269
Views
2
CrossRef citations to date
0
Altmetric
Review

Recent Advances in the Management of Hormone-Sensitive Oligometastatic Prostate Cancer

ORCID Icon, ORCID Icon, ORCID Icon, , , , , & ORCID Icon show all
Pages 89-101 | Published online: 06 Jan 2022

References

  • Tosoian JJ, Gorin MA, Ross AE, et al. Oligometastatic prostate cancer: definitions, clinical outcomes, and treatment considerations. Nat Rev Urol. 2017;14:15–25. doi:10.1038/nrurol.2016.175
  • Foster CC, Weichselbaum RR, Pitroda SP. Oligometastatic prostate cancer: reality or figment of imagination? Cancer. 2019;125:340–352. doi:10.1002/cncr.31860
  • Fraser M, Koontz B, Emmenegger U, et al. What Is Oligometastatic Prostate Cancer? Eur Urol Focus. 2019;5:159–161. doi:10.1016/j.euf.2018.12.009
  • Sumiyoshi T, Chi KN, Wyatt AW. Clinical implications of genomic alterations in metastatic prostate cancer. Prostate Cancer Prostatic Dis. 2021;24:310–322. doi:10.1038/s41391-020-00308-x
  • Kothari G, Ost P, Cheung P, et al. Trends in management of oligometastatic hormone-sensitive prostate cancer. Curr Oncol Rep. 2019;21(43). doi:10.1007/s11912-019-0791-5
  • Lievens Y, Guckenberger M, Gomez D, et al. Defining oligometastatic disease from a radiation oncology perspective: an ESTRO-ASTRO consensus document. Radiother Oncol J Eur Soc Ther Radiol Oncol. 2020;148:157–166. doi:10.1016/j.radonc.2020.04.003
  • APCCC 2019: oligometastatic Prostate Cancer – definitions and Concepts. Available from: https://www.urotoday.com/conference-highlights/apccc-2019/114616-apccc-2019-oligometastatic-prostate-cancer-definitions-and-concepts.html. Accessed December 10, 2021.
  • Rogowski P, Roach M, Schmidt-Hegemann NS, et al. Radiotherapy of oligometastatic prostate cancer: a systematic review. Radiat Oncol. 2021;16(50):1–6. doi:10.1186/s13014-021-01776-8
  • Marvaso G, Volpe S, Pepa M, et al. Oligorecurrent prostate cancer and stereotactic body radiotherapy: where are we now? A systematic review and meta-analysis of prospective studies. Eur Urol Open Sci. 2021;27:19–28. doi:10.1016/j.euros.2021.02.008
  • Mazzola R, Francolini G, Triggiani L, et al. Metastasis-directed Therapy (SBRT) Guided by PET-CT 18F-CHOLINE Versus PET-CT 68Ga-PSMA in Castration-sensitive Oligorecurrent Prostate Cancer: a Comparative Analysis of Effectiveness. Clin Genitourin Cancer. 2021;19:230–236. doi:10.1016/j.clgc.2020.08.002
  • Albisinni S, Tafuri A, Pirozzi M, et al. A systematic review of imaging-guided metastasis-directed therapy for oligorecurrent prostate cancer: revolution or devolution? Minerva Urol Nefrol;2020. 72. doi:10.23736/S0393-2249.19.03586-0
  • Sritharan K, Rieu R, Tree A. A narrative review of oligometastatic prostate cancer—an evolving paradigm. Ann Palliat Med. 2021;10:5969–5987. doi:10.21037/apm-20-1215
  • De Bleser E, Jereczek-Fossa BA, Pasquier D, et al. Metastasis-directed therapy in treating nodal oligorecurrent prostate cancer: a multi-institutional analysis comparing the outcome and toxicity of stereotactic body radiotherapy and elective nodal radiotherapy. Eur Urol. 2019;76:732–739. doi:10.1016/j.eururo.2019.07.009
  • Ost P, Reynders D, Decaestecker K, et al. Surveillance or metastasis-directed therapy for oligometastatic prostate cancer recurrence: a prospective, randomized, multicenter phase II trial. J Clin Oncol. 2018;36:446–453. doi:10.1200/JCO.2017.75.4853
  • Deek MP, Phillips RM, Tran PT. Local Therapies in Oligometastatic and Oligoprogressive Prostate Cancer. Semin Radiat Oncol. 2021;31:242–249. doi:10.1016/j.semradonc.2021.03.007
  • Perez-Lopez R, Tunariu N, Padhani AR, et al. Imaging diagnosis and follow-up of advanced prostate cancer: clinical perspectives and state of the art. Radiology. 2019;292:273–286. doi:10.1148/radiol.2019181931
  • Rao A, Vapiwala N, Schaeffer EM, Ryan CJ. Oligometastatic prostate cancer: a shrinking subset or an opportunity for cure? Am Soc Clin Oncol Educ Book. 2019;309–320. doi:10.1200/EDBK_239041
  • Hövels AM, Heesakkers RAM, Adang EM, et al. The diagnostic accuracy of CT and MRI in the staging of pelvic lymph nodes in patients with prostate cancer: a meta-analysis. Clin Radiol. 2008;63:387–395. doi:10.1016/j.crad.2007.05.022
  • Shen G, Deng H, Hu S, Jia Z. Comparison of choline-PET/CT, MRI, SPECT, and bone scintigraphy in the diagnosis of bone metastases in patients with prostate cancer: a meta-analysis. Skeletal Radiol. 2014;43:1503–1513. doi:10.1007/s00256-014-1903-9
  • Farolfi A, Koschel S, Murphy DG, Fanti S. PET imaging in urology: a rapidly growing successful collaboration. Curr Opin Urol. 2020;Publish Ahead of Print. doi:10.1097/MOU.0000000000000800
  • Lecouvet FE, Oprea-Lager DE, Liu Y, et al. Use of modern imaging methods to facilitate trials of metastasis-directed therapy for oligometastatic disease in prostate cancer: a consensus recommendation from the EORTC Imaging Group. Lancet Oncol. 2018;19:e534–e545. doi:10.1016/S1470-2045(18)30571-0
  • Farolfi A, Hadaschik B, Hamdy FC, et al. Positron emission tomography and whole-body magnetic resonance imaging for metastasis-directed therapy in hormone-sensitive oligometastatic prostate cancer after primary radical treatment: a systematic review. Eur Urol Oncol. 2021:S2588931121000377. doi:10.1016/j.euo.2021.02.003
  • Gillessen S, Attard G, Beer TM, et al. Management of Patients with Advanced Prostate Cancer: the Report of the Advanced Prostate Cancer Consensus Conference APCCC 2017. Eur Urol. 2018;73:178–211. doi:10.1016/j.eururo.2017.06.002
  • FDA approves new diagnostic imaging agent FLUCICLOVINE F-18 to detect recurrent prostate cancer « New Drug Approvals. Available from: https://newdrugapprovals.org/2016/05/28/fda-approves-new-diagnostic-imaging-agent-fluciclovine-f-18-to-detect-recurrent-prostate-cancer/. Accessed December 10, 2021.
  • Axumin® (fluciclovine F 18) injection | terms & Conditions. Available from: https://www.axumin.com/sites/default/files/2018-03/Axumin_PI_08_2016_Clean. Accessed December 10, 2021.
  • Kim EH, Siegel BA, Teoh EJ, Andriole GL. Prostate cancer recurrence in patients with negative or equivocal conventional imaging: a role for 18F-fluciclovine-PET/CT in delineating sites of recurrence and identifying patients with oligometastatic disease. Urol Oncol Semin Orig Investig. 2021;39:365.e9–365.e16.
  • Andriole GL, Kostakoglu L, Chau A, et al. The Impact of Positron Emission Tomography with 18F-Fluciclovine on the Treatment of Biochemical Recurrence of Prostate Cancer: results from the LOCATE Trial. J Urol. 2019;201:322–331. doi:10.1016/j.juro.2018.08.050
  • Woo S, Suh CH, Kim SY, Cho JY, Kim SH. The diagnostic performance of MRI for detection of lymph node metastasis in bladder and prostate cancer: an updated systematic review and diagnostic meta-analysis. Am J Roentgenol. 2018;210:W95–W109. doi:10.2214/AJR.17.18481
  • Padhani AR, Lecouvet FE, Tunariu N, et al. METastasis reporting and data system for prostate cancer: practical guidelines for acquisition, interpretation, and reporting of whole-body magnetic resonance imaging-based evaluations of multiorgan involvement in advanced prostate cancer. Eur Urol. 2017;71:81–92. doi:10.1016/j.eururo.2016.05.033
  • Sweeney CJ, Chen Y-H, Carducci M, et al. Chemohormonal Therapy in Metastatic Hormone-Sensitive Prostate Cancer. N Engl J Med. 2015;373:737–746. doi:10.1056/NEJMoa1503747
  • Gravis G, Fizazi K, Joly F, et al. Androgen-deprivation therapy alone or with docetaxel in non-castrate metastatic prostate cancer (GETUG-AFU 15): a randomised, open-label, Phase 3 trial. Lancet Oncol. 2013;14:149–158. doi:10.1016/S1470-2045(12)70560-0
  • Kyriakopoulos CE, Chen Y-H, Carducci MA, 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 Off J Am Soc Clin Oncol. 2018;36:1080–1087. doi:10.1200/JCO.2017.75.3657
  • Barata PC, Sartor AO. Metastatic castration‐sensitive prostate cancer: abiraterone, docetaxel, or …. Cancer. 2019;125:1777–1788. doi:10.1002/cncr.32039
  • Sweeney CJ, Martin AJ, Stockler MR, et al. Overall survival of men with metachronous metastatic hormone-sensitive prostate cancer treated with enzalutamide and androgen deprivation therapy. Eur Urol. 2021;80:275–279. doi:10.1016/j.eururo.2021.05.016
  • Davis ID, Martin AJ, Stockler MR, et al. Enzalutamide with standard first-line therapy in metastatic prostate cancer. N Engl J Med. 2019;381:121–131. doi:10.1056/NEJMoa1903835
  • Armstrong AJ, Szmulewitz RZ, Petrylak DP, et al. ARCHES: a randomized, phase iii study of androgen deprivation therapy with enzalutamide or placebo in men with metastatic hormone-sensitive prostate cancer. J Clin Oncol Off J Am Soc Clin Oncol. 2019;37:2974–2986. doi:10.1200/JCO.19.00799
  • Armstrong AJ, Shore ND, Szmulewitz RZ, et al. Efficacy of Enzalutamide plus Androgen Deprivation Therapy in Metastatic Hormone-Sensitive Prostate Cancer by Pattern of Metastatic Spread: ARCHES Post Hoc Analyses. J Urol. 2021;205:1361–1371. doi:10.1097/JU.0000000000001568
  • Sathianathen NJ, Koschel S, Thangasamy IA, et al. Indirect comparisons of efficacy between combination approaches in metastatic hormone-sensitive prostate cancer: a systematic review and network meta-analysis. Eur Urol. 2020;77:365–372. doi:10.1016/j.eururo.2019.09.004
  • Madan RA, Gulley JL. Finding an immunologic beachhead in the prostate cancer microenvironment. J Natl Cancer Inst. 2019;111:219–220. doi:10.1093/jnci/djy145
  • Zhao SG, Lehrer J, Chang SL, et al. The Immune Landscape of Prostate Cancer and Nomination of PD-L2 as a Potential Therapeutic Target. J Natl Cancer Inst. 2019;111:301–310. doi:10.1093/jnci/djy141
  • Patel D, McKay R, Parsons JK. Immunotherapy for Localized Prostate Cancer: the Next Frontier? Urol Clin North Am. 2020;47:443–456. doi:10.1016/j.ucl.2020.07.008
  • Hasan H, Deek MP, Phillips R, et al. A phase II randomized trial of RAdium-223 dichloride and SABR Versus SABR for oligomEtastatic prostate caNcerS (RAVENS). BMC Cancer. 2020;20(492). doi:10.1186/s12885-020-07000-2
  • Privé BM, Peters SMB, Muselaers CHJ, et al. Lutetium-177-PSMA-617 in Low-volume hormone-sensitive metastatic prostate cancer: a Prospective Pilot Study. Clin Cancer Res Off J Am Assoc Cancer Res. 2021;27:3595–3601. doi:10.1158/1078-0432.CCR-20-4298
  • Privé BM, Janssen MJR, van Oort IM, et al. Update to a randomized controlled trial of lutetium-177-PSMA in Oligo-metastatic hormone-sensitive prostate cancer: the BULLSEYE trial. Trials. 2021;22(768). doi:10.1186/s13063-021-05733-4
  • Kucharczyk MJ, Gravis G, Niazi T. The biology of oligometastatic prostate cancer: a different beast than polymetastatic prostate cancer. Eur Urol Focus. 2019;5:117–118. doi:10.1016/j.euf.2018.11.011
  • Lussier YA, Xing HR, Salama JK, et al. MicroRNA expression characterizes oligometastasis(es). PLoS One. 2011;6(e28650):e28650. doi:10.1371/journal.pone.0028650
  • Lussier YA, Khodarev NN, Regan K, et al. Oligo- and polymetastatic progression in lung metastasis(es) patients is associated with specific microRNAs. PLoS One. 2012;7(e50141):e50141. doi:10.1371/journal.pone.0050141
  • Joice GA, Rowe SP, Pienta KJ, Gorin MA. Oligometastatic prostate cancer: shaping the definition with molecular imaging and an improved understanding of tumor biology. Curr Opin Urol. 2017;27:533–541. doi:10.1097/MOU.0000000000000449
  • Dhondt B, De Bleser E, Claeys T, et al. Discovery and validation of a serum microRNA signature to characterize oligo- and polymetastatic prostate cancer: not ready for prime time. World J Urol. 2019;37:2557–2564. doi:10.1007/s00345-018-2609-8
  • Zhang W, Wu Y, Zhang Z, et al. Controlling Nutritional Status score: a new prognostic indicator for patients with oligometastatic prostate cancer. Curr Probl Cancer. 2019;43:461–470. doi:10.1016/j.currproblcancer.2019.02.001
  • Sonpavde G. The biology of prostate cancer metastases: does oligo differ from polymetastatic? Curr Opin Urol. 2017;27:542–546. doi:10.1097/MOU.0000000000000434
  • Li Y, Tang Z-Y, Ye S-L, et al. Establishment of cell clones with different metastatic potential from the metastatic hepatocellular carcinoma cell line MHCC97. World J Gastroenterol. 2001;7:630–636. doi:10.3748/wjg.v7.i5.630
  • Uppal A, Wightman SC, Mallon S, et al. 14q32-encoded microRNAs mediate an oligometastatic phenotype. Oncotarget. 2015;6:3540–3552. doi:10.18632/oncotarget.2920
  • Reyes DK, Pienta KJ. The biology and treatment of oligometastatic cancer. Oncotarget. 2015;6:8491–8524. doi:10.18632/oncotarget.3455
  • Arancio W, Belmonte B, Castiglia M, Di Napoli A, Tripodo C. Tissue Versus Liquid Biopsy: opposite or Complementary? In: Russo A, Giordano A, Rolfo C editors. Liquid Biopsy in Cancer Patients: The Hand Lens for Tumor Evolution. Springer International Publishing; 2017:41–49. doi:10.1007/978-3-319-55661-1_4
  • Ghosh RK, Pandey T, Dey P. Liquid biopsy: a new avenue in pathology. Cytopathol off J Br Soc Clin Cytol. 2019;30:138–143. doi:10.1111/cyt.12661
  • Stelcer E, Konkol M, Głȩboka A, Suchorska WM. Liquid Biopsy in Oligometastatic Prostate Cancer-A Biologist’s Point of View. Front Oncol. 2019;9(775). doi:10.3389/fonc.2019.00775
  • Wang J, Ni J, Beretov J, et al. Exosomal microRNAs as liquid biopsy biomarkers in prostate cancer. Crit Rev Oncol Hematol. 2020;145(102860):102860. doi:10.1016/j.critrevonc.2019.102860
  • Hegemann M, Stenzl A, Bedke J, et al. Liquid biopsy: ready to guide therapy in advanced prostate cancer? BJU Int. 2016;118:855–863. doi:10.1111/bju.13586
  • Gkountela S, Castro-Giner F, Szczerba BM, et al. Circulating Tumor Cell Clustering Shapes DNA Methylation to Enable Metastasis Seeding. Cell. 2019;176:98–112.e14. doi:10.1016/j.cell.2018.11.046
  • Faugeroux V, Lefebvre C, Pailler E, et al. An Accessible and Unique Insight into Metastasis Mutational Content Through Whole-exome Sequencing of Circulating Tumor Cells in Metastatic Prostate Cancer. Eur Urol Oncol. 2020;3:498–508. doi:10.1016/j.euo.2018.12.005
  • Mandel PC, Huland H, Tiebel A, et al. Enumeration and Changes in Circulating Tumor Cells and Their Prognostic Value in Patients Undergoing Cytoreductive Radical Prostatectomy for Oligometastatic Prostate Cancer-Translational Research Results from the Prospective ProMPT trial. Eur Urol Focus. 2021;7:55–62. doi:10.1016/j.euf.2019.05.008
  • Barriere G, Fici P, Gallerani G, et al. Circulating tumor cells and epithelial, mesenchymal and stemness markers: characterization of cell subpopulations. Ann Transl Med. 2014;2:109. doi:10.3978/j.issn.2305-5839.2014.10.04
  • Magbanua MJM, Solanki TI, Ordonez AD, Hsiao F, Park JW. Enumeration of Circulating Tumor Cells and Disseminated Tumor Cells in Blood and Bone Marrow by Immunomagnetic Enrichment and Flow Cytometry (IE/FC). Methods Mol Biol Clifton NJ. 1634;203–210:2017.
  • Togo S, Katagiri N, Namba Y, et al. Sensitive detection of viable circulating tumor cells using a novel conditionally telomerase-selective replicating adenovirus in non-small cell lung cancer patients. Oncotarget. 2017;8(21):34884–34895. doi:10.18632/oncotarget.16818
  • Mansilla C, Soria E, Ramírez N. The identification and isolation of CTCs: a biological Rubik’s cube. Crit Rev Oncol Hematol. 2018;126:129–134. doi:10.1016/j.critrevonc.2018.03.027
  • Lowes LE, Bratman S, Dittamore R, et al. Circulating Tumor Cells (CTC) and Cell-Free DNA (cfDNA) Workshop 2016: scientific Opportunities and Logistics for Cancer Clinical Trial Incorporation. Int J Mol Sci. 2016;17:E1505. doi:10.3390/ijms17091505
  • Elshimali YI, Khaddour H, Sarkissyan M, Wu Y, Vadgama JV. The clinical utilization of circulating cell free DNA (CCFDNA) in blood of cancer patients. Int J Mol Sci. 2013;14:18925–18958. doi:10.3390/ijms140918925
  • Salvi S, Gurioli G, De Giorgi U, et al. Cell-free DNA as a diagnostic marker for cancer: current insights. OncoTargets Ther. 2016;9:6549–6559. doi:10.2147/OTT.S100901
  • Esquela-Kerscher A, Slack FJ. Oncomirs - microRNAs with a role in cancer. Nat Rev Cancer. 2006;6:259–269. doi:10.1038/nrc1840
  • Zabolotneva AA, Zhavoronkov A, Garazha AV, Roumiantsev SA, Buzdin AA. Characteristic patterns of microRNA expression in human bladder cancer. Front Genet. 2012;3(310):2521.
  • Hessvik NP, Sandvig K, Llorente A. Exosomal miRNAs as Biomarkers for Prostate Cancer. Front Genet. 2013;4(36). doi:10.3389/fgene.2013.00036
  • Mulrane L, McGee SF, Gallagher WM, O’Connor D. miRNA dysregulation in breast cancer. Cancer Res. 2013;73:6554–6562. doi:10.1158/0008-5472.CAN-13-1841
  • Cheng HH, Plets M, Li H, et al. Circulating microRNAs and treatment response in the Phase II SWOG S0925 study for patients with new metastatic hormone-sensitive prostate cancer. The Prostate. 2018;78(2):121–127. doi:10.1002/pros.23452
  • Dhondt B, Rousseau Q, De Wever O, Hendrix A. Function of extracellular vesicle-associated miRNAs in metastasis. Cell Tissue Res. 2016;365:621–641. doi:10.1007/s00441-016-2430-x
  • Al-Kafaji G, Said HM, Alam MA, Al Naieb ZT. Blood-based microRNAs as diagnostic biomarkers to discriminate localized prostate cancer from benign prostatic hyperplasia and allow cancer-risk stratification. Oncol Lett. 2018;16:1357–1365. doi:10.3892/ol.2018.8778
  • Bhagirath D, Yang TL, Bucay N, et al. microRNA-1246 Is an Exosomal Biomarker for Aggressive Prostate Cancer. Cancer Res. 2018;78(7):1833–1844. doi:10.1158/0008-5472.CAN-17-2069
  • Paziewska A, Mikula M, Dabrowska M, et al. Candidate diagnostic miRNAs that can detect cancer in prostate biopsy. Prostate. 2018;78(3):178–185. doi:10.1002/pros.23427
  • Tu J, Peng Q, Shen Y, et al. Identification of biomarker microRNA-mRNA regulatory pairs for predicting the docetaxel resistance in prostate cancer. J Cancer. 2019;10:5469–5482. doi:10.7150/jca.29032
  • Cochetti G, Vermandois JA, Maulà V, et al. Role of miRNAs in prostate cancer: do we really know everything? Urol Oncol Semin Orig Investig. 2020;38:623–635.