Abstract
PURPOSE:
While 177Lu-PSMA-617 (LuPSMA) is an effective therapy for many patients with metastatic castration-resistant prostate cancer (mCRPC), biomarkers associated with outcomes are not well defined. We hypothesized that prostate cancer mutational profile may associate with clinical activity of LuPSMA. We devised a study to evaluate associations between mCRPC mutational profile with LuPSMA clinical outcomes.
PATIENTS AND METHODS:
This was a multicenter retrospective analysis of mCRPC patients with next-generation sequencing (NGS) who received LuPSMA. PSA50 response (i.e. ≥50% decline in PSA) rate, PSA progression free survival (PSA PFS) and overall survival (OS) were compared between genetically defined sub-groups.
RESULTS:
One hundred twenty-six patients with NGS results who received at least one cycle of LuPSMA were identified. The median age was 73 (IQR: 68 – 78), 124 (98.4%) received ≥1 prior androgen receptor-signaling inhibitor and 121 (96%) received ≥1 taxane-based chemotherapy regimen. Fifty-eight (46%) patients with a DNA damage repair gene mutation (DDR group) and fifty-nine (46.8%) with a mutation in TP53, RB1, or PTEN tumor suppressor genes (TSG group) were identified. After adjusting for relevant confounders, the presence of ≥1 TSG mutation was associated with shorter PSA PFS (HR=1.93; 95% CI 1.05, 3.54; p=0.034) and OS (HR=2.65; 95% CI 1.15, 6.11; p=0.023). There was improved OS favoring the DDR group (HR=0.37; 95% CI 0.14, 0.97, p=0.044) on multivariable analysis. Univariate analysis of patients with ATM mutations had significantly higher rates of PSA50 response, PSA PFS and OS.
CONCLUSIONS:
Outcomes on LuPSMA varied based on mutational profile. Prospective studies to define the clinical activity of LuPSMA in predefined genomic subgroups are justified.
Keywords: 177Lu-PSMA-617, ATM, CDK12, DNA damage repair, metastatic prostate cancer, Prostate-specific Membrane Antigen, tumor suppressor gene alterations
Introduction:
While a number of agents have been shown to improve overall survival (OS) for men with metastatic castration-resistant prostate cancer (mCRPC), there remains a great need for more effective targeted therapies1,2. Prostate-specific membrane antigen (PSMA) is a transmembrane protein that is highly expressed in most cells of prostatic origin, and has emerged as a selective target for both imaging and therapeutics. Currently, a number of agents directed towards PSMA-expressing cells are in development including radioligands, antibody drug conjugates, and CAR-T constructs3.
To date, 177Lu-PSMA-617 (LuPSMA) is the only agent approved for the therapeutic targeting of PSMA. It is composed of PSMA-617, a small molecule ligand with specificity for PSMA, conjugated to the beta emitter 177Lu. Its approval was based on the VISION trial, which tested LuPSMA against protocol-permitted standard of care in patients with mCRPC who had PSMA-expressing cancer (determined by PET imaging) and who had previously progressed on an androgen receptor (AR)-signaling inhibitor (ARSI) and taxane chemotherapy4. VISION demonstrated improvements in both OS and progression free survival (PFS); however, the clinical activity of LuPSMA was heterogeneous with a median PFS of only 9 months and objective responses in approximately half of patients. As such, additional biomarkers are needed to further refine patient selection strategies and to determine which patients stand to benefit from LuPSMA.
Unsurprisingly, baseline PSMA expression on PET imaging appears to associate with improved clinical outcomes to LuPSMA5,6. However, other factors associated with improved outcomes with LuPSMA are poorly defined, including the tumor molecular profile. To that end, only a handful of small, inconclusive studies have evaluated associations between prostate cancer molecular profile and outcomes following LuPSMA therapy7–9.
A recent study suggested that alterations in a variety of DDR pathway genes may correlate with higher PSMA expression10. In addition, DDR gene alterations may also increase sensitivity to radiation-mediated DNA damage11. Of note, a prior study has correlated the presences of these mutations with increased sensitivity to the alpha emitter Radium-22312. Conversely, loss-of-function mutations in tumor suppressor genes (TSGs) have been associated with neuroendocrine transdifferentiation as well as emergence of AR-null phenotypes, which often do not express PSMA and would therefore not be expected to respond to LuPSMA13,14. Loss of TSGs have also been associated with radio-resistance across cancer types and have been shown to mediate resistance to PSMA radioligand therapy in a mouse model15,16.
Given the limited data correlating genomics with LuPSMA clinical activity, we conducted a multi-institutional retrospective study aimed at addressing this knowledge gap. We hypothesized that i) patients with DDR gene alterations would have more favorable clinical outcomes to LuPSMA, and ii) loss-of-function mutations in TSGs would be associated with less favorable LuPSMA outcomes.
Methods:
Patients and Methods
Participating centers included University of Washington (UW), University of California San Francisco (UCSF), and University of Colorado (UC). IRB approval was obtained at each site. Patients were included in the primary analysis of outcomes to LuPSMA if they: i) had a diagnosis of mCRPC, ii) received at least one cycle of LuPSMA, and iii) had next-generation sequencing panel (NGS) testing performed on tumor material using a CLIA-approved clinical-grade assay. A germline alteration in a DDR gene was considered sufficient to classify a patient as having a DDR-deficient prostate cancer; however, in the absence of a germline DDR mutation, somatic testing was required for inclusion in this analysis.
A variety of sequencing platforms were used, including UW-OncoPlex, UCSF-500, Foundation One, and Tempus, and mutations reported as pathogenic on the sequencing report were considered for this analysis17–20. Tumor DNA analysis was performed on primary samples, metastatic tissue, or cell-free circulating tumor DNA (ctDNA). To avoid false negatives due to low tumor content, we required the presence of an identifiable somatic mutations when ctDNA was sequenced. Cases demonstrating alterations that are commonly attributed to clonal hematopoiesis of indeterminate potential (CHIP) (e.g., TET2, DNMT3A) reported at a variant allele frequency less than 1% were not included.
Cases with any pathogenic mutation involved in a DDR pathway, including those involved in homologous recombination repair or mismatch repair (MMR), were included in the DDR-deficient cohort given prior data implicating these pathways in repairing radiation induced DNA damage (supplemental table 1)21,22. The commercial sequencing assays used in this study did not consistently report zygosity status of affected DDR pathway genes. If a sequencing report indicated a pathogenic alteration in a DDR pathway was detected, this was considered sufficient for inclusion in the DDR-deficient cohort. However, when a sequencing report explicitly stated that a DDR gene mutation was monoallelic, that case was included in the DDR-intact cohort. Mutations in TP53, PTEN and/or RB1 were included in the TSG analyses.
Assessment of PSMA Expression
To explore associations between DDR gene mutational status and PSMA expression, we assembled an additional cohort of mCRPC patients who had undergone sequencing and PSMA PET imaging at UW. PSMA SUVmean was used as a global measure of PSMA expression and was determined using image analysis software (MIM 8.7.11; MIM Software, Cleveland, OH) from baseline PSMA PET images. Lesions with an SUV ≥3 were used to calculate the total tumor volume and determine the PSMA SUVmean as previously described13. PSMA-high disease was defined as PSMA SUVmean ≥10 while PSMA-low disease was defined as a PSMA SUVmean <10 because this threshold is associated with clinical outcomes on LuPSMA5,6.
Statistical Analyses
Clinical outcomes of interest included PSA50 response (i.e. ≥50% decline in PSA from baseline), PSA progression-free survival (PSA PFS), and overall survival (OS) from the time of initiating LuPSMA. PSA progression was defined as PSA ≥2 ng/mL and ≥25% above the nadir on two consecutive lab draws, as recommended by PCWG321. Given inconsistency of repeat imaging and differences in practice patterns, radiographic PFS could not be reliably assessed.
Differences in baseline patient characteristics were compared using the Wilcoxon rank sum - and chi-squared test. PSA PFS and OS were evaluated using the Kaplan-Meier method and compared using the log-rank test. Cox regression analyses were used to evaluate associations between any DDR or TSG status and PSA PFS or OS after adjusting for baseline PSMA SUVmean, prior receipt of PARP inhibitor (PARPi), and presence of liver metastases. Covariates were selected based on observed associations with clinical activity of LuPSMA in univariate analysis and/or prior data suggesting that covariates are associated with outcomes4–6. Analyses were performed using R 4.3.1.
Results:
Patient characteristics
A total of 126 patients who had undergone NGS and received at least one cycle of LuPSMA were identified (Supplemental Figure 1, Supplemental Table 2). The median age was 73 years (IQR: 68 – 78), 124 (98.4%) received ≥1 prior ARSI and 121 (96%) received ≥1 taxane-based chemotherapy regimen (Table 1). The PSMA SUVmean was 7.2 for 66 patients at UW who had baseline PSMA PET scans available for analysis. Fifty-eight (46%) patients with a DDR gene mutation (i.e. DDR-deficient) were identified. The cohorts were largely similar when stratified by DDR gene status; however, the DDR-deficient cohort was younger, had higher likelihood of receiving prior PARPi therapy, and were less likely to have ECOG 2 performance status (Supplemental table 3). After excluding six patients with germline only or limited panel sequencing, clinical data on 120 patients were available for analysis based on TSG status. Fifty-nine (49%) patients with a TSG mutation were identified. The TSG-altered cohort was more likely to have lower PSA at diagnosis and to have received more lines of ARSI therapy (Supplemental Table 4).
Table 1:
Baseline characteristics
Characteristic | N = 126 |
---|---|
DDR Status, n (%) | |
DDR deficient | 58 (46%) |
DDR intact | 68 (54%) |
TSG Status, n (%) | |
Any TSG | 59 (49%) |
No TSG | 61 (51%) |
Unknown | 6 |
Age, median (IQR) | 73 (68, 78) |
Race, n (%) | |
Asian | 5 (4.0%) |
Black | 5 (4.0%) |
Other | 8 (6.5%) |
White | 106 (85%) |
Unknown | 2 |
Gleason, n (%) | |
<8 | 26 (24%) |
8-10 | 83 (76%) |
Unknown | 17 |
ECOG PS, n (%) | |
0 | 46 (38%) |
1 | 63 (52%) |
2 | 12 (9.9%) |
Unknown | 5 |
PSA at Diagnosis, median (IQR) | 25 (8, 87) |
Unknown | 18 |
De Novo Metastatic, n (%) | 56 (44%) |
Local Therapy, n (%) | |
Radiation | 32 (25%) |
Surgery | 44 (35%) |
None | 50 (40%) |
Liver Metastases, n (%) | 20 (16%) |
Bone Metastases, n (%) | 117 (93%) |
Nodal Metastases, n (%) | 81 (64%) |
Lung Metastases, n (%) | 20 (16%) |
Lines of ARSI, n (%) | |
0 | 2 (1.6%) |
1 | 37 (29%) |
2 | 87 (69%) |
Lines of Taxane, n (%) | |
0 | 5 (4.0%) |
1 | 69 (55%) |
2 | 50 (40%) |
3 | 2 (1.6%) |
Radium-223 Therapy, n (%) | 17 (13%) |
Mutational Profile and Activity of LuPSMA
Overall, 64 out of 126 patients (50%) achieved a PSA50 response. The median duration of follow up was 9.43 months (95% CI: 8.77 – 10.5). Median PSA PFS was 5.72 months (95% CI: 4.76—7.82) and the median OS was 12.5 months (95% CI: 10.2— not reached) for the entire study cohort. There was no significant difference in PSA50 response rate between the DDR-deficient vs DDR-proficient cohort (57% vs 46%, P=0.3). The unadjusted survival outcomes between the DDR-deficient vs DDR-proficient groups were similar, with a median PSA-PFS of 6.2 months (95% CI 4.6, 9.1) and 5.8 (95% CI 4.2, 8.4) (p=0.3) for the DDR-deficient and DDR-proficient cohorts, respectively. The median OS was 23.8 months (95% CI 11.9, not reached) for the DDR-deficient cohort and 10.5 months (95% CI 8.1, not reached) (p = 0.18) for the DDR-proficient cohort (Figure 1).
Figure 1:
Unadjusted clinical outcomes for patients with vs. without DNA damage response (DDR) gene mutations.
We did not observe a difference in PSA50 response rates for patients whose tumors did vs did not have alterations in TP53 (53% vs 49%, p= 0.8), RB1 (50% vs. 51%, p>0.9) or in PTEN (58% vs. 50%, p=0.7) (Supplemental Figure 2). We also did not observe any differences in PSA50 response rates based on the number of TSG altered (Figure 2, Supplemental Table 3). Unadjusted survival outcomes were similar based on TSG status (Figure 2, Supplemental Figure 2).
Figure 2:
Unadjusted clinical outcomes based on mutations in tumor suppressor genes (TSG) (i.e. TP53, Rb1, PTEN).
A multivariable model including TSG status, DDR gene mutational status, liver metastases, receipt of prior PARPi and baseline PSMA SUVmean was constructed (Table 2). The presence of ≥1 TSG mutation was associated with shorter PSA PFS (HR=1.93; 95% CI 1.05, 3.54; p=0.034) and OS (HR=2.65; 95% CI 1.15, 6.11; p=0.023). While there was no difference in PSA-PFS based on DDR mutational status, OS was improved in the DDR-deficient cohort (HR=0.37; 95% CI 0.14, 0.97, p=0.044) after controlling for relevant covariates. The presence of liver metastases was also independently associated with shorter PSA PFS (HR=3.08; 95% CI 1.34, 7.09; p=0.008) and OS (HR=3.19; 95% CI 1.18, 8.60; p=0.022). A trend was observed for increased PSA PFS (p = 0.13) and OS (p = 0.08) for patients with high (PSMA SUVmean ≥10) vs. low (PSMA SUVmean <10) PSMA expression, although the sample size of this group was limited (N=66) (Supplemental Figure 3). Of note, we did not observe any difference in PSMA expression between DDR-deficient vs. DDR-proficient cohorts (Supplemental Figure 4) or the TSG-mutated vs. unmutated cohorts (Supplemental Figure 5).
Table 2:
Multivariable Cox regression model of PSA progression-free survival and overall survival
Characteristic | PSA Progression Free Survival | Overall Survival | ||||
---|---|---|---|---|---|---|
HR1 | 95% CI1 | p-value | HR1 | 95% CI1 | p-value | |
PSMA SUVmean > 10 | 0.59 | 0.25, 1.42 | 0.2 | 0.40 | 0.09, 1.71 | 0.2 |
DDR Alteration Present | 0.75 | 0.39, 1.45 | 0.4 | 0.37 | 0.14, 0.97 | 0.044 |
Liver Metastases | 3.08 | 1.34, 7.09 | 0.008 | 3.19 | 1.18, 8.60 | 0.022 |
PARPi Received | 1.44 | 0.70, 2.97 | 0.3 | 1.81 | 0.66, 4.94 | 0.2 |
TSG Alteration Present | 1.93 | 1.05, 3.54 | 0.034 | 2.65 | 1.15, 6.11 | 0.023 |
HR = Hazard Ratio, CI = Confidence Interval
Gene-Specific Analyses
Next, we evaluated clinical-genomic associations on an individual gene basis for DDR genes mutated in at least 10 patients (Figure 3, Table 3). We found substantial variation with regards to PSA50 responses between sub-groups. Twelve of 14 (86%) patients with an ATM alteration achieved a PSA50 response, which was significantly higher compared to the 52/112 (46%) PSA50 response rate in those without an ATM alteration (p=0.01). The PSA PFS for patients with vs. without an ATM alteration was also significantly longer, with a median PSA PFS of 9.6 months (95% CI 8.0—not reached) vs. 5.4 months (95% CI 4.1–7.4) (p=0.03). OS was also superior in the ATM altered group, where the median OS has not yet been reached compared to 11.9 months (95% CI 8.8, not reached) in the unaltered group (p=0.04). BRCA2 alterations were not associated with improved clinical outcomes. The PSA50 response rate for the BRCA2 group was 8/15 (53%) compared to 56/111 (50%) (p>0.9) in the unaltered group. There was also no difference in PSA PFS or OS for patients with vs. without a BRCA2 mutation. Patients with a CDK12 alteration had inferior outcomes, with 3/13 (23%) exhibiting a PSA50 response compared to 61/113 (54%, p = 0.07) for all other patients. These patients also had significantly decreased PSA PFS (p = 0.009) compared to the unaltered group, although OS was similar (p=0.4) between groups. PSMA expression did not vary significantly between genomic subgroups, although analysis was limited by sample size (Supplemental figure 6).
Figure 3:
Waterfall plot of maximum PSA response relative to baseline, PSA progression-free survival (PSA PFS), and overall survival (OS) for individual DNA damage response (DDR) mutations.
Table 3:
PSA response by individual DNA damage response (DDR) alterations.
Alteration | N | PSA50, N | PSA50, % |
---|---|---|---|
ATM | 10 | 8 | 80.00 |
ATM, CDK12 | 1 | 1 | 100.00 |
BRCA1 | 3 | 2 | 66.67 |
BRCA1, ATM | 1 | 1 | 100.00 |
BRCA2 | 13 | 6 | 46.15 |
BRCA2, ATM | 2 | 2 | 100.00 |
CDK12 | 12 | 2 | 16.67 |
CHEK2 | 3 | 3 | 100.00 |
FANCA | 1 | 1 | 100.00 |
PALB2 | 3 | 1 | 33.33 |
RAD51 | 1 | 0 | 0.00 |
RAD54L | 1 | 1 | 100.00 |
MMRd | 7 | 5 | 71.42 |
Discussion
To our knowledge, this is the largest study to correlate prostate cancer genomics with clinical outcomes to LuPSMA. After controlling for clinically relevant covariates, we found that mutations in DDR genes were associated with improved OS, while loss of function mutations in TSGs were associated with worse OS. Exploratory gene-by-gene analyses have also identified several clinically relevant subgroups that warrant further study. These include those with ATM mutations, who appear to have significantly better outcomes to LuPSMA, and those with CDK12 mutations, who appear to do worse.
Both clinical and preclinical studies have consistently demonstrated a correlation between PSMA expression and activity of LuPSMA5,6,22–24. As such, it is not surprising that loss of function mutations in TSGs, which are associated with the emergence of AR-null phenotypes that lack PSMA expression, would also correlate with poor outcomes to LuPSMA13,14. It is worth noting, however, that this association persisted after controlling for the presence of liver metastases and PSMA expression, which would also enrich for AR-null prostate cancers. As such, it is possible that these mutations may also influence the clinical activity of LuPSMA by inducing a radioresistant phenotype. Indeed, clinical data has also suggested that the presence of RB1 mutations may associate with poor outcomes to the non-PSMA targeted radiopharmaceutical radium-22325. Preclinical models have also implicated TSG mutations as a mediator of radio-resistance to PSMA targeted radioligand therapy15. It is also worth noting that two recent studies have reported similar genomic features to be associated with poor outcomes to LuPSMA, including TSG mutations and PI3K pathway alterations (including PTEN deletions)26,27.
A key challenge facing this, as well as other precision oncology studies, is the broad number of genes involved in DNA damage repair. Given the exploratory nature of this study, we opted for an inclusive approach toward defining our DDR-deficient cohort, including genes directly and indirectly involved in a number of DDR pathways. Overall, we observed improved survival following treatment with LuPSMA for the entire DDR-deficient cohort; however, within this cohort outcomes were heterogenous. While a biologic rationale can be made for why specific associations were observed, it is important to acknowledge that these findings need to be confirmed prospectively.
We observed excellent clinical outcomes in patients with ATM mutations who received LuPSMA. This may be explained by this gene’s role in mediating cellular response to radiation and is consistent with data showing that ATM deficient cells are highly radiosensitive. Some clinic data has also suggested that patients with homozygous germline defects in ATM (i.e. ataxia-telangectasia syndrome) are profoundly sensitive to adverse-effects of radiation28,29. Of relevance, a recent study found ATM-signaling to be one of the most significantly upregulated kinase pathways in response to LuPSMA therapy15. Interestingly, the data supporting ATM mutations as a predictive biomarker for response to PARPi monotherapy are conflicting30–32.
We also found that CDK12 mutations are associated with poor responses to LuPSMA. Of note, these mutations have not clearly been shown to predict response to PARPi either. Retrospective data has suggested that patients with CDK12 mutations appear to have a more aggressive clinical course, and translational work has shown that these alterations are associated with a focal tandem duplicator phenotype rather than the genomic signatures seen in tumors with deficiencies in homologous recombination repair33–35. Given the unclear role of CDK12 in the response to radiation and its association with worse prognosis in general, it is perhaps not surprising that this sub-group had inferior outcomes to LuPSMA. These data further highlight the need to develop new treatment strategies tailored toward this aggressive genomic subgroup.
We also found that outcomes to LuPSMA appeared favorable in patients with MMR deficient (MMRd) prostate cancer, although it is important to note that this group only included seven patients. While these mutations are not predictive for response to DNA damaging cytotoxics and PARP inhibitors, there is data showing that this pathway is involved in responding to DNA damage from ionizing radiation, which could explain the excellent outcomes in this group36. Overall, five (71%) of these patients had a PSA50 and the median PSA PFS and OS were 12 and 13 months, respectively (Supplemental figure 6). Notably, one patient with an MSH2 alteration and hypermutation had a complete response and currently off all treatment, including androgen deprivation therapy.
Our study has several important limitations. These include the retrospective nature of the study and small sample size of the analyzed genomic sub-groups. We were also only able to analyze PSMA SUVmean at one site, which limits our ability to form robust conclusions regarding the intersection between genomics, PSMA expression and clinical outcomes to LuPSMA. Another key limitation is the inability to clearly distinguish biallelic versus monoallelic alterations given that this is not consistently reported by most commercial assays – despite the fact that true monoallelic alterations are unlikely to associate with a DDR deficient phenotype37. We also note that, while all mCRPC patients should receive germline and tumor somatic genetic testing, this is still not uniformly done, and exclusion of patients who did not have sequencing may have led to selection bias. With these caveats, we believe this hypothesis generating study provides a framework for future work evaluating precision medicine approaches as they pertain to the use of LuPSMA.
Supplementary Material
Context Summary.
Key objective:
177Lu-PSMA-617 is a relatively new treatment option for men with metastatic castration resistant prostate cancer. However, about half of patients undergoing this treatment do not achieve objective tumor responses and biomarkers associated with response are lacking. This retrospective, multi-center study assessed 126 patients who had undergone next-generation sequencing as part of standard of care, and explored whether genomic alterations in prostate cancer may correlate with responses to 177Lu-PSMA-617 therapy.
Knowledge generated:
Patients with mutations in the DNA damage repair pathway have improved survival on 177Lu-PSMA-617 compared with those who do not. Exploratory gene-level analysis found that patients with ATM mutations appear to do particularly well with 177Lu-PSMA-617 therapy.
Relevance:
Our findings suggest that alterations in certain genes and pathways may hold prognostic significance for patients undergoing therapy with 177Lu-PSMA-617. Further validation in larger cohorts is essential to establish these genomic markers for refining patient selection.
Acknowledgements
This work was supported by the National Cancer Institute (award numbers P50CA097186, R50CA221836), and the NIH T32 CA009515-38 institutional training grant. CCP acknowledges support from SPORE CA097186, W81XWH-18-1-0756, and PC170510, W81XWH-21-1-0265 PC200262P1.
References:
- 1.Tannock IF et al. Docetaxel plus Prednisone or Mitoxantrone plus Prednisone for Advanced Prostate Cancer. New England Journal of Medicine 351, 1502–1512, doi: 10.1056/nejmoa040720 (2004). [DOI] [PubMed] [Google Scholar]
- 2.De Bono JS et al. Abiraterone and Increased Survival in Metastatic Prostate Cancer. New England Journal of Medicine 364, 1995–2005, doi: 10.1056/nejmoa1014618 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Jones W, Griffiths K, Barata PC & Paller CJ PSMA theranostics: review of the current status of PSMA-targeted imaging and radioligand therapy. Cancers 12, 1367 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Sartor O et al. Lutetium-177–PSMA-617 for Metastatic Castration-Resistant Prostate Cancer. New England Journal of Medicine 385, 1091–1103, doi: 10.1056/nejmoa2107322 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Kuo P et al. [68Ga] Ga-PSMA-11 PET baseline imaging as a prognostic tool for clinical outcomes to [177Lu] Lu-PSMA-617 in patients with mCRPC: A VISION substudy. (2022). [Google Scholar]
- 6.Buteau JP et al. PSMA and FDG-PET as predictive and prognostic biomarkers in patients given [177Lu] Lu-PSMA-617 versus cabazitaxel for metastatic castration-resistant prostate cancer (TheraP): a biomarker analysis from a randomised, open-label, phase 2 trial. The Lancet Oncology 23, 1389–1397 (2022). [DOI] [PubMed] [Google Scholar]
- 7.Privé BM et al. Impact of DNA damage repair defects on response to PSMA radioligand therapy in metastatic castration-resistant prostate cancer. Prostate Cancer and Prostatic Diseases 25, 71–78, doi: 10.1038/s41391-021-00424-2 (2022). [DOI] [PubMed] [Google Scholar]
- 8.Crumbaker M, Emmett L, Horvath LG & Joshua AM Exceptional response to 177Lutetium prostate-specific membrane antigen in prostate cancer harboring DNA repair defects. JCO Precision Oncology 3, 1–5 (2019). [DOI] [PubMed] [Google Scholar]
- 9.Ahmadzadehfar H, Gaertner F, Lossin PS, Schwarz B & Essler M BRCA2 mutation as a possible cause of poor response to 177Lu-PSMA therapy. Clinical Nuclear Medicine 43, 609–610 (2018). [DOI] [PubMed] [Google Scholar]
- 10.Paschalis A et al. Prostate-specific membrane antigen heterogeneity and DNA repair defects in prostate cancer. European urology 76, 469–478 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Schumann S et al. DNA damage in blood leucocytes of prostate cancer patients during therapy with 177Lu-PSMA. European Journal of Nuclear Medicine and Molecular Imaging 46, 1723–1732, doi: 10.1007/s00259-019-04317-4 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Velho PI et al. Efficacy of radium-223 in bone-metastatic castration-resistant prostate cancer with and without homologous repair gene defects. European urology 76, 170–176 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Beltran H et al. The role of lineage plasticity in prostate cancer therapy resistance. Clinical cancer research 25, 6916–6924 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Sayar E et al. Reversible epigenetic alterations mediate PSMA expression heterogeneity in advanced metastatic prostate cancer. JCI Insight 8, doi: 10.1172/jci.insight.162907 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Stuparu AD et al. Mechanisms of Resistance to Prostate-Specific Membrane Antigen–Targeted Radioligand Therapy in a Mouse Model of Prostate Cancer. Journal of Nuclear Medicine 62, 989–995 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.El-Deiry WS The role of p53 in chemosensitivity and radiosensitivity. Oncogene 22, 7486–7495 (2003). [DOI] [PubMed] [Google Scholar]
- 17.Pritchard CC et al. Validation and implementation of targeted capture and sequencing for the detection of actionable mutation, copy number variation, and gene rearrangement in clinical cancer specimens. The Journal of Molecular Diagnostics 16, 56–67 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Frampton GM et al. Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nature biotechnology 31, 1023–1031 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Beaubier N et al. Integrated genomic profiling expands clinical options for patients with cancer. Nature biotechnology 37, 1351–1360 (2019). [DOI] [PubMed] [Google Scholar]
- 20.Wagle N et al. High-throughput detection of actionable genomic alterations in clinical tumor samples by targeted, massively parallel sequencing. Cancer discovery 2, 82–93 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Scher HI et al. Trial Design and Objectives for Castration-Resistant Prostate Cancer: Updated Recommendations From the Prostate Cancer Clinical Trials Working Group 3. Journal of Clinical Oncology 34, 1402–1418, doi: 10.1200/jco.2015.64.2702 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Khreish F et al. Response Assessment and Prediction of Progression-Free Survival by 68Ga-PSMA-11 PET/CT Based on Tumor-to-Liver Ratio (TLR) in Patients with mCRPC Undergoing 177Lu-PSMA-617 Radioligand Therapy. Biomolecules 11, 1099, doi: 10.3390/biom11081099 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Seifert R et al. Analysis of PSMA expression and outcome in patients with advanced Prostate Cancer receiving 177Lu-PSMA-617 Radioligand Therapy. Theranostics 10, 7812–7820, doi: 10.7150/thno.47251 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Current K et al. Investigating PSMA-Targeted Radioligand Therapy Efficacy as a Function of Cellular PSMA Levels and Intratumoral PSMA HeterogeneityPSMA-RLT Efficacy as Function of PSMA Level and Heterogeneity. Clinical Cancer Research 26, 2946–2955 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Liu AJ et al. The impact of genetic aberrations on response to radium‐223 treatment for castration‐resistant prostate cancer with bone metastases. The Prostate 82, 1202–1209 (2022). [DOI] [PubMed] [Google Scholar]
- 26.Vanwelkenhuyzen J et al. AR and PI3K Genomic Profiling of Cell-free DNA Can Identify Poor Responders to Lutetium-177-PSMA Among Patients with Metastatic Castration-resistant Prostate Cancer. European Urology Open Science 53, 63–66 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Crumbaker M et al. Circulating Tumour DNA Biomarkers Associated with Outcomes in Metastatic Prostate Cancer Treated with Lutetium-177-PSMA-617. European Urology Open Science 57, 30–36 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Kühne M et al. A double-strand break repair defect in ATM-deficient cells contributes to radiosensitivity. Cancer research 64, 500–508 (2004). [DOI] [PubMed] [Google Scholar]
- 29.Meyn MS Ataxia-telangiectasia and cellular responses to DNA damage. Cancer research 55, 5991–6001 (1995). [PubMed] [Google Scholar]
- 30.Marshall CH et al. Differential response to olaparib treatment among men with metastatic castration-resistant prostate cancer harboring BRCA1 or BRCA2 versus ATM mutations. European urology 76, 452–458 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Abida W et al. Non-BRCA DNA damage repair gene alterations and response to the PARP inhibitor rucaparib in metastatic castration-resistant prostate cancer: analysis from the phase II TRITON2 study. Clinical Cancer Research 26, 2487–2496 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Schweizer MT, Cheng HH, Nelson PS & Montgomery RB Two steps forward and one step back for precision in prostate cancer treatment. Journal of Clinical Oncology 38, 3740 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Antonarakis ES et al. CDK12-altered prostate cancer: clinical features and therapeutic outcomes to standard systemic therapies, poly (ADP-ribose) polymerase inhibitors, and PD-1 inhibitors. JCO precision oncology 4, 370–381 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Schweizer MT et al. CDK12-mutated prostate cancer: clinical outcomes with standard therapies and immune checkpoint blockade. JCO precision oncology 4, 382–392 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Wu Y-M et al. Inactivation of CDK12 delineates a distinct immunogenic class of advanced prostate cancer. Cell 173, 1770–1782. e1714 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Martin LM et al. DNA mismatch repair and the DNA damage response to ionizing radiation: making sense of apparently conflicting data. Cancer treatment reviews 36, 518–527 (2010). [DOI] [PubMed] [Google Scholar]
- 37.Warner E et al. BRCA2, ATM, and CDK12 defects differentially shape prostate tumor driver genomics and clinical aggression. Clinical Cancer Research 27, 1650–1662 (2021). [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.