [Google Scholar]
- 13.Taylor BS, Schultz N, Hieronymus H, Gopalan A, Xiao Y, Carver BS, et al. Integrative genomic profiling of human prostate cancer. Cancer Cell. 2010;18:11–22. doi: 10.1016/j.ccr.2010.05.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Cancer Genome Atlas Research Network. The molecular taxonomy of primary prostate cancer. Cell. 2015;163:1011–1025. doi: 10.1016/j.cell.2015.10.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Robinson D, Van Allen EM, Wu YM, Schultz N, Lonigro RJ, Mosquera JM, et al. Integrative clinical genomics of advanced prostate cancer. Cell. 2015;161:1215–1228. doi: 10.1016/j.cell.2015.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.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 Commun. 2016;7:12008. doi: 10.1038/ncomms12008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Mayrhofer M, De Laere B, Whitington T, Van Oyen P, Ghysel C, Ampe J, et al. Cell-free DNA profiling of metastatic prostate cancer reveals microsatellite instability, structural rearrangements and clonal hematopoiesis. Genome Med. 2018;10:85. doi: 10.1186/s13073-018-0595-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Baca SC, Prandi D, Lawrence MS, Mosquera JM, Romanel A, Drier Y, et al. Punctuated evolution of prostate cancer genomes. Cell. 2013;153:666–677. doi: 10.1016/j.cell.2013.03.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Fraser M, Sabelnykova VY, Yamaguchi TN, Heisler LE, Livingstone J, Huang V, et al. Genomic hallmarks of localized, non-indolent prostate cancer. Nature. 2017;541:359–364. doi: 10.1038/nature20788. [DOI] [PubMed] [Google Scholar]
- 20.Barbieri CE, Baca SC, Lawrence MS, Demichelis F, Blattner M, Theurillat JP, et al. Exome sequencing identifies recurrent SPOP, FOXA1 and MED12 mutations in prostate cancer. Nat Genet. 2012;44:685–689. doi: 10.1038/ng.2279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2:401–404. doi: 10.1158/2159-8290.CD-12-0095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 2013;6:pl1. doi: 10.1126/scisignal.2004088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Abida W, Armenia J, Gopalan A, Brennan R, Walsh M, Barron D, et al. Prospective genomic profiling of prostate cancer across disease states reveals germline and somatic alterations that may affect clinical decision making. JCO Precis Oncol. 2017;2017:PO.17.00029. doi: 10.1200/PO.17.00029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Armenia J, Wankowicz SAM, Liu D, Gao J, Kundra R, Reznik E, et al. The long tail of oncogenic drivers in prostate cancer. Nat Genet. 2018;50:645–651. doi: 10.1038/s41588-018-0078-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Mateo J, Seed G, Bertan C, Rescigno P, Dolling D, Figueiredo I, et al. Genomics of lethal prostate cancer at diagnosis and castration resistance. J Clin Invest. 2020;130:1743–1751. doi: 10.1172/JCI132031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.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;10:5251. doi: 10.1038/s41467-019-13084-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Dess RT, Hartman HE, Mahal BA, Soni PD, Jackson WC, Cooperberg MR, et al. Association of black race with prostate cancer-specific and other-cause mortality. JAMA Oncol. 2019;5:975–983. doi: 10.1001/jamaoncol.2019.0826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Mahal BA, Alshalalfa M, Kensler KH, Chowdhury-Paulino I, Kantoff P, Mucci LA, et al. Racial differences in genomic profiling of prostate cancer. N Engl J Med. 2020;383:1083–1085. doi: 10.1056/NEJMc2000069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Kimura T. East meets West: ethnic differences in prostate cancer epidemiology between East Asians and Caucasians. Chin J Cancer. 2012;31:421–429. doi: 10.5732/cjc.011.10324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Li J, Xu C, Lee HJ, Ren S, Zi X, Zhang Z, et al. A genomic and epigenomic atlas of prostate cancer in Asian populations. Nature. 2020;580:93–99. doi: 10.1038/s41586-020-2135-x. [DOI] [PubMed] [Google Scholar]
- 31.Orikasa K, Fukushige S, Hoshi S, Orikasa S, Kondo K, Miyoshi Y, et al. Infrequent genetic alterations of the PTEN gene in Japanese patients with sporadic prostate cancer. J Hum Genet. 1998;43:228–230. doi: 10.1007/s100380050078. [DOI] [PubMed] [Google Scholar]
- 32.Mao X, Yu Y, Boyd LK, Ren G, Lin D, Chaplin T, et al. Distinct genomic alterations in prostate cancers in Chinese and Western populations suggest alternative pathways of prostate carcinogenesis. Cancer Res. 2010;70:5207–5212. doi: 10.1158/0008-5472.CAN-09-4074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Miyagi Y, Sasaki T, Fujinami K, Sano J, Senga Y, Miura T, et al. ETS family-associated gene fusions in Japanese prostate cancer: analysis of 194 radical prostatectomy samples. Mod Pathol. 2010;23:1492–1498. doi: 10.1038/modpathol.2010.149. [DOI] [PubMed] [Google Scholar]
- 34.Hieronymus H, Schultz N, Gopalan A, Carver BS, Chang MT, Xiao Y, et al. Copy number alteration burden predicts prostate cancer relapse. Proc Natl Acad Sci U S A. 2014;111:11139–11144. doi: 10.1073/pnas.1411446111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Wang L, Dehm SM, Hillman DW, Sicotte H, Tan W, Gormley M, et al. A prospective genome-wide study of prostate cancer metastases reveals association of wnt pathway activation and increased cell cycle proliferation with primary resistance to abiraterone acetate-prednisone. Ann Oncol. 2018;29:352–360. doi: 10.1093/annonc/mdx689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Abida W, Cyrta J, Heller G, Prandi D, Armenia J, Coleman I, et al. Genomic correlates of clinical outcome in advanced prostate cancer. Proc Natl Acad Sci U S A. 2019;116:11428–11436. doi: 10.1073/pnas.1902651116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Chen WS, Aggarwal R, Zhang L, Zhao SG, Thomas GV, Beer TM, et al. West Coast Prostate Cancer Dream Team. Genomic drivers of poor prognosis and enzalutamide resistance in metastatic castration-resistant prostate cancer. Eur Urol. 2019;76:562–571. doi: 10.1016/j.eururo.2019.03.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Stopsack KH, Nandakumar S, Wibmer AG, Haywood S, Weg ES, Barnett ES, et al. Oncogenic genomic alterations, clinical phenotypes, and outcomes in metastatic castration-sensitive prostate cancer. Clin Cancer Res. 2020;26:3230–3238. doi: 10.1158/1078-0432.CCR-20-0168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Deek MP, Van der Eecken K, Phillips R, Parikh NR, Isaacsson Velho P, Lotan TL, et al. The mutational landscape of metastatic castration-sensitive prostate cancer: the spectrum theory revisited. Eur Urol. 2021 doi: 10.1016/j.eururo.2020.12.040. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.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;76:89–97. doi: 10.1016/j.eururo.2018.11.045. [DOI] [PubMed] [Google Scholar]
- 41.Boysen G, Rodrigues DN, Rescigno P, Seed G, Dolling D, Riisnaes R, et al. SPOP-mutated/CHD1-deleted lethal prostate cancer and abiraterone sensitivity. Clin Cancer Res. 2018;24:5585–5593. doi: 10.1158/1078-0432.CCR-18-0937. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Swami U, Isaacsson Velho P, Nussenzveig R, Chipman J, Sacristan Santos V, Erickson S, et al. Association of SPOP mutations with outcomes in men with de novo metastatic castration-sensitive prostate cancer. Eur Urol. 2020;78:652–656. doi: 10.1016/j.eururo.2020.06.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.An J, Wang C, Deng Y, Yu L, Huang H. Destruction of full-length androgen receptor by wild-type SPOP, but not prostate-cancer-associated mutants. Cell Rep. 2014;6:657–669. doi: 10.1016/j.celrep.2014.01.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.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:djx118. doi: 10.1093/jnci/djx118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.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;21:162. doi: 10.1186/s13059-020-02045-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Faugeroux V, Lefebvre C, Pailler E, Pierron V, Marcaillou C, Tourlet S, et al. An accessible and unique insight into metastasis mutational content through whole-exome sequencing of circulating tumor cells in metastatic prostate cancer. Eur Urol Oncol. 2020;3:498–508. doi: 10.1016/j.euo.2018.12.005. [DOI] [PubMed] [Google Scholar]
- 47.Fan L, Fei X, Zhu Y, Pan J, Sha J, Chi C, et al. Comparative analysis of genomic alterations across castration sensitive and castration resistant prostate cancer via circulating tumor DNA sequencing. J Urol. 2021;205:461–469. doi: 10.1097/JU.0000000000001363. [DOI] [PubMed] [Google Scholar]
- 48.Heitzer E, Ulz P, Belic J, Gutschi S, Quehenberger F, Fischereder K, et al. Tumor-associated copy number changes in the circulation of patients with prostate cancer identified through whole-genome sequencing. Genome Med. 2013;5:30. doi: 10.1186/gm434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.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;75:667–675. doi: 10.1016/j.eururo.2018.12.042. [DOI] [PubMed] [Google Scholar]
- 50.Belic J, Graf R, Bauernhofer T, Cherkas Y, Ulz P, Waldispuehl-Geigl J, et al. Genomic alterations in plasma DNA from patients with metastasized prostate cancer receiving abiraterone or enzalutamide. Int J Cancer. 2018;143:1236–1248. doi: 10.1002/ijc.31397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Choudhury AD, Werner L, Francini E, Wei XX, Ha G, Freeman SS, et al. Tumor fraction in cell-free DNA as a biomarker in prostate cancer. JCI Insight. 2018;3:e122109. doi: 10.1172/jci.insight.122109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Goodall J, Mateo J, Yuan W, Mossop H, Porta N, Miranda S, et al. TOPARP-A investigators. TOPARP-A investigators. Circulating cell-free DNA to guide prostate cancer treatment with PARP inhibition. Cancer Discov. 2017;7:1006–1017. doi: 10.1158/2159-8290.CD-17-0261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Sumiyoshi T, Mizuno K, Yamasaki T, Miyazaki Y, Makino Y, Okasho K, et al. Clinical utility of androgen receptor gene aberrations in circulating cell-free DNA as a biomarker for treatment of castration-resistant prostate cancer. Sci Rep. 2019;9:4030. doi: 10.1038/s41598-019-40719-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.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;21:2315–2324. doi: 10.1158/1078-0432.CCR-14-2666. [DOI] [PubMed] [Google Scholar]
- 55.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;2:1598–1606. doi: 10.1001/jamaoncol.2016.0494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Conteduca V, Wetterskog D, Sharabiani MTA, Grande E, Fernandez-Perez MP, Jayaram A, et al. Androgen receptor gene status in plasma DNA associates with worse outcome on enzalutamide or abiraterone for castration-resistant prostate cancer: a multi-institution correlative biomarker study. Ann Oncol. 2017;28:1508–1516. doi: 10.1093/annonc/mdx155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Kohli M, Li J, Du M, Hillman DW, Dehm SM, Tan W, et al. Prognostic association of plasma cell-free DNA-based androgen receptor amplification and circulating tumor cells in prechemotherapy metastatic castration-resistant prostate cancer patients. Prostate Cancer Prostatic Dis. 2018;21:411–418. doi: 10.1038/s41391-018-0043-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.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;78:173–180. doi: 10.1016/j.eururo.2020.03.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Du M, Tian Y, Tan W, Wang L, Wang L, Kilari D, et al. Plasma cell-free DNA-based predictors of response to abiraterone acetate/prednisone and prognostic factors in metastatic castration-resistant prostate cancer. Prostate Cancer Prostatic Dis. 2020;23:705–713. doi: 10.1038/s41391-020-0224-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.De Laere B, van Dam PJ, Whitington T, Mayrhofer M, Diaz EH, Van den Eynden G, et al. Comprehensive profiling of the androgen receptor in liquid biopsies from castration-resistant prostate cancer reveals novel intra-AR structural variation and splice variant expression patterns. Eur Urol. 2017;72:192–200. doi: 10.1016/j.eururo.2017.01.011. [DOI] [PubMed] [Google Scholar]
- 61.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;75:368–373. doi: 10.1016/j.eururo.2018.09.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.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;8:444–457. doi: 10.1158/2159-8290.CD-17-0937. [DOI] [PubMed] [Google Scholar]
- 63.De Laere B, Oeyen S, Mayrhofer M, Whitington T, van Dam PJ, Van Oyen P, et al. TP53 outperforms other androgen receptor biomarkers to predict abiraterone or enzalutamide outcome in metastatic castration-resistant prostate cancer. Clin Cancer Res. 2019;25:1766–1773. doi: 10.1158/1078-0432.CCR-18-1943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Sonpavde G, Agarwal N, Pond GR, Nagy RJ, Nussenzveig RH, Hahn AW, et al. Circulating tumor DNA alterations in patients with metastatic castration-resistant prostate cancer. Cancer. 2019;125:1459–1469. doi: 10.1002/cncr.31959. [DOI] [PubMed] [Google Scholar]
- 65.Ritch E, Fu SYF, Herberts C, Wang G, Warner EW, Schönlau E, et al. Identification of hypermutation and defective mismatch repair in ctDNA from metastatic prostate cancer. Clin Cancer Res. 2020;26:1114–1125. doi: 10.1158/1078-0432.CCR-19-1623. [DOI] [PubMed] [Google Scholar]
- 66.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Wu J, Wei Y, Pan J, Jin S, Gu W, Gan H, et al. Prevalence of comprehensive DNA damage repair gene germline mutations in Chinese prostate cancer patients. Int J Cancer. 2021;148:673–681. doi: 10.1002/ijc.33324. [DOI] [PubMed] [Google Scholar]
- 68.Momozawa Y, Iwasaki Y, Hirata M, Liu X, Kamatani Y, Takahashi A, et al. Germline pathogenic variants in 7636 Japanese patients with prostate cancer and 12 366 controls. J Natl Cancer Inst. 2020;112:369–376. doi: 10.1093/jnci/djz124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Castro E, Goh C, Olmos D, Saunders E, Leongamornlert D, Tymrakiewicz M, et al. Germline BRCA mutations are associated with higher risk of nodal involvement, distant metastasis, and poor survival outcomes in prostate cancer. J Clin Oncol. 2013;31:1748–1757. doi: 10.1200/JCO.2012.43.1882. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Decker B, Karyadi DM, Davis BW, Karlins E, Tillmans LS, Stanford JL, et al. Biallelic BRCA2 mutations shape the somatic mutational landscape of aggressive prostate tumors. Am J Hum Genet. 2016;98:818–829. doi: 10.1016/j.ajhg.2016.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Na R, Zheng SL, Han M, Yu H, Jiang D, Shah S, et al. Germline mutations in ATM and BRCA1/2 distinguish risk for lethal and indolent prostate cancer and are associated with early age at death. Eur Urol. 2017;71:740–747. doi: 10.1016/j.eururo.2016.11.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Mijuskovic M, Saunders EJ, Leongamornlert DA, Wakerell S, Whitmore I, Dadaev T, et al. Rare germline variants in DNA repair genes and the angiogenesis pathway predispose prostate cancer patients to develop metastatic disease. Br J Cancer. 2018;119:96–104. doi: 10.1038/s41416-018-0141-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Castro E, Romero-Laorden N, Del Pozo A, Lozano R, Medina A, Puente J, et al. PROREPAIR-B: a prospective cohort study of the impact of germline DNA repair mutations on the outcomes of patients with metastatic castration-resistant prostate cancer. J Clin Oncol. 2019;37:490–503. doi: 10.1200/JCO.18.00358. [DOI] [PubMed] [Google Scholar]
- 74.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:e1042–e1050. doi: 10.1634/theoncologist.2019-0495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Cheng HH, Sokolova AO, Schaeffer EM, Small EJ, Higano CS. Germline and somatic mutations in prostate cancer for the clinician. J Natl Compr Canc Netw. 2019;17:515–521. doi: 10.6004/jnccn.2019.7307. [DOI] [PubMed] [Google Scholar]
- 76.Lord CJ, Ashworth A. PARP inhibitors: synthetic lethality in the clinic. Science. 2017;355:1152–1158. doi: 10.1126/science.aam7344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357:409–413. doi: 10.1126/science.aan6733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.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;5:471–478. doi: 10.1001/jamaoncol.2018.5801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Nava Rodrigues D, Rescigno P, Liu D, Yuan W, Carreira S, Lambros MB, et al. Immunogenomic analyses associate immunological alterations with mismatch repair defects in prostate cancer. J Clin Invest. 2018;128:4441–4453. doi: 10.1172/JCI121924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Lin J, Sampath D, Nannini MA, Lee BB, Degtyarev M, Oeh J, et al. Targeting activated Akt with GDC-0068, a novel selective Akt inhibitor that is efficacious in multiple tumor models. Clin Cancer Res. 2013;19:1760–1772. doi: 10.1158/1078-0432.CCR-12-3072. [DOI] [PubMed] [Google Scholar]
- 81.Conteduca V, Oromendia C, Eng KW, Bareja R, Sigouros M, Molina A, et al. Clinical features of neuroendocrine prostate cancer. Eur J Cancer. 2019;121:7–18. doi: 10.1016/j.ejca.2019.08.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Aggarwal R, Huang J, Alumkal JJ, Zhang L, Feng FY, Thomas GV, et al. Clinical and genomic characterization of treatment-emergent small-cell neuroendocrine prostate cancer: a multi-institutional prospective study. J Clin Oncol. 2018;36:2492–2503. doi: 10.1200/JCO.2017.77.6880. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Wang HT, Yao YH, Li BG, Tang Y, Chang JW, Zhang J. Neuroendocrine Prostate Cancer (NEPC) progressing from conventional prostatic adenocarcinoma: factors associated with time to development of NEPC and survival from NEPC diagnosis-a systematic review and pooled analysis. J Clin Oncol. 2014;32:3383–3390. doi: 10.1200/JCO.2013.54.3553. [DOI] [PubMed] [Google Scholar]
- 84.Beltran H, Romanel A, Conteduca V, Casiraghi N, Sigouros M, Franceschini GM, et al. Circulating tumor DNA profile recognizes transformation to castration-resistant neuroendocrine prostate cancer. J Clin Invest. 2020;130:1653–1668. doi: 10.1172/JCI131041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Allegretti M, Fabi A, Buglioni S, Martayan A, Conti L, Pescarmona E, et al. Tearing down the walls: FDA approves next generation sequencing (NGS) assays for actionable cancer genomic aberrations. J Exp Clin Cancer Res. 2018;37:47. doi: 10.1186/s13046-018-0702-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Zehir A, Benayed R, Shah RH, Syed A, Middha S, Kim HR, et al. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat Med. 2017;23:703–713. doi: 10.1038/nm.4333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Guyard A, Boyez A, Pujals A, Robe C, Tran Van Nhieu J, Allory Y, et al. DNA degrades during storage in formalin-fixed and paraffin-embedded tissue blocks. Virchows Arch. 2017;471:491–500. doi: 10.1007/s00428-017-2213-0. [DOI] [PubMed] [Google Scholar]
- 88.Boutros PC, Fraser M, Harding NJ, de Borja R, Trudel D, Lalonde E, et al. Spatial genomic heterogeneity within localized, multifocal prostate cancer. Nat Genet. 2015;47:736–745. doi: 10.1038/ng.3315. [DOI] [PubMed] [Google Scholar]
- 89.Espiritu SMG, Liu LY, Rubanova Y, Bhandari V, Holgersen EM, Szyca LM, et al. The evolutionary landscape of localized prostate cancers drives clinical aggression. Cell. 2018;173:1003–1013.e15. doi: 10.1016/j.cell.2018.03.029. [DOI] [PubMed] [Google Scholar]
- 90.Løvf M, Zhao S, Axcrona U, Johannessen B, Bakken AC, Carm KT, et al. Multifocal primary prostate cancer exhibits high degree of genomic heterogeneity. Eur Urol. 2019;75:498–505. doi: 10.1016/j.eururo.2018.08.009. [DOI] [PubMed] [Google Scholar]
- 91.Carreira S, Romanel A, Goodall J, Grist E, Ferraldeschi R, Miranda S, et al. Tumor clone dynamics in lethal prostate cancer. Sci Transl Med. 2014;6:254ra125. doi: 10.1126/scitranslmed.3009448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.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;22:369–378. doi: 10.1038/nm.4053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Foundation Medicine. FoundationOne® Liquid CDx: technical information [Internet] Cambridge (MA): Foundation Medicine; c2020. [cited 2021 Apr 30]. Available from: https://www.accessdata.fda.gov/cdrh_docs/pdf19/P190032C.pdf. [Google Scholar]
- 94.Zhang Y, Yao Y, Xu Y, Li L, Gong Y, Zhang K, et al. Pan-cancer circulating tumor DNA detection in over 10,000 Chinese patients. Nat Commun. 2021;12:11. doi: 10.1038/s41467-020-20162-8. [DOI] [PMC free article] [PubMed] [Google Scholar]