The PDE4D7 score is associated with survival outcomes in high-risk prostate cancer patients
All subjects enrolled in this study experienced post-surgical PSA relapse and were subsequently stratified to salvage radiation therapy (SRT). Pathological analysis of surgical specimens revealed pT3a or higher disease in nearly 80% of the 367 patients (Table 1). Extra-prostate extension and positive surgical resection margins were identified in 62.4% and 72.2% of cases, respectively. Based on the CAPRA-S risk score [24], most patients (85%) were classified as intermediate-risk (41.1%; CAPRA-S scores 2–5) or high-risk (44.1%; CAPRA-S scores > 5) for post-surgical disease progression. Over half of the patients (188/367) received ADT at some point during the study period. During a median follow-up of 103 months post-prostatectomy, 11.7% of subjects died from prostate cancer and 17.4% died from any cause (Table 1).
Table 1
Aggregated summary of the characteristics of the studied patient cohort. Demographics of the radical prostatectomy (RP) patient cohort including the N = 367 patients eligible for statistical data analysis. For patient age and preoperative PSA the min and max values in the cohort are shown; median and IQR are shown in parentheses. Post-surgical pathology is given (Note: extracapsular extension was derived from pathology stage information). The outcome category illustrates the cumulative events in terms of recurrence to clinical metastases (CR), salvage radiation therapy (SRT) or androgen deprivation therapy (ADT) after surgery. Mortality is shown as prostate cancer specific mortality (PCSM) as well as overall all-cause mortality (ACM); (N/A = not available).
# Patients
|
N = 367
|
Age range [years]
|
45.3–79.2 (median: 62.5; IQR: 8.1)
|
pre-operative PSA [ng/ml]
|
0.3–168 (median: 10.0; IQR: 9.3)
|
pT Stage
|
N
|
%
|
pT2
|
78
|
21.3%
|
pT3a
|
131
|
35.7%
|
pT3b
|
103
|
28.1%
|
pT3c
|
44
|
12.0%
|
pT4
|
11
|
3.0%
|
ISUP Gleason
|
|
|
Grade Group 1
|
36
|
9.8%
|
Grade Group 2
|
147
|
40.1%
|
Grade Group 3
|
109
|
29.7%
|
Grade Group 4
|
19
|
5.2%
|
Grade Group 5
|
55
|
15.0%
|
Not available
|
1
|
0.3%
|
CAPRA-S Score
|
|
|
CAPRA-S < = 2
|
39
|
10.6%
|
CAPRA-S 3–5
|
151
|
41.1%
|
CAPRA-S > = 6
|
162
|
44.1%
|
Not available
|
15
|
4.1%
|
EAU-BCR Risk Score
|
|
|
EAU-BCR Risk 0
|
185
|
50.4%
|
EAU-BCR Risk 1
|
163
|
44.4%
|
Not available
|
19
|
5.2%
|
Surgical Margin Status
|
|
|
Negative
|
100
|
27.2%
|
Positive
|
265
|
72.2%
|
Not available
|
2
|
0.5%
|
Extra-Prostatic Extension
|
|
|
Absent
|
135
|
36.8%
|
Present
|
229
|
62.4%
|
Not available
|
3
|
0.8%
|
Seminal Vesicle Invasion
|
|
|
Absent
|
244
|
66.5%
|
Present
|
123
|
33.5%
|
Lymph Node Invasion
|
|
|
Absent
|
334
|
91.0%
|
Present
|
33
|
9.0%
|
Clinical Metastasis
|
|
|
No event
|
182
|
49.6%
|
Event
|
128
|
34.9%
|
Unknown
|
57
|
15.5%
|
Salvage Radiation
|
|
|
Not administered
|
0
|
0%
|
Administered
|
367
|
100%
|
Androgen Deprivation Therapy
|
|
|
Not administered
|
162
|
44.1%
|
Administered
|
188
|
51.2%
|
Unknown
|
17
|
4.6%
|
Prostate Cancer-Specific Mortality
|
|
|
No event
|
324
|
88.3%
|
Event
|
43
|
11.7%
|
All-Cause Mortality
|
|
|
No event
|
303
|
82.6%
|
Event
|
64
|
17.4%
|
Median Follow-Up [post-RP]
|
|
|
|
103 months
|
Multivariate Cox regression analysis was conducted to assess the association between the PDE4D7 score and prostate cancer-specific mortality (PCSM) after SRT initiation, adjusting for pathological prognostic variables (i.e., ISUP Gleason grade group, pathological pT stage, status of extra-prostate extension and surgical margins, seminal vesicle involvement, and lymph node invasion). As previously reported for biochemical recurrence as a clinical endpoint, we observed a strong inverse association between the PDE4D7 score and PCSM after SRT initiation (HR = 0.37; 95% CI 0.23–0.58; p < 0.0001) in the multivariable model that included clinical prognostic parameters (Table 2). To determine whether the PDE4D7 score provided independent prognostic information in the context of the genomic GPS or CCP signatures, the respective RNAseq derived score (mxGPS, mxCCP) was included in the multivariable model. Interestingly, only the mxGPS signature demonstrated a significant association with PCSM in multivariable analysis (HR = 1.3; 95% CI 1.0-1.6; p = 0.03). In both analyses, the PDE4D7 score remained a significant variable (p = 0.01 and p = 0.007; Supplementary Tables 1 and 2, respectively). The PDE4D7 score was also tested in multivariable analysis for PCSM in combination with the clinical prognostic models CAPRA-S and EAU-BCR risk score [24, 25]. In both models, the PDE4D7 score remained the most significant predictor for PCSM after SRT (HR = 0.36, p < 0.0001; HR = 0.31, p < 0.0001, respectively; Supplementary Table 3).
Table 2
Multivariable Cox regression analysis of the PDE4D7 score with the clinical prognostic parameters in a N = 342 clinical high risk prostate cancer patient cohort. The tested endpoint was time to prostate cancer specific mortality (N = 42 events). The PDE4D7 score and the ISUP Gleason grade group were used as continuous variables; the pathology stage pT was used as categorical variable with pT2 as reference. Extra-Prostatic Extension (EPE), Seminal Vesicle Invasion (SVI), Surgical Margin Status (SMS), and Lymph Node Invasion (LNI) were used as binary variables (0=’no’; 1=’yes’). The Hazard Ratio (HR), 95% confidence interval (CI) of the HR, and the p-values are indicated.
N = 342 Patients
|
Multivariable
|
Variable
|
HR
|
95% CI
|
P
|
PDE4D7 score
|
0.37
|
0.23 to 0.58
|
< 0.0001
|
ISUP pGleason Grade Group
|
1.8
|
1.4 to 2.4
|
< 0.0001
|
pT_stage="pT3a"
|
0.45
|
0.14 to 1.4
|
0.19
|
pT_stage="pT3b"
|
0.1
|
0.01 to 0.9
|
0.04
|
pT_stage="pT3c"
|
0.01
|
0 to 0.28
|
0.006
|
pT_stage="pT4"
|
0.36
|
0.04 to 3.3
|
0.36
|
Extra-Prostatic Extension
|
1.6
|
0.3 to 5.9
|
0.63
|
Seminal Vesicle Invasion
|
17.1
|
1.4 to 205
|
0.02
|
Surgical Margin Status
|
0.45
|
0.23 to 0.93
|
0.03
|
Lymph Node Invasion
|
2.2
|
1.0 to 4.8
|
0.04
|
The Kaplan-Meier (KM) analysis of pPDE4D7 percentile scores stratified patients into three sub-cohorts with significantly different survival rates (HR intermed-high: NA; HR low-intermed: 4.9; 95% CI 2.1–11.3; logrank p < 0.0001; Fig. 1A). Patients in the high pPDE4D7 score class had 100% 10-year survival rate after SRT, while those in the intermediate and low score classes had a 10-year post-SRT survival rates of approximately 90% and below 50%, respectively (Fig. 1A). All-cause mortality exhibited similar trends in this patient cohort (Fig. 1B). The EAU-BCR Risk model stratified patients into low- and high-risk groups, with the low-risk group having a significantly better survival rate at 10 years post-radiation (HR = 3.7; 95% CI 2.0-6.8; logrank p < 0.0001; Fig. 1C). The survival rate in the low-risk group was greater than 90%, while it was approximately 72% in the high-risk group at 10 years post SRT. The CAPRA-S model did not show significant stratification between risk groups (not shown).
To assess the predictive ability of the PDE4D7 score for 5-year PCSM after SRT, we conducted receiver operating characteristic (ROC) curve analysis and calculated the corresponding area under the curve (AUC). We compared the PDE4D7 score to individual clinical parameters and clinical models, as well as combination models incorporating the PDE4D7 score with the EAU-BCR risk score and additional clinical variables (full risk model: PDE4D7 score, ISUP pGleason grade group, pT Stage, extra-prostatic extension and surgical margin status, SVI, and LNI). The individual clinical parameters and clinical models demonstrated AUCs below 0.7, while the PDE4D7 & clinical combination models exhibited AUCs of 0.81 (PDE4D7_EAU-BCR) and 0.88 (full risk model) (p < 0.0001). These results suggest that the PDE4D7 & clinical combination models have superior predictive ability for 5-year PCSM compared to the individual clinical parameters and models (Supplementary Table 4).
Finally, we applied KM survival analysis to investigate whether pPDE4D7 percentile score classes could stratify patients receiving anti-androgens in addition to SRT into distinct subgroups with varying risks of PCSM or ACM. Our results revealed a statistically significant difference in survival outcomes between pPDE4D7 score classes (HR intermed-high: NA; HR low-intermed: 4.3; 95% CI 1.9–9.6; logrank p < 0.0001; Fig. 1D). Specifically, patients in the low pPDE4D7 score class had a median survival of 89.6 months and an overall cancer-specific survival rate of 30% post-initiation of ADT, while those in the intermediate and high pPDE4D7 score classes had not reached median survival. A similar pattern was observed for ACM, with the overall survival rate dropping below 20% for the lowest pPDE4D7 class (Fig. 1E). In contrast, none of the other studied risk classification models (mxpGPS, EAU-BCR risk, CAPRA-S) exhibited the ability to stratify patients into different risk classes with significant performance in this setting (Supplementary Figs. 2–4).
Our data demonstrate that the PDE4D7 score is a significant predictor of disease specific (PCSM) and overall (ACM) mortality in patients with recurrent and progressive PCa following primary surgical resection. Patients with higher PDE4D7 scores had a more favourable long-term survival rate, while those with lower scores exhibited a poorer prognosis in terms of survival following post-surgical treatments. These findings suggest that the PDE4D7 score may be a useful predictor of patient outcomes in this patient population.
PDE4D7 knockdown is associated with loss of androgen signaling, enrichment of the mesenchymal epithelial transition, and neuroendocrine prostate cancer.
To further investigate the biological significance of PDE4D7 in PCa development and progression, we generated a derivative of the LNCaP (clone FCG) PCa cell line with selective knockdown of PDE4D7 expression using a lentivirus-mediated shRNA strategy (Supplementary Fig. 5A). Among several PDE4D-knockdown LNCaP clones, LNCaP_P1 cells exhibited the most significant downregulation of PDE4D7 mRNA expression, as confirmed by RT-qPCR (Supplementary Fig. 6). Western blot analysis and confocal microscopy further confirmed the reduction in PDE4D7 protein expression in LNCaP_P1 cells compared to the wildtype (LNCaP_WT) and a scrambled shRNA control (LNCaP_SC2) (Figs. 2A, 2B, and 2C). These findings demonstrate the successful generation of a PDE4D7 knockdown model in LNCaP cells.
We performed next-generation sequencing (NGS) RNA sequencing of the two control cell lines (LNCaP_WT and LNCaP_SC2) as well as three PDE4D7 knockdown clones (LNCaP_P1, LNCaP_w5.2 and LNCaP_w6.3) and used the corresponding gene expression count tables as input for gene set enrichment analysis (GSEA) of the 50 GSEA hallmark pathways (gsea-msigdb.org). For pathway analysis in phenotypes WT and SC2 vs. the PDE4D7 knockdown cell lines, we identified the AR response pathway as the most significantly depleted gene set in the shRNA knockdown cell lines (false discovery rate (FDR) q-value < 0.001; normalised enrichment score − 2.1; Figs. 2D, 2E, Supplementary Table 5, respectively). To confirm that RNAseq data was reflected at the protein level, western blotting for selected PCa-related proteins was performed, with significant downregulation observed in the P1 cell line compared to LNCaP WT (Fig. 2F). Importantly, the scrambled shRNA cell line SC2 did not show notable difference in expression of these genes (Supplementary Fig. 7). Amongst the most downregulated genes in the three PDE4D7 knockdown cell lines were the known AR-regulated kallikrein-related peptidases KLK2 and KLK3, the transmembrane serine protease 2 (TMPRSS2), and the transcription factor NK3 homeobox-1 (NKX3-1). Also, the AR itself – although not part of this hallmark AR response pathway – is strongly downregulated in the PDE4D7 knockdown cell line P1 (Fig. 2F). The described findings indicate that the selective depletion of the PDE4D7 transcript expression leads to a cellular phenotype which represents androgen-insensitive proliferation.
In contrast, we identified multiple hallmark pathways significantly enriched in the PDE4D7 knockdown LNCaP derivatives. Of those, the EMT pathway demonstrated the highest level of enrichment (FDR q-value < 0.001; normalised enrichment score 1.9; Figs. 2G, 2H, Supplementary Table 6, respectively). Other hallmark pathways with FDR q-values < 0.1 that may further contribute to the progressive phenotype of the PDE4D7-knockdown cells, and are potentially relevant in a clinical setting, are those representing WNT beta catenin and hedgehog signaling. Both pathways were previously linked to progression of PCa after the disease developed into hormone resistance [27, 28].
Neuroendocrine differentiation (NED) in PCa is a hallmark of aggressive, AR-independent and treatment-resistant disease [29]. It can occur in both primary and metastatic PCa [30]. NED is characterized by decreased expression of the AR and increased expression of neuroendocrine markers [31]. Given the importance of NED in aggressive PCa, we evaluated a range of genes altered in NEPC (Supplementary Table 7) for expression in our LNCaP models. This analysis revealed that many genes with previously reported induced expression levels in NEPC (Neuroendocrine Prostate Cancer) were up-regulated in the PDE4D7 knockdown models including classical NED markers like ENO2, SYP, AURKA, or NCAM1 (Fig. 2I).
To explore the potential clinical relevance of these findings, we also analysed expression of hallmark AR response genes and key markers for NED in RNAseq data of 533 human patient samples described previously by us [32] and found that all tested AR response genes were down-regulated along with reduced PDE4D7 expression in these patient tumours, while expression of the AR itself did not change (Supplementary Fig. 8). Similarly, we found several markers reported in NED with altered expression between low vs high PDE4D7 tumours (Supplementary Fig. 9) and the change of regulation was according to what was reported previously in NEPC [29, 33].
PDE4D7-knockdown in LNCaP cells alters phenotype and confers resistance to AR antagonist
Knockdown of PDE4D7 in LNCaP cell line P1 resulted in increased growth characteristics as confirmed by real-time growth assays comparing P1 LNCaP cells to WT and SC2 LNCaP cells (Fig. 3A). Importantly, no significant difference in growth characteristics were found between the WT and SC2 LNCaP cells.
Enzalutamide, an AR antagonist used to treat advanced PCa [34], was utilised to investigate the effects on growth of PDE4D7-knockdown PCa cells. Dose-response experiments using real-time growth assays showed a significant decrease in WT LNCaP proliferation at all concentrations tested (0.1–30 µM), with the most profound effects at 1 µM and above (Fig. 3B). In contrast, P1 LNCaP cells showed minimal reductions in proliferation upon treatment with enzalutamide, and even exhibited increased growth at higher concentrations (Fig. 3C). Re-expression of PDE4D7 in LNCaP P1 cells via a Tet-On inducible vector (Supplementary Fig. 5B) rescued the growth phenotype and restored sensitivity of P1 LNCaP cells to enzalutamide (Fig. 3D, 3E). This data strongly highlights that low PDE4D7 expression in PCa cells confers resistance to anti-androgens such as enzalutamide.
MR-L2, a PDE4 longform activator that binds allosterically to the PDE4 enzyme in order to mimic PKA phosphorylation in the UCR1 domain [35], was tested to rescue the enhanced growth phenotype caused by PDE4D7 knockdown in LNCaPs. Results showed inhibition of growth in P1 cells, but no effect on proliferation in WT LNCaPs (Fig. 3F, 3G). This data suggests that activation of PDE4 in PDE4D7-knockdown LNCaPs has an inhibitory effect on their proliferative potential and highlights again the importance of PDE4D7 in driving PCa.
PDE4D7 specific knockdown in LNCaPs is associated with expression changes in DNA damage repair genes
Multiple studies have conclusively demonstrated that genomic alterations in DNA damage repair (DDR) genes are prevalent in both primary and metastatic PCa tissue [36], leading to the clinical utilisation of PARP inhibitors (PARPi) in patients with DDR deficiencies [37]. However, the emergence of resistance mechanisms has been observed in patients receiving PARPi treatment [38]. To investigate the potential impact of PDE4D7 knockdown on DNA repair mechanisms, we used the REACTOME homology-directed repair (HDR) of DNA double strand breaks gene set (reactome.org: R-HSA-5693538) and expanded it with additional genes from other DDR pathways previously reported to be altered in PCa (Supplementary Table 8) [36]. Our analysis across the LNCaP WT and SC2 cell lines, as well as their PDE4D7 knockdown derivatives, revealed alterations in the expression of a subset of DDR genes in the knockdown cells, particularly among DDR genes that are commonly mutated in PCa.. Most of these genes are significantly enhanced in expression in the PDE4D7 knockdown cells (Fig. 4A), while we did not identify significant differences in single nucleotide variants between the parent and knockdown cell line (Supplementary Figs. 10A, 10B, respectively).
To further evaluate the correlation between low PDE4D7 expression and potential resistance to PARPi treatment, we re-analysed RNAseq data from 533 human clinical samples [32], focusing on the expression of PCa-relevant DDR genes. Our analysis revealed a most significant increase in BRCA2 expression with decreasing PDE4D7 expression in patient tumours (p < 0.0001) along with a range of other DDR-related genes (Fig. 4B; Supplementary Fig. 11). BRCA1, ATM and BRIP1 were also upregulated but at non-significant levels (not shown). Based on these findings, we posit that the altered expression of DDR genes with decreasing transcription of PDE4D7 may contribute to resistance to DDR targeting therapies, and that cell lines with low expression of PDE4D7 following selective knockdown may exhibit resistance to PARPi compounds such as Olaparib.
PDE4D7 knockdown in LNCaPs confers resistance to PARP-inhibition and is rescued upon reintroduction of PDE4D7
Olaparib is an FDA-approved PARPi that is effective against LNCaP cells [39]. Ceralasertib (AZD6738) is an ataxia-telangiectasia mutated and Rad3-related (ATR) inhibitor, which has been suggested to enhance synergistic effects in combination with Olaparib in metastatic castration resistant prostate cancer (CRPC) [40]. In light of this, we aimed to determine whether PDE4D7 knockdown affects LNCaP cells response to either of these treatments. Using real-time growth analysis, LNCaP WT proliferation decreased in a dose-dependent manner with Olaparib (Fig. 5A), whereas the effect on PDE4D7-knockdown LNCaP P1 was minimal (Fig. 5B). Evaluation of growth rate via slope analysis yielded similar data with significant reductions in WT growth at 3, 10 and 30 µM Olaparib, but with unexpected reduction in slope at low concentrations (0.3 and 1 µM) in cells with PDE4D7 knockdown. It is notable, however, that cells with reduced PDE4D7 expression were resistant to Olaparib at 10 and 30 µM (Fig. 5B). Next, we sought to determine whether we could rescue the phenotype through transient re-introduction of PDE4D7 into LNCaP P1. As with the TET-On induction of PDE4D7, transiently transfected PDE4D7 into P1 cells (P1-PDE4D7+) revealed significant downregulation in growth (Fig. 5C). Furthermore, P1-PDE4D7+ cells were re-sensitised to 10 µM Olaparib treatment (Fig. 5D), with a statistically significant downregulation in growth compared to both Olaparib-treated P1 cells, and DMSO-treated P1-PDE4D7+ cells. Additionally, proliferation of P1-PDE4D7+ cells decreased further with Olaparib treatment in comparison to DMSO (p = 0.057). This data re-confirms the role of PDE4D7 in blunting of PCa cell growth and promoting susceptibility to clinically relevant therapeutics.
PDE4D7-knockdown in LNCaP cells does not affect response to ATR inhibition yet may limit docetaxel treatment response
Docetaxel is a DNA-damage inducing drug which is clinically used in advanced PCa treatment. Compared to DMSO, Docetaxel significantly reduced growth of both WT and P1 LNCaPs (Supplementary Figs. 12A, 12B, respectively]. We then investigated how PDE4D7-reintroduction into P1 cells affected their response to this. As with Olaparib, P1-PDE4D7+ cells were more sensitive to Docetaxel treatment than P1 (Supplementary Fig. 12C), suggesting that a paucity of PDE4D7 promotes the ability to repair associated DNA damage and protects against induction of cell death. This is in line with the increased expression of several key DDR genes in PDE4D7-knockdown LNCaPs (Fig. 4A).
Interestingly, whilst PDE4D7-knockdown conferred resistance of LNCaP cells to Olaparib at 10–30 µM, these cells were extremely sensitive to ATR inhibition via Ceralasertib at the same concentrations. Growth curves for WT and P1 LNCaP cells treated with increasing concentrations of Ceralasertib showed similar trends in sensitivity to the compound (Figs. 5E, 5F, respectively). In both cases, the most significant growth inhibition occurred at 10–30 µM treatment. These results highlight that PDE4D7 knockdown confers resistance to PARP-inhibition via Olaparib, however these cells are highly susceptible to ATR-inhibition via Ceralasertib.
Cyclic AMP dynamics in response to PDE4D7 knockdown and effect of PDE4 activation
Using FRET microscopy [41], we investigated how disease-related PDE4D7 knockdown affects localized cAMP signaling in response to drug treatments or cellular conditions in PCa CUTie cAMP probes targeting the plasma membrane (AKAP-79) or cytosol (CYTO-pDUAL) were used [42]. Rolipram downregulated the FRET ratio in the membrane compartment of WT LNCaPs, indicating increased cAMP concentration [Fig. 6B]. Forskolin/IBMX treatment did not further affect these cells, indicating that PDE4 inhibition maximized cAMP concentration. P1 LNCaPs showed no significant difference in FRET ratio upon rolipram treatment, but forskolin/IBMX addition slightly increased intracellular cAMP [Fig. 6D]. PDE4D7 is substantially membrane-located [Fig. 4C], leading to increased cAMP at this locale in WT cells. PDE4D7 knockdown elevated basal cAMP levels, leading to no upregulation in P1 cells. No observable difference was seen in WT and P1 LNCaPs upon rolipram treatment in the cytosol, but forskolin/IBMX addition decreased FRET ratio/increased cAMP [Figs. 6A and 6B]. This data stipulates the importance of PDE4D7 as a key regulator of cAMP concentration in the membrane domain of prostate cancer cells.