Next Article in Journal
Current and Future Biomarkers for Immune Checkpoint Inhibitors in Head and Neck Squamous Cell Carcinoma
Previous Article in Journal
Minimally Invasive Interventional Procedures for Metastatic Bone Disease: A Comprehensive Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Case Report

A Case Study of Clinical Response to Rucaparib in a Patient with Metastatic Castration-Resistant Prostate Cancer and a RAD51B Alteration

1
Medical Oncology, University Hospital of Liège, CHU Sart Tilman, 4000 Liège, Belgium
2
Translational Medicine, Clovis Oncology Inc., Boulder, CO 08006, USA
3
Clinical Science, Clovis Oncology UK, Ltd., Cambridge CB21 6GP, UK
4
Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
*
Author to whom correspondence should be addressed.
Curr. Oncol. 2022, 29(6), 4178-4184; https://doi.org/10.3390/curroncol29060333
Submission received: 5 May 2022 / Revised: 28 May 2022 / Accepted: 6 June 2022 / Published: 8 June 2022
(This article belongs to the Section Genitourinary Oncology)

Abstract

:
PARP inhibitors, such as rucaparib, have been well characterized in metastatic castration-resistant prostate cancer (mCRPC) associated with BRCA alterations, and the clinical activity of these agents has also been evaluated in patients with mCRPC associated with alterations in other non-BRCA DNA damage repair (DDR) genes, including RAD51B. There is likely a differential sensitivity to PARP inhibition based on the specific DDR gene altered, but research in this area is limited because of the low frequency of alterations in these genes. Here, we describe a mCRPC patient with a truncating rearrangement of RAD51B who had a radiographic and PSA response when treated with the PARP inhibitor rucaparib within the TRITON2 trial. We investigated the patients’ response parameters, circulating tumor DNA (ctDNA) fraction and tumor genomics longitudinally, using next-generation sequencing (NGS) of tissue and plasma. ctDNA fraction correlates with radiographic and PSA response and is lower during times of response. NGS did not reveal any potential genomic mechanism of acquired drug resistance. This case shows evidence for rucaparib activity in a rare patient with mCRPC and a RAD51B truncation.

1. Introduction

DNA repair is mediated by various proteins such as the poly(adenosine diphosphate [ADP]-ribose) polymerase (PARP) enzymes, which are crucial for single-strand DNA break repair, and by BRCA1, BRCA2, and RAD51, which are involved in homologous recombination repair (HRR) [1,2,3]. In tumor cells with impaired HRR (e.g., through gene alteration), the enzymatic inhibition of PARP proteins results in the accumulation of DNA damage and cell death through an interaction known as synthetic lethality [4,5,6].
Rucaparib is a PARP inhibitor approved in the United States for the treatment of patients with deleterious BRCA1 or BRCA2 (BRCA) mutation-associated metastatic castration-resistant prostate cancer (mCRPC) who have been treated with androgen receptor-directed therapy and a taxane-based chemotherapy [7]. The accelerated approval of rucaparib as a treatment for patients with mCRPC was based on the efficacy results from TRITON2 (NCT02952534), an international, multicenter phase II study of rucaparib in patients with mCRPC and homologous recombination repair deficiency (HRD) [8].
In addition to evaluating patients with a BRCA alteration, TRITON2 investigated rucaparib treatment in a smaller cohort of patients with mCRPC and with a non-BRCA DNA damage repair (DDR) gene alteration [9], including RAD51B, one of the RAD51 paralogs involved in the HRR pathway [3,10]. Deleterious alterations of RAD51B in prostate cancer are rare and estimated to be present in roughly 0.56% of patients [11]. Here, we report a case study of a patient with mCRPC and a RAD51B rearrangement, which is of particular interest because data from patients with RAD51B alterations who were treated with a PARP inhibitor are limited due to the low prevalence.

2. Case Report

In April 2006, a 63-year-old man, a former smoker with no family history of cancer, was diagnosed with cT2 N0 M0 prostate cancer. The patient received a radical prostatectomy, and the pathology report classified the tumor as pT3a Nx adenocarcinoma with a Gleason score of 7 (3 + 4).
In May 2014, the patient was prescribed a short-course bicalutamide and started long-term triptorelin as an androgen deprivation therapy (ADT) for bone and lymph node metastases (Figure 1A). In February 2016, the confirmed diagnosis of mCRPC led to the addition of abiraterone to ADT. With this regimen, the patient had a best response of stable disease; however, the treatment was stopped in October 2017 (after 87 weeks of treatment) due to a rise in prostate-specific antigen (PSA) values (doubling time of 2 months), with confirmed progression in bone and lymph nodes. Docetaxel was initiated in November 2017, and the patient achieved a confirmed partial response per Response Evaluation Criteria In Solid Tumors, version 1.1 (RECIST), as well as a PSA decrease >50%. Treatment was discontinued in May 2018 (23 weeks, 8 cycles), and PSA values began to rise within 8 weeks, followed by pain that required palliative radiation (1 × 8 Gy) for a spinal cord compression in late July 2018. Nodal and bone disease progression was confirmed in August 2018 (Figure 2A).
In September 2018, the patient was enrolled in the TRITON2 study based on results from genomic testing with the FoundationOne next-generation sequencing (NGS) assay (Foundation Medicine, Cambridge, MA, USA) [12], which detected the presence of a truncating RAD51B rearrangement in an archival tumor tissue biopsy obtained at the time of diagnosis (June 2006; Table 1). The rearrangement was a fusion with ACTN1 resulting in the deletion of exons 3 through 11 of RAD51B, which has a total of 11 exons. In addition, a pathogenic TMPRSS2-ERG fusion and a deleterious RB1 rearrangement were detected. NGS of a plasma sample collected prior to rucaparib treatment using the FoundationOne Liquid CDx assay [13] detected the presence of additional somatic pathogenic alterations, among them 2 TP53 mutations (Table 1, Table S1).
No other potential drivers of the disease were identified, and all detected alterations were confirmed to be of somatic origin using the Color Hereditary Cancer Test [14].
The patient started rucaparib at the recommended dose of 600 mg twice daily, but hematologic toxicity, particularly anemia, led to several treatment interruptions with subsequent dose reductions to 200 mg twice daily; the patient ultimately received rucaparib for 107 weeks (Figure 1A). At the start of TRITON2, the patient had more than 10 bone-associated lesions and multiple soft-tissue lesions in the left shoulder, left scapula, and left axillary and latero aortic lymph nodes. Treatment with rucaparib resulted in a confirmed partial response per modified RECIST and/or Prostate Cancer Clinical Trials Working Group 3 criteria (maximum of 81% decrease in target lesion diameters; Figure 1B and Figure 2A) lasting 80 weeks (December 2018 to July 2020), with no confirmed progression in the bone. The patient also had a confirmed PSA response (≥50% decrease from baseline confirmed by a second measurement ≥3 weeks later) (Figure 1B) lasting 64 weeks (October 2018 to January 2020) with a maximum decrease from the baseline of 99%. In comparison, 15 of 27 TRITON2 patients with a BRCA mutation and a radiographic response had a duration of response ≥6 months, and the median time to PSA progression was 28 weeks in all TRITON2 BRCA-mutated patients [8]. The patient discontinued due to clinical disease progression at new sites in the left subclavicular and para-aortic areas in September 2020 (Figure 2B). Following discontinuation of rucaparib treatment, the patient received enzalutamide 160 mg every day from September 2020 until December 2020 and died in February 2021.
The longitudinal genomic profile of the patient was assessed through genomic testing with the GuardantOMNI assay (Guardant Health, Redwood City, CA, USA) [15] of plasma samples collected at the start of treatment (September 2018 [week 1]), at the nadir of response (October 2019 [week 60]), after a rise in PSA following the confirmed response (March 2020 [week 80]), and following progression (September 2020 [week 108]) to further explore the genomic landscape. The RAD51B truncation was found in the archival tumor sample obtained at the time of initial diagnosis, in the plasma obtained prior to rucaparib treatment, and in the plasma after progression; however, the on-treatment plasma samples taken around the time of best response contained less than 2% of cell-free tumor DNA, and the RAD51B rearrangement was not detected due to the low (<10%) tumor fraction. The tumor fraction of all plasma samples was low and ranged from 1.3% at the time of the best radiographic response to 10.3% at the time of treatment (Table 1). In the progression sample, no secondary alterations of RAD51B or other apparent mechanisms of reversion were detected.

3. Discussion

While rucaparib and other PARP inhibitors have been perhaps best characterized in mCRPC associated with BRCA alterations, the clinical activity of these agents has been evaluated in patients with mCRPC associated with alterations in other non-BRCA DDR genes, including RAD51B [16]. In the phase III PROfound study (NCT02987543), while median progression-free survival was significantly longer with olaparib versus control agents in men with mCRPC who had alterations in BRCA or ATM (7.4 vs. 3.6 months; hazard ratio, 0.34 [95% CI, 0.25–0.47]; n = 245), more modest effects were seen in the subgroup of patients with alterations in other DDR genes (4.8 vs. 3.3 months; hazard ratio, 0.88; n = 142) [16]. There is likely a differential sensitivity to PARP inhibition based on the specific DDR gene altered, but research in this area is hindered by the low frequency of these alterations: among the genomically selected PROfound patients, only 7 had an alteration in RAD51B, including one in the olaparib group with a co-occurring alteration in ATM and another in the control group with a co-occurring alteration in BRCA2. For patients who had a truncation RAD51B rearrangement without a co-occurring DDR gene alteration (n = 5), the median imaging-based progression-free survival was 10.9 months in the olaparib group (n = 4) and 1.8 months in the control group (n = 1) [16].
In the TRITON2 patient described here, we hypothesize the tumor response to rucaparib was likely driven by HRD caused by the truncating rearrangement of RAD51B. The RAD51B truncation was found in the archival tumor sample obtained at the time of initial diagnosis and in the plasma obtained prior to and post rucaparib treatment; however, the on-treatment plasma samples at time of response contained very little circulating tumor DNA, and the RAD51B rearrangement could not be detected. The tumor fraction of all plasma samples was low, and all plasma samples had reduced sensitivity for deletion calling. Therefore, the possibility of another undetected disease driver, such as homozygous BRCA loss, cannot definitively be ruled out.
Treatment with rucaparib resulted in radiographic and PSA responses, and following a dose reduction to mitigate adverse events, a continuation of treatment with rucaparib 200 mg twice daily allowed for the maintenance of the radiological response for over 1 year. The patient eventually developed new lesions in the left subclavicular and para-aortic areas. However, enlargement was not witnessed in the target lesions of the left axillary lymph nodes that were present at the start of rucaparib treatment, suggesting that the new lesions may have potentially been due to the emergence of new cancer clones. While the NGS analysis of postprogression plasma did not reveal any reversion mechanism or new genomic clones, in the absence of NGS data from these new lesions, it is not possible to confirm or reject this hypothesis.
In summary, this case shows evidence for rucaparib activity in the sole patient with a genomic alteration of RAD51B enrolled in TRITON2, adding further data to suggest that patients with mCRPC and alterations in DDR genes besides BRCA1 or BRCA2 may also experience clinical benefits from treatment with a PARP inhibitor.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/curroncol29060333/s1. Table S1. Supplemental Data.

Author Contributions

Conception and design: B.S., A.L., S.P.W., W.A. Acquisition of data (e.g., managed and treated patients): B.S., A.L., H.S. Analysis and interpretation of data: All authors. Writing, review, and/or revision of the manuscript: All authors. All authors have read and agreed to the published version of the manuscript.

Funding

TRITON2 was funded by Clovis Oncology. W.A. is supported in part by the National Cancer Institute (NCI) Cancer Center Support Grant P30 CA 008748, the NCI Prostate Specialized Program of Research Excellence (SPORE) Grant P50 CA092629-16, the Department of Defense Prostate Cancer Research Program Grant W81XWH-17-1-0124, and a Prostate Cancer Foundation Young Investigator Award.

Institutional Review Board Statement

TRITON2 was approved by national or local institutional review boards including the Ethics Committee from University Hospital of Liège protocol number 2017/85 on 10 October 2017 and performed in accordance with the Declaration of Helsinki and Good Clinical Practice guidelines of the International Council for Harmonisation.

Informed Consent Statement

The patient provided written informed consent including consent to publish information and/or images before participation.

Data Availability Statement

Requests for de-identified datasets for the results reported in this publication will be made available to qualified researchers following submission of a methodologically sound proposal to medinfo@clovisoncology.com. Data will be made available for such requests following online publication of this article and for 1 year thereafter in compliance with applicable privacy laws, data protection, and requirements for consent and anonymization. Data will be provided by Clovis Oncology. Clovis Oncology does not share identified participant data or a data dictionary.

Acknowledgments

Medical writing and editorial support funded by Clovis Oncology, Inc., were provided by Shelly Lim and Stephen Bublitz of Ashfield MedComms, an Ashfield Health company.

Conflicts of Interest

Authors’ disclosures of potential conflicts of interest: Brieuc Sautois, Consulting or Advisory Role: Clovis Oncology, Astellas, Janssen, and BMS Belgium, Honoraria: Janssen: MSD, Andrea Loehr, Employment: Clovis Oncology, Stock and Other Ownership Interests: Clovis Oncology, Simon P. Watkins, Employment: Clovis Oncology, Stock and Other Ownership Interests: Clovis Oncology, Wassim Abida, Honoraria: Aptitude Health, Clinical Education Alliance, Medscape, OncLive/MJH Life Sciences, and Roche, Consulting or Advisory Role: Clovis Oncology, AstraZeneca/MedImmune, Daiichi Sankyo, Janssen, and ORIC Pharmaceuticals, Research Funding: Clovis Oncology (Inst), AstraZeneca (Inst), Epizyme (Inst), ORIC Pharmaceuticals (Inst), Zenith Epigenetics (Inst), No other potential conflicts of interests were reported.

References

  1. Prakash, R.; Zhang, Y.; Feng, W.; Jasin, M. Homologous recombination and human health: The roles of BRCA1, BRCA2, and associated proteins. Cold Spring Harb. Perspect. Biol. 2015, 7, a016600. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Caldecott, K.W.; Aoufouchi, S.; Johnson, P.; Shall, S. XRCC1 polypeptide interacts with DNA polymerase beta and possibly poly (ADP-ribose) polymerase, and DNA ligase III is a novel molecular ’nick-sensor’ in vitro. Nucleic Acids Res. 1996, 24, 4387–4394. [Google Scholar] [CrossRef] [PubMed]
  3. Skidmore, C.J.; Davies, M.I.; Goodwin, P.M.; Halldorsson, H.; Louis, P.L.; Shall, S.; Ziicc, A. The involvement of poly(ADP-ribose) polymerase in the degradation of NAD caused by gamma-radiation and N-methyl-N-nitrosourea. Eur. J. Biochem. 1979, 101, 135–142. [Google Scholar] [CrossRef] [PubMed]
  4. Drew, Y.; Mulligan, E.A.; Vong, W.T.; Thomas, H.D.; Kahn, S.; Kyle, S.; Mukhopadhyay, A.; Los, J.; Hostomsky, Z.; Plummer, E.R.; et al. Therapeutic potential of poly(ADP-ribose) polymerase inhibitor AG014699 in human cancers with mutated or methylated BRCA1 or BRCA2. J. Natl. Cancer Inst. 2011, 103, 334–346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Nguyen, M.; Simmons, A.D.; Harding, T.C. Preclinical assessment of the PARP inhibitor rucaparib in homologous recombination deficient prostate cancer models. Cancer Res. 2017, 77 (Suppl. 13), 2476. [Google Scholar]
  6. Robillard, L.; Nguyen, M.; Harding, T.C.; Simmons, A.D. In vitro and in vivo assessment of the mechanism of action of the PARP inhibitor rucaparib. Cancer Res. 2017, 77 (Suppl. 13), 2475. [Google Scholar]
  7. Anscher, M.S.; Chang, E.; Gao, X.; Gong, Y.; Weinstock, C.; Bloomquist, E.; Adeniyi, O.; Charlab, R.; Zimmerman, S.; Serlemitsos-Day, M.; et al. FDA Approval Summary: Rucaparib for the Treatment of Patients with Deleterious BRCA-Mutated Metastatic Castrate-Resistant Prostate Cancer. Oncologist 2021, 26, 139–146. [Google Scholar] [CrossRef] [PubMed]
  8. Abida, W.; Patnaik, A.; Campbell, D.; Shapiro, J.; Bryce, A.H.; McDermott, R.; Sautois, B.; Vogelzang, N.J.; Bambury, R.M.; Voog, E.; et al. Rucaparib in men with metastatic castration-resistant prostate cancer harboring a BRCA1 or BRCA2 gene alteration. J. Clin. Oncol. 2020, 38, 3763–3772. [Google Scholar] [CrossRef] [PubMed]
  9. Abida, W.; Campbell, D.; Patnaik, A.; Shapiro, J.D.; Sautois, B.; Vogelzang, N.J.; Voog, E.G.; Bryce, A.H.; McDermott, R.; Ricci, F.; 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 2 TRITON2 study. Clin. Cancer Res. 2020, 26, 2487–2496. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Takata, M.; Sasaki, M.S.; Sonoda, E.; Fukushima, T.; Morrison, C.; Albala, J.S.; Swagemakers, S.M.; Kanaar, R.; Thompson, L.H.; Takeda, S. The Rad51 paralog Rad51B promotes homologous recombinational repair. Mol. Cell. Biol. 2000, 20, 6476–6482. [Google Scholar] [CrossRef] [PubMed]
  11. Cerami, E.; Gao, J.; Dogrusoz, U.; Gross, B.E.; Sumer, S.O.; Aksoy, B.A.; Jacobsen, A.; Byrne, C.J.; Heuer, M.L.; Larsson, E.; et al. The cBio cancer genomics portal: An open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012, 2, 401–404. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Frampton, G.M.; Fichtenholtz, A.; Otto, G.A.; Wang, K.; Downing, S.R.; He, J.; Schnall-Levin, M.; White, J.; Sanford, E.M.; An, P.; et al. Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nat. Biotechnol. 2013, 31, 1023–1031. [Google Scholar] [CrossRef] [PubMed]
  13. Clark, T.A.; Chung, J.H.; Kennedy, M.; Hughes, J.D.; Chennagiri, N.; Lieber, D.S.; Fendler, B.; Young, L.; Zhao, M.; Coyne, M.; et al. Analytical Validation of a Hybrid Capture–Based Next-Generation Sequencing Clinical Assay for Genomic Profiling of Cell-Free Circulating Tumor DNA. J. Mol. Diagn. 2018, 20, 686–702. [Google Scholar] [CrossRef] [PubMed]
  14. Crawford, B.; Adams, S.B.; Sittler, T.; van den Akker, J.; Chan, S.; Leitner, O.; Ryan, L.; Gil, E.; van’t Veer, L. Multi-gene panel testing for hereditary cancer predisposition in unsolved high-risk breast and ovarian cancer patients. Breast Cancer Res. Treat. 2017, 163, 383–390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Helman, E.; Artieri, C.; Vowles, J.V.; Yen, J.; Nance, T.; Sikora, M.; Gourneau, J.; Goel, M.; Mortimer, S.; Chudova, D.; et al. Analytical validation of a comprehensive 500-gene ctDNA panel designed for immuno-oncology and DNA damage research. Cancer Res. 2018, 78 (Suppl. 13), 5603. [Google Scholar]
  16. de Bono, J.; Mateo, J.; Fizazi, K.; Saad, F.; Shore, N.; Sandhu, S.; Chi, K.N.; Sartor, O.; Agarwal, N.; Olmos, D.; et al. Olaparib for Metastatic Castration-Resistant Prostate Cancer. N. Engl. J. Med. 2020, 382, 2091–2102. [Google Scholar] [CrossRef] [PubMed]
Figure 1. (A) The clinical course of the patient, and (B) the evolution of PSA and tumor measurements per RECIST with rucaparib. mCRPC, metastatic castration-resistant prostate cancer; mPC, metastatic prostate cancer; NGS, next-generation sequencing; PC, prostate cancer; PSA, prostate-specific antigen; RECIST, Response Evaluation Criteria In Solid Tumors, version 1.1; rPD, radiological progressive disease; rPR, radiological partial response.
Figure 1. (A) The clinical course of the patient, and (B) the evolution of PSA and tumor measurements per RECIST with rucaparib. mCRPC, metastatic castration-resistant prostate cancer; mPC, metastatic prostate cancer; NGS, next-generation sequencing; PC, prostate cancer; PSA, prostate-specific antigen; RECIST, Response Evaluation Criteria In Solid Tumors, version 1.1; rPD, radiological progressive disease; rPR, radiological partial response.
Curroncol 29 00333 g001
Figure 2. (A) Initial tumor scans (at start of rucaparib treatment) compared with scans at the time of best response (week 60 of rucaparib treatment) of nodal metastases and lytic bone metastases at the target lesions. (B) Scans at the time of best response (week 60 of rucaparib treatment) compared with scans after radiographic progression.
Figure 2. (A) Initial tumor scans (at start of rucaparib treatment) compared with scans at the time of best response (week 60 of rucaparib treatment) of nodal metastases and lytic bone metastases at the target lesions. (B) Scans at the time of best response (week 60 of rucaparib treatment) compared with scans after radiographic progression.
Curroncol 29 00333 g002aCurroncol 29 00333 g002b
Table 1. Summary Of Longitudinal Genomic Testing.
Table 1. Summary Of Longitudinal Genomic Testing.
Time PointSample TypeAssayctDNA FractionRAD51B/ACTN1 RearrangementTMPRSS2-ERG FusionTP53 Y220C
Archival TissueFoundationOneNADetectedDetectedNot detected
Pre-treatmentPlasmaFoundationOne Liquid15.5%DetectedDetectedDetected
Pre-treatmentPlasmaGuardant Omni10.3%DetectedDetectedDetected
On-treatment Week 60PlasmaGuardant Omni1.3%Not DetectedNot DetectedDetected
On-treatment Week 80PlasmaGuardant Omni2.1%Not DetectedNot DetectedDetected
Post-progression Week 106PlasmaGuardant Omni9.6%DetectedDetectedDetected
Abbreviations: AF, allelic fraction; ctDNA, circulating tumor DNA.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Sautois, B.; Loehr, A.; Watkins, S.P.; Schroeder, H.; Abida, W. A Case Study of Clinical Response to Rucaparib in a Patient with Metastatic Castration-Resistant Prostate Cancer and a RAD51B Alteration. Curr. Oncol. 2022, 29, 4178-4184. https://doi.org/10.3390/curroncol29060333

AMA Style

Sautois B, Loehr A, Watkins SP, Schroeder H, Abida W. A Case Study of Clinical Response to Rucaparib in a Patient with Metastatic Castration-Resistant Prostate Cancer and a RAD51B Alteration. Current Oncology. 2022; 29(6):4178-4184. https://doi.org/10.3390/curroncol29060333

Chicago/Turabian Style

Sautois, Brieuc, Andrea Loehr, Simon P. Watkins, Hélène Schroeder, and Wassim Abida. 2022. "A Case Study of Clinical Response to Rucaparib in a Patient with Metastatic Castration-Resistant Prostate Cancer and a RAD51B Alteration" Current Oncology 29, no. 6: 4178-4184. https://doi.org/10.3390/curroncol29060333

Article Metrics

Back to TopTop