Back to blog

Ushering in a New Era of Personalized Treatment for Advanced Prostate Cancer

Over the last several decades, we’ve experienced a massive shift in how we approach, diagnose and treat prostate cancer. Prostate cancer remains the second leading cause of cancer-related deaths in men, behind lung cancer.1 But progress is being made as the relative five-year survival rate for patients with distant metastatic prostate cancer is nearly 37%,2 up from 28.7% in the early 2000s.3 

The emergence of comprehensive genomic profiling (CGP) in the last 15 years has shown us that prostate cancer is an extremely diverse and complex disease.4 Given the complexities of the disease, it’s clear a “one-size-fits-all” approach is not beneficial. But thanks to continued progress in research, advancements in biomarker testing, and new targeted treatment options, a shift toward more personalized care is underway for how we treat and assess response in advanced prostate cancer.

Biomarker discoveries drive new targeted treatment options for advanced prostate cancer

The introduction of hormone therapy in the 1980s ushered in the first revolution in treatment for prostate cancer and quickly became the standard of care.5 However, most prostate cancers develop resistance to these treatments over time, necessitating new, innovative and targeted treatment options.

In the early 2000s, studies indicated that alterations in BRCA1 and BRCA2 and other homologous recombination repair (HRR) genes rendered tumors sensitive to poly-ADP ribose polymerase (PARP) inhibitors, which work by preventing cancer cells from repairing and growing.6 Nearly a quarter of prostate cancer patients have either germline or somatic HRR alterations,7 with BRCA1/2 alterations the most common, occurring in nearly 11% of metastatic prostate cancer cases.8 The presence of these mutations is also associated with more advanced disease at diagnosis and poorer clinical outcomes.9

It wasn’t until 2020 that the U.S. Food and Drug Administration (FDA) approved the first PARP inhibitors for the treatment of metastatic castration-resistant prostate cancer (mCRPC),10 ushering in a new era of targeted treatment. Since then, the FDA has approved multiple PARP inhibitors for patients with mCRPC.11

Outside of PARP inhibitors, patients with prostate cancer may be eligible for PD-1 checkpoint inhibitors after progression on standard-of-care options if their cancer has genomic features such as high microsatellite instability, a defect in a mismatch repair gene, or a high tumor mutational burden.12

As our understanding of actionable biomarkers evolves, access to high-quality comprehensive genomic profiling (CGP), or biomarker tests, like FoundationOne®CDx and FoundationOne®Liquid CDx,13 is key to informing treatment options. FoundationOne CDx and FoundationOne Liquid CDx CGP tests each detect all 27 guideline-recommended biomarkers for advanced prostate cancer14,15,16,17 and FoundationOne Liquid CDx is the only FDA-approved CGP liquid biopsy to detect all 19 HRR genes for approved therapies.18 Additionally, Foundation Medicine now has seven FDA-approved companion diagnostic indications for prostate cancer, the most of any company.19

Hear from John, who has been living with metastatic prostate cancer since 2014, on the important role biomarker testing played in his cancer care:

Emerging biomarkers to aid treatment decisions in advanced prostate cancer

With high-quality biomarker testing and more genomic data available, we’re also able to identify new biomarkers in prostate cancer that can help providers make informed treatment decisions.

To aid with decision-making around PARP inhibitors, we recently introduced our proprietary homologous recombination deficiency signature (HRDsig) biomarker20 reported on FoundationOne CDx. Our HRDsig biomarker is a scar-based genomic signature that gives a functional readout of HRD using genome-wide copy number (CN) features. In prostate cancer, HRDsig positivity was associated with improved time to treatment discontinuation and real-world overall survival on PARP inhibitors compared to patients without HRDsig detected.21

Biallelic loss of BRCA1/2 is also an important new biomarker for PARP inhibitor use in prostate cancer. New research indicates the prevalence of BRCA1/2 loss at approximately 3% in both primary prostate tumors and metastatic prostate tumors.21 Unlike other types of BRCA alterations, biallelic BRCA1/2 loss is irreversible and, therefore, not susceptible to secondary reversion mutations caused by PARP inhibitor use that can cause treatment resistance.22,23 As a result, those patients with biallelic loss of BRCA1/2 are more likely to have favorable outcomes and durable benefits from PARP inhibitors in clinical practice.21 Detecting biallelic copy loss for BRCA1/2 can be technically challenging,24 particularly via liquid biopsy since it requires a higher level of circulating tumor DNA (ctDNA).21 However, our portfolio of tissue- and liquid-based tests can help detect BRCA1/2 losses, and we observe them in primary tumors and metastatic samples.21

Additionally, new research is emerging that indicates SPOP mutations may be a potential new biomarker for patients with de novo metastatic castration-sensitive prostate cancer. SPOP mutations occur in approximately 8-10% of prostate cancer,25 and research demonstrates that patients with these mutations experience considerably more favorable outcomes on second-generation hormonal therapies compared to those without, but this effect was not seen with standard taxane chemotherapy. These observations, combined with a biological understanding of SPOP, suggest that SPOP is a predictive biomarker of extreme benefit from second-generation hormonal therapies in the hormone-sensitive setting. If physicians and patients prefer to try a chemotherapy-sparing option for initial treatment of de novo metastatic prostate cancer, SPOP serves as an emerging biomarker that can help aid in this clinical decision. With FoundationOne Liquid CDx, the only FDA-approved liquid biopsy that identifies SPOP mutations,26 healthcare providers have information to make informed decisions to support an upfront non-chemotherapy option, helping to preserve quality of life.

The value of liquid biopsy to inform treatment decisions and monitor prostate cancer progression

While tissue testing remains the gold standard for identifying genomic alterations, not all patients with mCRPC have sufficient tumor tissue available for testing, especially at the point of disease progression.27 That’s where a high-quality liquid biopsy like FoundationOne Liquid CDx can be particularly valuable.

One of the traditional challenges with liquid biopsy is the interpretation of a negative result. Is the negative result a lack of detectable tumor DNA in the blood sample or the lack of alterations in a sample with sufficient ctDNA? In the case of a negative liquid biopsy result, ctDNA tumor fraction can serve as a useful tool. Foundation Medicine’s ctDNA tumor fraction28 is a determination of the amount of tumor DNA in the blood, and FoundationOne Liquid CDx reports ctDNA tumor fraction – for greater confidence in negative results.28,29 When tumor fraction is low (<1%) and no drivers are detected by FoundationOne Liquid CDx, tumor tissue testing is more likely to detect alterations that can drive treatment decisions.

Outside of helping you interpret liquid biopsy results, ctDNA tumor fraction is emerging as a potentially powerful new biomarker in prostate cancer. Recent research found that ctDNA tumor fraction in prostate cancer provided complementary prognostic value to PSA testing and provided additional risk stratification for patients with clinically ambiguous results.30

Finally, longitudinal ctDNA tumor fraction monitoring can provide value in tracking tumor response to treatment in metastatic prostate cancer. Traditionally, PSA and radiographic imaging have been used to monitor disease response to treatment. With FoundationOne®Monitor,31 a tissue-naïve ctDNA monitoring assay currently available for research use, we can evaluate quantitative changes in ctDNA levels over time to understand how tumors are responding to treatment. Changes in ctDNA tumor fraction may complement imaging and PSA-based approaches to treatment response assessment, especially in ambiguous situations where changes in PSA no longer correlate with tumor burden or in cases of bone-only disease where bone scans may exhibit flair reactions that are difficult to interpret.  In addition to quantitative changes in ctDNA tumor fraction, FoundationOne®Monitor reports genomic alterations that can occur with acquired resistance to targeted therapies or disease progression, informing early resistance to treatment and future therapy options.  Longitudinal ctDNA monitoring holds great promise for impacting treatment decisions for patients with metastatic prostate cancer.

Hope for the future of prostate cancer treatment

In a few short years, CGP has revolutionized the way we detect and treat prostate cancer, offering profound value and promise to patients. I’m optimistic that CGP testing will continue to help us improve outcomes for those with advanced prostate cancer.

As our understanding of the disease continues to evolve through the discovery of new biomarkers, we remain committed to continuing to innovate our portfolio and unlocking the promise of precision oncology so that every patient can benefit from personalized, targeted treatments.

References

1SEER Cancer Statistics Factsheets: Common Cancer Sites. National Cancer Institute. Bethesda, MD, https://seer.cancer.gov/statfacts/html/common.html 

2SEER Cancer Stat Facts: Prostate Cancer. National Cancer Institute. Bethesda, MD, https://seer.cancer.gov/statfacts/html/prost.html

3Siegel DA, O’Neil ME, Richards TB, Dowling NF, Weir HK. Prostate Cancer Incidence and Survival, by Stage and Race/Ethnicity — United States, 2001–2017. MMWR Morb Mortal Wkly Rep 2020;69:1473–1480. DOI: http://dx.doi.org/10.15585/mmwr.mm6941a1

4Ikeda S, Elkin SK, Tomson BN, Carter JL, Kurzrock R. Next-generation sequencing of prostate cancer: genomic and pathway alterations, potential actionability patterns, and relative rate of use of clinical-grade testing. Cancer Biol Ther. 2018;20(2):219-226. https://doi.org/10.1080/15384047.2018.1523849 

5Lehtonen M, Kellokumpu-Lehtinen PL. The past and present of prostate cancer and its treatment and diagnostics: A historical review. SAGE Open Med. 2023;11. https://doi.org/10.1177/20503121231216837 

6McCabe N, Turner NC, Lord CJ, et al. Deficiency in the repair of DNA damage by homologous recombination and sensitivity to poly(ADP-ribose) polymerase inhibition. Cancer Res. 2006;66(16):8109–8115. https://doi.org/10.1158/0008-5472.CAN-06-0140 

7Chung JH, Dewal N, Sokol E, et al. Prospective comprehensive genomic profiling of primary and metastatic prostate tumors. JCO Precis Oncol. 2019;3:1-23. https://doi.org/10.1200/PO.18.00283

8Valsecchi AA, Dionisio R, Panepinto O, et al. Frequency of germline and somatic BRCA1 and BRCA2 mutations in prostate cancer: An updated systematic review and meta-analysis. Cancers. 2023;15(9):2435. https://doi.org/10.3390/cancers15092435 

9Messina C, Cattrini C, Soldato D, et al. BRCA Mutations in Prostate Cancer: Prognostic and Predictive Implications. J Oncol. 2020;2020(1) https://doi.org/10.1155/2020/4986365 

10National Cancer Institute. With Two FDA Approvals, Prostate Cancer Treatment Enters the PARP Era. Created June 11, 2020. Accessed July 26, 2024. https://www.cancer.gov/news-events/cancer-currents-blog/2020/fda-olaparib-rucaparib-prostate-cancer 

11National Cancer Institute. FDA Approves New Initial Treatment Option for Some Metastatic Prostate Cancers. Created August 4, 2023. Accessed July 26, 2024. https://www.cancer.gov/news-events/cancer-currents-blog/2023/fda-talazoparib-enzalutamide-prostate-cancer 

12American Cancer Society. Immunotherapy for Prostate Cancer. Updated November 22, 2023. Accessed July 26, 2024. https://www.cancer.org/cancer/types/prostate-cancer/treating/vaccine-treatment.html

13FoundationOne®CDx and FoundationOne®Liquid CDx are qualitative next-generation sequencing based in vitro diagnostic tests for advanced cancer patients with solid tumors and are for prescription use only. FoundationOne CDx utilizes FFPE tissue and analyzes 324 genes as well as genomic signatures. FoundationOne Liquid CDx analyzes 324 genes utilizing circulating cell-free DNA and is FDA-approved to report short variants in 311 genes. The tests are companion diagnostics to identify patients who may benefit from treatment with specific therapies in accordance with the therapeutic product labeling.  Additional genomic findings may be reported and are not prescriptive or conclusive for labeled use of any specific therapeutic product. Use of the tests does not guarantee a patient will be matched to a treatment. A negative result does not rule out the presence of an alteration. Some patients may require a biopsy for testing with FoundationOne CDx when archival tissue is not available which may pose a risk. Patients being considered for eligibility for therapy based on detection of NTRK1/2/3 and ROS1 fusions should only be tested if tissue is unavailable. Patients who are tested with FoundationOne Liquid CDx and are negative for other companion diagnostic mutations should be reflexed to tumor tissue testing and mutation status confirmed using an FDA-approved tumor tissue test, if feasible. For the complete label, including companion diagnostic indications and important risk information, please visit www.F1CDxLabel.com and www.F1LCDxLabel.com.

14Based on somatic testing guidelines in advanced prostate cancer, including 19 HRR genes, MSI/MSI-H, plus 7 clinically relevant biomarkers (TMB, MSH2, MSH6, PMS2, PTEN, RB1, TP53)

15NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for Prostate Cancer V4.2024. © 2024 National Comprehensive Cancer Network, Inc. All rights reserved. Accessed May 31 2024. To view the most recent and complete version of the guidelines, go online to NCCN.org.

16FoundationOne Liquid CDx Technical Specifications (Gene List). https://www.foundationmedicine.com/sites/default/files/media/documents/2023-10/F1LCDx_Technical_Specs_072021-2.pdf. Accessed February 13, 2024.

17FoundationOne CDx Technical Specifications (Gene List). https://www.foundationmedicine.com/sites/default/files/media/documents/2024-01/F1CDx%20Technical%20Specifications%20SPEC-01197.pdf. Accessed February 13, 2024

18Data on File, Foundation Medicine, Inc., 2024

19Data on File, Foundation Medicine, Inc., 2024

20HRDsig is reported as a laboratory professional service that has not been reviewed or approved by the FDA.

21Triner D, Graf RP, Madison RW, et al. Durable benefit from poly(ADP-ribose) polymerase inhibitors in metastatic prostate cancer in routine practice: biomarker associations and implications for optimal clinical next-generation sequencing testing. ESMO Open. 2024;9(9). https://doi.org/10.1016/j.esmoop.2024.103684 

22Loehr A, Hussain A, Patnaik A, et al. Emergence of BRCA reversion mutations in patients with metastatic castration-resistant prostate cancer after treatment with rucaparib. Eur Urol. 2023;83(3):200–209. https://doi.org/10.1016/j.eururo.2022.09.010 

23Carreira S, Porta N, Arce-Gallego S, et al. Biomarkers associating with PARP inhibitor benefit in prostate cancer in the TOPARP-B trial. Cancer Discov. 2021;11(11): 2812–2827. https://doi.org/10.1158/2159-8290.CD-21-0007

24Vandekerkhove G, Giri VA, Halabi S, et al. Toward informed selection and interpretation of clinical genomic tests in prostate cancer. JCO Precis Oncol. 2024;8. https://doi.org/10.1200/PO.23.00654

25Swami U, Graf RP, Nussenzveig RH, et al. SPOP mutations as a predictive biomarker for androgen receptor axis-targeted therapy in de novo metastatic castration-sensitive prostate cancer. Clin Cancer Res. 2022;28(22):4917-4925. https://doi.org/10.1158/1078-0432.ccr-22-2228

26Orme JJ, Taza F, De Sarkar N, et al. Co-occurring BRCA2/SPOP mutations predict exceptional poly (ADP-ribose) polymerase inhibitor sensitivity in metastatic castration-resistant prostate cancer. Eur Urol Oncol. 2024;7(4)877-887. https://doi.org/10.1016/j.euo.2023.11.014 

27Chi KM, Barnicle A, Sibilia C, et al. Detection of BRCA1, BRCA2 and ATM alterations in matched tumor tissue and circulating tumor DNA in patients with prostate cancer screened in PROfound. Clin Cancer Res. 2023;29(1):81-9. https://doi.org/10.1158/1078-0432.CCR-22-0931 

28ctDNA tumor fraction is reported as a laboratory professional service which has not been reviewed or approved by the FDA. Foundation Medicine’s ctDNA tumor fraction is a determination of the amount of circulating tumor DNA as a fraction of total cell free DNA in a blood sample that accounts for aneuploidy, variant allele frequency, fragment length information, clonal hematopoiesis predictions and known tumor-associated alterations.

29Data on File, Foundation Medicine, Inc., 2024.

30Sweeney CJ, Petry R, Xu C, et al. Circulating tumor DNA assessment for treatment monitoring adds value to PSA in metastatic castration-resistant prostate cancer. Clin Cancer Res. 2024. https://doi.org/10.1158/1078-0432.CCR-24-1096

31FoundationOne®Monitor is a laboratory developed test that was developed and its performance characteristics determined by Foundation Medicine. This test has not been cleared or approved by the U.S. Food and Drug Administration. FoundationOne Monitor is a test for patients with solid malignant neoplasms that detects and longitudinally tracks circulating tumor DNA (ctDNA), reported as ctDNA tumor fraction (a determination of the amount of ctDNA as a fraction of total cell free DNA in a blood sample) as a biomarker for tumor content in the blood. Treatment decisions are the responsibility of the treating physician. ctDNA tumor fraction determination sensitivity may be limited if blood collection occurs within two weeks of surgery. ctDNA not detected status does not definitively indicate the absence of cancer and declining ctDNA tumor fraction does not necessarily indicate a response to therapy. This test is not designed to detect or report germline variation, nor does it infer hereditary cancer risk for a patient. This test is expected to have limited sensitivity in some cancer types due to limited ctDNA shed.

September 30, 2024 Mia Levy, MD, PhD Chief Medical Officer, Foundation Medicine

Blog Subscription

Get notified about new articles