The Role of Liquid Biopsy in Metastatic Prostate Cancer

Authors

  • Joana Glória Instituto de Ciências Biomédicas Abel Salazar, Porto, Portugal
  • Diogo Nunes-Carneiro Departamento de Urologia do Hospital de Santo António - Centro Hospitalar e Universitário do Porto, Instituto de Ciências Biomédicas Abel Salazar, i3S/INEB - Universidade do Porto, Porto, Portugal https://orcid.org/0000-0001-5910-2018
  • Avelino Fraga Departamento de Urologia do Hospital de Santo António - Centro Hospitalar e Universitário do Porto, Instituto de Ciências Biomédicas Abel Salazar, i3S/INEB - Universidade do Porto, Porto, Portugal https://orcid.org/0000-0002-4824-8123

DOI:

https://doi.org/10.24915/aup.37.1-2.116

Keywords:

Biomarkers, Tumor, Liquid Biopsy, Prostatic Neoplasms

Abstract

Prostate cancer is the second most common malignancy in men. The high prevalence of prostate cancer, as well as its relatively indolent clinical course and the emerging therapeutic options, have led to the need to identify biomarkers that may aid in clinical decisions and may reflect response to treatments. “Liquid biopsy” is a generic term applied to the study of biomarkers attributed to tumors that are circulating in the body fluids of cancer patients. It relies on the principle that in these patients there are tumor cells and fragments with genomic content of tumor that circulate and can be detected and used as biomarkers for clinical application. Most of these biomarkers correlate with tumor burden and are most often identified in individuals with metastatic disease. The most studied biomarkers in prostate cancer patients can be subdivided into three major groups: circulating tumor cells, free genetic material (such as RNA and DNA - namely, microRNA and circulating free DNA) and extracellular vesicles (where there is inclusion of specific material and single tumor). This knowledge may allow the development of potential clinical applications from diagnosis to prognosis, as well as the development of predictive biomarkers of response to treatment and disease monitoring.

Downloads

Download data is not yet available.

References

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer

statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide

for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394-424.

Vandekerkhove G, Chi KN, Wyatt AW. Clinical utility of emerging liquid

biomarkers in advanced prostate cancer. Cancer Genet. 2018;228-9:151-8.

Morrison GJ, Goldkorn A. Development and Application of Liquid Biopsies in

Metastatic Prostate Cancer. Curr Oncol Rep. 2018;20:35.

Riaz IB, Wang L, Kohli M. Liquid biopsy approach in the management of

prostate cancer. TranslRes. 2018;201:60-70. doi: 10.1016/j.trsl.2018.05.004.

Hegemann M, Stenzl A, Bedke J, Chi KN, Black PC, Todenhofer T. Liquid

biopsy: ready to guide therapy in advanced prostate cancer? BJU Int. 2016;

:855-63.

Ashworth T. A case of cancer in which cells similar to those in the tumors were

seen in the blood after death. Aust Med J. 1869;14:146-9.

Zainfeld D, Goldkorn A. Liquid Biopsy in Prostate Cancer: Circulating Tumor

Cells and Beyond. Cancer Treat Res. 2018;175:87-104.

Alix-Panabieres C, Pantel K. Technologies for detection of circulating tumor

cells: facts and vision. Lab Chip. 2014;14:57-62.

de Bono JS, Scher HI, Montgomery RB, Parker C, Miller MC, Tissing H, et al.

Circulating tumor cells predict survival benefit from treatment in metastatic

castration-resistant prostate cancer. Clin Cancer Res. 2008;14:6302-9.

Goldkorn A PM, Agarwal N, Hussain M, Lara P, Vaena DA, et al. Circulating

tumor cells (CTCs) in SWOG S1216: A phase 3 multicenter trial in metastatic

hormone sensitive prostate cancer (mHSPC). J Clin Oncol. 2016;34.

Okegawa T, Nutahara K, Higashihara E. Immunomagnetic quantification of

circulating tumor cells as a prognostic factor of androgen deprivation responsiveness

in patients with hormone naive metastatic prostate cancer. J Urol.

;180:1342-7.

Goodman OB, Jr., Symanowski JT, Loudyi A, Fink LM, Ward DC, Vogelzang

NJ. Circulating tumor cells as a predictive biomarker in patients with hormonesensitive

prostate cancer. Clin Genitourin Cancer. 2011;9:31-8.

Lorente D, Olmos D, Mateo J, Bianchini D, Seed G, Fleisher M, et al. Decline in

Circulating Tumor Cell Count and Treatment Outcome in Advanced Prostate

Cancer. Eur Urol. 2016;70:985-92.

Goldkorn A, Ely B, Quinn DI, Tangen CM, Fink LM, Xu T, et al. Circulating tumor

cell counts are prognostic of overall survival in SWOG S0421: a phase III trial of

docetaxel with or without atrasentan for metastatic castration-resistant prostate

cancer. J Clin Oncol. 2014;32:1136-42.

Scher HI, Heller G, Molina A, Attard G, Danila DC, Jia X, et al. Circulating tumor

cell biomarker panel as an individual-level surrogate for survival in metastatic

castration-resistant prostate cancer. J Clin Oncol. 2015;33:1348-55.

Magbanua MJ, Sosa EV, Scott JH, Simko J, Collins C, Pinkel D, et al. Isolation

and genomic analysis of circulating tumor cells from castration resistant

metastatic prostate cancer.BMCCancer. 2012;12:78.

Jiang Y, Palma JF, Agus DB, Wang Y, Gross ME. Detection of androgen

receptor mutations in circulating tumor cells in castration-resistant prostate

cancer. Clin Chem. 2010;56:1492-5.

Darshan MS, Loftus MS, Thadani-Mulero M, Levy BP, Escuin D, Zhou XK, et al.

Taxane-induced blockade to nuclear accumulation of the androgen receptor

predicts clinical responses in metastatic prostate cancer. Cancer Res. 2011;

:6019-29.

Antonarakis ES, Lu C,Wang H, Luber B, Nakazawa M, Roeser JC, et al. AR-V7

and resistance to enzalutamide and abiraterone in prostate cancer. N Engl J

Med. 2014;371:1028-38.

De Laere B, van Dam PJ, Whitington T, Mayrhofer M, Diaz EH, Van den Eynden

G, et al. Comprehensive Profiling of the Androgen Receptor in Liquid Biopsies

from Castration-resistant Prostate Cancer Reveals Novel Intra-AR Structural

Variation and Splice Variant Expression Patterns. Eur Urol. 2017;72:192-200.

Antonarakis ES, Lu C, Luber B, Wang H, Chen Y, Nakazawa M, et al. Androgen

Receptor Splice Variant 7 and Efficacy of Taxane Chemotherapy in PatientsWith

MetastaticCastration-ResistantProstateCancer. JAMAOncol. 2015;1:582-91.

Antonarakis ES, Lu C, Luber B, Wang H, Chen Y, Zhu Y, et al. Clinical Significance

of Androgen Receptor Splice Variant-7 mRNA Detection in Circulating

Tumor Cells of Men With Metastatic Castration-Resistant Prostate

Cancer Treated With First- and Second-Line Abiraterone and Enzalutamide. J

Clin Oncol. 2017;35:2149-56.

McDaniel AS, Ferraldeschi R, Krupa R, Landers M, Graf R, Louw J, et al.

Phenotypic diversity of circulating tumour cells in patients with metastatic

castration-resistant prostate cancer. BJU Int. 2017;120:E30-E44.

Scher HI, Graf RP, Schreiber NA, McLaughlin B, Jendrisak A, Wang Y, et al.

Phenotypic Heterogeneity of Circulating Tumor Cells Informs Clinical Decisions

between AR Signaling Inhibitors and Taxanes in Metastatic Prostate Cancer.

Cancer Res. 2017;77:5687-98.

Attard G, Swennenhuis JF, Olmos D, Reid AH, Vickers E, A’Hern R, et al.

Characterization of ERG, AR and PTEN gene status in circulating tumor cells

from patients with castration-resistant prostate cancer. Cancer Res. 2009;69:

-8.

Reig O, Marin-Aguilera M, Carrera G, Jimenez N, Pare L, Garcia-Recio S, et al.

TMPRSS2-ERG in Blood and Docetaxel Resistance in Metastatic Castrationresistant

Prostate Cancer. Eur Urol. 2016;70:709-13.

Punnoose EA, Ferraldeschi R, Szafer-Glusman E, Tucker EK, Mohan S, Flohr P,

et al. PTEN loss in circulating tumour cells correlates with PTEN loss in fresh

tumour tissue from castration-resistant prostate cancer patients. Br J Cancer.

;113:1225-33.

Goldkorn A, Ely B, Tangen CM, Tai YC, Xu T, Li H, et al. Circulating tumor cell

telomerase activity as a prognostic marker for overall survival in SWOG 0421: a

phase III metastatic castration resistant prostate cancer trial. Int J Cancer.

;136:1856-62.

Xu T, Lu B, Tai YC, Goldkorn A. A cancer detection platform which measures

telomerase activity from live circulating tumor cells captured on a microfilter.

Cancer Res. 2010;70:6420-6.

Marrugo-Ramirez J, Mir M, Samitier J. Blood-Based Cancer Biomarkers in

Liquid Biopsy: A Promising Non-Invasive Alternative to Tissue Biopsy. Int J Mol

Sci. 2018;19 pii: E2877. doi: 10.3390/ijms19102877.

Di Meo A, Bartlett J, Cheng Y, Pasic MD, Yousef GM. Liquid biopsy: a step

forward towards precision medicine in urologic malignancies. Mol Cancer.

;16:80.

Sunami E, Shinozaki M, Higano CS, Wollman R, Dorff TB, Tucker SJ, et al.

Multimarker circulating DNA assay for assessing blood of prostate cancer

patients. Clin Chem. 2009;55:559-67.

Schutz E, Akbari MR, Beck J, Urnovitz H, Zhang WW, Bornemann-Kolatzki K,

et al. Chromosomal instability in cell-free DNA is a serum biomarker for prostate

cancer. Clin Chem. 2015;61:239-48.

Chun FK, Muller I, Lange I, Friedrich MG, Erbersdobler A, Karakiewicz PI, et al.

Circulating tumour-associated plasma DNA represents an independent and

informative predictor of prostate cancer. BJU Int. 2006;98:544-8.

Altimari A, Grigioni AD, Benedettini E, Gabusi E, Schiavina R, Martinelli A, et al.

Diagnostic role of circulating free plasma DNA detection in patients with

localized prostate cancer.AmJ Clin Pathol. 2008;129:756-62.

Horning AM, Awe JA,Wang CM, Liu J, Lai Z,Wang VY, et al. DNA methylation

screening of primary prostate tumors identifies SRD5A2 and CYP11A1 as

candidate markers for assessing risk of biochemical recurrence. Prostate.

;75:1790-801.

Romanel A, Gasi Tandefelt D, Conteduca V, Jayaram A, Casiraghi N, Wetterskog

D, et al. Plasma AR and abiraterone-resistant prostate cancer. Sci Transl

Med. 2015;7:312re10.

Kwee S, Song M-A, Cheng I, Loo L, Tiirikainen M. Measurement of Circulating

Cell-Free DNA in Relation to 18F-Fluorocholine PET/CT Imaging in Chemotherapy-

Treated Advanced Prostate Cancer. Clinical and Translational Science.

;5:65-70.

Kienel A, Porres D, Heidenreich A, Pfister D. cfDNA as a Prognostic Marker of

Response to Taxane Based Chemotherapy in Patients with Prostate Cancer. J

Urol. 2015;194:966-71.

Ulz P, Belic J, Graf R, Auer M, Lafer I, Fischereder K, et al. Whole-genome

plasma sequencing reveals focal amplifications as a driving force in metastatic

prostate cancer. Nat Commun. 2016;7:12008.

Heitzer E, Ulz P, Belic J, Gutschi S, Quehenberger F, Fischereder K, et al.

Tumor-associated copy number changes in the circulation of patients with

prostate cancer identified through whole-genome sequencing. Genome Med.

;5:30.

Wyatt AW, Azad AA, Volik SV, Annala M, Beja K, McConeghy B, et al. Genomic

Alterations in Cell-Free DNA and Enzalutamide Resistance in Castration-

Resistant Prostate Cancer. JAMA Oncol. 2016;2:1598-606.

Azad AA, Volik SV, Wyatt AW, Haegert A, Le Bihan S, Bell RH, et al. Androgen

Receptor Gene Aberrations in Circulating Cell-Free DNA: Biomarkers of

Therapeutic Resistance in Castration-Resistant Prostate Cancer. Clin Cancer

Res. 2015;21:2315-24.

Lallous N, Volik SV, Awrey S, Leblanc E, Tse R, Murillo J, et al. Functional

analysis of androgen receptor mutations that confer anti-androgen resistance

identified in circulating cell-free DNA from prostate cancer patients. Genome

Biol. 2016;17:10.

Salvi S, Casadio V, Conteduca V, Burgio SL, Menna C, Bianchi E, et al.

Circulating cell-free AR and CYP17A1 copy number variations may associate

with outcome of metastatic castration-resistant prostate cancer patients

treated with abiraterone. Br J Cancer. 2015;112:1717-24.

Carreira S, Romanel A, Goodall J, Grist E, Ferraldeschi R, Miranda S, et al.

Tumor clone dynamics in lethal prostate cancer. Sci Transl Med. 2014;6:

ra125.

Conteduca V,Wetterskog D, Sharabiani MTA, Grande E, Fernandez-Perez MP,

Jayaram A, et al. Androgen receptor gene status in plasma DNA associates

with worse outcome on enzalutamide or abiraterone for castration-resistant

prostate cancer: a multi-institution correlative biomarker study. Ann Oncol.

;28:1508-16.

Mahon KL, Qu W, Devaney J, Paul C, Castillo L, Wykes RJ, et al. Methylated

Glutathione S-transferase 1 (mGSTP1) is a potential plasma free DNA

epigenetic marker of prognosis and response to chemotherapy in castrateresistant

prostate cancer. Br J Cancer. 2014;111:1802-9.

Wroclawski ML, Serpa-Neto A, Fonseca FL, Castro-Neves-Neto O, Pompeo

AS, Machado MT, et al. Cell-free plasma DNA as biochemical biomarker for the

diagnosis and follow-up of prostate cancer patients. Tumour Biol. 2013;34:

-7.

Wyatt AW, Annala M, Aggarwal R, Beja K, Feng F, Youngren J, et al.

Concordance of Circulating Tumor DNA and Matched Metastatic Tissue

Biopsy in Prostate Cancer. J Natl Cancer Inst. 2017;109. doi: 10.1093/jnci/

djx118.

Mateo J, Carreira S, Sandhu S, Miranda S, Mossop H, Perez-Lopez R, et al.

DNA-Repair Defects and Olaparib in Metastatic Prostate Cancer. N Engl J

Med. 2015;373:1697-708.

Pritchard CC, Mateo J, Walsh MF, De Sarkar N, Abida W, Beltran H, et al.

Inherited DNA-Repair Gene Mutations in Men with Metastatic Prostate Cancer.

NEngl J Med. 2016;375:443-53.

Xia S, Kohli M, Du M, Dittmar RL, Lee A, Nandy D, et al. Plasma genetic and

genomic abnormalities predict treatment response and clinical outcome in

advanced prostate cancer. Oncotarget. 2015;6:16411-21.

Sonpavde G, Agarwal N, Pond GR, Nagy RJ, Nussenzveig RH, Hahn AW, et al.

Circulating tumor DNA alterations in patients with metastatic castrationresistant

prostate cancer. Cancer. 2019;125:1459-69. doi: 10.1002/cncr.

Robinson D, Van Allen EM,Wu YM, Schultz N, Lonigro RJ, Mosquera JM, et al.

Integrative clinical genomics of advanced prostate cancer. Cell.

;161:1215-28.

Kumar A, Coleman I, Morrissey C, Zhang X, True LD, Gulati R, et al. Substantial

interindividual and limited intraindividual genomic diversity among tumors from

men with metastatic prostate cancer. Nat Med. 2016;22:369-78.

Annala M, Struss WJ, Warner EW, Beja K, Vandekerkhove G, Wong A, et al.

Treatment Outcomes and Tumor Loss of Heterozygosity in Germline DNA

Repair-deficient Prostate Cancer. Eur Urol. 2017;72:34-42.

Fong PC, Boss DS, Yap TA, Tutt A,Wu P, Mergui-Roelvink M, et al. Inhibition of

poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N Engl J

Med. 2009;361:123-34.

Goodall J, Mateo J, Yuan W, Mossop H, Porta N, Miranda S, et al. Circulating

Cell-Free DNA to Guide Prostate Cancer Treatment with PARP Inhibition.

Cancer Discov. 2017;7:1006-17.

Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4

encodes small RNAs with antisense complementarity to lin-14. Cell. 1993;75:

-54.

Kozomara A, Birgaoanu M, Griffiths-Jones S. miRBase: from microRNA

sequences to function. Nucleic Acids Res. 2019;47:D155-D62.

Ambros V. The functions of animal microRNAs. Nature. 2004;431:350-5.

Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A, Petrocca F, et al. A microRNA

expression signature of human solid tumors defines cancer gene targets. Proc

Natl Acad SciUS A. 2006;103:2257-61.

Zidan HE, Abdul-Maksoud RS, Elsayed WSH, Desoky EAM. Diagnostic and

prognostic value of serum miR-15a and miR-16-1 expression among egyptian

patients with prostate cancer. IUBMB Life. 2018;70:437-44.

Musumeci M, Coppola V, Addario A, Patrizii M, Maugeri-Sacca M, Memeo L, et

al. Control of tumor and microenvironment cross-talk by miR-15a and miR-16

in prostate cancer. Oncogene. 2011;30:4231-42.

Bonci D, De Maria R. miR-15/miR-16 loss, miR-21 upregulation, or deregulation

of their target genes predicts poor prognosis in prostate cancer patients.

Mol Cell Oncol. 2016;3:e1109744.

Jin W, Chen F, Wang K, Song Y, Fei X, Wu B. miR-15a/miR-16 cluster inhibits

invasion of prostate cancer cells by suppressing TGF-beta signaling pathway.

Biomed Pharmacother. 2018;104:637-44.

Hasegawa T, Glavich GJ, Pahuski M, Short A, Semmes OJ, Yang L, et al.

Characterization and Evidence of the miR-888 Cluster as a Novel Cancer

Network in Prostate. Mol Cancer Res. 2018;16:669-81.

Yaman Agaoglu F, Kovancilar M, Dizdar Y, Darendeliler E, Holdenrieder S, Dalay

N, et al. Investigation of miR-21, miR-141, and miR-221 in blood circulation of

patients with prostate cancer. Tumour Biol. 2011;32:583-8.

Shen J, Hruby GW, McKiernan JM, Gurvich I, Lipsky MJ, Benson MC, et al.

Dysregulation of circulating microRNAs and prediction of aggressive prostate

cancer. Prostate. 2012;72:1469-77.

Zhang HL, Yang LF, Zhu Y, Yao XD, Zhang SL, Dai B, et al. Serum miRNA-21:

elevated levels in patients with metastatic hormone-refractory prostate cancer

and potential predictive factor for the efficacy of docetaxel-based chemotherapy.

Prostate. 2011;71:326-31.

Lin HM, Castillo L, Mahon KL, Chiam K, Lee BY, Nguyen Q, et al. Circulating

microRNAs are associated with docetaxel chemotherapy outcome in castration-

resistant prostate cancer. Br J Cancer. 2014;110:2462-71.

Hoey C, Liu SK. Circulating blood miRNAs for prostate cancer risk stratification:

miRroring the underlying tumor biology with liquid biopsies. Res Rep

Urol. 2019;11:29-42.

Kelly BD, Miller N, Sweeney KJ, Durkan GC, Rogers E, Walsh K, et al. A

Circulating MicroRNA Signature as a Biomarker for Prostate Cancer in a High

Risk Group. J Clin Med. 2015;4:1369-79.

Brase JC, Johannes M, Schlomm T, Falth M, Haese A, Steuber T, et al.

Circulating miRNAs are correlated with tumor progression in prostate cancer.

Int J Cancer. 2011;128:608-16.

Porzycki P, Ciszkowicz E, Semik M, Tyrka M. Combination of three miRNA

(miR-141, miR-21, and miR-375) as potential diagnostic tool for prostate

cancer recognition. Int Urol Nephrol. 2018;50:1619-26.

Moltzahn F, Olshen AB, Baehner L, Peek A, Fong L, Stoppler H, et al. Microfluidic-

based multiplex qRT-PCR identifies diagnostic and prognostic micro-

RNA signatures in the sera of prostate cancer patients. Cancer Res. 2011;

:550-60.

Alhasan AH, Scott AW,Wu JJ, Feng G, Meeks JJ, Thaxton CS, et al. Circulating

microRNA signature for the diagnosis of very high-risk prostate cancer. Proc

Natl Acad SciUS A. 2016;113:10655-60.

Sharova E, Grassi A, Marcer A, Ruggero K, Pinto F, Bassi P, et al. A circulating

miRNA assay as a first-line test for prostate cancer screening. Br J Cancer.

;114:1362-6.

Zhang HL, Qin XJ, Cao DL, Zhu Y, Yao XD, Zhang SL, et al. An elevated serum

miR-141 level in patients with bone-metastatic prostate cancer is correlated

with more bone lesions. Asian J Androl. 2013;15:231-5.

Nguyen HC, Xie W, Yang M, Hsieh CL, Drouin S, Lee GS, et al. Expression

differences of circulating microRNAs in metastatic castration resistant prostate

cancer and low-risk, localized prostate cancer. Prostate. 2013;73:346-54.

Liu RSC, Olkhov-Mitsel E, Jeyapala R, Zhao F, Commisso K, Klotz L, et al.

Assessment of Serum microRNA Biomarkers to Predict Reclassification

of Prostate Cancer in Patients on Active Surveillance. J Urol. 2018;199:1475-

-81.

Larne O, Martens-Uzunova E, Hagman Z, Edsjo A, Lippolis G, den Berg MS, et

al. miQ—a novel microRNA based diagnostic and prognostic tool for prostate

cancer. Int J Cancer. 2013;132:2867-75.

Minciacchi VR, Zijlstra A, Rubin MA, Di Vizio D. Extracellular vesicles for liquid

biopsy in prostate cancer: where are we and where are we headed? Prostate

Cancer Prostatic Dis. 2017;20:251-8.

Shah R, Patel T, Freedman JE. Circulating Extracellular Vesicles in Human

Disease.NEngl J Med. 2018;379:958-66.

Pan B-T, Johnstone RM. Fate of the transferrin receptor during maturation of

sheep reticulocytes in vitro: Selective externalization of the receptor. Cell.

;33(3):967-78.

O’Driscoll L. Expanding on exosomes and ectosomes in cancer. N Engl J Med.

;372:2359-62.

Giulietti M, Santoni M, Cimadamore A, Carrozza F, Piva F, Cheng L, et al.

Exploring Small Extracellular Vesicles for Precision Medicine in Prostate

Cancer. Front Oncol. 2018;8:221.

Sanchez CA, Andahur EI, Valenzuela R, Castellon EA, Fulla JA, Ramos CG, et

al. Exosomes from bulk and stem cells from human prostate cancer have a

differential microRNA content that contributes cooperatively over local and

pre-metastatic niche. Oncotarget. 2016;7:3993-4008.

Huang X, Yuan T, Liang M, Du M, Xia S, Dittmar R, et al. Exosomal miR-1290

and miR-375 as prognostic markers in castration-resistant prostate cancer.

Eur Urol. 2015;67:33-41.

Bhagirath D, Yang TL, Bucay N, Sekhon K, Majid S, Shahryari V, et al.

microRNA-1246 Is an Exosomal Biomarker for Aggressive Prostate Cancer.

Cancer Res. 2018;78:1833-44.

Kharaziha P, Chioureas D, Rutishauser D, Baltatzis G, Lennartsson L, Fonseca

P, et al. Molecular profiling of prostate cancer derived exosomes may reveal a

predictive signature for response to docetaxel.Oncotarget. 2015;6:21740-54.

Published

2020-07-17