Prostate cancer: Early detection, diagnosis, and advances in treatment- Review of updated data for healthcare providers

https://doi.org/10.53730/ijhs.v3nS1.15235

Authors

  • Sahar Adi Albogamy KSA, National Guard Health Affairs
  • ‏Maram Jaafar Alnemer KSA, National Guard Health Affairs
  • ‏Ahmad Alhelo Alanazi KSA, National Guard Health Affairs
  • ‏Mona Mudlah Alsaadi KSA, National Guard Health Affairs
  • ‏Mohammed Abdullah Alzaher KSA, National Guard Health Affairs
  • ‏Hussain Mahdi Aljawad KSA, National Guard Health Affairs
  • ‏Ali Suliman Al Aloula KSA, National Guard Health Affairs
  • ‏Barakat Shumailan Alazmiy KSA, National Guard Health Affairs
  • ‏Fahad Abdulaziz Bahaidarah KSA, National Guard Health Affairs
  • Mohammad Raja Aljehani KSA, National Guard Health Affairs
  • Fouad Hamed Alamri KSA, National Guard Health Affairs

Keywords:

Prostate cancer, metastatic castration-resistant prostate cancer, immunotherapy, early detection, clinical trials, vaccines, immune checkpoint inhibitors

Abstract

Background: Prostate cancer (PCa) is the most commonly diagnosed cancer among men in the U.S. and the second leading cause of cancer-related deaths. Standard treatment options include radical prostatectomy and radiation for localized PCa, while androgen ablation therapy is used for recurrent or advanced stages. However, almost all patients progress to metastatic castration-resistant prostate cancer (mCRPC), with limited treatment options that offer only modest survival improvements. Immunotherapy has shown promise in addressing this challenge. Aim: This review aims to discuss the current advancements in the early detection, diagnosis, and treatment of PCa, with a focus on immunotherapy and ongoing clinical trials. Methods: A comprehensive literature review was conducted to analyze data from recent clinical trials, focusing on the efficacy of current treatments such as vaccines, immune checkpoint inhibitors (ICIs), cell-based therapies, and DNA-based immunotherapy approaches. Various ongoing clinical trials in the field were also reviewed. Results: Current FDA-approved treatments for mCRPC, such as docetaxel, abiraterone, and Sipuleucel-T, have shown survival advantages of 2-4 months. Immunotherapeutic approaches, including Sipuleucel-T, have demonstrated improved overall survival in clinical trials. Numerous ongoing trials are investigating new combinations of immunotherapies, vaccines, and immune modulators to enhance treatment efficacy. 

Downloads

Download data is not yet available.

References

Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. (2018) 68:7–30. doi: 10.3322/caac.21442 DOI: https://doi.org/10.3322/caac.21442

Heidenreich A, Bastian PJ, Bellmunt J, Bolla M, Joniau S, van der Kwast T, et al. EAU guidelines on prostate cancer. Part II: treatment of advanced, relapsing, and castration-resistant prostate cancer. Eur Urol. (2014) 65:467–79. doi: 10.1016/j.eururo.2013.11.002 DOI: https://doi.org/10.1016/j.eururo.2013.11.002

Shelley M, Harrison C, Coles B, Staffurth J, Wilt TJ, Mason MD. Chemotherapy for hormone-refractory prostate cancer. Cochrane database Syst Rev. (2006) 8:CD005247. doi: 10.1002/14651858.CD005247.pub2 DOI: https://doi.org/10.1002/14651858.CD005247.pub2

Petrylak DP, Tangen CM, Hussain MHA, Lara PNJ, Jones JA, Taplin ME, et al. Docetaxel and estramustine compared with mitoxantrone and prednisone for advanced refractory prostate cancer. N Engl J Med. (2004) 351:1513–20. doi: 10.1056/NEJMoa041318 DOI: https://doi.org/10.1056/NEJMoa041318

de Bono JS, Logothetis CJ, Molina A, Fizazi K, North S, Chu L, et al. Abiraterone and increased survival in metastatic prostate cancer. N Engl J Med. (2011) 364:1995–2005. doi: 10.1056/NEJMoa1014618 DOI: https://doi.org/10.1056/NEJMoa1014618

Scher HI, Fizazi K, Saad F, Taplin M-E, Sternberg CN, Miller K, et al. Increased survival with enzalutamide in prostate cancer after chemotherapy. N Engl J Med. (2012) 367:1187–97. doi: 10.1056/NEJMoa1207506 DOI: https://doi.org/10.1056/NEJMoa1207506

Parker C, Nilsson S, Heinrich D, Helle SI, O'Sullivan JM, Fossa SD, et al. Alpha emitter radium-223 and survival in metastatic prostate cancer. N Engl J Med. (2013) 369:213–23. doi: 10.1056/NEJMoa1213755 DOI: https://doi.org/10.1056/NEJMoa1213755

Patel SA, Hoffman-Censits J. Cabazitaxel in the treatment of metastatic castration-resistant prostate cancer: patient selection and special considerations. Onco Targets Ther. (2017) 10:4089–98. doi: 10.2147/OTT.S103532 DOI: https://doi.org/10.2147/OTT.S103532

Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. (2010) 363:411–22. doi: 10.1056/NEJMoa1001294 DOI: https://doi.org/10.1056/NEJMoa1001294

Moreira DM, Howard LE, Sourbeer KN, Amarasekara HS, Chow LC, Cockrell DC, et al. Predicting time from metastasis to overall survival in castration-resistant prostate cancer: results from SEARCH. Clin Genitourin Cancer (2017) 15:60–6.e2. doi: 10.1016/j.clgc.2016.08.018 DOI: https://doi.org/10.1016/j.clgc.2016.08.018

Beer TM, Armstrong AJ, Rathkopf DE, Loriot Y, Sternberg CN, Higano CS, et al. Enzalutamide in metastatic prostate cancer before chemotherapy. N Engl J Med. (2014) 371:424–33. doi: 10.1056/NEJMoa1405095 DOI: https://doi.org/10.1056/NEJMoa1405095

Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. (2010) 363:711–23. doi: 10.1056/NEJMoa1003466 DOI: https://doi.org/10.1056/NEJMoa1003466

Tang J, Yu JX, Hubbard-Lucey VM, Neftelinov ST, Hodge JP, Lin Y. The clinical trial landscape for PD1/PDL1 immune checkpoint inhibitors. Nat Rev Drug Discov. (2018) 17:854. doi: 10.1038/nrd.2018.210 DOI: https://doi.org/10.1038/nrd.2018.210

Zhang, J., Quadri, S., Wolfgang, C. L., & Zheng, L. (2018). New development of biomarkers for gastrointestinal cancers: From neoplastic cells to tumor microenvironment. Biomedicines, 6(3), 87. DOI: https://doi.org/10.3390/biomedicines6030087

Cole G, McCaffrey J, Ali AA, McCarthy HO. DNA vaccination for prostate cancer: key concepts and considerations. Cancer Nanotechnol. (2015) 6:2. doi: 10.1186/s12645-015-0010-5 DOI: https://doi.org/10.1186/s12645-015-0010-5

Yang B, Jeang J, Yang A, Wu TC, Hung C-F. DNA vaccine for cancer immunotherapy. Hum Vaccin Immunother. (2014) 10:3153–64. doi: 10.4161/21645515.2014.980686 DOI: https://doi.org/10.4161/21645515.2014.980686

Zahm CD, Colluru VT, McNeel DG. DNA vaccines for prostate cancer. Pharmacol Ther. (2017) 174:27–42. doi: 10.1016/j.pharmthera.2017.02.016 DOI: https://doi.org/10.1016/j.pharmthera.2017.02.016

Alam S, McNeel DG. DNA vaccines for the treatment of prostate cancer. Exp Rev Vaccines. (2010) 9:731–45. doi: 10.1586/erv.10.64 DOI: https://doi.org/10.1586/erv.10.64

Kiessling A, Wehner R, Fussel S, Bachmann M, Wirth MP, Schmitz M. Tumor-associated antigens for specific immunotherapy of prostate cancer. Cancers. (2012) 4:193–217. doi: 10.3390/cancers4010193 DOI: https://doi.org/10.3390/cancers4010193

Gati A, Lajmi N, Derouiche A, Marrakchi R, Chebil M, Benammar-Elgaaied A. NY-ESO-1 expression and immunogenicity in prostate cancer patients. Tunis Med. (2011) 89:779–83.

McNeel DG, Dunphy EJ, Davies JG, Frye TP, Johnson LE, Staab MJ, et al. Safety and immunological efficacy of a DNA vaccine encoding prostatic acid phosphatase in patients with stage D0 prostate cancer. J Clin Oncol. (2009) 27:4047–54. doi: 10.1200/JCO.2008.19.9968 DOI: https://doi.org/10.1200/JCO.2008.19.9968

Becker JT, Olson BM, Johnson LE, Davies JG, Dunphy EJ, McNeel DG. DNA vaccine encoding prostatic acid phosphatase. (PAP) elicits long-term T-cell responses in patients with recurrent prostate cancer. J Immunother. (2010) 33:639–47. doi: 10.1097/CJI.0b013e3181dda23e DOI: https://doi.org/10.1097/CJI.0b013e3181dda23e

Guo Y, Lei K, Tang L. Neoantigen Vaccine delivery for personalized anticancer immunotherapy. Front Immunol. (2018) 9:1499. doi: 10.3389/fimmu.2018.01499 DOI: https://doi.org/10.3389/fimmu.2018.01499

Carreno BM, Magrini V, Becker-Hapak M, Kaabinejadian S, Hundal J, Petti AA, et al. Cancer immunotherapy. A dendritic cell vaccine increases the breadth and diversity of melanoma neoantigen-specific T cells. Science. (2015) 348:803–8. doi: 10.1126/science.aaa3828 DOI: https://doi.org/10.1126/science.aaa3828

Sahin U, Derhovanessian E, Miller M, Kloke B-P, Simon P, Lower M, et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature. (2017) 547:222–6.

Ott PA, Hu Z, Keskin DB, Shukla SA, Sun J, Bozym DJ, et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature. (2017) 547:217–21. doi: 10.1038/nature22991 DOI: https://doi.org/10.1038/nature22991

Small EJ, Sacks N, Nemunaitis J, Urba WJ, Dula E, Centeno AS, et al. Granulocyte macrophage colony-stimulating factor–secreting allogeneic cellular immunotherapy for hormone-refractory prostate cancer. Clin Cancer Res. (2007) 13:3883–91. doi: 10.1158/1078-0432.CCR-06-2937 DOI: https://doi.org/10.1158/1078-0432.CCR-06-2937

Demkow EST, Gerritsen W. A phase III trial of GVAX immunotherapy for prostate cancer in combination with docetaxel versus docetaxel plus prednisone in symptomatic, castration-resistant prostate cancer (CRPC). In Proc 2009 Genitourin Cancer Symp. Orlando, FL (2009).

Higano CS, Corman JM, Smith DC, Centeno AS, Steidle CP, Gittleman M, et al. Phase 1/2 dose-escalation study of a GM-CSF-secreting, allogeneic, cellular immunotherapy for metastatic hormone-refractory prostate cancer. Cancer. (2008) 113:975–84. doi: 10.1002/cncr.23669 DOI: https://doi.org/10.1002/cncr.23669

Simons JW, Mikhak B, Chang JF, DeMarzo AM, Carducci MA, Lim M, et al. Induction of immunity to prostate cancer antigens: results of a clinical trial of vaccination with irradiated autologous prostate tumor cells engineered to secrete granulocyte-macrophage colony-stimulating factor using ex vivo gene transfer. Cancer Res. (1999) 59:5160–8.

Mo L, Zhang X, Shi X, Wei L, Zheng D, Li H, et al. Norcantharidin enhances antitumor immunity of GM-CSF prostate cancer cells vaccine by inducing apoptosis of regulatory T cells. Cancer Sci. (2018) 109:2109–18. doi: 10.1111/cas.13639 DOI: https://doi.org/10.1111/cas.13639

Burch PA, Breen JK, Buckner JC, Gastineau DA, Kaur JA, Laus RL, et al. Priming tissue-specific cellular immunity in a phase I trial of autologous dendritic cells for prostate cancer. Clin Cancer Res. (2000) 6:2175–82.

Anassi E, Ndefo UA. Sipuleucel-T. (provenge) injection: the first immunotherapy agent. (vaccine) for hormone-refractory prostate cancer. P T. (2011) 36:197–202.

Small EJ, Schellhammer PF, Higano CS, Redfern CH, Nemunaitis JJ, Valone FH, et al. Placebo-controlled phase III trial of immunologic therapy with sipuleucel-T. (APC8015) in patients with metastatic, asymptomatic hormone refractory prostate cancer. J Clin Oncol. (2006) 24:3089–94. doi: 10.1200/JCO.2005.04.5252 DOI: https://doi.org/10.1200/JCO.2005.04.5252

Higano CS, Schellhammer PF, Small EJ, Burch PA, Nemunaitis J, Yuh L, et al. Integrated data from 2 randomized, double-blind, placebo-controlled, phase 3 trials of active cellular immunotherapy with sipuleucel-T in advanced prostate cancer. Cancer. (2009) 115:3670–9. doi: 10.1002/cncr.24429 DOI: https://doi.org/10.1002/cncr.24429

Hall SJ, Klotz L, Pantuck AJ, George DJ, Whitmore JB, Frohlich MW, et al. Integrated safety data from 4 randomized, double-blind, controlled trials of autologous cellular immunotherapy with sipuleucel-T in patients with prostate cancer. J Urol. (2011) 186:877–81. doi: 10.1016/j.juro.2011.04.070 DOI: https://doi.org/10.1016/j.juro.2011.04.070

Holko P, Kawalec P. Economic evaluation of sipuleucel-T immunotherapy in castration-resistant prostate cancer. Expert Rev Anticancer Ther. (2014) 14:63–73. doi: 10.1586/14737140.2014.856270 DOI: https://doi.org/10.1586/14737140.2014.856270

Sadelain M, Brentjens R, Riviere I. The basic principles of chimeric antigen receptor design. Cancer Discov. (2013) 3:388–98. doi: 10.1158/2159-8290.CD-12-0548 DOI: https://doi.org/10.1158/2159-8290.CD-12-0548

Maude SL, Teachey DT, Porter DL, Grupp SA. CD19-targeted chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Blood. (2015) 125:4017–23. doi: 10.1182/blood-2014-12-580068 DOI: https://doi.org/10.1182/blood-2014-12-580068

Priceman SJ, Gerdts EA, Tilakawardane D, Kennewick KT, Murad JP, Park AK, et al. Co-stimulatory signaling determines tumor antigen sensitivity and persistence of CAR T cells targeting PSCA+ metastatic prostate cancer. Oncoimmunology. (2018) 7:e1380764. doi: 10.1080/2162402X.2017.1380764 DOI: https://doi.org/10.1080/2162402X.2017.1380764

Noguchi M, Sasada T, Itoh K. Personalized peptide vaccination: a new approach for advanced cancer as therapeutic cancer vaccine. Cancer Immunol Immunother. (2013) 62:919–29. doi: 10.1007/s00262-012-1379-1 DOI: https://doi.org/10.1007/s00262-012-1379-1

Inoue Y, Takaue Y, Takei M, Kato K, Kanai S, Harada Y, et al. Induction of tumor specific cytotoxic t lymphocytes in prostate cancer using prostatic acid phosphatase derived hla-a2402 binding peptide. J Urol. (2001) 166:1508–13. doi: 10.1016/S0022-5347(05)65821-1 DOI: https://doi.org/10.1016/S0022-5347(05)65821-1

Harada M, Kobayashi K, Matsueda S, Nakagawa M, Noguchi M, Itoh K. Prostate-specific antigen-derived epitopes capable of inducing cellular and humoral responses in HLA-A24+ prostate cancer patients. Prostate. (2003) 57:152–9. doi: 10.1002/pros.10280 DOI: https://doi.org/10.1002/pros.10280

Kobayashi K, Noguchi M, Itoh K, Harada M. Identification of a prostate-specific membrane antigen-derived peptide capable of eliciting both cellular and humoral immune responses in HLA-A24+ prostate cancer patients. Cancer Sci. (2003) 94:622–7. doi: 10.1111/j.1349-7006.2003.tb01493.x DOI: https://doi.org/10.1111/j.1349-7006.2003.tb01493.x

Noguchi M, Kakuma T, Uemura H, Nasu Y, Kumon H, Hirao Y, et al. A randomized phase II trial of personalized peptide vaccine plus low dose estramustine phosphate. (EMP) versus standard dose EMP in patients with castration resistant prostate cancer. Cancer Immunol Immunother. (2010) 59:1001–9. doi: 10.1007/s00262-010-0822-4 DOI: https://doi.org/10.1007/s00262-010-0822-4

Noguchi M, Moriya F, Suekane S, Matsuoka K, Arai G, Matsueda S, et al. Phase II study of personalized peptide vaccination for castration-resistant prostate cancer patients who failed in docetaxel-based chemotherapy. Prostate. (2012) 72:834–45. doi: 10.1002/pros.21485 DOI: https://doi.org/10.1002/pros.21485

Lilleby W, Gaudernack G, Brunsvig PF, Vlatkovic L, Schulz M, Mills K, et al. Phase I/IIa clinical trial of a novel hTERT peptide vaccine in men with metastatic hormone-naive prostate cancer. Cancer Immunol Immunother. (2017) 66:891–901. doi: 10.1007/s00262-017-1994-y DOI: https://doi.org/10.1007/s00262-017-1994-y

Sultan H, Fesenkova VI, Addis D, Fan AE, Kumai T, Wu J, et al. Designing therapeutic cancer vaccines by mimicking viral infections. Cancer Immunol Immunother. (2017) 66:203–13. doi: 10.1007/s00262-016-1834-5 DOI: https://doi.org/10.1007/s00262-016-1834-5

Madan RA, Bilusic M, Heery C, Schlom J, Gulley JL. Clinical evaluation of TRICOM vector therapeutic cancer vaccines. Semin Oncol. (2012) 39:296–304. doi: 10.1053/j.seminoncol.2012.02.010 DOI: https://doi.org/10.1053/j.seminoncol.2012.02.010

Kantoff PW, Schuetz TJ, Blumenstein BA, Glode LM, Bilhartz DL, Wyand M, et al. Overall survival analysis of a phase II randomized controlled trial of a Poxviral-based PSA-targeted immunotherapy in metastatic castration-resistant prostate cancer. J Clin Oncol. (2010) 28:1099–105. doi: 10.1200/JCO.2009.25.0597 DOI: https://doi.org/10.1200/JCO.2009.25.0597

Gulley JL, Arlen PM, Madan RA, Tsang K-Y, Pazdur MP, Skarupa L, et al. Immunologic and prognostic factors associated with overall survival employing a poxviral-based PSA vaccine in metastatic castrate-resistant prostate cancer. Cancer Immunol Immunother. (2010) 59:663–74. doi: 10.1007/s00262-009-0782-8 DOI: https://doi.org/10.1007/s00262-009-0782-8

Hossain, M. K., Nahar, K., Donkor, O., & Apostolopoulos, V. (2018). Immune-based therapies for metastatic prostate cancer: an update. Immunotherapy, 10(4), 283-298. DOI: https://doi.org/10.2217/imt-2017-0123

Lubaroff DM, Konety BR, Link B, Gerstbrein J, Madsen T, Shannon M, et al. Phase I clinical trial of an adenovirus/prostate-specific antigen vaccine for prostate cancer: safety and immunologic results. Clin Cancer Res. (2009) 15:7375–80. doi: 10.1158/1078-0432.CCR-09-1910 DOI: https://doi.org/10.1158/1078-0432.CCR-09-1910

Lubaroff DM, Williams RD, Vaena D, Joudi F, Brown J, Smith M, et al. An ongoing phase II trial of an adenovirus/PSA vaccine for prostate cancer. In: 103rd Annu Meet Am Assoc Cancer Res. Chicago, IL (2012). DOI: https://doi.org/10.1158/1538-7445.AM2012-2692

Wei SC, Duffy CR, Allison JP. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov. (2018) 8:1069–86. doi: 10.1158/2159-8290.CD-18-0367 DOI: https://doi.org/10.1158/2159-8290.CD-18-0367

Slovin SF, Higano CS, Hamid O, Tejwani S, Harzstark A, Alumkal JJ, et al. Ipilimumab alone or in combination with radiotherapy in metastatic castration-resistant prostate cancer: results from an open-label, multicenter phase I/II study. Ann Oncol Off J Eur Soc Med Oncol. (2013) 24:1813–21. doi: 10.1093/annonc/mdt107 DOI: https://doi.org/10.1093/annonc/mdt107

Kwon ED, Drake CG, Scher HI, Fizazi K, Bossi A, van den Eertwegh AJM, et al. Ipilimumab versus placebo after radiotherapy in patients with metastatic castration-resistant prostate cancer that had progressed after docetaxel chemotherapy. (CA184-043): a multicentre, randomised, double-blind, phase 3 trial. Lancet Oncol. (2014) 15:700–712. doi: 10.1016/S1470-2045(14)70189-5 DOI: https://doi.org/10.1016/S1470-2045(14)70189-5

Beer TM, Kwon ED, Drake CG, Fizazi K, Logothetis C, Gravis G, et al. Randomized, double-blind, phase III Trial of ipilimumab versus placebo in asymptomatic or minimally symptomatic patients with metastatic chemotherapy-naive castration-resistant prostate cancer. J Clin Oncol. (2017) 35:40–7. doi: 10.1200/JCO.2016.69.1584 DOI: https://doi.org/10.1200/JCO.2016.69.1584

Gong J, Chehrazi-Raffle A, Reddi S, Salgia R. Development of PD-1 and PD-L1 inhibitors as a form of cancer immunotherapy: a comprehensive review of registration trials and future considerations. J Immunother cancer. (2018) 6:8. doi: 10.1186/s40425-018-0316-z DOI: https://doi.org/10.1186/s40425-018-0316-z

Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. (2012) 366:2443–54. doi: 10.1056/NEJMoa1200690 DOI: https://doi.org/10.1056/NEJMoa1200690

Graff JN, Alumkal JJ, Drake CG, Thomas GV, Redmond WL, Farhad M, et al. Early evidence of anti-PD-1 activity in enzalutamide-resistant prostate cancer. Oncotarget. (2016) 7:52810–7. doi: 10.18632/oncotarget.10547 DOI: https://doi.org/10.18632/oncotarget.10547

Hansen AR, Massard C, Ott PA, Haas NB, Lopez JS, Ejadi S, et al. Pembrolizumab for advanced prostate adenocarcinoma: findings of the KEYNOTE-028 study. Ann Oncol Off J Eur Soc Med Oncol. (2018) 29:1807–13. doi: 10.1093/annonc/mdy232 DOI: https://doi.org/10.1093/annonc/mdy232

Kim JW, Shaffer DR, Massard C, Powles T, Harshman LC, Braiteh FS, et al. A phase Ia study of safety and clinical activity of atezolizumab. (atezo) in patients. (pts) with metastatic castration-resistant prostate cancer. (mCRPC). J Clin Oncol. (2018) 36:187. doi: 10.1200/JCO.2018.36.6_suppl.187 DOI: https://doi.org/10.1200/JCO.2018.36.6_suppl.187

Chapoval AI, Ni J, Lau JS, Wilcox RA, Flies DB, Liu D, et al. B7-H3: A costimulatory molecule for T cell activation and IFN-γ production. Nat Immunol. (2001) 2:269–74. doi: 10.1038/85339 DOI: https://doi.org/10.1038/85339

Castellanos JR, Purvis IJ, Labak CM, Guda MR, Tsung AJ, Velpula KK, et al. B7-H3 role in the immune landscape of cancer. Am J Clin Exp Immunol. (2017) 6:66–75.

Picarda E, Ohaegbulam KC, Zang X. Molecular pathways: targeting B7-H3. (CD276) for human cancer immunotherapy. Clin Cancer Res. (2016) 22:3425–31. doi: 10.1158/1078-0432.CCR-15-2428 DOI: https://doi.org/10.1158/1078-0432.CCR-15-2428

Benzon B, Zhao SG, Haffner MC, Takhar M, Erho N, Yousefi K, et al. Correlation of B7-H3 with androgen receptor, immune pathways and poor outcome in prostate cancer: an expression-based analysis. Prostate Cancer Prostatic Dis. (2017) 20:28–35. doi: 10.1038/pcan.2016.49 DOI: https://doi.org/10.1038/pcan.2016.49

Shenderov E, Demarzo A, Boudadi K, Allaf M, Wang H, Chapman C, et al. Phase II neoadjuvant and immunologic study of B7-H3 targeting with enoblituzumab in localized intermediate- and high-risk prostate cancer. J Clin Oncol. (2018) 36:TPS5099. doi: 10.1200/JCO.2018.36.15_suppl.TPS5099 DOI: https://doi.org/10.1200/JCO.2018.36.15_suppl.TPS5099

Lee P, Gujar S. Potentiating prostate cancer immunotherapy with oncolytic viruses. Nat Rev Urol. (2018) 15:235–50. doi: 10.1038/nrurol.2018.10 DOI: https://doi.org/10.1038/nrurol.2018.10

Bartlett DL, Liu Z, Sathaiah M, Ravindranathan R, Guo Z, He Y, et al. Oncolytic viruses as therapeutic cancer vaccines. Mol Cancer. (2013) 12:103. doi: 10.1186/1476-4598-12-103 DOI: https://doi.org/10.1186/1476-4598-12-103

Russell SJ, Barber GN. Oncolytic viruses as antigen-agnostic cancer vaccines. Cancer Cell. (2018) 33:599–605. doi: 10.1016/j.ccell.2018.03.011 DOI: https://doi.org/10.1016/j.ccell.2018.03.011

Taguchi S, Fukuhara H, Homma Y, Todo T. Current status of clinical trials assessing oncolytic virus therapy for urological cancers. Int J Urol. (2017) 24:342–51. doi: 10.1111/iju.13325 DOI: https://doi.org/10.1111/iju.13325

Rogulski KR, Wing MS, Paielli DL, Gilbert JD, Kim JH, Freytag SO. Double suicide gene therapy augments the antitumor activity of a replication-competent lytic adenovirus through enhanced cytotoxicity and radiosensitization. Hum Gene Ther. (2000) 11:67–76. doi: 10.1089/10430340050016166 DOI: https://doi.org/10.1089/10430340050016166

Freytag SO, Khil M, Stricker H, Peabody J, Menon M, DePeralta-Venturina M, et al. Phase I study of replication-competent adenovirus-mediated double suicide gene therapy for the treatment of locally recurrent prostate cancer. Cancer Res. (2002) 62:4968–76.

Freytag SO, Movsas B, Aref I, Stricker H, Peabody J, Pegg J, et al. Phase I trial of replication-competent adenovirus-mediated suicide gene therapy combined with IMRT for prostate cancer. Mol Ther. (2007) 15:1016–23. doi: 10.1038/mt.sj.6300120 DOI: https://doi.org/10.1038/mt.sj.6300120

Freytag SO, Barton KN, Zhang Y. Efficacy of oncolytic adenovirus expressing suicide genes and interleukin-12 in preclinical model of prostate cancer. Gene Ther. (2013) 20:1131–9. doi: 10.1038/gt.2013.40 DOI: https://doi.org/10.1038/gt.2013.40

Coffey MC, Strong JE, Forsyth PA, Lee PW. Reovirus therapy of tumors with activated Ras pathway. Science. (1998) 282:1332–4. doi: 10.1126/science.282.5392.1332 DOI: https://doi.org/10.1126/science.282.5392.1332

Phillips MB, Stuart JD, Rodríguez Stewart RM, Berry JT, Mainou BA, Boehme KW. Current understanding of reovirus oncolysis mechanisms. Oncolyt Virother. (2018) 7:53–63. doi: 10.2147/OV.S143808 DOI: https://doi.org/10.2147/OV.S143808

Gujar SA, Pan DA, Marcato P, Garant KA, Lee PWK. Oncolytic virus-initiated protective immunity against prostate cancer. Mol Ther. (2011) 19:797–804. doi: 10.1038/mt.2010.297 DOI: https://doi.org/10.1038/mt.2010.297

Thirukkumaran CM, Nodwell MJ, Hirasawa K, Shi Z-Q, Diaz R, Luider J, et al. Oncolytic viral therapy for prostate cancer: efficacy of reovirus as a biological therapeutic. Cancer Res. (2010) 70:2435–44. doi: 10.1158/0008-5472.CAN-09-2408 DOI: https://doi.org/10.1158/0008-5472.CAN-09-2408

Vidal L, Pandha HS, Yap TA, White CL, Twigger K, Vile RG, et al. A phase I study of intravenous oncolytic reovirus type 3 Dearing in patients with advanced cancer. Clin Cancer Res. (2008) 14:7127–37. doi: 10.1158/1078-0432.CCR-08-0524 DOI: https://doi.org/10.1158/1078-0432.CCR-08-0524

Williams ES, Rodriguez-Bravo V, Chippada-Venkata U, De Ia Iglesia-Vicente J, Gong Y, Galsky M, et al. Generation of prostate cancer patient derived xenograft models from circulating tumor cells. J Vis Exp. (2015) 20:53182. doi: 10.3791/53182 DOI: https://doi.org/10.3791/53182-v

van den Eertwegh AJM, Versluis J, van den Berg HP, Santegoets SJAM, van Moorselaar RJA, van der Sluis TM, et al. Combined immunotherapy with granulocyte-macrophage colony-stimulating factor-transduced allogeneic prostate cancer cells and ipilimumab in patients with metastatic castration-resistant prostate cancer: a phase 1 dose-escalation trial. Lancet Oncol. (2012) 13:509–17. doi: 10.1016/S1470-2045(12)70007-4 DOI: https://doi.org/10.1016/S1470-2045(12)70007-4

McNeel DG, Eickhoff JC, Wargowski E, Zahm C, Staab MJ, Straus J, et al. Concurrent, but not sequential, PD-1 blockade with a DNA vaccine elicits anti-tumor responses in patients with metastatic, castration-resistant prostate cancer. Oncotarget. (2018) 9:25586–96. doi: 10.18632/oncotarget.25387 DOI: https://doi.org/10.18632/oncotarget.25387

Mercader M, Bodner BK, Moser MT, Kwon PS, Park ES, Manecke RG, et al. T cell infiltration of the prostate induced by androgen withdrawal in patients with prostate cancer. Proc Natl Acad Sci USA. (2001) 98:14565–70. doi: 10.1073/pnas.251140998 DOI: https://doi.org/10.1073/pnas.251140998

Antonarakis ES, Kibel AS, Yu EY, Karsh LI, Elfiky A, Shore ND, et al. Sequencing of sipuleucel-T and androgen deprivation therapy in men with hormone-sensitive biochemically recurrent prostate cancer: a phase II randomized trial. Clin Cancer Res. (2017) 23:2451–9. doi: 10.1158/1078-0432.CCR-16-1780 DOI: https://doi.org/10.1158/1078-0432.CCR-16-1780

Published

15-01-2019

How to Cite

Albogamy, S. A., Alnemer, ‏Maram J., Alanazi, ‏Ahmad A., Alsaadi, ‏Mona M., Alzaher, ‏Mohammed A., Aljawad, ‏Hussain M., Al Aloula, ‏Ali S., Alazmiy, ‏Barakat S., Bahaidarah, ‏Fahad A., Aljehani, M. R., & Alamri, F. H. (2019). Prostate cancer: Early detection, diagnosis, and advances in treatment- Review of updated data for healthcare providers. International Journal of Health Sciences, 3(S1), 369–385. https://doi.org/10.53730/ijhs.v3nS1.15235

Issue

Section

Peer Review Articles

Most read articles by the same author(s)