Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Neoadjuvant hormonal therapy before radical prostatectomy in high-risk prostate cancer

Subjects

Abstract

Patients with high-risk prostate cancer treated with curative intent are at an increased risk of biochemical recurrence, metastatic progression and cancer-related death compared with patients treated for low-risk or intermediate-risk disease. Thus, these patients often need multimodal therapy to achieve complete disease control. Over the past two decades, multiple studies on the use of neoadjuvant treatment have been performed using conventional androgen deprivation therapy, which comprises luteinizing hormone-releasing hormone agonists or antagonists and/or first-line anti-androgens. However, despite results from these studies demonstrating a reduction in positive surgical margins and tumour volume, no benefit has been observed in hard oncological end points, such as cancer-related death. The introduction of potent androgen receptor signalling inhibitors (ARSIs), such as abiraterone, apalutamide, enzalutamide and darolutamide, has led to a renewed interest in using neoadjuvant hormonal treatment in high-risk prostate cancer. The addition of ARSIs to androgen deprivation therapy has demonstrated substantial survival benefits in the metastatic castration-resistant, non-metastatic castration-resistant and metastatic hormone-sensitive settings. Intuitively, a similar survival effect can be expected when applying ARSIs as a neoadjuvant strategy in high-risk prostate cancer. Most studies on neoadjuvant ARSIs use a pathological end point as a surrogate for long-term oncological outcome. However, no consensus yet exists regarding the ideal definition of pathological response following neoadjuvant hormonal therapy and pathologists might encounter difficulties in determining pathological response in hormonally treated prostate specimens. The neoadjuvant setting also provides opportunities to gain insight into resistance mechanisms against neoadjuvant hormonal therapy and, consequently, to guide personalized therapy.

Key points

  • Patients with high-risk prostate cancer are at an increased risk of biochemical recurrence, metastatic progression and cancer-related death following primary treatment compared with patients with low-risk or intermediate-risk disease.

  • Multiple studies on neoadjuvant treatment using conventional androgen deprivation therapy before radical prostatectomy have reported reduced positive surgical margins and tumour volume but no benefits in hard oncological end points.

  • Androgen receptor signalling inhibitors have demonstrated substantial survival benefits in metastatic and non-metastatic castration-resistant and metastatic hormone-sensitive prostate cancer.

  • The importance of pathological response following neoadjuvant hormonal therapy is unclear, and the effect of hormonal therapy on the prostate tissue renders pathological assessment difficult.

  • The presence of castration-resistant prostate cancer cells in early disease stages or rapid therapy-induced resistance might be responsible for the low proportion of pathological complete response (10%) in neoadjuvant androgen receptor signalling inhibitor studies.

  • Longitudinal imaging could help to identify early responders by assessing the effect of neoadjuvant hormonal therapy on the local tumour and possible micrometastases.

  • Ongoing and future neoadjuvant studies should include translational end points to identify early resistance mechanisms and to develop novel biomarkers, enabling personalized neoadjuvant hormonal treatment for patients with high-risk prostate cancer.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Steroidogenesis and mechanisms of hormonal therapies.
Fig. 2: Difficulties pathologists might encounter when determining minimal residual disease defined as remaining maximal tumour diameter <5 mm following neoadjuvant treatment.
Fig. 3: Histological features following neoadjuvant hormonal therapy.
Fig. 4: Possible resistance mechanisms to neoadjuvant hormone-based therapy.
Fig. 5: Different phases in the development of predictive biomarkers in the neoadjuvant setting.
Fig. 6: Future perspectives of biomarker-guided studies.

Similar content being viewed by others

References

  1. Sung, H. et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 71, 209–249 (2021).

    PubMed  Google Scholar 

  2. Cooperberg, M. R., Broering, J. M. & Carroll, P. R. Time trends and local variation in primary treatment of localized prostate cancer. J. Clin. Oncol. 28, 1117–1123 (2010).

    PubMed  PubMed Central  Google Scholar 

  3. Zelic, R. et al. Predicting prostate cancer death with different pretreatment risk stratification tools: a head-to-head comparison in a nationwide cohort study. Eur. Urol. 77, 180–188 (2020).

    PubMed  Google Scholar 

  4. Joniau, S. et al. Stratification of high-risk prostate cancer into prognostic categories: a European multi-institutional study. Eur. Urol. 67, 157–164 (2015).

    PubMed  Google Scholar 

  5. D’Amico, A. V. et al. Biochemical outcome after radical prostatectomy, external beam radiation therapy, or interstitial radiation therapy for clinically localized prostate cancer. JAMA 280, 969–974 (1998).

    PubMed  Google Scholar 

  6. Sanda, M. G. et al. Clinically localized prostate cancer: AUA/ASTRO/SUO guideline. Part I: risk stratification, shared decision making, and care options. J. Urol. 199, 683–690 (2018).

    PubMed  Google Scholar 

  7. Dasgupta, P., Davis, J. & Hughes, S. NICE guidelines on prostate cancer 2019. BJU Int. 124, 1 (2019).

    PubMed  Google Scholar 

  8. Mottet, N. et al. EAU-EANM-ESTRO-ESUR-SIOG guidelines on prostate cancer — 2020 Update. Part 1: screening, diagnosis, and local treatment with curative intent. Eur. Urol. 79, 243–262 (2021).

    CAS  PubMed  Google Scholar 

  9. Albertsen, P. C., Hanley, J. A. & Fine, J. 20-Year outcomes following conservative management of clinically localized prostate cancer. JAMA 293, 2095 (2005).

    CAS  PubMed  Google Scholar 

  10. Akre, O. et al. Mortality among men with locally advanced prostate cancer managed with noncurative intent: a nationwide study in PCBaSe Sweden. Eur. Urol. 60, 554–563 (2011).

    PubMed  Google Scholar 

  11. Rider, J. R. et al. Long-term outcomes among noncuratively treated men according to prostate cancer risk category in a nationwide, population-based study. Eur. Urol. 63, 88–96 (2013).

    PubMed  Google Scholar 

  12. Moris, L. et al. Benefits and risks of primary treatments for high-risk localized and locally advanced prostate cancer: an international multidisciplinary systematic review. Eur. Urol. 77, 614–627 (2020).

    CAS  PubMed  Google Scholar 

  13. Van den Broeck, T. et al. Prognostic value of biochemical recurrence following treatment with curative intent for prostate cancer: a systematic review. Eur. Urol. 75, 967–987 (2018).

    PubMed  Google Scholar 

  14. Briganti, A. et al. Natural history of surgically treated high-risk prostate cancer. Urol. Oncol. Semin. Orig. Investig. 33, 163.e7–163.e13 (2015).

    Google Scholar 

  15. Jackson, W. C. et al. Intermediate endpoints after postprostatectomy radiotherapy: 5-year distant metastasis to predict overall survival. Eur. Urol. 74, 413–419 (2018).

    PubMed  Google Scholar 

  16. Bolla, M. et al. Postoperative radiotherapy after radical prostatectomy for high-risk prostate cancer: Long-term results of a randomised controlled trial (EORTC trial 22911). Lancet 380, 2018–2027 (2012).

    PubMed  Google Scholar 

  17. Kent, E. C. & Hussain, M. H. Neoadjuvant therapy for prostate cancer: an Oncologist’s perspective. Rev. Urol. 5, S28–S37 (2003).

    PubMed  PubMed Central  Google Scholar 

  18. Petrelli, F. et al. Correlation of pathologic complete response with survival after neoadjuvant chemotherapy in bladder cancer treated with cystectomy: a meta-analysis. Eur. Urol. 65, 350–357 (2014).

    PubMed  Google Scholar 

  19. Berger, A. C. et al. Complete response to neoadjuvant chemoradiotherapy in esophageal carcinoma is associated with significantly improved survival. J. Clin. Oncol. 23, 4330–4337 (2005).

    PubMed  Google Scholar 

  20. Amiri-Kordestani, L. et al. First FDA approval of neoadjuvant therapy for breast cancer: Pertuzumab for the treatment of patients with HER2-positive breast cancer. Clin. Cancer Res. 20, 5359–5364 (2014).

    CAS  PubMed  Google Scholar 

  21. Kidane, B., Coughlin, S., Vogt, K. & Malthaner, R. Preoperative chemotherapy for resectable thoracic esophageal cancer. Cochrane Database of Syst. Rev. 2015, CD001556 (2015).

    Google Scholar 

  22. Crawford, E. D. et al. Androgen-targeted therapy in men with prostate cancer: evolving practice and future considerations. Prostate Cancer Prostatic Dis. 22, 24–38 (2018).

    PubMed  PubMed Central  Google Scholar 

  23. Bianco, F. J. et al. Proliferation of prostate cancer cells in the bone marrow predicts recurrence in patients with localized prostate cancer. Prostate 49, 235–242 (2001).

    PubMed  Google Scholar 

  24. Wood, D. P. & Banerjee, M. Presence of circulating prostate cells in the bone marrow of patients undergoing radical prostatectomy is predictive of disease-free survival. J. Clin. Oncol. 15, 3451–3457 (1997).

    PubMed  Google Scholar 

  25. Köllermann, J. et al. Prognostic significance of disseminated tumor cells in the bone marrow of prostate cancer patients treated with neoadjuvant hormone treatment. J. Clin. Oncol. 26, 4928–4933 (2008).

    PubMed  Google Scholar 

  26. Cher, M. L. et al. Cellular proliferation and prevalence of micrometastatic cells in the bone marrow of patients with clinically localized prostate cancer. Clin. Cancer Res. 5, 2421–2425 (1999).

    CAS  PubMed  Google Scholar 

  27. Morgan, T. M. et al. Disseminated tumor cells in prostate cancer patients after radical prostatectomy and without evidence of disease predicts biochemical recurrence. Clin. Cancer Res. 15, 677–683 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Melchior, S. W. et al. Early tumor cell dissemination in patients with clinically localized carcinoma of the prostate. Clin. Cancer Res. 3, 249–256 (1997).

    CAS  PubMed  Google Scholar 

  29. Berg, A. et al. Impact of disseminated tumor cells in bone marrow at diagnosis in patients with nonmetastatic prostate cancer treated by definitive radiotherapy. Int. J. Cancer 120, 1603–1609 (2007).

    CAS  PubMed  Google Scholar 

  30. Davey, R. A. & Grossmann, M. Androgen receptor structure, function and biology: from bench to bedside. Clin. Biochem. Rev. 37, 3 (2016).

    PubMed  PubMed Central  Google Scholar 

  31. Dai, C., Heemers, H. & Sharifi, N. Androgen signaling in prostate cancer. Cold Spring Harb. Perspect. Med. 7, a030452 (2017).

    PubMed  PubMed Central  Google Scholar 

  32. Mostaghel, E. A. Steroid hormone synthetic pathways in prostate cancer. Transl Androl. Urol. 2, 212–227 (2013).

    PubMed  PubMed Central  Google Scholar 

  33. Mostaghel, E. A. & Nelson, P. S. Intracrine androgen metabolism in prostate cancer progression: mechanisms of castration resistance and therapeutic implications. Best Pract. Res.Clin. Endocrinol. Metab. 22, 243–258 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Cattrini, C. et al. Current treatment options for metastatic hormone-sensitive prostate cancer. Cancers 11, 1355 (2019).

    CAS  PubMed Central  Google Scholar 

  35. Wadosky, K. M. & Koochekpour, S. Therapeutic rationales, progresses, failures, and future directions for advanced prostate cancer. Int. J. Biol. Sci. 12, 409–426 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Ku, S. Y., Gleave, M. E. & Beltran, H. Towards precision oncology in advanced prostate cancer. Nat. Rev. Urol. 16, 645–654 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Tran, C. et al. Development of a second-generation antiandrogen for treatment of advanced prostate cancer. Science 324, 787–790 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Rice, M. A., Malhotra, S. V. & Stoyanova, T. Second-generation antiandrogens: from discovery to standard of care in castration resistant prostate cancer. Front. Oncol. 10, 801 (2019).

    Google Scholar 

  39. Ryan, C. J. et al. Abiraterone acetate plus prednisone versus placebo plus prednisone in chemotherapy-naive men with metastatic castration-resistant prostate cancer (COU-AA-302): Final overall survival analysis of a randomised, double-blind, placebo-controlled phase 3 study. Lancet Oncol. 16, 152–160 (2015).

    CAS  PubMed  Google Scholar 

  40. de Bono, J. S. et al. Abiraterone and increased survival in metastatic prostate cancer. N. Engl. J. Med. 364, 1995–2005 (2011).

    PubMed  PubMed Central  Google Scholar 

  41. Beer, T. M. et al. Enzalutamide in metastatic prostate cancer before chemotherapy. N. Engl. J. Med. 371, 424–433 (2014).

    PubMed  PubMed Central  Google Scholar 

  42. Scher, H. I. et al. Increased survival with enzalutamide in prostate cancer after chemotherapy. N. Engl. J. Med. 367, 1187–1197 (2012).

    CAS  PubMed  Google Scholar 

  43. Smith, M. R. et al. Apalutamide treatment and metastasis-free survival in prostate cancer. N. Engl. J. Med. 378, 1408–1418 (2018).

    CAS  PubMed  Google Scholar 

  44. Hussain, M. et al. PROSPER: a phase 3, randomized, double-blind, placebo (PBO)-controlled study of enzalutamide (ENZA) in men with nonmetastatic castration-resistant prostate cancer (M0 CRPC). J. Clin. Oncol. 36, 3–3 (2018).

    Google Scholar 

  45. Fizazi, K. et al. Darolutamide in nonmetastatic, castration-resistant prostate cancer. N. Engl. J. Med. 380, 1235–1246 (2019).

    CAS  PubMed  Google Scholar 

  46. Fizazi, K. et al. Abiraterone acetate plus prednisone in patients with newly diagnosed high-risk metastatic castration-sensitive prostate cancer (LATITUDE): final overall survival analysis of a randomised, double-blind, phase 3 trial. Artic. Lancet Oncol. 20, 686–700 (2019).

    CAS  Google Scholar 

  47. Armstrong, A. J. et al. Arches: a randomized, phase III study of androgen deprivation therapy with enzalutamide or placebo in men with metastatic hormone-sensitive prostate cancer. J Clin Oncol 37, 2974–2986 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Chi, K. N. et al. Apalutamide for metastatic, castration-sensitive prostate cancer. N. Engl. J. Med. 381, 13–24 (2019).

    CAS  PubMed  Google Scholar 

  49. Carceles-Cordon, M. et al. Cellular rewiring in lethal prostate cancer: the architect of drug resistance. Nat. Rev. Urol. 17, 292–307 (2020).

    PubMed  PubMed Central  Google Scholar 

  50. Goldie, J. H. & Coldman, A. J. A mathematic model for relating the drug sensitivity of tumors to their spontaneous mutation rate. Cancer Treat. Rep. 63, 1727–1733 (1979).

    CAS  PubMed  Google Scholar 

  51. Goldie, J. H. Mathematical models of drug resistance and chemotherapy effects. Cancer Treat. Res. 48, 13–26 (1989).

    CAS  PubMed  Google Scholar 

  52. Bolla, M. et al. External irradiation with or without long-term androgen suppression for prostate cancer with high metastatic risk: 10-year results of an EORTC randomised study. Lancet Oncol. 11, 1066–1073 (2010).

    CAS  PubMed  Google Scholar 

  53. Iversen, P. et al. Antiandrogen monotherapy in patients with localized or locally advanced prostate cancer: final results from the bicalutamide Early Prostate Cancer programme at a median follow-up of 9.7 years. BJU Int. 105, 1074–1081 (2010).

    CAS  PubMed  Google Scholar 

  54. Bolla, M. et al. Long-term results with immediate androgen suppression and external irradiation in patients with locally advanced prostate cancer (an EORTC study): A phase III randomised trial. Lancet 360, 103–108 (2002).

    CAS  PubMed  Google Scholar 

  55. See, W. A. & Tyrrell, C. J. The addition of bicalutamide 150 mg to radiotherapy significantly improves overall survival in men with locally advanced prostate cancer. J. Cancer Res. Clin. Oncol. 132, 7–16 (2006).

    Google Scholar 

  56. Denham, J. W. et al. Short-term androgen suppression and radiotherapy versus intermediate-term androgen suppression and radiotherapy, with or without zoledronic acid, in men with locally advanced prostate cancer (TROG 03.04 RADAR): 10-year results from a randomised, phase 3, factorial trial. Lancet Oncol. 20, 267–281 (2019).

    CAS  PubMed  Google Scholar 

  57. Roach, M. et al. Short-term neoadjuvant androgen deprivation therapy and external-beam radiotherapy for locally advanced prostate cancer: long-term results of RTOG 8610. J. Clin. Oncol. 26, 585–591 (2008).

    PubMed  Google Scholar 

  58. Spratt, D. E. et al. Androgen receptor upregulation mediates radioresistance after ionizing radiation. Cancer Res. 75, 4688–4696 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Bartek, J., Mistrik, M. & Bartkova, J. Androgen receptor signaling fuels DNA repair and radioresistance in prostate cancer. Cancer Discov. 3, 1222–1224 (2013).

    CAS  PubMed  Google Scholar 

  60. Polkinghorn, W. R. et al. Androgen receptor signaling regulates DNA repair in prostate cancers. Cancer Discov. 3, 1245–1253 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Boevé, L. M. S. et al. Effect on survival of androgen deprivation therapy alone compared to androgen deprivation therapy combined with concurrent radiation therapy to the prostate in patients with primary bone metastatic prostate cancer in a prospective randomised clinical trial: data from the HORRAD trial. Eur. Urol. 75, 410–418 (2019).

    PubMed  Google Scholar 

  62. Parker, C. C. et al. Radiotherapy to the primary tumour for newly diagnosed, metastatic prostate cancer (STAMPEDE): a randomised controlled phase 3 trial. Lancet 392, 2353–2366 (2018).

    PubMed  PubMed Central  Google Scholar 

  63. Burdett, S. et al. Prostate radiotherapy for metastatic hormone-sensitive prostate cancer: A STOPCAP systematic review and meta-analysis. Eur. Urol. 76, 115–124 (2019).

    PubMed  PubMed Central  Google Scholar 

  64. Sooriakumaran, P. et al. TRoMbone: Testing radical prostatectomy in men with oligo metastatic prostate cancer that has spread to the bone - a randomized controlled feasibility trial [abstract]. Eur. Urol. Suppl. 18, e2199 (2019).

    Google Scholar 

  65. Isbarn, H. et al. Androgen deprivation therapy for the treatment of prostate cancer: consider both benefits and risks. Eur. Urol. 55, 62–75 (2009).

    CAS  PubMed  Google Scholar 

  66. Sonpavde, G. & Sternberg, C. N. Neoadjuvant systemic therapy for urological malignancies. BJU Int. 106, 6–22 (2009).

    Google Scholar 

  67. van der Kwast, T. H. et al. Prolonged neoadjuvant combined androgen blockade leads to a further reduction of prostatic tumor volume: three versus six months of endocrine therapy. Urology 53, 523–529 (1999).

    PubMed  Google Scholar 

  68. Soloway, M. S. et al. Neoadjuvant androgen ablation before radical prostatectomy in cT2bNxMo prostate cancer: 5-year results. J. Urol. 167, 112–116 (2002).

    PubMed  Google Scholar 

  69. Fair, W. R. et al. The indications, rationale, and results of neoadjuvant androgen deprivation in the treatment of prostatic cancer: Memorial Sloan-Kettering Cancer Center results. Urology 49, 46–55 (1997).

    CAS  PubMed  Google Scholar 

  70. Meyer, F. et al. Neoadjuvant hormonal therapy before radical prostatectomy and risk of prostate specific antigen failure. J. Urol. 162, 2024–2028 (1999).

    CAS  PubMed  Google Scholar 

  71. Berglund, R. K. et al. Ten-year follow-up of neoadjuvant therapy with goserelin acetate and flutamide before radical prostatectomy for clinical T3 and T4 prostate cancer: update on Southwest Oncology Group Study 9109. Urology 79, 633–637 (2012).

    PubMed  Google Scholar 

  72. Goldenberg, S. L. et al. Randomized, prospective, controlled study comparing radical prostatectomy alone and neoadjuvant androgen withdrawal in the treatment of localized prostate cancer. J. Urol. 156, 873–877 (1996).

    CAS  PubMed  Google Scholar 

  73. Labrie, F. et al. Neoadjuvant hormonal therapy: the Canadian experience. Urology 49, 56–64 (1997).

    CAS  PubMed  Google Scholar 

  74. Selli, C. et al. Effects of complete androgen blockade for 12 and 24 weeks on the pathological stage and resection margin status of prostate cancer. J. Clin. Pathol. 55, 508–513 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Gleave, M. E. et al. Randomized comparative study of 3 versus 8-month neoadjuvant hormonal therapy before radical prostatectomy: biochemical and pathological effects. J. Urol. 166, 500–507 (2001).

    CAS  PubMed  Google Scholar 

  76. Prezioso, D., Lotti, T., Polito, M. & Montironi, R. Neoadjuvant hormone treatment with leuprolide acetate depot 3.75 mg and Cyproterone acetate, before radical prostatectomy: a randomized study. Urol. Int. 72, 189–195 (2004).

    CAS  PubMed  Google Scholar 

  77. Dalkin, B. L., Ahmann, F. R., Nagle, R. & Johnson, C. S. Randomized study of neoadjuvant testicular androgen ablation therapy before radical prostatectomy in men with clinically localized prostate cancer. J. Urol. 155, 1357–1360 (1996).

    CAS  PubMed  Google Scholar 

  78. Klotz, L. H. et al. Long-term followup of a randomized trial of 0 versus 3 months of neoadjuvant androgen ablation before radical prostatectomy. J. Urol. 170, 791–794 (2003).

    CAS  PubMed  Google Scholar 

  79. Van Poppel, H. et al. Neoadjuvant hormonal therapy before radical prostatectomy decreases the number of positive surgical margins in stage T2 prostate cancer: interim results of a prospective randomized trial. The Belgian Uro-Oncological Study Group. J.Urol. 154, 429–434 (1995).

    PubMed  Google Scholar 

  80. Aus, G. et al. Three-month neoadjuvant hormonal therapy before radical prostatectomy: a 7-year follow-up of a randomized controlled trial. BJU Int. 90, 561–566 (2002).

    CAS  PubMed  Google Scholar 

  81. Schulman, C. C. et al. 4-Year follow-up results of a European prospective randomized study on neoadjuvant hormonal therapy prior to radical prostatectomy in T2-3N0M0 prostate cancer. Eur. Urol. 38, 706–713 (2000).

    CAS  PubMed  Google Scholar 

  82. Shelley, M. D. et al. A systematic review and meta-analysis of randomised trials of neo-adjuvant hormone therapy for localised and locally advanced prostate carcinoma. Cancer Treat. Rev. 35, 9–17 (2009).

    CAS  PubMed  Google Scholar 

  83. Lou, D. Y. & Fong, L. Neoadjuvant therapy for localized prostate cancer: examining mechanism of action and efficacy within the tumor. Urol. Oncol. 34, 182–192 (2016).

    CAS  PubMed  Google Scholar 

  84. Mostaghel, E. A. et al. Intraprostatic androgens and androgen-regulated gene expression persist after testosterone suppression: therapeutic implications for castration-resistant prostate cancer. Cancer Res. 67, 5033–5041 (2007).

    CAS  PubMed  Google Scholar 

  85. Mostaghel, E. A. et al. Targeted androgen pathway suppression in localized prostate cancer: a pilot study. J. Clin. Oncol. 32, 229–237 (2014).

    PubMed  Google Scholar 

  86. Gregory, C. W., Johnson, R. T., Mohler, J. L., French, F. S. & Wilson, E. M. Androgen receptor stabilization in recurrent prostate cancer is associated with hypersensitivity to low androgen. Cancer Res. 61, 2892–2898 (2001).

    CAS  PubMed  Google Scholar 

  87. McKay, R. R. et al. Evaluation of intense androgen deprivation before prostatectomy: a randomized phase II trial of enzalutamide and leuprolide with or without abiraterone. J. Clin. Oncol. 11, 1–10 (2019).

    Google Scholar 

  88. Montgomery, B. et al. Neoadjuvant enzalutamide prior to prostatectomy. Clin. Cancer Res. 23, 2169–2176 (2017).

    CAS  PubMed  Google Scholar 

  89. Graham, L. S. et al. Targeting backdoor androgen synthesis through AKR1C3 inhibition: A presurgical hormonal ablative neoadjuvant trial in high-risk localized prostate cancer. Prostate 81, 418–426 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Taplin, M. E. et al. Intense androgen-deprivation therapy with abiraterone acetate plus leuprolide acetate in patients with localized high-risk prostate cancer: results of a randomized phase II neoadjuvant study. J. Clin. Oncol. 32, 3705–3715 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Efstathiou, E. et al. Neoadjuvant apalutamide (APA) plus leuprolide (LHRHa) with or without abiraterone (AA) in localized high-risk prostate cancer (LHRPC). J. Clin.Oncol. 38, Abstr. 5504–5504 (2020).

    Google Scholar 

  92. Efstathiou, E. et al. Clinical and biological characterisation of localised high-risk prostate cancer: results of a randomised preoperative study of a luteinising hormone-releasing hormone agonist with or without abiraterone acetate plus prednisone. Eur. Urol. 76, 418–424 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Aslim, E. J. et al. Neoadjuvant apalutamide (arn-509) and radical prostatectomy in treatment of intermediate to high risk prostate cancer (NEAR) — initial results of a phase II trial [abstract]. Eur. Urol. Suppl. 18, e1394 (2019).

    Google Scholar 

  94. Corcoran, N. et al. The predictive value of ARv7 expression in localized prostate caner treated with abiraterone, degarelix, and bicalutamide. J. Clin. Oncol. 33, Abstr. 71 (2015).

    Google Scholar 

  95. McKay, R. R. et al. Results of a randomized phase II trial of intense androgen deprivation therapy prior to radical prostatectomy in men with high-risk localized prostate cancer. J. Urol. 206, 80–87 (2021).

    PubMed  Google Scholar 

  96. Karzai, F. et al. Sequential prostate magnetic resonance imaging in newly diagnosed high-risk prostate cancer treated with neoadjuvant enzalutamide is predictive of therapeutic response. Clin. Cancer Res. 27, 429–437 (2021).

    CAS  PubMed  Google Scholar 

  97. Wilkinson, S. et al. Nascent prostate cancer heterogeneity drives evolution and resistance to intense hormonal therapy. Eur. Urol. https://doi.org/10.1016/j.eururo.2021.03.009 (2021).

    Article  PubMed  Google Scholar 

  98. O’Donnell, A. et al. Hormonal impact of the 17α-hydroxylase/C17,20-lyase inhibitor abiraterone acetate (CB7630) in patients with prostate cancer. Br. J. Cancer 90, 2317–2325 (2004).

    PubMed  PubMed Central  Google Scholar 

  99. Chen, M. E. et al. A streamlined three-dimensional volume estimation method accurately classifies prostate tumors by volume. Am. J. Surg. Pathol. 27, 1291–1301 (2003).

    PubMed  Google Scholar 

  100. Symmans, W. F. et al. Measurement of residual breast cancer burden to predict survival after neoadjuvant chemotherapy. J. Clin. Oncol. 25, 4414–4422 (2007).

    PubMed  Google Scholar 

  101. Yang, X. Y. et al. Initial patient-reported outcomes of a phase II neoadjuvant apalutamide (ARN-509) and radical prostatectomy in treatment of intermediate to high risk prostate cancer (NEAR) trial [abstract]. Eur. Urol. Suppl. 18, e1402 (2019).

    Google Scholar 

  102. Tamae, D. et al. The DHEA-sulfate depot following P450c17 inhibition supports the case for AKR1C3 inhibition in high risk localized and advanced castration resistant prostate cancer. Chem. Biol. Interact. 234, 332–338 (2015).

    CAS  PubMed  Google Scholar 

  103. Hofland, J. et al. Evidence of limited contributions for intratumoral steroidogenesis in prostate cancer. Cancer Res. 70, 1256–1264 (2010).

    CAS  PubMed  Google Scholar 

  104. Locke, J. A. et al. Androgen levels increase by intratumoral de novo steroidogenesis during progression of castration-resistant prostate cancer. Cancer Res. 68, 6407–6415 (2008).

    CAS  PubMed  Google Scholar 

  105. Liu, C. et al. Intracrine androgens and AKR1C3 activation confer resistance to enzalutamide in prostate cancer. Cancer Res. 75, 1413–1422 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Sowalsky, A. G. et al. Neoadjuvant-intensive androgen deprivation therapy selects for prostate tumor foci with diverse subclonal oncogenic alterations. Cancer Res. 78, 4716–4730 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Deng, Q. & Tang, D. G. Androgen receptor and prostate cancer stem cells: Biological mechanisms and clinical implications. Endocr. Relat. Cancer 22, T209–T220 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Eigl, B. J. C. et al. Timing is everything: preclinical evidence supporting simultaneous rather than sequential chemohormonal therapy for prostate cancer. Clin. Cancer Res. 11, 4905–4911 (2005).

    CAS  PubMed  Google Scholar 

  109. Fizazi, K. et al. Androgen deprivation therapy plus docetaxel and estramustine versus androgen deprivation therapy alone for high-risk localised prostate cancer (GETUG 12): a phase 3 randomised controlled trial. Lancet Oncol. 16, 787–794 (2015).

    CAS  PubMed  Google Scholar 

  110. Rosenthal, S. A. et al. Effect of chemotherapy with docetaxel with androgen suppression and radiotherapy for localized high-risk prostate cancer: the randomized phase III NRG Oncology RTOG 0521 Trial. J. Clin. Oncol. 37, 1159–1168 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Kyriakopoulos, C. E. et al. Chemohormonal therapy in metastatic hormone-sensitive prostate cancer: long-term survival analysis of the randomized phase III E3805 CHAARTED trial. J. Clin. Oncol. 36, 1080–1087 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Thalgott, M. et al. Long-term results of a phase II study with neoadjuvant docetaxel chemotherapy and complete androgen blockade in locally advanced and high-risk prostate cancer. J. Hematol. Oncol. 7, 20 (2014).

    PubMed  PubMed Central  Google Scholar 

  113. Silberstein, J. L. et al. Long-term oncological outcomes of a phase II trial of neoadjuvant chemohormonal therapy followed by radical prostatectomy for patients with clinically localised, high-risk prostate cancer. BJU Int. 116, 50–56 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Koie, T. et al. Safety and effectiveness of neoadjuvant luteinizing hormone-releasing hormone agonist plus low-dose estramustine phosphate in high-risk prostate cancer: a prospective single-arm study. Prostate Cancer Prostatic Dis. 15, 397–401 (2012).

    CAS  PubMed  Google Scholar 

  115. Narita, S. et al. Short-term clinicopathological outcome of neoadjuvant chemohormonal therapy comprising complete androgen blockade, followed by treatment with docetaxel and estramustine phosphate before radical prostatectomy in Japanese patients with high-risk localized prostate cancer. World J. Surg. Oncol. 10, 1 (2012).

    PubMed  PubMed Central  Google Scholar 

  116. Zurita, A. J. et al. Integrating chemohormonal therapy and surgery in known or suspected lymph node metastatic prostate cancer. Prostate Cancer Prostatic Dis. 18, 276–280 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Nosov, A. et al. Safety and efficacy of neoadjuvant chemohormonal and hormonal treatment followed by radical prostatectomy for patients with high- and very high risk prostate cancer: Initial results of prospective, randomized, phase III clinical trial [abstract]. Eur. Urol. Suppl. 15, e1193 (2016).

    Google Scholar 

  118. Chi, K. N. et al. Multicenter phase II study of combined neoadjuvant docetaxel and hormone therapy before radical prostatectomy for patients with high risk localized prostate cancer. J. Urol. 180, 565–570 (2008).

    CAS  PubMed  Google Scholar 

  119. Eastham, J. A. et al. Cancer and Leukemia Group B 90203 (Alliance): radical prostatectomy with or without neoadjuvant chemohormonal therapy in localized, high-risk prostate cancer. J. Clin. Oncol. 38, 3042–3050 (2020).

    PubMed  PubMed Central  Google Scholar 

  120. Gharzai, L. A. et al. Intermediate clinical endpoints for surrogacy in localised prostate cancer: an aggregate meta-analysis. Lancet Oncol. 22, 402–410 (2021).

    PubMed  Google Scholar 

  121. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT03436654 (2020).

  122. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/results/NCT03279250 (2021).

  123. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT03821246 (2021).

  124. US National Library of Medicine. ClinicalTrials.gov https://www.clinicaltrials.gov/ct2/show/NCT04356430 (2020).

  125. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02949284 (2021).

  126. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02903368 (2020).

  127. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02770391 (2021).

  128. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02789878 (2021).

  129. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT03767244 (2021).

  130. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT03080116 (2020).

  131. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02543255 (2021).

  132. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT03297385 (2017).

  133. Wilkinson, S. & Sowalsky, A. G. Battling the two-headed dragon of prostate cancer targeted therapy. Mol. Cell Oncol. 7, 1745037 (2020).

    PubMed  PubMed Central  Google Scholar 

  134. Mckay, R. R. et al. Post prostatectomy outcomes of patients with high-risk prostate cancer treated with neoadjuvant androgen blockade. Prostate Cancer Prostatic Dis. 21, 364–372 (2018).

    PubMed  Google Scholar 

  135. McKay, R. R. et al. Outcomes post neoadjuvant intense hormone therapy and surgery for patients with high-risk localized prostate cancer: results of a pooled analysis of contemporary clinical trials. J. Urol. 205, 1689–1697 (2021).

    PubMed  Google Scholar 

  136. Rödel, C. et al. Prognostic significance of tumor regression after preoperative chemoradiotherapy for rectal cancer. J. Clin. Oncol. 23, 8688–8696 (2005).

    PubMed  Google Scholar 

  137. Grossman, H. B. et al. Neoadjuvant chemotherapy plus cystectomy compared with cystectomy alone for locally advanced bladder cancer. N. Engl. J. Med. 349, 859–866 (2003).

    CAS  PubMed  Google Scholar 

  138. Gianni, L. et al. Efficacy and safety of neoadjuvant pertuzumab and trastuzumab in women with locally advanced, inflammatory, or early HER2-positive breast cancer (NeoSphere): a randomised multicentre, open-label, phase 2 trial. Lancet Oncol. 13, 25–32 (2012).

    CAS  PubMed  Google Scholar 

  139. Cortazar, P. et al. Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis. Lancet 384, 164–172 (2014).

    PubMed  Google Scholar 

  140. Peintinger, F. et al. Reproducibility of residual cancer burden for prognostic assessment of breast cancer after neoadjuvant chemotherapy. Mod. Pathol. 28, 913–920 (2015).

    PubMed  PubMed Central  Google Scholar 

  141. Kollermann, J. et al. Prognosis of stage pT0 after prolonged neoadjuvant endocrine therapy of prostate cancer: a matched-pair analysis. Eur. Urol. 45, 42–45 (2004).

    PubMed  Google Scholar 

  142. Barry, M., Perner, S., Demichelis, F. & Rubin, M. A. TMPRSS2-ERG fusion heterogeneity in multifocal prostate cancer: clinical and biologic implications. Urology 70, 630–633 (2007).

    PubMed  Google Scholar 

  143. Meiers, I., Waters, D. J. & Bostwick, D. G. Preoperative prediction of multifocal prostate cancer and application of focal therapy: review 2007. Urology 70, 3–8 (2007).

    PubMed  Google Scholar 

  144. Fraser, M. et al. Genomic hallmarks of localized, non-indolent prostate cancer. Nature 541, 359–364 (2017).

    CAS  PubMed  Google Scholar 

  145. Espiritu, S. M. G. et al. The evolutionary landscape of localized prostate cancers drives clinical aggression. Cell 173, 1003–1013.e15 (2018).

    CAS  PubMed  Google Scholar 

  146. Neri, A. et al. Clinical significance of multifocal and multicentric breast cancers and choice of surgical treatment: a retrospective study on a series of 1158 cases. BMC Surg. 15, 1–10 (2015).

    PubMed  PubMed Central  Google Scholar 

  147. Joergensen, L. E., Gunnarsdottir, K. A., Lanng, C., Moeller, S. & Rasmussen, B. B. Multifocality as a prognostic factor in breast cancer patients registered in Danish Breast Cancer Cooperative Group (DBCG) 1996–2001. Breast 17, 587–591 (2008).

    CAS  PubMed  Google Scholar 

  148. Ogston, K. N. et al. A new histological grading system to assess response of breast cancers to primary chemotherapy: prognostic significance and survival. Breast 12, 320–327 (2003).

    PubMed  Google Scholar 

  149. Smith, I. C. et al. Neoadjuvant chemotherapy in breast cancer: significantly enhanced response with docetaxel. J. Clin. Oncol. 20, 1456–1466 (2002).

    CAS  PubMed  Google Scholar 

  150. Evans, A. J. Treatment effects in prostate cancer. Mod. Pathol. 31, 110–121 (2018).

    Google Scholar 

  151. Têtu, B. Morphological changes induced by androgen blockade in normal prostate and prostatic carcinoma. Best Pract. Res. 22, 271–283 (2008).

    Google Scholar 

  152. Thomsen, O. Ø. et al. A comparison of budesonide and mesalamine for active Crohn’s disease. N. Engl. J. Med. 339, 370–374 (1998).

    CAS  PubMed  Google Scholar 

  153. Penault-Llorca, F. et al. Comparison of the prognostic significance of Chevallier and Sataloff’s pathologic classifications after neoadjuvant chemotherapy of operable breast cancer. Hum. Pathol. 39, 1221–1228 (2008).

    PubMed  Google Scholar 

  154. Pinder, S. E. et al. Macroscopic handling and reporting of breast cancer specimens pre- and post-neoadjuvant chemotherapy treatment: review of pathological issues and suggested approaches. Histopathology 67, 279–293 (2015).

    PubMed  Google Scholar 

  155. Pinder, S. E., Provenzano, E., Earl, H. & Ellis, I. O. Laboratory handling and histology reporting of breast specimens from patients who have received neoadjuvant chemotherapy. Histopathology 50, 409–417 (2007).

    CAS  PubMed  Google Scholar 

  156. Epstein, J. I. et al. The 2005 International Society of Urological Pathology (ISUP) consensus conference on Gleason grading of prostatic carcinoma. Am. J. Surg. Pathol. 29, 1228–1242 (2005).

    PubMed  Google Scholar 

  157. Efstathiou, E. et al. Morphologic characterization of preoperatively treated prostate cancer: toward a post-therapy histologic classification. Eur. Urol. 57, 1030–1038 (2010).

    PubMed  Google Scholar 

  158. Murphy, C. et al. A novel system for estimating residual disease and pathologic response to neoadjuvant treatment of prostate cancer. Prostate 76, 1285–1292 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  159. Dogdas, B. et al. Computational pathological identification of prostate cancer following neoadjuvant treatment. J. Clin. Oncol. 38, Abstr. e14052–e14052 (2020).

    Google Scholar 

  160. Gold, S. A. et al. mpMRI preoperative staging in men treated with antiandrogen and androgen deprivation therapy before robotic prostatectomy. Urol. Oncol. Semin. Orig. Investig. 37, 352.e25–352.e30 (2019).

    CAS  Google Scholar 

  161. Hövels, A. M. et al. The diagnostic accuracy of CT and MRI in the staging of pelvic lymph nodes in patients with prostate cancer: a meta-analysis. Clin. Radiol. 63, 387–395 (2008).

    PubMed  Google Scholar 

  162. Maurer, T., Eiber, M., Schwaiger, M. & Gschwend, J. E. Current use of PSMA–PET in prostate cancer management. Nat. Rev. Urol. 13, 226–235 (2016).

    CAS  PubMed  Google Scholar 

  163. Perera, M. et al. Sensitivity, specificity, and predictors of positive 68 Ga–prostate-specific membrane antigen positron emission tomography in advanced prostate cancer: a systematic review and meta-analysis. Eur. Urol. 70, 926–937 (2016).

    PubMed  Google Scholar 

  164. Calais, J. et al. 68Ga-PSMA-11 PET/CT mapping of prostate cancer biochemical recurrence after radical prostatectomy in 270 patients with a PSA level of less than 1.0 ng/ml: impact on salvage radiotherapy planning. J. Nucl. Med. 59, 230–237 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  165. McCarthy, M., Francis, R., Tang, C., Watts, J. & Campbell, A. A multicenter prospective clinical trial of 68 gallium PSMA HBED-CC PET-CT restaging in biochemically relapsed prostate carcinoma: oligometastatic rate and distribution compared with standard imaging. Int. J. Radiat. Oncol. Biol. Phys. 104, 801–808 (2019).

    PubMed  Google Scholar 

  166. Hofman, M. S. et al. Prostate-specific membrane antigen PET-CT in patients with high-risk prostate cancer before curative-intent surgery or radiotherapy (proPSMA): a prospective, randomised, multicentre study. Lancet 395, 1208–1216 (2020).

    CAS  PubMed  Google Scholar 

  167. Tosco, L. et al. Neoadjuvant degarelix with or without apalutamide followed by radical prostatectomy for intermediate and high-risk prostate cancer: ARNEO, a randomized, double blind, placebo-controlled trial. BMC Cancer 18, 354 (2018).

    PubMed  PubMed Central  Google Scholar 

  168. Cho, S. et al. Preliminary analysis of PSMA-targeted 18F-DCFPyL PET/MRI for assessment of response to neo-adjuvant chemohormonal therapy in men with high-risk primary prostate cancer. J. Nucl. Med. 61, 1264 (2020).

    Google Scholar 

  169. Chen, M. et al. Can 68 Ga-PSMA-11 positorn emission technology/computerized tomography predict pathologic response of primary prostate cancer to neoadjuvant androgen deprivation therapy?: A Pilot Study. J. Urol. 205, 1082–1089 (2020).

    PubMed  Google Scholar 

  170. Vaz, S. et al. Influence of androgen deprivation therapy on PSMA expression and PSMA-ligand PET imaging of prostate cancer patients. Eur. J. Nucl. Med. Mol. Imaging 47, 9–15 (2020).

    CAS  PubMed  Google Scholar 

  171. Batra, J. S., Pienta, K. J., Pomper, M. G., Gorin, M. A. & Rowe, S. P. Can the interplay between androgen signaling and PSMA expression be leveraged for theranostic applications? Transl Androl. Urol. 8, S263–S264 (2019).

    PubMed  PubMed Central  Google Scholar 

  172. Evans, M. J. et al. Noninvasive measurement of androgen receptor signaling with a positron-emitting radiopharmaceutical that targets prostate-specific membrane antigen. Proc. Natl Acad. Sci. USA 108, 9578–9582 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  173. Spans, L. et al. The genomic landscape of prostate cancer. Int. J. Mol. Sci. 14, 10822–10851 (2013).

    PubMed  PubMed Central  Google Scholar 

  174. Devlies, W. et al. Clinical actionability of the genomic landscape of metastatic castration resistant prostate cancer. Cells 9, 2494 (2020).

    CAS  PubMed Central  Google Scholar 

  175. Beltran, H. et al. Impact of therapy on genomics and transcriptomics in high-risk prostate cance treated with neoadjuvant docetaxel and androgen deprivation therapy. Clin. Cancer Res. 23, 6802–6811 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  176. Handle, F. & Claessens, F. AR variants: lost in translation to clinical practice? Nat. Rev. Urol. 16, 451–452 (2019).

    PubMed  Google Scholar 

  177. Antonarakis, E. S. et al. AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer. N. Engl. J. Med. 371, 1028–1038 (2014).

    PubMed  PubMed Central  Google Scholar 

  178. Arora, V. K. et al. Glucocorticoid receptor confers resistance to antiandrogens by bypassing androgen receptor blockade. Cell 155, 1309–1322 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  179. Puhr, M. et al. The glucocorticoid receptor is a key player for prostate cancer cell survival and a target for improved antiandrogen therapy. Clin. Cancer Res. 24, 927–938 (2018).

    CAS  PubMed  Google Scholar 

  180. Claessens, F., Joniau, S. & Helsen, C. Comparing the rules of engagement of androgen and glucocorticoid receptors. Cell. Mol. Life Sci. 74, 2217–2228 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  181. Xie, N. et al. The expression of glucocorticoid receptor is negatively regulated by active androgen receptor signaling in prostate tumors. Int. J. Cancer 136, E27–E38 (2015).

    CAS  PubMed  Google Scholar 

  182. Li, J. et al. Aberrant corticosteroid metabolism in tumor cells enables GR takeover in enzalutamide resistant prostate cancer. eLife 6, e20183 (2017).

    PubMed  PubMed Central  Google Scholar 

  183. Kaochar, S. & Mitsiades, N. Glucocorticoids mediate adverse events of deep androgen receptor axis inhibition in prostate cancer patients. Ann. Oncol. 31, 323–325 (2020).

    CAS  PubMed  Google Scholar 

  184. Taplin, M. E. et al. A phase II study of mifepristone (RU-486) in castration-resistant prostate cancer, with a correlative assessment of androgen-related hormones. BJU Int. 101, 1084–1089 (2008).

    CAS  PubMed  Google Scholar 

  185. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT02012296 (2018).

  186. Gao, S. et al. ErbB2 signaling increases androgen receptor expression in abiraterone-resistant prostate cancer. Clin. Cancer Res. 22, 3672–3682 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  187. Miller, D. R., Ingersoll, M. A. & Lin, M. F. ErbB-2 signaling in advanced prostate cancer progression and potential therapy. Endocr. Rel. Cancer 26, R195–R209 (2018).

    Google Scholar 

  188. Shiota, M. et al. Inhibition of the HER2-YB1-AR axis with lapatinib synergistically enhances enzalutamide anti-tumor efficacy in castration resistant prostate cancer. Oncotarget 6, 9086–9098 (2015).

    PubMed  PubMed Central  Google Scholar 

  189. Bishop, J. L. et al. PD-L1 is highly expressed in enzalutamide resistant prostate cancer. Oncotarget 6, 234–242 (2015).

    PubMed  Google Scholar 

  190. Liu, J. et al. p300/CBP inhibition enhances the efficacy of programmed death-ligand 1 blockade treatment in prostate cancer. Oncogene 39, 3939–3951 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  191. Lieping, C. & Xue, H. Anti-PD-1/PD-L1 therapy of human cancer: past, present, and future. J. Clin. Invest. 125, 3384–3391 (2015).

    Google Scholar 

  192. Calagua, C. et al. Expression of PD-L1 in hormone-naïve and treated prostate cancer patients receiving neoadjuvant abiraterone acetate plus prednisone and leuprolide. Clin. Cancer Res. 23, 6812–6822 (2017).

    CAS  PubMed  Google Scholar 

  193. Sharma, A. et al. The retinoblastoma tumor suppressor controls androgen signaling and human prostate cancer progression. J. Clin. Invest. 120, 4478–4492 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  194. Tan, H. L. et al. Rb loss is characteristic of prostatic small cell neuroendocrine carcinoma. Clin. Cancer Res. 20, 890–903 (2014).

    CAS  PubMed  Google Scholar 

  195. Beltran, H. et al. Divergent clonal evolution of castration-resistant neuroendocrine prostate cancer. Nat. Med. 22, 298–305 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  196. Wyatt, A. W. et al. Genomic alterations in cell-free DNA and enzalutamide resistance in castration-resistant prostate cancer. JAMA Oncol. 2, 1598–1606 (2016).

    PubMed  PubMed Central  Google Scholar 

  197. Abida, W. et al. Genomic correlates of clinical outcome in advanced prostate cancer. Proc. Natl Acad. Sci. USA 116, 11428–11436 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  198. Carver, B. S. et al. Reciprocal feedback regulation of PI3K and androgen receptor signaling in PTEN-deficient prostate cancer. Cancer Cell 19, 575–586 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  199. Mateo, J. et al. Genomics of lethal prostate cancer at diagnosis and castration-resistance. J. Clin. Invest. 130, 1743–1751 (2019).

    Google Scholar 

  200. Yu, J. et al. An integrated network of androgen receptor, polycomb, and TMPRSS2-ERG gene fusions in prostate cancer progression. Cancer Cell 17, 443–454 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  201. De Laere, B. et al. TP53 outperforms other androgen receptor biomarkers to predict abiraterone or enzalutamide outcome in metastatic castration-resistant prostate cancer. Clin. Cancer Res. 25, 1766–1773 (2019).

    PubMed  Google Scholar 

  202. Aparicio, A. M. et al. Combined tumor suppressor defects characterize clinically defined aggressive variant prostate cancers. Clin. Cancer Res. 22, 1520–1530 (2016).

    CAS  PubMed  Google Scholar 

  203. Castro, E. et al. Germline BRCA mutations are associated with higher risk of nodal involvement, distant metastasis, and poor survival outcomes in prostate cancer. J. Clin. Oncol. 31, 1748–1757 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  204. Marshall, C. H. et al. Prevalence of DNA repair gene mutations in localized prostate cancer according to clinical and pathologic features: association of Gleason score and tumor stage. Prostate Cancer Prostatic Dis. 22, 59–65 (2019).

    CAS  PubMed  Google Scholar 

  205. Isaacsson Velho, P. et al. Molecular characterization and clinical outcomes of primary gleason pattern 5 prostate cancer after radical prostatectomy. JCO Precis. Oncol. 3, 1–13 (2019).

    Google Scholar 

  206. Isaacsson Velho, P. et al. Intraductal/ductal histology and lymphovascular invasion are associated with germline DNA-repair gene mutations in prostate cancer. Prostate 78, 401–407 (2018).

    CAS  PubMed  Google Scholar 

  207. Berchuck, J. E. et al. Impact of pathogenic germline DNA damage repair alterations on response to intense neoadjuvant androgen deprivation therapy in high-risk localized prostate cancer. Eur. Urol. 80, 295–303 (2021).

    CAS  PubMed  Google Scholar 

  208. Scher, H. I. et al. Association of AR-V7 on circulating tumor cells as a treatment-specific biomarker with outcomes and survival in castration-resistant prostate cancer. JAMA Oncol. 2, 1441–1449 (2016).

    PubMed  PubMed Central  Google Scholar 

  209. Antonarakis, E. S. et al. Androgen receptor splice variant 7 and efficacy of taxane chemotherapy in patients with metastatic castration-resistant prostate cancer. JAMA Oncol. 1, 582–591 (2015).

    PubMed  PubMed Central  Google Scholar 

  210. Le, D. T. et al. PD-1 blockade in tumors with mismatch-repair deficiency. N. Engl. J. Med. 372, 2509–2520 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  211. Clarke, N. et al. Olaparib combined with abiraterone in patients with metastatic castration-resistant prostate cancer: a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Oncol. 19, 975–986 (2018).

    CAS  PubMed  Google Scholar 

  212. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT04030559 (2021).

  213. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT04565496 (2020).

  214. Tucker, M. D. et al. Pembrolizumab in men with heavily treated metastatic castration-resistant prostate cancer (mCRPC). J. Clin. Oncol. 37, 172–172 (2019).

    Google Scholar 

  215. Antonarakis, E. S. et al. Clinical features and therapeutic outcomes in men with advanced prostate cancer and DNA mismatch repair gene mutations. Eur. Urol. 75, 378–382 (2019).

    CAS  PubMed  Google Scholar 

  216. Graff, J. N. et al. Early evidence of anti-PD-1 activity in enzalutamide-resistant prostate cancer. Oncotarget 7, 52810–52817 (2016).

    PubMed  PubMed Central  Google Scholar 

  217. Abida, W. et al. Analysis of the prevalence of microsatellite instability in prostate cancer and response to immune checkpoint blockade. JAMA Oncol. 5, 471–478 (2019).

    PubMed  Google Scholar 

  218. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT04737109 (2021).

  219. De Bono, J. S. et al. Randomized phase II study evaluating AKT blockade with ipatasertib, in combination with abiraterone, in patients with metastatic prostate cancer with and without PTEN loss. Clin. Cancer Res. 25, 928–936 (2019).

    PubMed  Google Scholar 

  220. Wang, Z. et al. The diverse roles of SPOP in prostate cancer and kidney cancer. Nat. Rev. Urol. 17, 339–350 (2020).

    CAS  PubMed  Google Scholar 

  221. Boysen, G. et al. SpoP-mutated/CHD1-deleted lethal prostate cancer and abiraterone sensitivity. Clin. Cancer Res. 24, 5585–5593 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  222. Blattner, M. et al. SPOP mutations in prostate cancer across demographically diverse patient cohorts. Neoplasia 16, 14–20 (2014).

    PubMed  PubMed Central  Google Scholar 

  223. McKay, R. R. et al. Results of a phase II trial of intense androgen deprivation therapy prior to radical prostatectomy (RP) in men with high-risk localized prostate cancer (PC). J. Clin. Oncol. 38, 5503–5503 (2020).

    Google Scholar 

  224. Shi, Q. et al. Prostate cancer-associated SPOP mutations enhance cancer cell survival and docetaxel resistance by upregulating Caprin1-dependent stress granule assembly. Mol. Cancer 18, 170 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  225. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT04812366 (2021).

Download references

Acknowledgements

G.D. is a PhD fellow of the Research Foundation Flanders (FWO). W.D. is a recipient of the Emmanuel van der Schueren scholarship of “Kom Op Tegen Kanker”. W.E. is a Senior Clinical Investigator of the FWO. S.J. is a Senior Clinical Investigator of the FWO. This work was supported by the ‘Jozef De Wever Fonds voor prostaatkankerpreventie’ of the KU Leuven.

Author information

Authors and Affiliations

Authors

Contributions

G.D., W.D., F.C. and S.J. researched data for the article. G.D., W.D., G.D.M., M.B., F.C. and S.J. made a substantial contribution to discussion of content. G.D., W.D., M.B., F.C. and S.J. wrote the article. G.D., W.D., G.D.M., T.G., L.M., D.M., H.V.P., C.B., W.E., F.C. and S.J. reviewed and/or edited the manuscript before submission.

Corresponding author

Correspondence to Gaëtan Devos.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Urology thanks W. Dahut, E. Klein and J. Parsons for their contribution to the peer review of this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Glossary

Biochemical recurrence

(BCR). Rising PSA following treatment with curative intent (surgery or radiotherapy) for localized prostate cancer. Often defined as a (confirmed) rise in PSA to 0.2 ng/ml or more following radical prostatectomy, or a rise of 2 ng/ml or more above the PSA nadir after radiation therapy for localized prostate cancer.

Hotspot mutations

Gain-of-function mutations.

Poly(ADP-ribose) polymerase (PARP) inhibitor

A family of nuclear proteins involved in the single-strand break DNA repair pathway. Inhibition of poly(ADP-ribose) polymerase (PARP) is effective in men with mutations of the homologous recombination DNA repair pathway (such as BRCA1 and BRCA2), which promotes DNA double-strand break repair. PARP inhibition blocks single-strand break repair that evolves in a double-strand break, which cannot be repaired because of a deficiency of the homologous recombination DNA repair pathway.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Devos, G., Devlies, W., De Meerleer, G. et al. Neoadjuvant hormonal therapy before radical prostatectomy in high-risk prostate cancer. Nat Rev Urol 18, 739–762 (2021). https://doi.org/10.1038/s41585-021-00514-9

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41585-021-00514-9

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing