Purpose:

To identify the safety of niraparib, a PARP inhibitor, in combination with Radium-223 for the treatment of metastatic castrate-resistant prostate cancer (mCRPC) in men without known BRCA mutations.

Patients and Methods:

Men with progressive mCPRC following ≥1 line of androgen receptor (AR)-targeted therapy and bone metastases but no documented BRCA-1 or BRCA-2 alterations or bulky visceral disease were included. Niraparib dose was escalated in combination with standard dosing of Radium-223 using a time-to-event continual reassessment method. The highest dose level with a DLT probability <20% was defined as MTD. Secondary endpoints included PSA change and progression-free survival. Exploratory analyses included assessing DNA mutations found in ctDNA as well as gene expression changes assessed in whole blood samples.

Results:

Thirty patients were treated with niraparib and radium-223: 13 patients received 100 mg, 12 received 200 mg, and 5 patients received 300 mg of niraparib. There were six DLT events: two (13%) for neutropenia, two (13%) for thrombocytopenia, whereas fatigue and nausea each occurred once (3%). Anemia (2/13%) and neutropenia (2/13%) were the most common grade 3 adverse events. For patients with prior chemotherapy exposure, the MTD was 100 mg, whereas the MTD for chemotherapy naïve patients was 200 mg. Whole blood gene expression of PAX5 and CD19 was higher in responders and ARG-1, IL2R, and FLT3 expression was higher in nonresponders.

Conclusions:

Combining niraparib with Radium-223 in patients with mCRPC was safe; however, further studies incorporating biomarkers will better elucidate the role of combinations of PARP inhibitors with DNA damaging and other agents.

Translational Relevance

PARP inhibitors have been demonstrated to be effective in treating metastatic castrate-resistant prostate cancer (mCRPC) that harbors mutations in DNA damage repair (DDR) pathways. However, their effectiveness outside this population is limited, and investigation into their role in combination with other therapies is prudent. In addition, advances in radiopharmaceuticals in mCRPC has created a need for trials evaluating their safety with other approved agents. This novel designed phase Ib study evaluated the safety and tolerability of niraparib combined with Radium-223 in treatment of men with mCRPC. We were able to demonstrate the utility of a time-to-event continuous reassessment method of enrollment, and the combination of therapies proved to be manageable with a 200 mg dose of niraparib. Although the combination was safe, no increase in clinical activity was found. We demonstrated that gene expression profiling on whole blood was feasible and observed different patterns in expression of immunomodulatory genes between responders and nonresponders to therapy.

An estimated 250,000 men were diagnosed with prostate cancer in 2021, which remains the second most common cause of cancer death for men in the United States (1). Prostate cancer cells are dependent on androgen stimulation and the androgen axis for growth and androgen deprivation therapy is the cornerstone of treatment (2). Despite androgen deprivation, progression of disease occurs over time, in part, through mechanisms that re-activate the androgen receptor (AR) independent of androgen stimulation, leading to castrate-resistant prostate cancer (3). There are currently several different therapies for mCRPC that are approved which include: taxane chemotherapy, androgen axis inhibitors such has enzalutamide, apalutamide and abiraterone, sipuleucel-T, Radium-223, and in select populations, PARP inhibitors Olaparib or Rucaparib and Pembrolizumab, an anti-PD1 antibody (4–11).

Radium-223 is an alpha particle-emitting, radioactive agent that is FDA approved for treatment of patients with mCRPC and symptomatic bone metastases (12). In comparison to gamma radiation, alpha particle radiation produces higher rates of single-strand DNA breaks that are more frequently converted to double-strand breaks (13). The agent targets calcium hydroxyapatite in bone and accumulates in regions of osteoblastic activity, emitting alpha particles that have low penetrance depth and high linear energy transfer, making its effect localized to the cortical bone and minimizing bone marrow toxicity (14, 15). This localized effect and minimal systemic toxicity creates the potential for Radium-223 to be combined safely with other agents.

PARP-1 is a nuclear enzyme that recruits proteins that impact the DNA damage repair (DDR) pathways. Inhibitors of PARP-1 have demonstrated clinical activity in mCRPC when concurrent mutations in genes that code for DNA repair proteins are identified (10, 11, 16). PARP may also play an important role in prostate cancer through interaction with the AR (17–20). Niraparib is an orally available, highly selective PARP-1/PARP-2 inhibitor that has shown activity in tumors with DNA repair deficiencies and is currently approved, at a dose of 200 to 300 mg daily, for clinical use in ovarian cancer (21–23). Niraparib has also been found to be effective in treating mCRPC with known DNA repair gene defects (24, 25). The benefit of PARP inhibitors may also extend to patients with mCRPC without deficiencies in DDR pathways, as evidenced by the phase III results from the PROpel study, which demonstrated an improvement in PFS for patients without mutations in homologous recombination repair (HRR) genes (26).

We hypothesize that for patients with mCRPC, regardless of status of DNA repair mutational status, PARP inhibitors may prove effective when given in combination with agents that stimulate DNA damage. Given that Radium-223 creates high rates of DNA single-strand breaks as a mechanism for cell death, and PARP is primarily responsible for repair of such breaks (27) there is the potential for synergy between these agents when treating mCRPC. In addition, the potential for niraparib to further suppress the AR pathway, suggests another potential mechanism for synergy when combined with Radium-223. We believe these interactions could extend benefit to patients without somatic or germline DNA repair mutations.

We conducted a phase IB study to determine the MTD and recommended phase II dosing (RP2D) of niraparib combined with standard dosing of Radium-223 in the treatment of patients with metastatic mCRPC in men with and without prior chemotherapy exposure. Additional correlative studies were conducted to evaluate oncogenic genetic mutations at baseline and gene expression and pathway profile changes induced by therapy.

Study design and participants

This was a multicenter, phase IB dose escalation trial (ClinicalTrials.gov ID NCT03076203) that was approved by the institutional review boards at all participating institutions. Patients were provided written informed consent prior to starting on trial and enrollment followed international standards of good clinical practice. There were three dose levels of niraparib evaluated along with standard of care dosing of Radium-223 in two cohorts (chemotherapy naïve and prior chemotherapy) of men with mCRPC. The primary objective was to determine the MTD and RP2D of niraparib combined with Radium-223. The MTD was defined as the highest dose level at which the probability of a dose-limiting toxicity (DLT) was less than 20%.

Because the toxicity of radium-223 is often delayed, a time-to-event continuous reassessment method (TITE-CRM; ref. 28) design was used to identify the MTD based on toxicities observed over 12 weeks of treatment (three cycles of Radium-223). A DLT was defined as any treatment-related Grade ≥3 nonhematologic clinical that required medical intervention, or persisted for ≥7 days; any grade 4 hematologic toxicity with the exception of neutropenia Grade 4 lasting for <7 days and not associated with fever >38.5°C and/or infection; a dose interruption for a non-DLT laboratory abnormality lasting ≥14 days or a dose interruption for nonhematologic AE leading to <80% of an intended dose of niraparib being administered. Patients were followed from time of enrollment until progression of disease or death. The trial was conducted under the auspices of the Prostate Cancer Clinical Trials Consortium.

Eligible patients had histologic or cytologic diagnosis of adenocarcinoma of the prostate without neuroendocrine or small cell features, bone metastases detected by conventional bone scans and evidence of progressive disease after receiving at least 1 line of AR-targeted therapy in the hormone sensitive or castrate-resistant setting. Each patient was required to have serum testosterone levels in the castrate range (less than 50 ng/dL) and this was measured at time of screening. Patients were ineligible if they had: previously received a PARP inhibitor, more than one prior line of AR-targeted therapy or more than one chemotherapy agent. Patients who were previously identified to be carriers of a pathogenic germline or somatic mutation in BRCA-1 or BRCA-2 were excluded; however, additional testing to identify BRCA or other DDR pathway mutations was not conducted prior to enrollment. In addition, patients with bulky visceral disease (defined as >4 cm), brain or leptomeningeal disease or impending spinal cord compression were excluded.

Treatment

Patients were assigned to one of the three different treatment arms based using TITE-CRM (28) design, which allowed for three different dose levels of niraparib to be tested simultaneously with Radium-223. Subjects were given niraparib at doses of 100, 200, or 300 mg oral daily in combination with standard dosing of Radium-223, 55 kBq per kg body weight, given at 4-week intervals for a total of six injections. Following six cycles, subjects were to continue on niraparib alone until objective progression, intolerance of therapy, or withdrawal of consent. Enrollment was stratified by prior chemotherapy use and we aimed to enroll up to 30 patients total, with no more than 15 patients in each arm.

The TITE-CRM model was specifically chosen to improve the time to completion of the study, given that toxicities to Radium-223 are often delayed by several weeks. In the TITE-CRM, dose levels were assigned to each newly enrolled subject based on DLT data from subjects already enrolled on trial, which were weighted to account for the proportion of the observation period that each enrolled subject had been observed. Dose interruptions and reductions of niraparib were allowed for management of AEs per study protocol. The dose of Radium-223 was not adjusted, both agents were held in the event of a DLT and Radium-223 was not continued as a single agent in the event of niraparib discontinuation. Patients were removed from the study if they had an AE on the lowest dose of niraparib (100 mg) or if the subject required a dose interruption of more than 28 days. The study treatment also stopped for any patient who had clinical disease progression, intercurrent illness that made further treatment unsafe, developed myelodysplastic syndrome (MDS) or acute myeloid leukemia (AML), or withdrew of the consent.

Endpoints and clinical assessments

The primary outcome of this phase IB study was determination of the MTD and RP2D of niraparib combined with standard dosing of Radium-223 in the treatment of patients with mCRPC. Secondary endpoints included proportion of subjects with 50% prostate-specific antigen (PSA) reduction at 12 weeks, radiographic progression-free survival (rPFS) at 6 months and long-term safety and tolerability of treatment combination. AEs were defined by the NCI's Common Terminology Criteria for Adverse Events version 4.0. Subjects were re-assessed for any AEs weekly for the first 4 weeks, then bi-weekly for the next 8 weeks and finally monthly until disease progression or death. Any individual who was withdrawn from the study due to an AE was followed until event had stabilized or resolved. Disease status (response or progression) was assessed every 12 weeks and were determined using a combination of the revised RECIST v1.1 criteria and the guideline for prostate cancer endpoints developed by the Prostate Cancer Clinical Trials Working Group 3 (29, 30).

Statistical analysis

The TITE-CRM statistical design was used, assuming prior probabilities of toxicity for the three doses of 0.04, 0.08, and 0.12, with a target probability of toxicity for the optimal dose of 0.20, and a DLT evaluation time of 84 days. Calculation of the next dose to be assigned and the final toxicity probability estimates were performed using the SAS macro developed by the University of Michigan Comprehensive Cancer Center Biostatistics Unit (31). Patients were stratified within group by prior chemotherapy exposure and analyses were performed separately by strata. rPFS was estimated using the Kaplan–Meier method. Summary statistics were used to assess PSA change and to describe translational results. Time was calculated from day of first treatment until incidence of toxicity, progression, or death.

Exploratory correlative endpoints

Gene expression changes in whole blood samples at baseline and during treatment (cycle 1, day 15 and cycle 3, day 15) were evaluated in 18 of 30 patients. RNA from whole blood, collected from participants, was isolated using the Qiagen miRNA Isolation Kit (32) and was analyzed using the PanCancer Immune Pathways and PanCancer Driver Pathways panels, as described previously (33). The PanCaner Driver and Immune Panels included 770 genes each from 24 different driver pathways and 13 cancer associated canonical pathways, respectively.

In addition, plasma samples were collected at baseline in 15 patients and analyzed for circulating tumor DNA (ctDNA) using the Guardant 360 Liquid Biopsy platform (34). Each sample was assessed for different somatic and germline mutations including SNVs, insertions, deletions, gene fusions, and copy-number variants (CNV). For the 15 other participants, there were not viable samples to run such analysis.

Data availability

Data from this study is available through the Prostate Cancer Clinical Trials Consortium by emailing [email protected].

Patients

From May 2018 to January 2020 30 patients were enrolled at six different clinical sites in the United States and patient demographics are included in Table 1. Two patients were withdrawn from the study early. One patient withdrew shortly after randomization and never received a dose of either trial medication. One patient in the prior chemotherapy cohort at dose level 2 (Niraparib 200 mg) was removed from the study due prolonged thrombocytopenia. The remainder of the patients continued therapy until progression of disease or death. All patients were included in treatment analysis. The median age of patients was 69 years. Twenty-five patients were Caucasian, 5 patients identified as black or African American. For patients who had previously received chemotherapy, the median time from completion of chemotherapy to treatment start date was 86 weeks (range 21–193 weeks).

Table 1.

Patient, tumor, and treatment characteristics.

Study arm100 mg (N = 13)200 mg (N = 12)300 mg (N = 5)
Patient characteristics 
 Age (median, years) 71 66 73 
 ECOG performance status 
  0 6 (46%) 8 (67%) 3 (60%) 
  1 7 (54%) 4 (33%) 2 (40%) 
 Race 
  Caucasian 11 (85%) 10 (83%) 4 (80%) 
  African American 2 (15%) 2 (17%) 1 (20%) 
 Baseline hemoglobin (median, g/dL) 11.7 12.4 13.1 
 Baseline platelet count (median, B/L) 212 217 244 
Disease characteristics 
 Total Gleason score at diagnosis 
  6 3 (25%) 1 (20%) 
  7 3 (23%) 1 (8.3%) 1 (20%) 
  8–10 10 (76.9%) 8 (66.7%) 3 (60%) 
 Sites of metastases 
  Bone only 6 (46%) 6 (50%) 2 (40%) 
  Bone and lymph node only 3 (23%) 3 (25%) 3 (60%) 
  Visceral metastases 2 (15%) 2 (16%) 
Treatment characteristics 
 Prior prostatectomy 1 (8%) 2 (17%) 2 (40%) 
 Prior definitive radiotherapy 5 (38%) 3 (25%) 1 (20%) 
 Prior AR signaling inhibitors 
  Abiraterone 7 (54%) 8 (67%) 2 (40%) 
  Enzalutamide 6 (46%) 4 (33%) 2 (40%) 
 Prior chemotherapy 10 (77%) 5 (42%) 
  Docetaxel 9 (69%) 5 (42%) 
  Cabazitaxel 1 (8%) 
Study arm100 mg (N = 13)200 mg (N = 12)300 mg (N = 5)
Patient characteristics 
 Age (median, years) 71 66 73 
 ECOG performance status 
  0 6 (46%) 8 (67%) 3 (60%) 
  1 7 (54%) 4 (33%) 2 (40%) 
 Race 
  Caucasian 11 (85%) 10 (83%) 4 (80%) 
  African American 2 (15%) 2 (17%) 1 (20%) 
 Baseline hemoglobin (median, g/dL) 11.7 12.4 13.1 
 Baseline platelet count (median, B/L) 212 217 244 
Disease characteristics 
 Total Gleason score at diagnosis 
  6 3 (25%) 1 (20%) 
  7 3 (23%) 1 (8.3%) 1 (20%) 
  8–10 10 (76.9%) 8 (66.7%) 3 (60%) 
 Sites of metastases 
  Bone only 6 (46%) 6 (50%) 2 (40%) 
  Bone and lymph node only 3 (23%) 3 (25%) 3 (60%) 
  Visceral metastases 2 (15%) 2 (16%) 
Treatment characteristics 
 Prior prostatectomy 1 (8%) 2 (17%) 2 (40%) 
 Prior definitive radiotherapy 5 (38%) 3 (25%) 1 (20%) 
 Prior AR signaling inhibitors 
  Abiraterone 7 (54%) 8 (67%) 2 (40%) 
  Enzalutamide 6 (46%) 4 (33%) 2 (40%) 
 Prior chemotherapy 10 (77%) 5 (42%) 
  Docetaxel 9 (69%) 5 (42%) 
  Cabazitaxel 1 (8%) 

MTD

DLTs were evaluated in all three treatment groups and stratified by prior chemotherapy exposure (Table 2). There were no DLT events for patients who received the 100 mg dose of niraparib and no patients were removed from the study due to a treatment related adverse event. In the cohort that had never received chemotherapy, 7 patients received a 200 mg dose of niraparib and there was 1 DLT event, leading to an estimated probability of DLT at this dose level of 0.156 with a 95% confidence interval of 0.042 to 0.385. There were two DLT events in the 5 patients who received a 300 mg dose of niraparib, leading to an estimated probability of DLT for this cohort of 0.214. Because the probability of DLT was greater than 0.2 in the 300 mg cohort, the 200 mg dose of niraparib was confirmed as the MTD and RP2D in chemotherapy naïve patients. There were three DLT events in 5 patients who had previously received chemotherapy for the 200 mg treatment group, leading to an unacceptable DLT probability and a recommendation of 100 mg niraparib as the MTD and RP2D for patients with prior chemotherapy exposure.

Table 2.

Probabilities of dose-limiting toxicities.

Chemotherapy naïve
Dose levelPatients treatedDLTsDLT probability
100 mg 0.089 (0.019–0.281) 
200 mg 0.156 (0.042–0.385) 
300 mg 0.214 (0.068–0.455) 
Chemotherapy naïve
Dose levelPatients treatedDLTsDLT probability
100 mg 0.089 (0.019–0.281) 
200 mg 0.156 (0.042–0.385) 
300 mg 0.214 (0.068–0.455) 
Prior chemotherapy exposure
DosePatients treatedDLTsDLT probability
100 mg 10 0.127 (0.029–0.352) 
200 mg 0.207 (0.061–0.455) 
300 mg 0.272 (0.095–0.521) 
Prior chemotherapy exposure
DosePatients treatedDLTsDLT probability
100 mg 10 0.127 (0.029–0.352) 
200 mg 0.207 (0.061–0.455) 
300 mg 0.272 (0.095–0.521) 

Safety

There were 297 recorded adverse events that occurred during the trial, 153 of which were deemed attributable to study medications (Table 3). Most commonly, low-grade disturbances to the gastrointestinal system occurred: with nausea (60%), diarrhea (30%), and constipation (27%) observed most frequently. Constitutional complaints such as fatigue (53%) and decreased appetite (27%) were also frequently observed. Significant hematologic toxicities were encountered infrequently, with only 3 patients with grade 3 or higher neutropenia. The average time to neutrophil recovery was 5.5 days, whereas the average time to platelet recovery was 18 days. There were no deaths considered related to study drug. One patient died secondary to widespread progression of disease and this event was not considered to be related to either study medication.

Table 3.

Most frequently observed treatment-related adverse events.

Prior chemotherapy exposureChemotherapy naïve
Dose level100 mg200 mg100 mg200 mg300 mg
Patients105375
Max Grade 2 TRAE 
Hematologic 
 Anemia 1 (10%) 1 (20%) — 1 (14%) 1 (20%) 
 Thrombocytopenia 2 (20%) 1 (20%) — 2 (29%) — 
 Neutropenia 2 (20%) 1 (20%) — — — 
Gastrointestinal 
 Diarrhea 2 (20%) 1 (20%) — 3 (43%) 3 (60%) 
 Constipation 3 (30%) — — 2 (29%) 3 (60%) 
 Dry mouth 1 (10%) — — 2 (29%) 1 (20%) 
 Nausea 4 (40%) 4 (80%) 1 (33%) 5 (71%) 3 (60%) 
 Vomiting — 1 (20%)  2 (29%) 1 (20%) 
Other 
 Hypertension 2 (20%) — 1 (33%) — — 
 Fatigue 4 (40%) 3 (60%) — 3 (43%) 5 (100%) 
 Decrease appetite 3 (30%) 1 (20%) 2 (67%) 2 (29%) — 
Max Grade 4 TRAE 
Hematologic 
 Anemia — 1 (20%) — 1 (14%) 1 (20%) 
 Thrombocytopenia — 1 (20%) — 1 (14%) — 
 Neutropenia — 2 (40%) — — 1 (20%) 
Gastrointestinal 
 Nausea — — — — 1 (20%) 
Other 
 Hypertension 1 (10%) 1 (20%) 1 (33%) — 1 (20%) 
Prior chemotherapy exposureChemotherapy naïve
Dose level100 mg200 mg100 mg200 mg300 mg
Patients105375
Max Grade 2 TRAE 
Hematologic 
 Anemia 1 (10%) 1 (20%) — 1 (14%) 1 (20%) 
 Thrombocytopenia 2 (20%) 1 (20%) — 2 (29%) — 
 Neutropenia 2 (20%) 1 (20%) — — — 
Gastrointestinal 
 Diarrhea 2 (20%) 1 (20%) — 3 (43%) 3 (60%) 
 Constipation 3 (30%) — — 2 (29%) 3 (60%) 
 Dry mouth 1 (10%) — — 2 (29%) 1 (20%) 
 Nausea 4 (40%) 4 (80%) 1 (33%) 5 (71%) 3 (60%) 
 Vomiting — 1 (20%)  2 (29%) 1 (20%) 
Other 
 Hypertension 2 (20%) — 1 (33%) — — 
 Fatigue 4 (40%) 3 (60%) — 3 (43%) 5 (100%) 
 Decrease appetite 3 (30%) 1 (20%) 2 (67%) 2 (29%) — 
Max Grade 4 TRAE 
Hematologic 
 Anemia — 1 (20%) — 1 (14%) 1 (20%) 
 Thrombocytopenia — 1 (20%) — 1 (14%) — 
 Neutropenia — 2 (40%) — — 1 (20%) 
Gastrointestinal 
 Nausea — — — — 1 (20%) 
Other 
 Hypertension 1 (10%) 1 (20%) 1 (33%) — 1 (20%) 

Clinical outcome

PSA data were recorded for 28 of 30 patients. Of these, 9 patients had a PSA decline, of whom 3 had a PSA decline of >50% (Supplementary Fig. S1). For five participants, the PSA assessment was made prior to the 12th week, given that progressive disease had already occurred.

The median rPFS for all patients included in analysis was 7.1 months (CI, 2.7–9.3 months) with an estimated 6-month rPFS of 51% (CI, 31–67%). For patients who were chemotherapy naïve (Fig. 1A), the median rPFS was 2.9 months (CI, 2.5–10.8 months) and for patients with prior chemotherapy exposure the median rPFS was 8.0 months (CI, 2.7–10.3 months). The estimated 6 month rPFS for these groups was 47% (CI, 21–69%) and 54% (CI, 23–77%). rPFS distributions were similar by dose overall (Fig. 1B) and in chemotherapy naïve (Fig. 1C) and prior chemotherapy cohorts (Fig. 1D).

Figure 1.

rPFS. A, Overall rPFS stratified by prior chemotherapy exposure. B, Overall rPFS stratified by dose cohort. C, rPFS of the chemotherapy naïve cohort, stratified by dose cohort. D, rPFS of the prior chemotherapy exposed cohort stratified by dose cohort.

Figure 1.

rPFS. A, Overall rPFS stratified by prior chemotherapy exposure. B, Overall rPFS stratified by dose cohort. C, rPFS of the chemotherapy naïve cohort, stratified by dose cohort. D, rPFS of the prior chemotherapy exposed cohort stratified by dose cohort.

Close modal

Correlative endpoints

We performed gene expression profiling in total RNA from blood obtained from patients at the stated time points, with the hypothesis that potential differences in gene expression would be observed between patients experiencing longer duration on treatment as a surrogate marker for response. Indeed, several intriguing changes in gene expression profiles between patients based on treatment duration (median 21 weeks duration for the 18 patients that had viable RNA available for analysis) were detected. At baseline, paired box protein 5 (PAX5) and CD-19 expression were higher in the group that was on treatment for longer than 21 weeks in comparison with patients that stayed on treatment for less than 21 weeks (Fig. 2A). By cycle 3 day 15, expression of both PAX5 (linear fold change = −5.08) and CD19 (linear fold change = −3.82) was more attenuated in the longer treatment duration group in comparison with the shorter treatment duration group.

Figure 2.

Gene expression differences in blood from enrolled patients with treatment duration greater or less than median 21 weeks. A, CD19 and PAX5 expression appears to be higher at baseline and decrease with treatment in patients treated longer than median 21 weeks compared with patients treated less than 21 weeks. B, IL1R2, ARG1, and FLT3 expression increases over time in patients with duration of treatment shorter than median 21 weeks but does not in patients with treatment duration longer than median 21 weeks.

Figure 2.

Gene expression differences in blood from enrolled patients with treatment duration greater or less than median 21 weeks. A, CD19 and PAX5 expression appears to be higher at baseline and decrease with treatment in patients treated longer than median 21 weeks compared with patients treated less than 21 weeks. B, IL1R2, ARG1, and FLT3 expression increases over time in patients with duration of treatment shorter than median 21 weeks but does not in patients with treatment duration longer than median 21 weeks.

Close modal

In contrast, gene expression patterns for IL-1R2, Arginase 1 (ARG1), and FMS like tyrosine kinase 3 (FLT3) increased in the group treated for less than 21 weeks in comparison with the group treated for longer than 21 weeks (Fig. 2B). Specifically, by cycle 3 day 15, ARG1 (linear fold change = 3.32), FLT3 (linear fold change = 3.32), and IL-1R2 (linear fold change = 2.80) had all markedly increased in patients who experienced a relatively faster time to progression. These changes in gene expression are intriguing and need further investigation in future studies.

ctDNA was detected in 14 of 15 samples collected at baseline. Of these, seven had AR amplification and four instances of point mutations within the gene were detected (Table 4). Somatic mutations in TP53 and PIK3CA were detected in 7 and 4 patients respectively as well. Deletions in RB1 and BRCA2 were also detected via copy-number variations in 7 and 4 patients, respectively. There were six mutations found in BRCA2, two of which were germline SNVs, and one mutation found in ATM and CDK12 genes, respectively. Notably, the median time to progression for 7 patients with HRR variants was only 21 weeks; however, the median time to progression for the 2 patients with germline BRCA2 mutations was 43 weeks, significantly longer than the median duration for the entire population.

Table 4.

Baseline ctDNA results for samples with relevant co-occurring alterations.

Patient numberSNV/InDelCNVDose and cohortTime to progression (weeks)
CDK12 K480  100 mg - C 19 
 CDK12 L867P    
 AR L702H    
 AR amplification 200 mg - C 30 
  RB1 homozygous del   
PIK3CA N345K BRCA2 LOH deletion 200 mg - N 13 
 ATM M2405V RB1 deletion   
 AR L702H AR amplification   
 AR amplification 200 mg - N 31 
  BRCA2 homozygous del   
  RB1 LOH deletion   
PIK3CA N345K PIK3CA amplification 100 mg - N 15 
  AR amplification   
  RB1 homozygous del   
AR W742C RB1 homozygous del 100 mg - N 32 
 EGFR D837N EGFR amplification   
AR F877L ATM LOH deletion 100 mg - N 21 
 PIK3CA H1047R BRCA2 LOH deletion   
  RB1 deletion   
BRAF K601E  300 mg - N 38 
 AR T878A    
 BRCA2 H1003fs (g)    
 BRCA2 Q2859fs    
 RB1 L572fs    
 BRCA2 deletion 200 mg - C 12 
  RB1 LOH deletion   
  PIK3CA amplification   
  AR amplification   
10 BRCA2 c682–1G>A  100 mg - C 49 
 BRCA2 K2849fs (g)    
11 PIK3CA H1047R AR amplification 100 mg - C 18 
Patient numberSNV/InDelCNVDose and cohortTime to progression (weeks)
CDK12 K480  100 mg - C 19 
 CDK12 L867P    
 AR L702H    
 AR amplification 200 mg - C 30 
  RB1 homozygous del   
PIK3CA N345K BRCA2 LOH deletion 200 mg - N 13 
 ATM M2405V RB1 deletion   
 AR L702H AR amplification   
 AR amplification 200 mg - N 31 
  BRCA2 homozygous del   
  RB1 LOH deletion   
PIK3CA N345K PIK3CA amplification 100 mg - N 15 
  AR amplification   
  RB1 homozygous del   
AR W742C RB1 homozygous del 100 mg - N 32 
 EGFR D837N EGFR amplification   
AR F877L ATM LOH deletion 100 mg - N 21 
 PIK3CA H1047R BRCA2 LOH deletion   
  RB1 deletion   
BRAF K601E  300 mg - N 38 
 AR T878A    
 BRCA2 H1003fs (g)    
 BRCA2 Q2859fs    
 RB1 L572fs    
 BRCA2 deletion 200 mg - C 12 
  RB1 LOH deletion   
  PIK3CA amplification   
  AR amplification   
10 BRCA2 c682–1G>A  100 mg - C 49 
 BRCA2 K2849fs (g)    
11 PIK3CA H1047R AR amplification 100 mg - C 18 

Note: C, prior chemotherapy; N, chemotherapy naïve; CNV, copy-number variant; InDel, insertion/deletion variant; LOH, loss of heterozygosity; del, deletion; fs, frameshift; (g), germline.

This multicenter phase IB trial was the first to assess the combination of a PARP inhibitor with Radium-223 in patients with mCRPC without known BRCA mutations. The combination of 200 mg of niraparib with standard dosing of Radium-223 was found to be tolerable in a subset of patients who were chemotherapy naïve, whereas a dose of 100 mg niraparib was tolerable for patients who had experienced prior treatment with taxane therapy. Currently, the combination of olaparib with Radium-223 is under investigation and investigators used a 3+3 trial design to evaluate 12 patients at two different dose levels of olaparib, determining 200 mg twice daily as their R2PD (35). Importantly, our study demonstrates the feasibility and utility of the TITE-CRM statistical design when investigating therapeutics that have a delayed toxicity profile like radio-ligands. We were able to safely evaluate 30 patients over three different dose levels simultaneously, which accelerated trial accrual without exposing participants to excess risk. This trial can serve as a framework for the development of other combined modality therapies using radio-ligands, which may prove pivotal with the recent approval of Lu 177 vipivotide tetraxetan for patients with mCRPC (36).

By downregulating several DNA repair pathways, PARP inhibitors have gained interest as radiosensitizers in many different cancer models and recent studies have demonstrated safety of PARP inhibitors with other radiation modalities (37–39). Our study adds to these findings as we were able to show tolerability of 200 mg of niraparib with standard of care dosing of Radium-223 in patients without prior chemotherapy exposure. The majority of adverse events related to therapy were either grade 1 or 2, with nausea, diarrhea or constipation and fatigue reported as the most common symptoms. Despite potential overlapping toxicities to the bone marrow, we did not observe significant issues with impaired hematopoiesis and there were few grade 3 events of cytopenia. In all but one instance, the cytopenia resolved after holding niraparib. There were no instances of grade 3 infections and there were no deaths attributable to study medications.

Currently, niraparib is approved as a monotherapy for treatment of ovarian cancer at 300 mg daily and studies are ongoing in biomarker selected patients with metastatic prostate cancer at 200 mg daily in combination with abiraterone acetate 1,000 mg. In regards to combination with radiotherapy, studies evaluating Olaparib in lung and glioblastoma have concluded that, when combined with radiation, the MTD of Olaparib needs to be reduced from the recommend monotherapy dose to limit hematologic toxicity (37, 40). In addition, trials are currently enrolling that are testing reduced doses, 100 or 200 mg, of Niraparib in combination with radiotherapy for high-risk early-stage prostate cancer (41, 42). Recently, de Haan and colleagues were able to show that a 25 mg dose of Olaparib was able to effectively reduce baseline poly ADP-ribose (PAR) levels along with radiation induced PARylation in lung tumor tissue and peripheral blood mononuclear cells, concluding that a reduced dose of a PARP inhibitor still has biologic activity in combination with radiotherapy (37). Taken together, our findings that using either 100 or 200 mg doses of Niraparib in combination with radiotherapy are not likely to drastically reduce the clinical activity of the PARP inhibitor.

Our translational studies demonstrate that using the NanoString platform to perform gene expression profiling on whole blood samples was feasible. Although one limitation to testing whole blood is the uncertainty of the source of RNA, whether it is tumor-derived versus from normal cells (43).

We observed differences in PAX5 and CD19 expression when comparing patients who had a response to therapy to those without a response. At baseline, RNA transcripts were higher for both of these targets in responders compared with nonresponders and a statistically significant reduction in mRNA level was observed with treatment in those who remained on study for longer than 21 weeks. PAX5 is an oncogene that encodes a potent transcription factor leading to B-cell development and aberrant expression has been observed frequently with development of aggressive B-cell malignancies (44). CD19, a cell surface marker expressed on all B lymphocytes, is critical for cell function and CD19 expression is in part regulated by PAX5 (45). In addition, significant PAX5 expression has also been demonstrated in nonlymphoid malignancies, including breast, small cell lung cancer, and pediatric tumors and is believed to play a role in cell survival and metastatic potential (44). One downstream target of PAX5 is c-MET, a tyrosine kinase receptor for which mutations and aberrant expression are known promote oncogenesis (46).

B cells in the tumor microenvironment can promote tumorigenesis and increase metastatic potential by causing antibody mediated T-cell suppression, promoting lymphangiogenesis and producing immunomodulatory cytokines that suppress the antitumor immune response (47). In prostate cancer, it has been observed that B-cell infiltration is higher in high-grade tumors and in tumors that eventually recurred or progressed in comparison with low or intermediate grade tumors (48). In addition, infiltrating B cells were shown to support castrate resistant cell growth by secreting lymphotoxin, which lead to cell survival (49).

ARG1 expression was similar at baseline when comparing patients who responded to therapy to those who did not; however, in the patients who did not respond to therapy, by cycle 3 day 15, ARG1 expression was nearly four times higher. These suggest that expression of ARG1 could possibly serve as a protective mechanism, limiting the effect of treatment. ARG1 expression by tumor-associated macrophages (TAM) has been demonstrated as a mechanism to suppress the effect of infiltrating T cells by limiting the amount of L-arginine available for T-cell function (50). Interestingly, we observed expansion of FLT3 transcripts mirrored ARG1 in patients who had relatively faster time to progression. FLT3 critically drives dendritic cell maturation and activity in the tumor microenvironment. This may either lead to an amplified immune response, through increased antigen presentation to infiltrating cytotoxic T cells, or diminished local immune response, by expansion of T regulatory cells (Treg; ref. 51).

With these hypothesis-generating observations in whole blood RNA expression, a speculative mechanism for the effect of niraparib with Radium-223 may be through the immune response within the tumor microenvironment. In patients who stayed on treatment for longer than 21 weeks we observed changes indicative of decreased B-cell function which may allow for increased immune activity within the microenvironment. Conversely, resistance to therapy may be mediated through decreased local T-cell activity, by increased ARG1 expression by TAMs and FLT3 expression driving Treg expansion. Further investigation is needed to explore this phenomenon, but if the mechanism is valid, one could speculate that adding therapies to improve T-cell-mediated immune response could augment this therapeutic approach. Notably, recent phase III trials have reported improved outcomes in mCRPC when combining PARP inhibitors with abiraterone acetate plus prednisone in those with germline/somatic homologous recombination defects, with potentially a less robust benefit in unselected patients (25, 52). Trials are also evaluating the combination of PARP inhibitors with other androgen inhibitors, immune checkpoint inhibitors, and Lu-PSMA-617 (53–55). Thus, the optimal combination partner remains to be determined.

Our exploratory analysis of baseline ctDNA discovered 4 patients with somatic BRCA variants and 2 patients that had germline BRCA2 mutations. These 2 patients were in the 300 and 100 mg cohorts and had dramatically longer times to progression of 38 and 49 weeks, respectively. In addition, the patient in the 100 mg cohort had previously received chemotherapy. These outcomes are concordant with prior observations of favorable responses to Radium-223 in patients with known BRCA mutations (56–58), in addition to the apparent effect of PARP inhibitors in this population. Given the known efficacy of PARP inhibitors in patients with germline/somatic BRCA mutations, we intentionally excluded patients with prior record of these alterations at trial enrollment. However, this expected finding highlights the impact of ctDNA analysis and further demonstrates the importance of screening patients for molecular biomarkers before choosing therapies.

This study was not powered nor designed to evaluated efficacy, but we were able to collect data on PSA change and rPFS to help describe trends. PSA increased in the majority of patients on study; however, rapid PSA response is not a hallmark of Radium-223 therapy, as Radium-223 can cause an early PSA flare in patients who are responding to therapy (59, 60). Nearly 30% of patients had a PSA decline, however only 10% had a PSA decline of >50% at week 12. Three of the 9 patients with a PSA decline were found to have BRCA mutations on ctDNA; 5 of the remaining 6 patients did not have blood samples evaluated for ctDNA. In regards to rPFS, there did not appear to be a statistically significant difference between patients who had prior chemotherapy exposure and those who were chemotherapy naïve, with wide overlapping confidence intervals ranging from 2.5 to 10.8 months.

In conclusion, niraparib in combination with Radium-223 was demonstrated to be safe in combination for the treatment of mCRPC for patients who have previously progressed on AR therapies. We intend to continue investigation with a prospective phase II trial that will better evaluate efficacy endpoints and assess potential long-term toxicities and will serve as a companion to the ongoing trial of olaparib with Radium-223 in unselected patients with mCRPC (35).

B. Leiby reports grants from NCI during the conduct of the study as well as personal fees from Bayer Healthcare outside the submitted work. G. Sonpavde reports personal fees from Janssen during the conduct of the study as well as grants from Sanofi, Helsinn, and Jazz; grants and personal fees from AstraZeneca, Gilead, and EMD Serono/Pfizer; personal fees and nonfinancial support from Lucence; personal fees from Seagen/Astellas, Merck, Bicycle Therapeutics, G1 Therapeutics, Eli Lilly/Loxo, Immunovaccine Technology (IMV), Elsevier Practiceupdate, Mereo, Vial, and Suba Therapeutics; other support from Myriad; and nonfinancial support from BMS outside the submitted work. A.D. Choudhury reports grants and personal fees from Bayer and personal fees from Janssen during the conduct of the study as well as personal fees from Pfizer, Astellas, Eli Lilly, AstraZeneca, Clovis, Dendreon, and Blue Earth outside the submitted work. C. Sweeney reports grants and personal fees from Janssen, Astellas, Pfizer, Bayer, Lilly, AstraZeneca, and CellCentric outside the submitted work. D. Einstein reports grants from Bristol-Myers Squibb, Puma Biotechnology, Cardiff Oncology, and MiNK Therapeutics and other support from Foundation Medicine outside the submitted work. R. Szmulewitz reports personal fees from Janssen and Astra Zeneca and grants and personal fees from Astellas outside the submitted work. O. Sartor reports grants and personal fees from Advanced Accelerator Applications, AstraZeneca, Bayer, Constellation, Janssen, Merck, Progenics, and Tenebio; personal fees from Astellas, Blue Earth Diagnostics, Inc., Bavarian Nordic, Bristol Myers Squibb, Clarity Pharmaceuticals, Clovis, EMD Serono, Dendreon, Fusion, Isotopen Technologien Meunchen, Moyvant, Myriad, Noria Therapeutics, Noxopharm, Novartis, Northstar, POINT Biopharma, Pfizer, Sanofi, Telix, and Theragnostics; and grants from Amgen, Endocyte, Invitae, and Lantheus outside the submitted work. K. Knudsen reports grants from NCI and Prostate Cancer Foundation during the conduct of the study as well as the following relationships over the past 3 years: research support (Novartis, CellCentric) and consultancy/advisory (CellCentric, Sanofi, Celgene, Janssen, Genentech). E.S.-H. Yang reports personal fees from Clovis, Bayer, and AstraZeneca and grants from Eli Lilly and PUMA outside the submitted work. W.K. Kelly reports other support from Janssen and Bayer during the conduct of the study as well as nonfinancial support from Janssen and Bayer outside the submitted work. No disclosures were reported by the other authors.

Z. Quinn: Formal analysis, writing–original draft, writing–review and editing. B. Leiby: Data curation, formal analysis, methodology. G. Sonpavde: Conceptualization, investigation. A.D. Choudhury: Conceptualization, supervision, investigation. C. Sweeney: Investigation. D. Einstein: Investigation, writing–review and editing. R. Szmulewitz: Conceptualization. O. Sartor: Conceptualization, formal analysis, investigation, writing–review and editing. K. Knudsen: Conceptualization, methodology, writing–review and editing. E.S.-H. Yang: Conceptualization, formal analysis, investigation. W.K. Kelly: Conceptualization, formal analysis, supervision, investigation, writing–review and editing.

The authors would like to thank Ms. Deborah Della Manna and Dr. Jianqing Zhang for performing the NanoString experiments. In addition, we thank Jake Vinson, Garrett Abrams, Sarah Wise, and other members of the Prostate Cancer Clinical Trials Consortium for their ongoing support.

The publication costs of this article were defrayed in part by the payment of publication fees. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/).

1.
Cancer Stat Facts: Prostate Cancer
.
National Cancer Institute: Surveillance, Epidemiology, and End Results Program
;
2021
.
2.
Dai
C
,
Heemers
H
,
Sharifi
N
.
Androgen signaling in prostate cancer
.
Cold Spring Harb Perspect Med
2017
;
7
.
3.
Scher
HI
,
Sawyers
CL
.
Biology of progressive, castration-resistant prostate cancer: directed therapies targeting the androgen-receptor signaling axis
.
J Clin Oncol
2005
;
23
:
8253
61
.
4.
Berthold
DR
,
Pond
GR
,
Soban
F
,
de Wit
R
,
Eisenberger
M
,
Tannock
IF
.
Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer: updated survival in the TAX 327 study
.
J Clin Oncol
2008
;
26
:
242
5
.
5.
Eisenberger
M
,
Hardy-Bessard
AC
,
Kim
CS
,
Geczi
L
,
Ford
D
,
Mourey
L
, et al
.
Phase III study comparing a reduced dose of cabazitaxel (20 mg/m(2)) and the currently approved dose (25 mg/m(2)) in postdocetaxel patients with metastatic castration-resistant prostate cancer-PROSELICA
.
J Clin Oncol
2017
;
35
:
3198
206
.
6.
Ryan
CJ
,
Smith
MR
,
de Bono
JS
,
Molina
A
,
Logothetis
CJ
,
de Souza
P
, et al
.
Abiraterone in metastatic prostate cancer without previous chemotherapy
.
N Engl J Med
2013
;
368
:
138
48
.
7.
Beer
TM
,
Armstrong
AJ
,
Rathkopf
D
,
Loriot
Y
,
Sternberg
CN
,
Higano
CS
, et al
.
Enzalutamide in men with chemotherapy-naive metastatic castration-resistant prostate cancer: extended analysis of the phase 3 PREVAIL study
.
Eur Urol
2017
;
71
:
151
4
.
8.
Kantoff
PW
,
Higano
CS
,
Shore
ND
,
Berger
ER
,
Small
EJ
,
Penson
DF
, et al
.
Sipuleucel-T immunotherapy for castration-resistant prostate cancer
.
N Engl J Med
2010
;
363
:
411
22
.
9.
Hansen
AR
,
Massard
C
,
Ott
PA
,
Haas
NB
,
Lopez
JS
,
Ejadi
S
, et al
.
Pembrolizumab for advanced prostate adenocarcinoma: findings of the KEYNOTE-028 study
.
Ann Oncol
2018
;
29
:
1807
13
.
10.
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
102
.
11.
Abida
W
,
Patnaik
A
,
Campbell
D
,
Shapiro
J
,
Bryce
AH
,
McDermott
R
, et al
.
Rucaparib in men with metastatic castration-resistant prostate cancer harboring a BRCA1 or BRCA2 gene alteration
.
J Clin Oncol
2020
;
38
:
3763
72
.
12.
Parker
C
,
Nilsson
S
,
Heinrich
D
,
Helle
SI
,
O'Sullivan
JM
,
Fossa
SD
, et al
.
Alpha emitter radium-223 and survival in metastatic prostate cancer
.
N Engl J Med
2013
;
369
:
213
23
.
13.
Hodgkins
PS
,
O'Neil
P
,
Stevens
D
,
Fairman
MP
.
The severity of alpha-particle-induced DNA damage is revealed by exposure to cell-free extracts
.
Radiat Res
1996
;
146
:
660
7
.
14.
Morris
MJ
,
Corey
E
,
Guise
TA
,
Gulley
JL
,
Kevin Kelly
W
,
Quinn
DI
, et al
.
Radium-223 mechanism of action: implications for use in treatment combinations
.
Nature Reviews Urology
2019
;
16
:
745
56
.
15.
Nilsson
S
.
Alpha-emitter radium-223 in the management of solid tumors: current status and future directions
.
Am Soc Clin Oncol Educ Book
2014
(
34
):
e132
e9
.
16.
Rose
M
,
Burgess
JT
,
O'Byrne
K
,
Richard
DJ
,
BEPARP
Inhibitors
:
Clinical relevance, mechanisms of action and tumor resistance
.
Front Cell Dev Biol
2020
;
8
:
564601
.
17.
Masson
M
,
Niedergang
C
,
Schreiber
V
,
Muller
S
,
Menissier-de Murcia
J
,
de Murcia
G
.
XRCC1 is specifically associated with poly(ADP-ribose) polymerase and negatively regulates its activity following DNA damage
.
Mol Cell Biol
1998
;
18
:
3563
71
.
18.
Polkinghorn
WR
,
Parker
JS
,
Lee
MX
,
Kass
EM
,
Spratt
DE
,
Iaquinta
PJ
, et al
.
Androgen receptor signaling regulates DNA repair in prostate cancers
.
Cancer Discov
2013
;
3
:
1245
53
.
19.
Li
L
,
Chang
W
,
Yang
G
,
Ren
C
,
Park
S
,
Karantanos
T
, et al
.
Targeting poly(ADP-ribose) polymerase and the c-Myb-regulated DNA damage response pathway in castration-resistant prostate cancer
.
Sci Signal
2014
;
7
:
ra47
.
20.
Goodwin
JF
,
Schiewer
MJ
,
Dean
JL
,
Schrecengost
RS
,
de Leeuw
R
,
Han
S
, et al
.
A hormone-DNA repair circuit governs the response to genotoxic insult
.
Cancer Discov
2013
;
3
:
1254
71
.
21.
Jones
P
,
Altamura
S
,
Boueres
J
,
Ferrigno
F
,
Fonsi
M
,
Giomini
C
, et al
.
Discovery of 2-{4-[(3S)-piperidin-3-yl]phenyl}-2H-indazole-7-carboxamide (MK-4827): a novel oral poly(ADP-ribose)polymerase (PARP) inhibitor efficacious in BRCA-1 and -2 mutant tumors
.
J Med Chem
2009
;
52
:
7170
85
.
22.
Moore
KN
,
Secord
AA
,
Geller
MA
,
Miller
DS
,
Cloven
N
,
Fleming
GF
, et al
.
Niraparib monotherapy for late-line treatment of ovarian cancer (QUADRA): a multicentre, open-label, single-arm, phase 2 trial
.
Lancet Oncol
2019
;
20
:
636
48
.
23.
Gonzalez-Martin
A
,
Pothuri
B
,
Vergote
I
,
DePont Christensen
R
,
Graybill
W
,
Mirza
MR
, et al
.
Niraparib in patients with newly diagnosed advanced ovarian cancer
.
N Engl J Med
2019
;
381
:
2391
402
.
24.
Smith
MR
,
Scher
HI
,
Sandhu
S
,
Efstathiou
E
,
Lara
PN
Jr
,
Yu
EY
, et al
.
Niraparib in patients with metastatic castration-resistant prostate cancer and DNA repair gene defects (GALAHAD): a multicentre, open-label, phase 2 trial
.
Lancet Oncol
2022
;
23
:
362
73
.
25.
Chi
KN
,
Rathkopf
DE
,
Smith
MR
,
Efstathiou
E
,
Attard
G
,
Olmos
D
, et al
.
Phase 3 MAGNITUDE study: First results of niraparib (NIRA) with abiraterone acetate and prednisone (AAP) as first-line therapy in patients (pts) with metastatic castration-resistant prostate cancer (mCRPC) with and without homologous recombination repair (HRR) gene alterations
.
J Clin Oncol
2022
;
40
(
6_suppl
):
12
.
26.
Clarke
NW
,
Armstrong
AJ
,
Thiery-Vuillemin
A
,
Oya
M
,
Shore
N
,
Loredo
E
, et al
.
Abiraterone and olaparib for metastatic castration-resistant prostate cancer
.
NEJM Evidence
;
0
:
EVIDoa2200043
.
27.
Zheng
F
,
Zhang
Y
,
Chen
S
,
Weng
X
,
Rao
Y
,
Fang
H
.
Mechanism and current progress of Poly ADP-ribose polymerase (PARP) inhibitors in the treatment of ovarian cancer
.
Biomed Pharmacother
2020
;
123
:
109661
.
28.
Normolle
D
,
Lawrence
T
.
Designing dose-escalation trials with late-onset toxicities using the time-to-event continual reassessment method
.
J Clin Oncol
2006
;
24
:
4426
33
.
29.
Eisenhauer
EA
,
Therasse
P
,
Bogaerts
J
,
Schwartz
LH
,
Sargent
D
,
Ford
R
, et al
.
New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1)
.
Eur J Cancer
2009
;
45
:
228
47
.
30.
Scher
HI
,
Morris
MJ
,
Stadler
WM
,
Higano
C
,
Basch
E
,
Fizazi
K
, et al
.
Trial design and objectives for castration-resistant prostate cancer: updated recommendations from the prostate cancer clinical trials working group 3
.
J Clin Oncol
2016
;
34
:
1402
18
.
31.
Biostatistics TCFC
2009 TITE-CRM RESOURCES. University of Michigan School of Public Health
.
<
https://sph.umich.edu/ccb/tite-resources.html>.
33.
Yang
ES
,
Nassar
AH
,
Adib
E
,
Jegede
OA
,
Alaiwi
SA
,
Manna
DLD
, et al
.
Gene expression signature correlates with outcomes in metastatic renal cell carcinoma patients treated with everolimus alone or with a vascular disrupting agent
.
Mol Cancer Ther
2021
;
20
:
1454
61
.
34.
Inc GH
.
Guardant360 CDx Gene List
.
2021
.
35.
McKay
RR
,
Xie
W
,
Ajmera
A
,
Saraiya
B
,
Parikh
M
,
Folefac
E
, et al
.
A phase 1/2 study of olaparib and radium-223 in men with metastatic castration-resistant prostate cancer (mCRPC) with bone metastases (COMRADE): results of the phase 1 study
.
J Clin Oncol
2021
;
39
:
e17020
.
36.
2022 Novartis Pluvicto™ approved by FDA as first targeted radioligand therapy for treatment of progressive, PSMA positive metastatic castration-resistant prostate cancer
. <https://www.novartis.com/news/media-releases/novartis-pluvictotm-approved-fda-first-targeted-radioligand-therapy-treatment-progressive-psma-positive-metastatic-castration-resistant-prostate-cancer>.
37.
de Haan
R
,
van den Heuvel
MM
,
van Diessen
J
,
Peulen
HMU
,
van Werkhoven
E
,
de Langen
AJ
, et al
.
Phase I and pharmacologic study of olaparib in combination with high-dose radiotherapy with and without concurrent cisplatin for non-small cell lung cancer
.
Clin Cancer Res
2021
;
27
:
1256
66
.
38.
Karam
SD
,
Reddy
K
,
Blatchford
PJ
,
Waxweiler
T
,
DeLouize
AM
,
Oweida
A
, et al
.
Final report of a phase I trial of olaparib with cetuximab and radiation for heavy smoker patients with locally advanced head and neck cancer
.
Clin Cancer Res
2018
;
24
:
4949
59
.
39.
Chabot
P
,
Hsia
TC
,
Ryu
JS
,
Gorbunova
V
,
Belda-Iniesta
C
,
Ball
D
, et al
.
Veliparib in combination with whole-brain radiation therapy for patients with brain metastases from non-small cell lung cancer: results of a randomized, global, placebo-controlled study
.
J Neurooncol
2017
;
131
:
105
15
.
40.
Chalmers
AJ
,
Short
S
,
Watts
C
,
Herbert
C
,
Morris
A
,
Stobo
J
, et al
.
Phase I clinical trials evaluating olaparib in combination with radiotherapy (RT) and/or temozolomide (TMZ) in glioblastoma patients: results of OPARATIC and PARADIGM phase I and early results of PARADIGM-2
.
J Clin Oncol
2018
;
36
(
15_suppl
):
2018
-.
41.
A multi-center trial of androgen suppression with abiraterone acetate, leuprolide, PARP inhibition and stereotactic body radiotherapy in prostate cancer
. https://ClinicalTrials.gov/show/NCT04194554.
42.
Niraparib with standard combination radiation therapy and androgen deprivation therapy in treating patients with high risk prostate cancer
. https://ClinicalTrials.gov/show/NCT04037254.
43.
Jensen
K
,
Konnick
EQ
,
Schweizer
MT
,
Sokolova
AO
,
Grivas
P
,
Cheng
HH
, et al
.
Association of clonal hematopoiesis in DNA repair genes with prostate cancer plasma cell-free DNA testing interference
.
JAMA Oncol
2021
;
7
:
107
10
.
44.
O'Brien
P
,
Morin
P
Jr
,
Ouellette
RJ
,
Robichaud
GA
.
The Pax-5 gene: a pluripotent regulator of B-cell differentiation and cancer disease
.
Cancer Res
2011
;
71
:
7345
50
.
45.
Chung
EY
,
Psathas
JN
,
Yu
D
,
Li
Y
,
Weiss
MJ
,
Thomas-Tikhonenko
A
.
CD19 is a major B cell receptor–independent activator of MYC-driven B-lymphomagenesis
.
J Clin Invest
2012
;
122
:
2257
66
.
46.
Kanteti
R
,
Nallasura
V
,
Loganathan
S
,
Tretiakova
M
,
Kroll
T
,
Krishnaswamy
S
, et al
.
PAX5 is expressed in small-cell lung cancer and positively regulates c-Met transcription
.
Lab Invest
2009
;
89
:
301
14
.
47.
Yuen
GJ
,
Demissie
E
,
Pillai
S
.
B lymphocytes and cancer: a love-hate relationship
.
Trends Cancer
2016
;
2
:
747
57
.
48.
Woo
JR
,
Liss
MA
,
Muldong
MT
,
Palazzi
K
,
Strasner
A
,
Ammirante
M
, et al
.
Tumor infiltrating B-cells are increased in prostate cancer tissue
.
J Transl Med
2014
;
12
:
30
.
49.
Ammirante
M
,
Luo
JL
,
Grivennikov
S
,
Nedospasov
S
,
Karin
M
.
B-cell-derived lymphotoxin promotes castration-resistant prostate cancer
.
Nature
2010
;
464
:
302
5
.
50.
Korrer
MJ
,
Zhang
Y
,
Routes
JM
.
Possible role of arginase-1 in concomitant tumor immunity
.
PLoS One
2014
;
9
:
e91370
.
51.
Wilson
KR
,
Villadangos
JA
,
Mintern
JD
.
Dendritic cell Flt3 - regulation, roles and repercussions for immunotherapy
.
Immunol Cell Biol
2021
;
99
:
962
71
.
52.
Saad
F
,
Armstrong
AJ
,
Thiery-Vuillemin
A
,
Oya
M
,
Loredo
E
,
Procopio
G
, et al
.
PROpel: Phase III trial of olaparib (ola) and abiraterone (abi) versus placebo (pbo) and abi as first-line (1L) therapy for patients (pts) with metastatic castration-resistant prostate cancer (mCRPC)
.
J Clin Oncol
2022
;
40
(
6_suppl)
:
11
.
53.
A study of olaparib and durvalumab in prostate cancer
. https://ClinicalTrials.gov/show/NCT03810105.
54.
Study of pembrolizumab (MK-3475) plus olaparib versus abiraterone acetate or enzalutamide in metastatic castration-resistant prostate cancer (mCRPC) (MK-7339–010/KEYLYNK-010)
. https://ClinicalTrials.gov/show/NCT03834519.
55.
177Lu-PSMA-617 therapy and olaparib in patients with metastatic castration resistant prostate cancer
. https://ClinicalTrials.gov/show/NCT03874884.
56.
Isaacsson Velho
P
,
Qazi
F
,
Hassan
S
,
Carducci
MA
,
Denmeade
SR
,
Markowski
MC
, et al
.
Efficacy of radium-223 in bone-metastatic castration-resistant prostate cancer with and without homologous repair gene defects
.
Eur Urol
2019
;
76
:
170
6
.
57.
Steinberger
AE
,
Cotogno
P
,
Ledet
EM
,
Lewis
B
,
Sartor
O
.
Exceptional duration of radium-223 in prostate cancer with a BRCA2 mutation
.
Clin Genitourin Cancer
2017
;
15
:
e69
71
.
58.
van der Doelen
MJ
,
Isaacsson Velho
P
,
Slootbeek
PHJ
,
Pamidimarri Naga
S
,
Bormann
M
,
van Helvert
S
, et al
.
Impact of DNA damage repair defects on response to radium-223 and overall survival in metastatic castration-resistant prostate cancer
.
Eur J Cancer
2020
;
136
:
16
24
.
59.
Alva
A
,
Nordquist
L
,
Daignault
S
,
George
S
,
Ramos
J
,
Albany
C
, et al
.
Clinical correlates of benefit from Radium-223 therapy in metastatic castration resistant prostate cancer
.
Prostate
2017
;
77
:
479
88
.
60.
Castello
A
,
Macapinlac
HA
,
Lopci
E
,
Santos
EB
.
Prostate-specific antigen flare induced by (223)RaCl2 in patients with metastatic castration-resistant prostate cancer
.
Eur J Nucl Med Mol Imaging
2018
;
45
:
2256
63
.

Supplementary data