Nanoparticles as theranostic vehicles in prostate cancer
Editorial Commentary

Nanoparticles as theranostic vehicles in prostate cancer

Rafael Morales-Barrera, Cristina Suárez, Joaquín Mateo, Macarena González, Joan Carles

Vall d’Hebron Institute of Oncology, Vall d’ Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain

Correspondence to: Joan Carles, MD, PhD. Director GU, CNS and Sarcoma Program, Department of Medical Oncology, Vall d’Hebron University Hospital, Passeig Vall d’Hebron 119-129, 08035-Barcelona, Catalonia, Spain. Email: jcarles@vhio.net.

Provenance: This is an invited article commissioned by the Section Editor Peng Zhang, MD, PhD (Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China).

Comment on: Autio KA, Dreicer R, Anderson J, et al. Safety and Efficacy of BIND-014, a Docetaxel Nanoparticle Targeting Prostate-Specific Membrane Antigen for Patients With Metastatic Castration-Resistant Prostate Cancer: A Phase 2 Clinical Trial. JAMA Oncol 2018;4:1344-51.


Submitted Jan 23, 2019. Accepted for publication Jan 28, 2019.

doi: 10.21037/atm.2019.01.77


Prostate cancer (PCa) is the second most common tumor in men with more than 1.3 million cases diagnosed and more than 350,000 deaths estimated to occur in 2018 (1). Androgen deprivation therapy (ADT) is the cornerstone in the management for metastatic PCa. However, ADT is not curative and most of the patients will develop metastatic castration-resistant prostate cancer (mCRPC) during the next two years after ADT. Approved drugs for mCRPC include second-generation androgen receptor inhibitors (abiraterone acetate and enzalutamide) (2-5), immunotherapy like sipuleucel-T (6), alpha-emitting radiotherapeutic drug (Radium 223) (7) and chemotherapies including docetaxel and cabazitaxel (8-10).

Docetaxel, a cytotoxic taxane, is an antimicrotubule agent that promotes and stabilizes microtubule assembly, disrupting microtubule dynamics, which are sufficient to induce mitotic arrest in G2/M (8). Also, it has been reported that docetaxel is able to modulate androgen receptor (AR) expression and modulate its trafficking from the cytoplasm to the nucleus (11). In 2004, docetaxel plus prednisone was the first FDA approved therapy for the treatment of mCRPC improving overall survival (OS) and pain palliation compared with mitoxantrone plus prednisone in two large phase III studies. (8,9). Also in the last years, it has been demonstrated the survival benefit when docetaxel is added to ADT in patients with metastatic hormone-sensitive PCa patients (12,13).

Over the last decades, the nanothecnology in cancer therapy is providing significant opportunities to develop novel and effective treatments, particularly improving drug delivery and tumor drug-exposure. Among a wide variety of nanosystems, Doxil®, Myocet®, Depocyt®, Genexol-PM® are approved for use in the treatment of cancer (14). BIND-014 is a targeted nanoparticle (NP) with diameter of 100nm improving the delivery of docetaxel in cancer cells. BIND-014 binding to prostate-specific membrane antigen (PSMA), a cell-surface protein that is overexpressed on PCa cells (15).

Nanothecnology is an emerging and promising field for PCa. Autio et al. conducted a phase II clinical trial among patients with mCRPC treated with BIND-014, the first targeted polymeric nanoparticle containing docetaxel (16). The median radiographic progression free survival (rPFS) was 9.9 months (mo) (95% CI, 7.1–12.6 mo) that was better than the prespecified in the trial design (rPFS ≥6 mo). Despite the potential benefits of nanoparticles, the safety profile of BIND-014 is similar to docetaxel in terms of rates of grade 3/4 hematoxicity, fatigue and neuropathy, and no unexpected toxicities were observed (8,16). However, BIND-014 is associated with less nonhematological (diarrhea, decreased appetite). Tumor responses were higher with BIND-014 (32%) compared with docetaxel every three weeks (12%), whereas PSA response observed with BIND-014 were lower (30% vs. 45%) (8,16). The role of circulating tumor cells (CTCs) as independent adverse prognostic factor of OS in mCPRC has been demonstrated in two prospective phase 3 studies evaluating docetaxel (17,18). Autio et al., reported a conversion from unfavorable CTC (≥5 cells per 7.5 mL) counts to favorable counts (<5 cells per 7.5 mL) in 50% of the patients, which is comparable to data from the SWOG S4021 trial. Interestingly, they evaluated 18 patients with CTCs that expressed PSMA (based on the Epic Sciences platform), 61% of those patients had PSMA-positive CTCs. High PSMA-positive CTC counts at 3 and 9 weeks were associated with worse rPFS (1.84 vs. 5.82 mo). These findings suggest the potential role of PSMA-positive CTCs in identifying patients who are not getting benefit of BIND-014.

This study provides an excellent clinical utility of nanoparticle therapeutics for PCa therapy specifically PSMA overexpression on the surface on PCa cells. On the other hand, the characterization of PSMA-positive CTCs can be a useful tool for the identification of patients who respond or not to PSMA-targeted therapies. However, the lower prevalence of PSMA expression in CTC (16), the intrapatient heterogeneity (14) and the effect of ADT on PSMA PET/CT is highly dependent on the castration sensitivity (19) are the main challenges for the development of this diagnostic test that can provide useful information about treatment decision in real life evaluations.

The implementation of radiolabelled compounds targeting (PSMA) for diagnostic and therapeutic approaches is a key challenge to overcome in the management for PCa patients. Recently, the correlation between PSMA PET/CT in metastatic PCa patients and clinical response to chemotherapy showed promising results for evaluation of treatment response (20,21).

Expression of PSMA is an important prerequisite for future prospective clinical trials in mCRPC patients and opens a window of opportunity of how we should evaluate the clinical impact of PSMA expression in CTC for patients with advanced PCa and/or early stage of the disease.


Acknowledgements

None.


Footnote

Conflicts of Interest: R Morales-Barrera has served in a consulting or advisory role and/or on speakers bureaus for Sanofi Aventis, Bayer, Janssen, AstraZeneca, Merck Sharp & Dohme, Asofarma and received travel and accommodations expenses from Roche, Sanofi Aventis, Astellas, Janssen, Merck Sharp & Dohme, Bayer, Pharmacyclics, Clovis Oncology, and Lilly. C Suárez has served in a consulting or advisory role and/or on speakers bureaus and received travel or acommodations expeneses from Astellas, Astrazeneca, BMS, IPSEN, Novartis, Pfizer, Roche and Sanofi. M González has served in a consulting or advisory role for Roche and received travel and accommodations expenses from Astellas, Bayer, Pand Lilly, Roche and Amgen. J Mateo has participated in paid advisory boards or speaker bureaus for AstraZeneca, Roche, Astellas, Sanofi and Janssen. J Carles reports personal fees from Bayer, personal fees from Johnson & Johnson, personal fees from Brystol-Myers Squibb, personal fees from Astellas Pharma personal fees from Pfizer, personal fees from Sanofi, personal fees from MSD Oncology, personal fees from Roche, personal fees from Astra Zeneca, personal fees from Asofarma, outside the submitted work.


References

  1. Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 38 cancers in 185 countries. CA Cancer J Clin 2018;68:394-424. [Crossref] [PubMed]
  2. de Bono JS, Logothetis CJ, Molina A, et al. Abiraterone and increased survival in metastatic prostate cancer. N Engl J Med 2011;364:1995-2005. [Crossref] [PubMed]
  3. Ryan CJ, Smith MR, de Bono JS, et al. Abiraterone in metastatic prostate cancer without previous chemotherapy. N Engl J Med 2013;368:138-48. [Crossref] [PubMed]
  4. Scher HI, Fizazi K, Saad F, et al. Increased survival with enzalutamide in prostate cancer after chemotherapy. N Engl J Med 2012;367:1187-97. [Crossref] [PubMed]
  5. Beer TM, Armstrong AJ, Rathkopf DE, et al. Enzalutamide in metastatic prostate cancer before chemotherapy. N Engl J Med 2014;371:424-33. [Crossref] [PubMed]
  6. Kantoff PW, Higano CS, Shore ND, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med 2010;363:411-22. [Crossref] [PubMed]
  7. Parker C, Nilsson S, Heinrich D, et al. Alpha emitter radium-223 and survival in metastatic prostate cancer. N Engl J Med 2013;369:213-23. [Crossref] [PubMed]
  8. Tannock IF, de Wit R, Berry WR, et al. Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. N Engl J Med 2004;351:1502-12. [Crossref] [PubMed]
  9. Petrylak DP, Tangen CM, Hussain MH, et al. Docetaxel and estramustine compared with mitoxantrone and prednisone for advanced refractory prostate cancer. N Engl J Med 2004;351:1513-20. [Crossref] [PubMed]
  10. de Bono JS, Oudard S, Ozguroglu M, et al. Prednisone plus cabazitaxel or mitoxantrone for metastatic castration-resistant prostate cancer progressing after docetaxel treatment: a randomised open-label trial. Lancet 2010;376:1147-54. [Crossref] [PubMed]
  11. Tinzl M, Chen B, Chen SY, et al. Interaction between c-jun and androgen receptor determines the outcome of taxane therapy in castration resistant prostate cancer. PLoS One 2013;8:e79573. [Crossref] [PubMed]
  12. Sweeney CJ, Chen YH, Carducci M, et al. Chemohormonal Therapy in Metastatic Hormone-Sensitive Prostate Cancer. N Engl J Med 2015;373:737-46. [Crossref] [PubMed]
  13. James ND, Sydes MR, Clarke NW, et al. Addition of docetaxel, zoledronic acid, or both to first-line long-term hormone therapy in prostate cancer (STAMPEDE): survival results from an adaptive, multiarm, multistage, platform randomised controlled trial. Lancet 2016;387:1163-77. [Crossref] [PubMed]
  14. Sanna V, Sechi M. Nanoparticle therapeutics for prostate cancer treatment. Nanomedicine 2012;8 Suppl 1:S31-6. [Crossref] [PubMed]
  15. Von Hoff DD, Mita MM, Ramanathan RK, et al. Phase I Study of PSMA-Targeted Docetaxel-Containing Nanoparticle BIND-014 in Patients with Advanced Solid Tumors. Clin Cancer Res 2016;22:3157-63. [Crossref] [PubMed]
  16. Autio KA, Dreicer R, Anderson J, et al. Safety and Efficacy of BIND-014, a Docetaxel Nanoparticle Targeting Prostate-Specific Membrane Antigen for Patients With Metastatic Castration-Resistant Prostate Cancer: A Phase 2 Clinical Trial. JAMA Oncol 2018;4:1344-51. [Crossref] [PubMed]
  17. Goldkorn A, Ely B, Quinn DI, et al. Circulating tumor cell counts are prognostic of overall survival in SWOG S0421: a phase III trial of docetaxel with or without atrasentan for metastatic castration-resistant prostate cancer. J Clin Oncol 2014;32:1136-42. [Crossref] [PubMed]
  18. Vogelzang NJ, Fizazi K, Burke JM, et al. Circulating Tumor Cells in a Phase 3 Study of Docetaxel and Prednisone with or without Lenalidomide in Metastatic Castration-resistant Prostate Cancer. Eur Urol 2017;71:168-71. [Crossref] [PubMed]
  19. Emmett LM, Yin C, Crumbaker M, et al. Rapid modulation of PSMA expression by Androgen deprivation: Serial 68Ga PSMA-11 PET in men with hormone sensitive and castrate resistant prostate cancer commencing androgen blockade. J Nucl Med 2018. [Epub ahead of print]. [Crossref] [PubMed]
  20. Seitz AK, Rauscher I, Haller B, et al. Preliminary results on response assessment using 68Ga-HBED-CC-PSMA PET/CT in patients with metastatic prostate cancer undergoing docetaxel chemotherapy. Eur J Nucl Med Mol Imaging 2018;45:602-12. [Crossref] [PubMed]
  21. Schmidkonz C, Cordes M, Schmidt D, et al. 68Ga-PSMA-11 PET/CT-derived metabolic parameters for determination of whole-body tumor burden and treatment response in prostate cancer. Eur J Nucl Med Mol Imaging 2018;45:1862-72. [Crossref] [PubMed]
Cite this article as: Morales-Barrera R, Suárez C, Mateo J, González M, Carles J. Nanoparticles as theranostic vehicles in prostate cancer. Ann Transl Med 2019;7(Suppl 1):S29. doi: 10.21037/atm.2019.01.77

Download Citation