Emerging therapeutic agents for genitourinary cancers

Kevin Zarrabi, Azzam Paroya, Shenhong Wu, Kevin Zarrabi, Azzam Paroya, Shenhong Wu

Abstract

The treatment of genitourinary malignancies has dramatically evolved over recent years. Renal cell carcinoma, urothelial carcinoma of the bladder, and prostate adenocarcinoma are the most commonly encountered genitourinary malignancies and represent a heterogeneous population of cancers, in both histology and approach to treatment. However, all three cancers have undergone paradigm shifts in their respective therapeutic landscapes due to a greater understanding of their underlying molecular mechanisms and oncogenic drivers. The advance that has gained the most recent traction has been the advent of immunotherapies, particularly immune checkpoint inhibitors. Immunotherapy has increased overall survival and even provided durable responses in the metastatic setting in some patients. The early success of immune checkpoint inhibitors has led to further drug development with the emergence of novel agents which modulate the immune system within the tumor microenvironment. Notwithstanding immunotherapy, investigators are also developing novel agents tailored to a variety of targets including small-molecule tyrosine kinase inhibitors, mTOR inhibitors, and novel fusion proteins to name a few. Erdafitinib has become the first targeted therapy approved for metastatic bladder cancer. Moreover, the combination therapy of immune checkpoint inhibitors with targeted agents such as pembrolizumab or avelumab with axitinib has demonstrated both safety and efficacy and just received FDA approval for their use. We are in an era of rapid progression in drug development with multiple exciting trials and ongoing pre-clinical studies. We highlight many of the promising new emerging therapies that will likely continue to improve outcomes in patients with genitourinary malignancies.

Keywords: Bladder cancer; Immunotherapy; Prostate cancer; Renal cell carcinoma.

Conflict of interest statement

S. Wu is a speaker for Exelixis, Novartis, and Pfizer. The other authors declare that they have no competing interests.

References

    1. Mei MTP. Madame Curie Bioscience Database. Austin: Landes Bioscience; 2000. Metastatic Genitourinary Malignancies.
    1. Vogelzang NJ. Future directions for gemcitabine in the treatment of genitourinary cancer. Semin Oncol. 2002;29(1 Suppl 3):40–45. doi: 10.1053/sonc.2002.30755.
    1. Zarrabi K, Fang C, Wu S. New treatment options for metastatic renal cell carcinoma with prior anti-angiogenesis therapy. J Hematol Oncol. 2017;10(1):38. doi: 10.1186/s13045-016-0374-y.
    1. Atkins MB, Larkin J. Immunotherapy combined or sequenced with targeted therapy in the treatment of solid tumors: Current Perspectives. J Natl Cancer Inst. 2016;108(6):djv414. doi: 10.1093/jnci/djv414.
    1. Antoni S, Ferlay J, Soerjomataram I, Znaor A, Jemal A, Bray F. Bladder cancer incidence and mortality: a global overview and recent trends. Eur Urol. 2017;71(1):96–108. doi: 10.1016/j.eururo.2016.06.010.
    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin. 2019;69(1):7–34. doi: 10.3322/caac.21551.
    1. Howlader N, Noone AM. In: SEER Cancer Statistics Review, 1975-2014. Krapcho M, Miller D, Bishop K, Kosary CL, Yu M, Ruhl J, Tatalovich Z, Mariotto A, Lewis DR, Chen HS, Feuer EJ, Cronin KA, editors. Bethesda: Institute NC; 2016.
    1. El-Achkar A, Souhami L, Kassouf W. Bladder preservation therapy: review of literature and future directions of trimodal therapy. Curr Urol Rep. 2018;19(12):108. doi: 10.1007/s11934-018-0859-z.
    1. Galsky MD, Hahn NM, Rosenberg J, Sonpavde G, Hutson T, Oh WK, Dreicer R, Vogelzang N, Sternberg C, Bajorin DF, et al. A consensus definition of patients with metastatic urothelial carcinoma who are unfit for cisplatin-based chemotherapy. Lancet Oncol. 2011;12(3):211–214. doi: 10.1016/S1470-2045(10)70275-8.
    1. Lattanzi M, Balar AV. Current status and future direction of immunotherapy in urothelial carcinoma. Curr Oncol Rep. 2019;21(3):24. doi: 10.1007/s11912-019-0775-5.
    1. Johnson SB, Yu JB. Bladder preserving trimodality therapy for muscle-invasive bladder cancer. Curr Oncol Rep. 2018;20(9):66. doi: 10.1007/s11912-018-0711-0.
    1. Ploussard G, Shariat SF, Dragomir A, Kluth LA, Xylinas E, Masson-Lecomte A, Rieken M, Rink M, Matsumoto K, Kikuchi E, et al. Conditional survival after radical cystectomy for bladder cancer: evidence for a patient changing risk profile over time. Eur Urol. 2014;66(2):361–370. doi: 10.1016/j.eururo.2013.09.050.
    1. Donat SM, Shabsigh A, Savage C, Cronin AM, Bochner BH, Dalbagni G, Herr HW, Milowsky MI. Potential impact of postoperative early complications on the timing of adjuvant chemotherapy in patients undergoing radical cystectomy: a high-volume tertiary cancer center experience. Eur Urol. 2009;55(1):177–185. doi: 10.1016/j.eururo.2008.07.018.
    1. Bellmunt J, von der Maase H, Mead GM, Skoneczna I, De Santis M, Daugaard G, Boehle A, Chevreau C, Paz-Ares L, Laufman LR, et al. Randomized phase III study comparing paclitaxel/cisplatin/gemcitabine and gemcitabine/cisplatin in patients with locally advanced or metastatic urothelial cancer without prior systemic therapy: EORTC Intergroup Study 30987. J Clin Oncol. 2012;30(10):1107–1113. doi: 10.1200/JCO.2011.38.6979.
    1. Hermans TJN, Fransen van de Putte EE, Horenblas S, Lemmens V, Aben K, van der Heijden MS, Beerepoot LV, Verhoeven RH, van Rhijn BWG. Perioperative treatment and radical cystectomy for bladder cancer--a population based trend analysis of 10,338 patients in the Netherlands. Eur J Cancer. 2016;54:18–26. doi: 10.1016/j.ejca.2015.11.006.
    1. Godwin JL, Hoffman-Censits J, Plimack E. Recent developments in the treatment of advanced bladder cancer. Urol Oncol. 2018;36(3):109–114. doi: 10.1016/j.urolonc.2017.12.018.
    1. Vaughn DJ, Broome CM, Hussain M, Gutheil JC, Markowitz AB. Phase II trial of weekly paclitaxel in patients with previously treated advanced urothelial cancer. J Clin Oncol. 2002;20(4):937–940. doi: 10.1200/JCO.2002.20.4.937.
    1. Balar AV, Castellano D, O'Donnell PH, Grivas P, Vuky J, Powles T, Plimack ER, Hahn NM, de Wit R, Pang L, et al. First-line pembrolizumab in cisplatin-ineligible patients with locally advanced and unresectable or metastatic urothelial cancer (KEYNOTE-052): a multicentre, single-arm, phase 2 study. Lancet Oncol. 2017;18(11):1483–1492. doi: 10.1016/S1470-2045(17)30616-2.
    1. Sharma P, Retz M, Siefker-Radtke A, Baron A, Necchi A, Bedke J, Plimack ER, Vaena D, Grimm MO, Bracarda S, et al. Nivolumab in metastatic urothelial carcinoma after platinum therapy (CheckMate 275): a multicentre, single-arm, phase 2 trial. Lancet Oncol. 2017;18(3):312–322. doi: 10.1016/S1470-2045(17)30065-7.
    1. Bellmunt J, de Wit R, Vaughn DJ, Fradet Y, Lee JL, Fong L, Vogelzang NJ, Climent MA, Petrylak DP, Choueiri TK, et al. Pembrolizumab as second-line therapy for advanced urothelial carcinoma. N Engl J Med. 2017;376(11):1015–1026. doi: 10.1056/NEJMoa1613683.
    1. Prescott S, Jackson AM, Hawkyard SJ, Alexandroff AB, James K. Mechanisms of action of intravesical bacille Calmette-Guerin: local immune mechanisms. Clin Infect Dis. 2000;31(Suppl 3):S91–S93. doi: 10.1086/314066.
    1. Rao A, Patel MR. A review of avelumab in locally advanced and metastatic bladder cancer. Ther Adv Urol. 2019;11:1756287218823485. doi: 10.1177/1756287218823485.
    1. Pignot G, Loriot Y, Kamat AM, Shariat SF, Plimack ER. Effect of immunotherapy on local treatment of genitourinary malignancies. Eur Urol Oncol. 2019;2(4):355–364. doi: 10.1016/j.euo.2019.01.002.
    1. Balar AV, Galsky MD, Rosenberg JE, Powles T, Petrylak DP, Bellmunt J, Loriot Y, Necchi A, Hoffman-Censits J, Perez-Gracia JL, et al. Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: a single-arm, multicentre, phase 2 trial. Lancet. 2017;389(10064):67–76. doi: 10.1016/S0140-6736(16)32455-2.
    1. FDA Alters Approved Use of Two Checkpoint Inhibitors for Bladder Cancer. []. Accessed 20 Apr 2019.
    1. Harris J. FDA requires PD-L1 testing prior to administration of immunotherapy agents for urothelial cancer. Target Oncol. 2018. . Accessed 4 Apr 2019.
    1. Balar AMA, Grande E, Villalobos V, Salas S, Kang TW, Kim SE, Powles T, Tsai F, Razak A, et al. American Association for Cancer Research: July 1, 2018. Philadelphia: Procedings of the American Association for Cancer Research Annual Meeting; 2018. Abstract CT112: Durvalumab + tremelimumab in patients with metastatic urothelial cancer; p. 78.
    1. Bentebibel SE, Hurwitz ME, Bernatchez C, Haymaker C, Hudgens CW, Kluger HM, Tetzlaff MT, Tagliaferri MA, Zalevsky J, Hoch U, et al. A first-in-human study and biomarker analysis of NKTR-214, a novel IL-2-receptor beta/gamma (betagamma)-biased cytokine, in patients with advanced or metastatic solid tumors. Cancer Discov. 2019.
    1. Arlene O, Siefker-Radtke MNF, Balar AV, Grignani G, Diab A, Gao J, Tagliaferri MA, Hannah AL, Karski EE, Zalevsky J, Hoch U, Rizwan AN, Bilen MA. Journal of Clinical Oncology. 2019. NKTR-214 + nivolumab in first-line advanced/metastatic urothelial carcinoma (mUC): Updated results from PIVOT-02; p. 388.
    1. Balar AV. Recent clinical trials explore immunotherapies for urothelial carcinoma. Oncology (Williston Park) 2019;33(4):132–136.
    1. Knudson KM, Hicks KC, Luo X, Chen JQ, Schlom J, Gameiro SR. M7824, a novel bifunctional anti-PD-L1/TGFbeta trap fusion protein, promotes anti-tumor efficacy as monotherapy and in combination with vaccine. Oncoimmunology. 2018;7(5):e1426519. doi: 10.1080/2162402X.2018.1426519.
    1. Strauss Julius, Heery Christopher R., Schlom Jeffrey, Madan Ravi A., Cao Liang, Kang Zhigang, Lamping Elizabeth, Marté Jennifer L., Donahue Renee N., Grenga Italia, Cordes Lisa, Christensen Olaf, Mahnke Lisa, Helwig Christoph, Gulley James L. Phase I Trial of M7824 (MSB0011359C), a Bifunctional Fusion Protein Targeting PD-L1 and TGFβ, in Advanced Solid Tumors. Clinical Cancer Research. 2018;24(6):1287–1295. doi: 10.1158/1078-0432.CCR-17-2653.
    1. Fisher TS, Kamperschroer C, Oliphant T, Love VA, Lira PD, Doyonnas R, Bergqvist S, Baxi SM, Rohner A, Shen AC, et al. Targeting of 4-1BB by monoclonal antibody PF-05082566 enhances T-cell function and promotes anti-tumor activity. Cancer Immunol Immunother. 2012;61(10):1721–1733. doi: 10.1007/s00262-012-1237-1.
    1. Tolcher AW, Sznol M, Hu-Lieskovan S, Papadopoulos KP, Patnaik A, Rasco DW, Di Gravio D, Huang B, Gambhire D, Chen Y, et al. Phase Ib study of utomilumab (PF-05082566), a 4-1BB/CD137 agonist, in combination with pembrolizumab (MK-3475) in patients with advanced solid tumors. Clin Cancer Res. 2017;23(18):5349–5357. doi: 10.1158/1078-0432.CCR-17-1243.
    1. Aznar MA, Labiano S, Diaz-Lagares A, Molina C, Garasa S, Azpilikueta A, Etxeberria I, Sanchez-Paulete AR, Korman AJ, Esteller M, et al. CD137 (4-1BB) costimulation modifies DNA methylation in CD8(+) T cell-relevant genes. Cancer Immunol Res. 2018;6(1):69–78. doi: 10.1158/2326-6066.CIR-17-0159.
    1. Marin-Acevedo JA, Dholaria B, Soyano AE, Knutson KL, Chumsri S, Lou Y. Next generation of immune checkpoint therapy in cancer: new developments and challenges. J Hematol Oncol. 2018;11(1):39. doi: 10.1186/s13045-018-0582-8.
    1. Croft M, So T, Duan W, Soroosh P. The significance of OX40 and OX40L to T-cell biology and immune disease. Immunol Rev. 2009;229(1):173–191. doi: 10.1111/j.1600-065X.2009.00766.x.
    1. Hamid O, Ros W, Thompson JA, Hu-Lieskovan S, Eskens FALM, Diab A, Doi T, Wasser J, Spano J-P, Rizvi NA, Angevin E, Chiappori A, Ott PA, Ganguly BJ, Fleener C, Dell V, Liao K, Joh T, Chou J, El-Khoueiry A. Safety, pharmacokinetics (PK) and pharmacodynamics (PD) data from a phase I dose-escalation study of OX40 agonistic monoclonal antibody (mAb) PF-04518600 (PF-8600) in combination with utomilumab, a 4-1BB agonistic mAb. Ann Oncol. 2017;28(suppl_5).
    1. El-Khoueiry A. The relationship of pharmacodynamics (PD) and pharmacokinetics (PK) to clinical outcomes in a phase I study of OX40 agonistic monoclonal antibody (mAb) PF-04518600 (PF-8600) J Clin Oncol. 2017;35(15_suppl):3027. doi: 10.1200/JCO.2017.35.15_suppl.3027.
    1. Selvan SR, Dowling JP, Kelly WK, Lin J. Indoleamine 2,3-dioxygenase (IDO): biology and target in cancer immunotherapies. Curr Cancer Drug Targets. 2016;16(9):755–764. doi: 10.2174/1568009615666151030102250.
    1. Liu M, Wang X, Wang L, Ma X, Gong Z, Zhang S, Li Y. Targeting the IDO1 pathway in cancer: from bench to bedside. J Hematol Oncol. 2018;11(1):100. doi: 10.1186/s13045-018-0644-y.
    1. Mitchell TC, Hamid O, Smith DC, Bauer TM, Wasser JS, Olszanski AJ, Luke JJ, Balmanoukian AS, Schmidt EV, Zhao Y, et al. Epacadostat plus pembrolizumab in patients with advanced solid tumors: phase i results from a multicenter, open-label phase I/II trial (ECHO-202/KEYNOTE-037). Am J Clin Oncol. 2018:Jco2018789602.
    1. Smith D. Epacadostat plus pembrolizumab in patients with advanced urothelial carcinoma: preliminary phase I/II results of ECHO-202/KEYNOTE-037. J Clin Oncol. 2017;35(15):4503. doi: 10.1200/JCO.2017.35.15_suppl.4503.
    1. Smith A. Unique immunotherapy combinations provide an optimistic outlook in early urothelial carcinoma trials. In: Targeted Therapies in Oncology: Targeted Oncology; 2019.
    1. Hsu MM, Balar AV. PD-1/PD-L1 combinations in advanced urothelial cancer: rationale and current clinical trials. Clin Genitourin Cancer. 2019;17(3):e618–e626. doi: 10.1016/j.clgc.2019.03.009.
    1. Motz GT, Coukos G. The parallel lives of angiogenesis and immunosuppression: cancer and other tales. Nat Rev Immunol. 2011;11(10):702–711. doi: 10.1038/nri3064.
    1. Sankhwar M, Sankhwar SN, Abhishek A, Rajender S. Clinical significance of the VEGF level in urinary bladder carcinoma. Cancer Biomark. 2015;15(4):349–355. doi: 10.3233/CBM-150478.
    1. Cumberbatch K, He T, Thorogood Z, Gartrell BA. Emerging drugs for urothelial (bladder) cancer. Expert Opin Emerg Drugs. 2017;22(2):149–164. doi: 10.1080/14728214.2017.1336536.
    1. Wallin JJ, Bendell JC, Funke R, Sznol M, Korski K, Jones S, Hernandez G, Mier J, He X, Hodi FS, et al. Atezolizumab in combination with bevacizumab enhances antigen-specific T-cell migration in metastatic renal cell carcinoma. Nat Commun. 2016;7:12624. doi: 10.1038/ncomms12624.
    1. Petrylak D. A multicohort phase I study of ramucirumab (R) plus pembrolizumab (P): Interim safety and clinical activity in patients with urothelial carcinoma. J Clin Oncol. 2017;35(6):suppl, 349. doi: 10.1200/JCO.2017.35.6_suppl.349.
    1. Petrylak DP, de Wit R, Chi KN, Drakaki A, Sternberg CN, Nishiyama H, Castellano D, Hussain S, Flechon A, Bamias A, et al. Ramucirumab plus docetaxel versus placebo plus docetaxel in patients with locally advanced or metastatic urothelial carcinoma after platinum-based therapy (RANGE): a randomised, double-blind, phase 3 trial. Lancet. 2017;390(10109):2266–2277. doi: 10.1016/S0140-6736(17)32365-6.
    1. Choueiri TK, Escudier B, Powles T, Tannir NM, Mainwaring PN, Rini BI, Hammers HJ, Donskov F, Roth BJ, Peltola K, et al. Cabozantinib versus everolimus in advanced renal cell carcinoma (METEOR): final results from a randomised, open-label, phase 3 trial. Lancet Oncol. 2016;17(7):917–927. doi: 10.1016/S1470-2045(16)30107-3.
    1. Apolo A. A phase I study of cabozantinib plus nivolumab (CaboNivo) and cabonivo plus ipilimumab (CaboNivoIpi) in patients (pts) with refractory metastatic (m) urothelial carcinoma (UC) and other genitourinary (GU) tumors. J Clin Oncol. 2017;35(15_suppl):4562. doi: 10.1200/JCO.2017.35.15_suppl.4562.
    1. Nadal R. Results of phase I plus expansion cohorts of cabozantinib (Cabo) plus nivolumab (Nivo) and CaboNivo plus ipilimumab (Ipi) in patients (pts) with with metastatic urothelial carcinoma (mUC) and other genitourinary (GU) malignancies. J Clin Oncol. 2018;36(16_suppl):515. doi: 10.1200/JCO.2018.36.6_suppl.515.
    1. Nishiwada S, Sho M, Yasuda S, Shimada K, Yamato I, Akahori T, Kinoshita S, Nagai M, Konishi N, Nakajima Y. Nectin-4 expression contributes to tumor proliferation, angiogenesis and patient prognosis in human pancreatic cancer. J Exp Clin Cancer Res. 2015;34:30. doi: 10.1186/s13046-015-0144-7.
    1. Challita-Eid PM, Satpayev D, Yang P, An Z, Morrison K, Shostak Y, Raitano A, Nadell R, Liu W, Lortie DR, et al. Enfortumab vedotin antibody-drug conjugate targeting Nectin-4 Is a highly potent therapeutic agent in multiple preclinical cancer models. Cancer Res. 2016;76(10):3003–3013. doi: 10.1158/0008-5472.CAN-15-1313.
    1. Marin-Acevedo JA, Soyano AE, Dholaria B, Knutson KL, Lou Y. Cancer immunotherapy beyond immune checkpoint inhibitors. J Hematol Oncol. 2018;11(1):8. doi: 10.1186/s13045-017-0552-6.
    1. Petrylak D. A phase I study of enfortumab vedotin (ASG-22 CE; ASG-22ME): updated analysis of patients with metastatic urothelial cancer. J Clin Oncol. 2017;35(15_suppl):106. doi: 10.1200/JCO.2017.35.15_suppl.106.
    1. Rosenberg J. Updated results from the enfortumab vedotin phase 1 (EV-101) study in patients with metastatic urothelial cancer (mUC) J Clin Oncol. 2018;36(15_suppl):4504. doi: 10.1200/JCO.2018.36.15_suppl.4504.
    1. Hoimes C. EV-103 study: A phase 1b dose-escalation and dose-expansion study of enfortumab vedotin in combination with immune checkpoint inhibitor (CPI) therapy for treatment of patients with locally advanced or metastatic urothelial cancer. J Clin Oncol. 2018;36(6_suppl):TPS532. doi: 10.1200/JCO.2018.36.6_suppl.TPS532.
    1. Petrylak D. EV-301: Phase III study to evaluate enfortumab vedotin (EV) versus chemotherapy in patients with previously treated locally advanced or metastatic urothelial cancer (la/mUC) J Clin Oncol. 2019;37(7_supple):TPS497. doi: 10.1200/JCO.2019.37.7_suppl.TPS497.
    1. Petrylak DP. EV-201: Results of enfortumab vedotin monotherapy for locally advanced or metastatic urothelial cancer previously treated with platinum and immune checkpoint inhibitors. J Clin Oncol. 2019;37(18_suppl):4505. doi: 10.1200/JCO.2019.37.18_suppl.LBA4505.
    1. Koshkin VODP, Yu E, Grivas P. Systematic review: HER2 in bladder cancer. Bladder Cancer. 2019;5(1):1–12. doi: 10.3233/BLC-180196.
    1. Baselga J, Swain SM. Novel anticancer targets: revisiting ERBB2 and discovering ERBB3. Nat Rev Cancer. 2009;9(7):463–475. doi: 10.1038/nrc2656.
    1. Yan M, Schwaederle M, Arguello D, Millis SZ, Gatalica Z, Kurzrock R. HER2 expression status in diverse cancers: review of results from 37,992 patients. Cancer Metastasis Rev. 2015;34(1):157–164. doi: 10.1007/s10555-015-9552-6.
    1. Balversa (erdafitinib) full prescribing information. In. Edited by Administration. USFaD. Janssen Pharmaceutical Companies; 2019. . Accessed 9 May 2019.
    1. Pal SK, Rosenberg JE, Hoffman-Censits JH, Berger R, Quinn DI, Galsky MD, Wolf J, Dittrich C, Keam B, Delord JP, et al. Efficacy of BGJ398, a fibroblast growth factor receptor 1-3 inhibitor, in patients with previously treated advanced urothelial carcinoma with FGFR3 alterations. Cancer Dis. 2018;8(7):812–821. doi: 10.1158/-18-0229.
    1. Collin MP, Lobell M, Hubsch W, Brohm D, Schirok H, Jautelat R, Lustig K, Bomer U, Vohringer V, Heroult M, et al. Discovery of rogaratinib (BAY 1163877): a pan-FGFR Inhibitor. ChemMedChem. 2018;13(5):437–445. doi: 10.1002/cmdc.201700718.
    1. Xu KY, Wu S. Update on the treatment of metastatic clear cell and non-clear cell renal cell carcinoma. Biomark Res. 2015;3:5. doi: 10.1186/s40364-015-0030-7.
    1. Vitale MG, Carteni G. Recent developments in second and third line therapy of metastatic renal cell carcinoma. Expert Rev Anticancer Ther. 2016;16(5):469–471. doi: 10.1586/14737140.2016.1168696.
    1. Reed JP, Posadas EM, Figlin RA. Developments in the use of tyrosine kinase inhibitors in the treatment of renal cell carcinoma. Expert Rev Anticancer Ther. 2019;19(3):259–271. doi: 10.1080/14737140.2019.1573678.
    1. Williamson SR, Taneja K, Cheng L. Renal cell carcinoma staging: pitfalls, challenges, and updates. Histopathology. 2019;74(1):18–30. doi: 10.1111/his.13743.
    1. Fyfe G, Fisher RI, Rosenberg SA, Sznol M, Parkinson DR, Louie AC. Results of treatment of 255 patients with metastatic renal cell carcinoma who received high-dose recombinant interleukin-2 therapy. J Clin Oncol Off J Am Soc Clin Oncol. 1995;13(3):688–696. doi: 10.1200/JCO.1995.13.3.688.
    1. De Meerleer G, Khoo V, Escudier B, Joniau S, Bossi A, Ost P, Briganti A, Fonteyne V, Van Vulpen M, Lumen N, et al. Radiotherapy for renal-cell carcinoma. Lancet Oncol. 2014;15(4):e170–e177. doi: 10.1016/S1470-2045(13)70569-2.
    1. Ross K, Jones RJ. Immune checkpoint inhibitors in renal cell carcinoma. Clin Sci (Lond) 2017;131(21):2627–2642. doi: 10.1042/CS20160894.
    1. Negrier S, Maral J, Drevon M, Vinke J, Escudier B, Philip T. Long-term follow-up of patients with metastatic renal cell carcinoma treated with intravenous recombinant interleukin-2 in Europe. Cancer J Sci Am. 2000;6(Suppl 1):S93–S98.
    1. Ravaud A, Motzer RJ, Pandha HS, George DJ, Pantuck AJ, Patel A, Chang YH, Escudier B, Donskov F, Magheli A, et al. Adjuvant sunitinib in high-risk renal-cell carcinoma after nephrectomy. N Engl J Med. 2016;375(23):2246–2254. doi: 10.1056/NEJMoa1611406.
    1. Zarrabi K, Wu S. Current and emerging therapeutic targets for metastatic renal cell carcinoma. Curr Oncol Rep. 2018;20(5):41. doi: 10.1007/s11912-018-0684-z.
    1. Choueiri TK, Figueroa DJ, Fay AP, Signoretti S, Liu Y, Gagnon R, Deen K, Carpenter C, Benson P, Ho TH, et al. Correlation of PD-L1 tumor expression and treatment outcomes in patients with renal cell carcinoma receiving sunitinib or pazopanib: results from COMPARZ, a randomized controlled trial. Clin Cancer Res. 2015;21(5):1071–1077. doi: 10.1158/1078-0432.CCR-14-1993.
    1. Brahmer JR, Drake CG, Wollner I, Powderly JD, Picus J, Sharfman WH, Stankevich E, Pons A, Salay TM, McMiller TL, et al. Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol. 2010;28(19):3167–3175. doi: 10.1200/JCO.2009.26.7609.
    1. Chedgy EC, Black PC. Nivolumab: the new second line treatment for advanced renal-cell carcinoma commentary on: nivolumab versus everolimus in advanced renal-cell carcinoma. Urology. 2016;89:8–9. doi: 10.1016/j.urology.2015.12.003.
    1. Motzer RJ, Tannir NM, McDermott DF, Aren Frontera O, Melichar B, Choueiri TK, Plimack ER, Barthelemy P, Porta C, George S, et al. Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma. N Engl J Med. 2018;378(14):1277–1290. doi: 10.1056/NEJMoa1712126.
    1. Calvo E, Porta C, Grunwald V, Escudier B. The current and evolving landscape of first-line treatments for advanced renal cell carcinoma. Oncologist. 2019;24(3):338–348. doi: 10.1634/theoncologist.2018-0267.
    1. Motzer RJ, Penkov K, Haanen J, Rini B, Albiges L, Campbell MT, Venugopal B, Kollmannsberger C, Negrier S, Uemura M, et al. Avelumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. N Engl J Med. 2019;380(12):1103–1115. doi: 10.1056/NEJMoa1816047.
    1. Immunotherapy and TKI Combinations Are Emerging as Standard of Care in Advanced RCC. . Accessed 11 May 2019.
    1. Rini BI, Plimack ER, Stus V, Gafanov R, Hawkins R, Nosov D, Pouliot F, Alekseev B, Soulieres D, Melichar B, et al. Pembrolizumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. N Eng J Med. 2019;380(12):1116–1127. doi: 10.1056/NEJMoa1816714.
    1. Motzer R. IMmotion151: a randomized phase III study of atezolizumab plus bevacizumab vs sunitinib in untreated metastatic renal cell carcinoma (mRCC) J Clin Oncol. 2019;36(6_suppl):578. doi: 10.1200/JCO.2018.36.6_suppl.578.
    1. Network NCC . Version 1.2020 edn. 2019. Kidney Cancer, NCCN Clinical Practice Guidelines in Oncology.
    1. Huynh H, Ngo VC, Fargnoli J, Ayers M, Soo KC, Koong HN, Thng CH, Ong HS, Chung A, Chow P, et al. Brivanib alaninate, a dual inhibitor of vascular endothelial growth factor receptor and fibroblast growth factor receptor tyrosine kinases, induces growth inhibition in mouse models of human hepatocellular carcinoma. Clin Cancer Res. 2008;14(19):6146–6153. doi: 10.1158/1078-0432.CCR-08-0509.
    1. de Vinuesa AG, Bocci M, Pietras K, Ten Dijke P. Targeting tumour vasculature by inhibiting activin receptor-like kinase (ALK)1 function. Biochem Soc Trans. 2016;44(4):1142–1149. doi: 10.1042/BST20160093.
    1. Voss M. The DART Study: Part 1 results from the dalantercept plus axitinib dose escalation and expansion cohorts in patients with advanced renal cell carcinoma (RCC) J Clin Oncol. 2015;33(15):4567. doi: 10.1200/jco.2015.33.15_suppl.4567.
    1. Voss MH, Bhatt RS, Vogelzang NJ, Fishman M, Alter RS, Rini BI, Beck JT, Joshi M, Hauke R, Atkins MB, et al. A phase 2, randomized trial evaluating the combination of dalantercept plus axitinib in patients with advanced clear cell renal cell carcinoma. Cancer. 2019;125(14):2400–2408. doi: 10.1002/cncr.32061.
    1. Melao A. Acceleron discontinues development of dalantercept for advanced renal cell carcinoma therapy. In: immuno-oncologynews, vol. 2019: : Bionews Feeds; 2017.
    1. Hahn AW, Pal SK, Agarwal N. Targeting endoglin to treat metastatic renal cell carcinoma: lessons from Osler-Weber-Rendu syndrome. Oncologist. 2019;24(2):143–145. doi: 10.1634/theoncologist.2018-0443.
    1. Choueiri TK, Michaelson MD, Posadas EM, Sonpavde GP, McDermott DF, Nixon AB, Liu Y, Yuan Z, Seon BK, Walsh M, et al. An open label phase Ib dose escalation study of TRC105 (anti-endoglin antibody) with axitinib in patients with metastatic renal cell carcinoma. Oncologist. 2019;24(2):202–210. doi: 10.1634/theoncologist.2018-0299.
    1. Ueda R. Clinical application of anti-CCR4 monoclonal antibody. Oncology. 2015;89(Suppl 1):16–21. doi: 10.1159/000431059.
    1. Cheng X, Wu H, Jin ZJ, Ma D, Yuen S, Jing XQ, Shi MM, Shen BY, Peng CH, Zhao R, et al. Up-regulation of chemokine receptor CCR4 is associated with Human Hepatocellular Carcinoma malignant behavior. Sci Rep. 2017;7(1):12362. doi: 10.1038/s41598-017-10267-4.
    1. Maxwell PH, Wiesener MS, Chang GW, Clifford SC, Vaux EC, Cockman ME, Wykoff CC, Pugh CW, Maher ER, Ratcliffe PJ. The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature. 1999;399(6733):271–275. doi: 10.1038/20459.
    1. Schoffski P, Wozniak A, Escudier B, Rutkowski P, Anthoney A, Bauer S, Sufliarsky J, van Herpen C, Lindner LH, Grunwald V, et al. Crizotinib achieves long-lasting disease control in advanced papillary renal-cell carcinoma type 1 patients with MET mutations or amplification. EORTC 90101 CREATE trial. Eur J Cancer. 2017;87:147–163. doi: 10.1016/j.ejca.2017.10.014.
    1. Sawyers CL. Will mTOR inhibitors make it as cancer drugs? Cancer Cell. 2003;4(5):343–348. doi: 10.1016/S1535-6108(03)00275-7.
    1. Hudes G, Carducci M, Tomczak P, Dutcher J, Figlin R, Kapoor A, Staroslawska E, Sosman J, McDermott D, Bodrogi I, et al. Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N Eng J Med. 2007;356(22):2271–2281. doi: 10.1056/NEJMoa066838.
    1. Knox JJ, Barrios CH, Kim TM, Cosgriff T, Srimuninnimit V, Pittman K, Sabbatini R, Rha SY, Flaig TW, Page RD, et al. Final overall survival analysis for the phase II RECORD-3 study of first-line everolimus followed by sunitinib versus first-line sunitinib followed by everolimus in metastatic RCC. Ann Oncol. 2017;28(6):1339–1345. doi: 10.1093/annonc/mdx075.
    1. Amaravadi RK, Lippincott-Schwartz J, Yin XM, Weiss WA, Takebe N, Timmer W, DiPaola RS, Lotze MT, White E. Principles and current strategies for targeting autophagy for cancer treatment. Clin Cancer Res. 2011;17(4):654–666. doi: 10.1158/1078-0432.CCR-10-2634.
    1. Mauthe M, Orhon I, Rocchi C, Zhou X, Luhr M, Hijlkema KJ, Coppes RP, Engedal N, Mari M, Reggiori F. Chloroquine inhibits autophagic flux by decreasing autophagosome-lysosome fusion. Autophagy. 2018;14(8):1435–1455. doi: 10.1080/15548627.2018.1474314.
    1. Haas NB, Appleman LJ, Stein M, Redlinger M, Wilks M, Xu X, Onorati A, Kalavacharla A, Kim T, Zhen CJ, et al. Autophagy inhibition to augment mTOR inhibition: a Phase I/II trial of everolimus and hydroxychloroquine in patients with previously treated renal cell carcinoma. Clin Cancer Res. 2019;25(7):2080–2087. doi: 10.1158/1078-0432.CCR-18-2204.
    1. Choueiri T. A phase 3, randomized, open-label study of nivolumab combined with cabozantinib vs sunitinib in patients with previously untreated advanced or metastatic renal cell carcinoma (RCC; CheckMate 9ER) J Clin Oncol. 2018;36(15_cuppl):TPS4598. doi: 10.1200/JCO.2018.36.15_suppl.TPS4598.
    1. Keeler M. Pembrolizumab (pembro) and cabozantinib (cabo) in patients (pts) with metastatic renal cell carcinoma (mRCC): Phase I results. J Clin Oncol. 2019;37(7_suppl):600. doi: 10.1200/JCO.2019.37.7_suppl.600.
    1. Rini BI, Stein M, Shannon P, Eddy S, Tyler A, Stephenson JJ, Jr, Catlett L, Huang B, Healey D, Gordon M. Phase 1 dose-escalation trial of tremelimumab plus sunitinib in patients with metastatic renal cell carcinoma. Cancer. 2011;117(4):758–767. doi: 10.1002/cncr.25639.
    1. Labriola MK, Batich KA, Zhu J, McNamara MA, Harrison MR, Armstrong AJ, George DJ, Zhang T. Immunotherapy is changing first-line treatment of metastatic renal-cell carcinoma. Clin Genitourin Cancer. 2019;17(3):e513–e521. doi: 10.1016/j.clgc.2019.01.017.
    1. Zang X, Allison JP. The B7 family and cancer therapy: costimulation and coinhibition. Clin Cancer Res. 2007;13(18 Pt 1):5271–5279. doi: 10.1158/1078-0432.CCR-07-1030.
    1. Janakiram M, Chinai JM, Fineberg S, Fiser A, Montagna C, Medavarapu R, Castano E, Jeon H, Ohaegbulam KC, Zhao R, et al. Expression, clinical significance, and receptor identification of the newest B7 family member HHLA2 protein. Clin Cancer Res. 2015;21(10):2359–2366. doi: 10.1158/1078-0432.CCR-14-1495.
    1. Janakiram M, Shah UA, Liu W, Zhao A, Schoenberg MP, Zang X. The third group of the B7-CD28 immune checkpoint family: HHLA2, TMIGD2, B7x, and B7-H3. Immunol Rev. 2017;276(1):26–39. doi: 10.1111/imr.12521.
    1. Chen D, Chen W, Xu Y, Zhu M, Xiao Y, Shen Y, Zhu S, Cao C, Xu X. Upregulated immune checkpoint HHLA2 in clear cell renal cell carcinoma: a novel prognostic biomarker and potential therapeutic target. J Med Genet. 2019;56(1):43–49. doi: 10.1136/jmedgenet-2018-105454.
    1. Amato RJ. Vaccine therapy for renal cell carcinoma. Rev Urol. 2003;5(2):65–71.
    1. Li Z, Song W, Rubinstein M, Liu D. Recent updates in cancer immunotherapy: a comprehensive review and perspective of the 2018 China Cancer Immunotherapy Workshop in Beijing. J Hematol Oncol. 2018;11(1):142. doi: 10.1186/s13045-018-0684-3.
    1. Srivastava PK, Amato RJ. Heat shock proteins: the ‘Swiss Army Knife’ vaccines against cancers and infectious agents. Vaccine. 2001;19(17-19):2590–2597. doi: 10.1016/S0264-410X(00)00492-8.
    1. Rini BI, Stenzl A, Zdrojowy R, Kogan M, Shkolnik M, Oudard S, Weikert S, Bracarda S, Crabb SJ, Bedke J, et al. IMA901, a multipeptide cancer vaccine, plus sunitinib versus sunitinib alone, as first-line therapy for advanced or metastatic renal cell carcinoma (IMPRINT): a multicentre, open-label, randomised, controlled, phase 3 trial. Lancet Oncol. 2016;17(11):1599–1611. doi: 10.1016/S1470-2045(16)30408-9.
    1. Amin A, Dudek AZ, Logan TF, Lance RS, Holzbeierlein JM, Knox JJ, Master VA, Pal SK, Miller WH, Jr, Karsh LI, et al. Survival with AGS-003, an autologous dendritic cell-based immunotherapy, in combination with sunitinib in unfavorable risk patients with advanced renal cell carcinoma (RCC): Phase 2 study results. J Immunother Cancer. 2015;3:14. doi: 10.1186/s40425-015-0055-3.
    1. Wallgren AC, Andersson B, Backer A, Karlsson-Parra A. Direct allorecognition promotes activation of bystander dendritic cells and licenses them for Th1 priming: a functional link between direct and indirect allosensitization. Scand J Immunol. 2005;62(3):234–242. doi: 10.1111/j.1365-3083.2005.01663.x.
    1. Laurell A, Lonnemark M, Brekkan E, Magnusson A, Tolf A, Wallgren AC, Andersson B, Adamson L, Kiessling R, Karlsson-Parra A. Intratumorally injected pro-inflammatory allogeneic dendritic cells as immune enhancers: a first-in-human study in unfavourable risk patients with metastatic renal cell carcinoma. J Immunother Cancer. 2017;5:52. doi: 10.1186/s40425-017-0255-0.
    1. Negoita S, Feuer EJ, Mariotto A, Cronin KA, Petkov VI, Hussey SK, Benard V, Henley SJ, Anderson RN, Fedewa S, et al. Annual Report to the Nation on the Status of Cancer, part II: recent changes in prostate cancer trends and disease characteristics. Cancer. 2018;124(13):2801–2814. doi: 10.1002/cncr.31549.
    1. Mottet N, Bellmunt J, Bolla M, Briers E, Cumberbatch MG, De Santis M, Fossati N, Gross T, Henry AM, Joniau S, et al. EAU-ESTRO-SIOG Guidelines on Prostate Cancer. Part 1: screening, diagnosis, and local treatment with curative intent. Eur Urol. 2017;71(4):618–629. doi: 10.1016/j.eururo.2016.08.003.
    1. Hu R, Dunn TA, Wei S, Isharwal S, Veltri RW, Humphreys E, Han M, Partin AW, Vessella RL, Isaacs WB, et al. Ligand-independent androgen receptor variants derived from splicing of cryptic exons signify hormone-refractory prostate cancer. Cancer Res. 2009;69(1):16–22. doi: 10.1158/0008-5472.CAN-08-2764.
    1. Scher HI, Solo K, Valant J, Todd MB, Mehra M. Prevalence of prostate cancer clinical states and mortality in the United States: estimates using a dynamic progression model. PLoS One. 2015;10(10):e0139440. doi: 10.1371/journal.pone.0139440.
    1. Sharifi N, Dahut WL, Steinberg SM, Figg WD, Tarassoff C, Arlen P, Gulley JL. A retrospective study of the time to clinical endpoints for advanced prostate cancer. BJU Int. 2005;96(7):985–989. doi: 10.1111/j.1464-410X.2005.05798.x.
    1. Berthold DR, Pond GR, Soban F, de Wit R, Eisenberger M, Tannock IF. Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer: updated survival in the TAX 327 study. J Clin Oncol. 2008;26(2):242–245. doi: 10.1200/JCO.2007.12.4008.
    1. Faiena I, Salmasi A, Pantuck AJ, Drakaki A. Re: abiraterone for prostate cancer not previously treated with hormone therapy. Eur Urol. 2018;73(6):981. doi: 10.1016/j.eururo.2018.01.023.
    1. Fizazi K, Tran N, Fein L, Matsubara N, Rodriguez-Antolin A, Alekseev BY, Ozguroglu M, Ye D, Feyerabend S, Protheroe A, 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. Lancet Oncol. 2019;20(5):686–700. doi: 10.1016/S1470-2045(19)30082-8.
    1. Scher HI, Fizazi K, Saad F, Taplin ME, Sternberg CN, Miller K, de Wit R, Mulders P, Chi KN, Shore ND, et al. Increased survival with enzalutamide in prostate cancer after chemotherapy. N Eng J Med. 2012;367(13):1187–1197. doi: 10.1056/NEJMoa1207506.
    1. Caffo O, De Giorgi U, Fratino L, Alesini D, Zagonel V, Facchini G, Gasparro D, Ortega C, Tucci M, Verderame F, et al. Clinical outcomes of castration-resistant prostate cancer treatments administered as third or fourth line following failure of docetaxel and other second-line treatment: results of an Italian multicentre study. Eur Urol. 2015;68(1):147–153. doi: 10.1016/j.eururo.2014.10.014.
    1. Virgo KS, Basch E, Loblaw DA, Oliver TK, Rumble RB, Carducci MA, Nordquist L, Taplin ME, Winquist E, Singer EA. Second-line hormonal therapy for men with chemotherapy-naive, castration-resistant prostate cancer: American Society of Clinical Oncology Provisional Clinical Opinion. J Clin Oncol. 2017;35(17):1952–1964. doi: 10.1200/JCO.2017.72.8030.
    1. Jung ME, Ouk S, Yoo D, Sawyers CL, Chen C, Tran C, Wongvipat J. Structure-activity relationship for thiohydantoin androgen receptor antagonists for castration-resistant prostate cancer (CRPC) J Med Chem. 2010;53(7):2779–2796. doi: 10.1021/jm901488g.
    1. Clegg NJ, Wongvipat J, Joseph JD, Tran C, Ouk S, Dilhas A, Chen Y, Grillot K, Bischoff ED, Cai L, et al. ARN-509: a novel antiandrogen for prostate cancer treatment. Cancer Res. 2012;72(6):1494–1503. doi: 10.1158/0008-5472.CAN-11-3948.
    1. Smith MR, Saad F, Chowdhury S, Oudard S, Hadaschik BA, Graff JN, Olmos D, Mainwaring PN, Lee JY, Uemura H, et al. Apalutamide treatment and metastasis-free survival in prostate cancer. N Eng J Med. 2018;378(15):1408–1418. doi: 10.1056/NEJMoa1715546.
    1. Beer TM, Armstrong AJ, Rathkopf DE, Loriot Y, Sternberg CN, Higano CS, Iversen P, Bhattacharya S, Carles J, Chowdhury S, et al. Enzalutamide in metastatic prostate cancer before chemotherapy. N Eng J Med. 2014;371(5):424–433. doi: 10.1056/NEJMoa1405095.
    1. Hussain M, Fizazi K, Saad F, Rathenborg P, Shore N, Ferreira U, Ivashchenko P, Demirhan E, Modelska K, Phung, et al. Enzalutamide in men with nonmetastatic, castration-resistant prostate cancer. N Engl J Med. 2018;378(26):2465–2474. doi: 10.1056/NEJMoa1800536.
    1. Chi KN, Chowdhury S, Radziszewski P, et al. TITAN: a randomized, double-blind, placebo-controlled, phase 3 trial of apalutamide (ARN-509) plus androgen deprivation therapy (ADT) in metastatic hormone-sensitive prostate cancer (mHSPC) Ann Oncol. 2016;27(6):771TiP. doi: 10.1093/annonc/mdw068.
    1. Janssen Announces ERLEADA®(apalutamide) Phase 3 TITAN Study unblinded as dual primary endpoints achieved in clinical program evaluating treatment of patients with metastatic castration-sensitive prostate cancer. Janssen. 2019. . Accessed 3 Apr 2019.
    1. Janssen Submits Application to U.S. FDA seeking approval of ERLEADA®(apalutamide) for patients with metastatic castration-sensitive prostate cancer. In.: Janssen. ; 2019.
    1. Chi KN, Agarwal N, Bjartell A, Chung BH, Pereira de Santana Gomes AJ, Given R, Juarez Soto A, Merseburger AS, Ozguroglu M, Uemura H, et al. Apalutamide for metastatic, castration-sensitive prostate cancer. N Engl J Med. 2019;381(1):13–24. doi: 10.1056/NEJMoa1903307.
    1. Rathkopf DE, Antonarakis ES, Shore ND, Tutrone RF, Alumkal JJ, Ryan CJ, Saleh M, Hauke RJ, Bandekar R, Maneval EC, et al. Safety and Antitumor Activity of Apalutamide (ARN-509) in metastatic castration-resistant prostate cancer with and without prior abiraterone acetate and prednisone. Clin Cancer Res. 2017;23(14):3544–3551. doi: 10.1158/1078-0432.CCR-16-2509.
    1. Rexer H, Graefen M. Phase III study for local or locally advanced prostate cancer : randomized, double-blind, placebo-controlled phase 3 study of apalutamide in patients with local high-risk prostate cancer or locally advanced prostate cancer receiving primary radiotherapy (ATLAS) - study AP 90/15 of the AUO. Urologe A. 2017;56(2):243–244. doi: 10.1007/s00120-017-0329-0.
    1. Borgmann H, Lallous N, Ozistanbullu D, Beraldi E, Paul N, Dalal K, Fazli L, Haferkamp A, Lejeune P, Cherkasov A, et al. Moving towards precision urologic oncology: targeting enzalutamide-resistant prostate cancer and mutated forms of the androgen receptor using the novel inhibitor darolutamide (ODM-201) Eur Urol. 2018;73(1):4–8. doi: 10.1016/j.eururo.2017.08.012.
    1. Joseph JD, Lu N, Qian J, Sensintaffar J, Shao G, Brigham D, Moon M, Maneval EC, Chen I, Darimont B, et al. A clinically relevant androgen receptor mutation confers resistance to second-generation antiandrogens enzalutamide and ARN-509. Cancer Dis. 2013;3(9):1020–1029. doi: 10.1158/-13-0226.
    1. Sugawara T, Baumgart SJ, Nevedomskaya E, Reichert K, Steuber H, Lejeune P, Mumberg D, Haendler B. Darolutamide is a potent androgen receptor antagonist with strong efficacy in prostate cancer models. Int J Cancer. 2019;145(5):1382–1394.
    1. Hodgson MC, Shen HC, Hollenberg AN, Balk SP. Structural basis for nuclear receptor corepressor recruitment by antagonist-liganded androgen receptor. Mol Cancer Ther. 2008;7(10):3187–3194. doi: 10.1158/1535-7163.MCT-08-0461.
    1. Girard BJ, Daniel AR, Lange CA, Ostrander JH. PELP1: a review of PELP1 interactions, signaling, and biology. Mol Cell Endocrinol. 2014;382(1):642–651. doi: 10.1016/j.mce.2013.07.031.
    1. Singh SS, Li Y, Ford OH, Wrzosek CS, Mehedint DC, Titus MA, Mohler JL. Thioredoxin reductase 1 expression and castration-recurrent growth of prostate cancer. Transl Oncol. 2008;1(3):153–157. doi: 10.1593/tlo.08145.
    1. Shore ND, Tammela TL, Massard C, Bono P, Aspegren J, Mustonen M, Fizazi K. Safety and antitumour activity of ODM-201 (BAY-1841788) in chemotherapy-naive and CYP17 inhibitor-naive patients: follow-up from the ARADES and ARAFOR Trials. Eur Urol Focus. 2018;4(4):547–553. doi: 10.1016/j.euf.2017.01.015.
    1. Fizazi K, Shore N, Tammela TL, Ulys A, Vjaters E, Polyakov S, Jievaltas M, Luz M, Alekseev B, Kuss I, et al. Darolutamide in nonmetastatic, castration-resistant prostate cancer. N Eng J Med. 2019;380(13):1235–1246. doi: 10.1056/NEJMoa1815671.
    1. Rafferty SW, Eisner JR, Moore WR, Schotzinger RJ, Hoekstra WJ. Highly-selective 4-(1,2,3-triazole)-based P450c17a 17,20-lyase inhibitors. Bioorg Med Chem Lett. 2014;24(11):2444–2447. doi: 10.1016/j.bmcl.2014.04.024.
    1. Norris JD, Ellison SJ, Baker JG, Stagg DB, Wardell SE, Park S, Alley HM, Baldi RM, Yllanes A, Andreano KJ, et al. Androgen receptor antagonism drives cytochrome P450 17A1 inhibitor efficacy in prostate cancer. J Clin Invest. 2017;127(6):2326–2338. doi: 10.1172/JCI87328.
    1. Attard G, Reid AH, Auchus RJ, Hughes BA, Cassidy AM, Thompson E, Oommen NB, Folkerd E, Dowsett M, Arlt W, et al. Clinical and biochemical consequences of CYP17A1 inhibition with abiraterone given with and without exogenous glucocorticoids in castrate men with advanced prostate cancer. J Clin Endocrinol Metab. 2012;97(2):507–516. doi: 10.1210/jc.2011-2189.
    1. Boudadi K, Antonarakis ES. Resistance to novel antiandrogen therapies in metastatic castration-resistant prostate cancer. Clin Med Insights Oncol. 2016;10(Suppl 1):1–9.
    1. Gupta S, Nordquist LT, Fleming MT, Berry WR, Zhang J, Ervin SL, Eisner JR, Baskin-Bey ES, Shore ND. Phase I study of seviteronel, a selective CYP17 lyase and androgen receptor inhibitor, in men with castration-resistant prostate cancer. Clin Cancer Res. 2018;24(21):5225–5232. doi: 10.1158/1078-0432.CCR-18-0564.
    1. Anassi E, Ndefo UA. Sipuleucel-T (provenge) injection: the first immunotherapy agent (vaccine) for hormone-refractory prostate cancer. P T. 2011;36(4):197–202.
    1. Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, Redfern CH, Ferrari AC, Dreicer R, Sims RB, et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Eng J Med. 2010;363(5):411–422. doi: 10.1056/NEJMoa1001294.
    1. Noguchi M, Moriya F, Koga N, Matsueda S, Sasada T, Yamada A, Kakuma T, Itoh K. A randomized phase II clinical trial of personalized peptide vaccination with metronomic low-dose cyclophosphamide in patients with metastatic castration-resistant prostate cancer. Cancer Immunol. 2016;65(2):151–160. doi: 10.1007/s00262-015-1781-6.
    1. Cordes LM, Gulley JL, Madan RA. The evolving role of immunotherapy in prostate cancer. Curr Opin Oncol. 2016;28(3):232–240. doi: 10.1097/CCO.0000000000000281.
    1. Kantoff PW, Schuetz TJ, Blumenstein BA, Glode LM, Bilhartz DL, Wyand M, Manson K, Panicali DL, Laus R, Schlom J, 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(7):1099–1105. doi: 10.1200/JCO.2009.25.0597.
    1. van den Eertwegh AJ, Versluis J, van den Berg HP, Santegoets SJ, van Moorselaar RJ, van der Sluis TM, Gall HE, Harding TC, Jooss K, Lowy I, 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(5):509–517. doi: 10.1016/S1470-2045(12)70007-4.
    1. Madan RA, Gulley JL. Prospects for the future of prostate cancer vaccines. Expert Rev Vacc. 2016;15(3):271–274. doi: 10.1586/14760584.2015.1118348.
    1. Gao J, Ward JF, Pettaway CA, Shi LZ, Subudhi SK, Vence LM, Zhao H, Chen J, Chen H, Efstathiou E, et al. VISTA is an inhibitory immune checkpoint that is increased after ipilimumab therapy in patients with prostate cancer. Nat Med. 2017;23(5):551–555. doi: 10.1038/nm.4308.
    1. Anti-PD-1-CTLA4 combo hits prostate cancer. Cancer Dscov. 2019;9(5):569–70.
    1. McNeel DG, Smith HA, Eickhoff JC, Lang JM, Staab MJ, Wilding G, Liu G. Phase I trial of tremelimumab in combination with short-term androgen deprivation in patients with PSA-recurrent prostate cancer. Cancer Immunol Immunother. 2012;61(7):1137–1147. doi: 10.1007/s00262-011-1193-1.
    1. Hansen AR, Massard C, Ott PA, Haas NB, Lopez JS, Ejadi S, Wallmark JM, Keam B, Delord JP, Aggarwal R, et al. Pembrolizumab for advanced prostate adenocarcinoma: findings of the KEYNOTE-028 study. Ann Oncol. 2018;29(8):1807–1813. doi: 10.1093/annonc/mdy232.
    1. Bono JD. KEYNOTE-199: Pembrolizumab (pembro) for docetaxel-refractory metastatic castration-resistant prostate cancer (mCRPC) J Clin Oncol. 2018;36(15_suppl):5007. doi: 10.1200/JCO.2018.36.15_suppl.5007.
    1. Dziadkowiec KN, Gasiorowska E, Nowak-Markwitz E, Jankowska A. PARP inhibitors: review of mechanisms of action and BRCA1/2 mutation targeting. Prz Menopauzalny. 2016;15(4):215–219.
    1. Martin GA, Chen AH, Parikh K. A novel use of olaparib for the treatment of metastatic castration-recurrent prostate cancer. Pharmacotherapy. 2017;37(11):1406–1414. doi: 10.1002/phar.2027.
    1. Mateo J, Carreira S, Sandhu S, Miranda S, Mossop H, Perez-Lopez R, Nava Rodrigues D, Robinson D, Omlin A, Tunariu N, et al. DNA-repair defects and olaparib in metastatic prostate cancer. N Eng J Med. 2015;373(18):1697–1708. doi: 10.1056/NEJMoa1506859.
    1. Clarke N, Wiechno P, Alekseev B, Sala N, Jones R, Kocak I, Chiuri VE, Jassem J, Flechon A, Redfern C, 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. 2018;19(7):975–986. doi: 10.1016/S1470-2045(18)30365-6.
    1. Yu EMC, Retz M, Tafreshi A, Galceran JC, Hammerer P, Fong P, Shore N, et al. Keynote-365 cohort a: pembrolizumab (pembro) plus olaparib in docetaxel-pretreated patients (pts) with metastatic castrate-resistant prostate cancer (mCRPC) J Clin Oncol. 2019;37(7_supple):145. doi: 10.1200/JCO.2019.37.7_suppl.145.
    1. Armenia J, Wankowicz SAM, Liu D, Gao J, Kundra R, Reznik E, Chatila WK, Chakravarty D, Han GC, Coleman I, et al. The long tail of oncogenic drivers in prostate cancer. Nat Genet. 2018;50(5):645–651. doi: 10.1038/s41588-018-0078-z.
    1. Cancer Genome Atlas Research N The molecular taxonomy of primary prostate cancer. Cell. 2015;163(4):1011–1025. doi: 10.1016/j.cell.2015.10.025.
    1. Castro E, Romero-Laorden N, Del Pozo A, Lozano R, Medina A, Puente J, Piulats JM, Lorente D, Saez MI, Morales-Barrera R, et al. PROREPAIR-B: a prospective cohort study of the impact of germline dna repair mutations on the outcomes of patients with metastatic castration-resistant prostate cancer. J Clin Oncol. 2019;37(6):490–503. doi: 10.1200/JCO.18.00358.
    1. Annala M, Struss WJ, Warner EW, Beja K, Vandekerkhove G, Wong A, Khalaf D, Seppala IL, So A, Lo G, et al. Treatment outcomes and tumor loss of heterozygosity in germline DNA repair-deficient prostate cancer. Eur Urol. 2017;72(1):34–42. doi: 10.1016/j.eururo.2017.02.023.
    1. Mateo J. 2019 American Society of Clinical Oncology Annual Meeting. Chicago: Journal of Clinical Oncology; 2019. TOPARP-B: A phase II randomized trial of the poly(ADP)-ribose polymerase (PARP) inhibitor olaparib for metastatic castration resistant prostate cancers (mCRPC) with DNA damage repair (DDR) alterations.
    1. Robinson D, Van Allen EM, Wu YM, Schultz N, Lonigro RJ, Mosquera JM, Montgomery B, Taplin ME, Pritchard CC, Attard G, et al. Integrative clinical genomics of advanced prostate cancer. Cell. 2015;161(5):1215–1228. doi: 10.1016/j.cell.2015.05.001.
    1. Nombela Paz, Lozano Rebeca, Aytes Alvaro, Mateo Joaquin, Olmos David, Castro Elena. BRCA2 and Other DDR Genes in Prostate Cancer. Cancers. 2019;11(3):352. doi: 10.3390/cancers11030352.
    1. Pomerantz MM, Spisak S, Jia L, Cronin AM, Csabai I, Ledet E, Sartor AO, Rainville I, O'Connor EP, Herbert ZT, et al. The association between germline BRCA2 variants and sensitivity to platinum-based chemotherapy among men with metastatic prostate cancer. Cancer. 2017;123(18):3532–3539. doi: 10.1002/cncr.30808.
    1. Francini E, Sweeney CJ. Docetaxel activity in the era of life-prolonging hormonal therapies for metastatic castration-resistant prostate cancer. European urology. 2016;70(3):410–412. doi: 10.1016/j.eururo.2016.05.002.
    1. Pienta KJ. Preclinical mechanisms of action of docetaxel and docetaxel combinations in prostate cancer. Seminars in oncology. 2001;28(4 Suppl 15):3–7. doi: 10.1016/S0093-7754(01)90148-4.
    1. Miller K. Abiraterone plus prednisone in patients with newly diagnosed high-risk metastatic hormone-sensitive prostate cancer (mHSPC). Aktuelle Urol. 2019.
    1. Caffo A. A multicentric phase II randomized trial of docetaxel (D) plus enzalutamide (E) versus docetaxel (D) as first-line chemotherapy for patients (pts) with metastatic castration-resistant prostate cancer (mCRPC): CHEIRON study. J Clin Oncol. 2019;37(7_suppl):148. doi: 10.1200/JCO.2019.37.7_suppl.148.
    1. Kyriakopoulos CE, Chen YH, Carducci MA, Liu G, Jarrard DF, Hahn NM, Shevrin DH, Dreicer R, Hussain M, Eisenberger M, 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. 2018;36(11):1080–1087. doi: 10.1200/JCO.2017.75.3657.
    1. Sternberg CN, Petrylak DP, Sartor O, Witjes JA, Demkow T, Ferrero JM, Eymard JC, Falcon S, Calabro F, James N, et al. Multinational, double-blind, phase III study of prednisone and either satraplatin or placebo in patients with castrate-refractory prostate cancer progressing after prior chemotherapy: the SPARC trial. J Clin Oncol. 2009;27(32):5431–5438. doi: 10.1200/JCO.2008.20.1228.
    1. Cheng HH, Pritchard CC, Boyd T, Nelson PS, Montgomery B. Biallelic inactivation of BRCA2 in platinum-sensitive metastatic castration-resistant prostate cancer. Eur Urol. 2016;69(6):992–995. doi: 10.1016/j.eururo.2015.11.022.
    1. Zafeiriou Z, Bianchini D, Chandler R, Rescigno P, Yuan W, Carreira S, Barrero M, Petremolo A, Miranda S, Riisnaes R, et al. Genomic analysis of three metastatic prostate cancer patients with exceptional responses to carboplatin indicating different types of DNA repair deficiency. Eur Urol. 2019;75(1):184–192. doi: 10.1016/j.eururo.2018.09.048.
    1. Bubendorf L, Schopfer A, Wagner U, Sauter G, Moch H, Willi N, Gasser TC, Mihatsch MJ. Metastatic patterns of prostate cancer: an autopsy study of 1,589 patients. Hum Pathol. 2000;31(5):578–583. doi: 10.1053/hp.2000.6698.
    1. Hegemann M, Bedke J, Stenzl A, Todenhofer T. Denosumab treatment in the management of patients with advanced prostate cancer: clinical evidence and experience. Ther Adv Urol. 2017;9(3-4):81–88. doi: 10.1177/1756287216686018.
    1. Prelaj A, Rebuzzi SE, Buzzacchino F, Pozzi C, Ferrara C, Frantellizzi V, Follacchio GA, Civitelli L, De Vincentis G, Tomao S, et al. Radium-223 in patients with metastatic castration-resistant prostate cancer: Efficacy and safety in clinical practice. Oncol Lett. 2019;17(2):1467–1476.
    1. Smith M, Parker C, Saad F, Miller K, Tombal B, Ng QS, Boegemann M, Matveev V, Piulats JM, Zucca LE, et al. Addition of radium-223 to abiraterone acetate and prednisone or prednisolone in patients with castration-resistant prostate cancer and bone metastases (ERA 223): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2019;20(3):408–419. doi: 10.1016/S1470-2045(18)30860-X.
    1. Rajasekaran AK, Anilkumar G, Christiansen JJ. Is prostate-specific membrane antigen a multifunctional protein? Am J Physiol Cell Physiol. 2005;288(5):C975–C981. doi: 10.1152/ajpcell.00506.2004.
    1. Wright GL, Jr, Haley C, Beckett ML, Schellhammer PF. Expression of prostate-specific membrane antigen in normal, benign, and malignant prostate tissues. Urol Oncol. 1995;1(1):18–28. doi: 10.1016/1078-1439(95)00002-Y.
    1. Liu H, Rajasekaran AK, Moy P, Xia Y, Kim S, Navarro V, Rahmati R, Bander NH. Constitutive and antibody-induced internalization of prostate-specific membrane antigen. Cancer Res. 1998;58(18):4055–4060.
    1. Emmett L, Willowson K, Violet J, Shin J, Blanksby A, Lee J. Lutetium (177) PSMA radionuclide therapy for men with prostate cancer: a review of the current literature and discussion of practical aspects of therapy. J Med Radiat Sci. 2017;64(1):52–60. doi: 10.1002/jmrs.227.
    1. Violet J, Jackson P, Ferdinandus J, Sandhu S, Akhurst T, Iravani A, Kong G, Kumar AR, Thang SP, Eu P, et al. Dosimetry of (177)Lu-PSMA-617 in metastatic castration-resistant prostate cancer: correlations between pretherapeutic imaging and whole-body tumor dosimetry with treatment outcomes. J Nucl Med. 2019;60(4):517–523. doi: 10.2967/jnumed.118.219352.
    1. Halliday PR, Blakely CM, Bivona TG. Emerging targeted therapies for the treatment of non-small cell lung cancer. Curr Oncol Rep. 2019;21(3):21. doi: 10.1007/s11912-019-0770-x.
    1. Guo J, Jin J, Oya M, Uemura H, Takahashi S, Tatsugami K, Rha SY, Lee JL, Chung J, Lim HY, et al. Safety of pazopanib and sunitinib in treatment-naive patients with metastatic renal cell carcinoma: Asian versus non-Asian subgroup analysis of the COMPARZ trial. J Hematol Oncol. 2018;11(1):69. doi: 10.1186/s13045-018-0617-1.
    1. Duma N, Abdel-Ghani A, Yadav S, Hoversten KP, Reed CT, Sitek AN, Enninga EAL, Paludo J, Aguilera JV, Leventakos K, et al. Sex differences in tolerability to anti-programmed cell death protein 1 therapy in patients with metastatic melanoma and non-small cell lung cancer: are we all equal? Oncologist. 2019.
    1. Betof AS, Nipp RD, Giobbie-Hurder A, Johnpulle RAN, Rubin K, Rubinstein SM, Flaherty KT, Lawrence DP, Johnson DB, Sullivan RJ. Impact of age on outcomes with immunotherapy for patients with melanoma. Oncologist. 2017;22(8):963–971. doi: 10.1634/theoncologist.2016-0450.
    1. Ghatalia P, Yang ES, Lasseigne BN, Ramaker RC, Cooper SJ, Chen D, Sudarshan S, Wei S, Guru AS, Zhao A, et al. Kinase gene expression profiling of metastatic clear cell renal cell carcinoma tissue identifies potential new therapeutic targets. PLoS One. 2016;11(8):e0160924. doi: 10.1371/journal.pone.0160924.
    1. Ayers M, Lunceford J, Nebozhyn M, Murphy E, Loboda A, Kaufman DR, Albright A, Cheng JD, Kang SP, Shankaran V, et al. IFN-gamma-related mRNA profile predicts clinical response to PD-1 blockade. J Clin Invest. 2017;127(8):2930–2940. doi: 10.1172/JCI91190.
    1. Vasyutin I, Zerihun L, Ivan C, Atala A. Bladder organoids and spheroids: potential tools for normal and diseased tissue modelling. Anticancer Res. 2019;39(3):1105–1118. doi: 10.21873/anticanres.13219.
    1. Maia MC, Grivas P, Agarwal N, Pal SK. Circulating tumor DNA in bladder cancer: novel applications and future directions. Eur Urol. 2018;73(4):541–542. doi: 10.1016/j.eururo.2017.10.011.
    1. Thalgott M, Rack B, Maurer T, Souvatzoglou M, Eiber M, Kress V, Heck MM, Andergassen U, Nawroth R, Gschwend JE, et al. Detection of circulating tumor cells in different stages of prostate cancer. J Cancer Res Clin Oncol. 2013;139(5):755–763. doi: 10.1007/s00432-013-1377-5.

Source: PubMed

3
Se inscrever