Circulating Tumor DNA in Genitourinary Cancers: Detection, Prognostics, and Therapeutic Implications
Abstract
:Simple Summary
Abstract
1. Introduction
2. Detection Targets and Methods
2.1. Digital PCR and BEAMing
2.2. Ability to Detect Novel Genomic Alterations: Next-Generation Sequencing Methods
2.3. Detection of Epigenetic Modifications: Targeting Histone Modification and Regulatory Elements
2.4. Non-Invasive Detection of Evolving Genetic Profiles
3. Clinical Significance
3.1. Prostate Cancer
3.1.1. Prostate Cancer Detection
3.1.2. Prostate Cancer Prognostics
3.1.3. Prostate Cancer Therapeutic Implications
Androgen Receptor Alterations
DNA Repair Defects and PARP Inhibitor Response
PTEN-PI3K-AKT Pathway Detection and Ipatasertib Response
Microsatellite Instability and Immune Checkpoint Inhibitor Response
3.1.4. Prostate Cancer Resistance, Relapse, and Residual Disease
3.2. Bladder Cancer
3.2.1. Bladder Cancer Detection
3.2.2. Bladder Cancer Prognostics
3.2.3. Bladder Cancer Therapeutic Implications
Immune-Modulating Therapies
FGFR Mutations
DNA Damage and Nucleotide Excision Repair Alterations and Platinum Sensitivity
3.2.4. Bladder Cancer Resistance, Recurrence, and Residual Disease Monitoring
3.3. Renal Cell Carcinoma
3.3.1. Renal Cell Carcinoma Detection
3.3.2. Renal Cell Carcinoma Prognostics
3.3.3. Renal Cell Carcinoma Therapeutic Implications
3.3.4. Renal Cell Carcinoma Resistance, Relapse, and Residual Disease Monitoring
3.4. Penile, Adrenal, and Testicular Cancer
3.4.1. Penile, Adrenal, and Testicular Cancer Detection
3.4.2. Penile, Adrenal, and Testicular Cancer Prognostics
4. Limitations and Future Directions
5. Conclusions
Author Contributions
Funding
Conflicts of Interest
Abbreviations
References
- Wan, J.C.M.; Massie, C.; Garcia-Corbacho, J.; Mouliere, F.; Brenton, J.D.; Caldas, C.; Pacey, S.; Baird, R.; Rosenfeld, N. Liquid biopsies come of age: Towards implementation of circulating tumour DNA. Nat. Rev. Cancer 2017, 17, 223–238. [Google Scholar] [CrossRef] [PubMed]
- Watanabe, K.; Nakamura, Y.; Low, S.-K. Clinical implementation and current advancement of blood liquid biopsy in cancer. J. Hum. Genet. 2021, 66, 909–926. [Google Scholar] [CrossRef] [PubMed]
- Bettegowda, C.; Sausen, M.; Leary, R.J.; Kinde, I.; Wang, Y.; Agrawal, N.; Bartlett, B.R.; Wang, H.; Luber, B.; Alani, R.M.; et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci. Transl. Med. 2014, 6, 224ra24. [Google Scholar] [CrossRef] [PubMed]
- Rose, K.M.; Huelster, H.L.; Meeks, J.J.; Faltas, B.M.; Sonpavde, G.P.; Lerner, S.P.; Ross, J.S.; Spiess, P.E.; Grass, G.D.; Jain, R.K.; et al. Circulating and urinary tumour DNA in urothelial carcinoma—Upper tract, lower tract and metastatic disease. Nat. Rev. Urol. 2023, 20, 406–419. [Google Scholar] [CrossRef] [PubMed]
- Trujillo, B.; Wu, A.; Wetterskog, D.; Attard, G. Blood-based liquid biopsies for prostate cancer: Clinical opportunities and challenges. Br. J. Cancer 2022, 127, 1394–1402. [Google Scholar] [CrossRef] [PubMed]
- Tivey, A.; Church, M.; Rothwell, D.; Dive, C.; Cook, N. Circulating tumour DNA—Looking beyond the blood. Nat. Rev. Clin. Oncol. 2022, 19, 600–612. [Google Scholar] [CrossRef] [PubMed]
- Odegaard, J.I.; Vincent, J.J.; Mortimer, S.; Vowles, J.V.; Ulrich, B.C.; Banks, K.C.; Fairclough, S.R.; Zill, O.A.; Sikora, M.; Mokhtari, R.; et al. Validation of a Plasma-Based Comprehensive Cancer Genotyping Assay Utilizing Orthogonal Tissue- and Plasma-Based Methodologies. Clin. Cancer Res. 2018, 24, 3539–3549. [Google Scholar] [CrossRef] [PubMed]
- García-Foncillas, J.; Alba, E.; Aranda, E.; Díaz-Rubio, E.; López-López, R.; Tabernero, J.; Vivancos, A. Incorporating BEAMing technology as a liquid biopsy into clinical practice for the management of colorectal cancer patients: An expert taskforce review. Ann. Oncol. 2017, 28, 2943–2949. [Google Scholar] [CrossRef]
- Nikanjam, M.; Kato, S.; Kurzrock, R. Liquid biopsy: Current technology and clinical applications. J. Hematol. Oncol. 2022, 15, 131. [Google Scholar] [CrossRef]
- Phallen, J.; Sausen, M.; Adleff, V.; Leal, A.; Hruban, C.; White, J.; Anagnostou, V.; Fiksel, J.; Cristiano, S.; Papp, E.; et al. Direct detection of early-stage cancers using circulating tumor DNA. Sci. Transl. Med. 2017, 9, eaan2415. [Google Scholar] [CrossRef]
- Chen, M.; Zhao, H. Next-generation sequencing in liquid biopsy: Cancer screening and early detection. Hum. Genom. 2019, 13, 34. [Google Scholar] [CrossRef] [PubMed]
- Ewalt, M.D.; West, H.; Aisner, D.L. Next Generation Sequencing—Testing Multiple Genetic Markers at Once. JAMA Oncol. 2019, 5, 1076. [Google Scholar] [CrossRef] [PubMed]
- García-Pardo, M.; Czarnecka-Kujawa, K.; Law, J.H.; Salvarrey, A.M.; Fernandes, R.; Fan, Z.J.; Waddell, T.K.; Yasufuku, K.; Liu, G.; Donahoe, L.L.; et al. Association of Circulating Tumor DNA Testing before Tissue Diagnosis with Time to Treatment among Patients with Suspected Advanced Lung Cancer: The ACCELERATE Nonrandomized Clinical Trial. JAMA Netw. Open 2023, 6, e2325332. [Google Scholar] [CrossRef] [PubMed]
- Baca, S.C.; Seo, J.-H.; Davidsohn, M.P.; Fortunato, B.; Semaan, K.; Sotudian, S.; Lakshminarayanan, G.; Diossy, M.; Qiu, X.; El Zarif, T.; et al. Liquid biopsy epigenomic profiling for cancer subtyping. Nat. Med. 2023, 29, 2737–2741. [Google Scholar] [CrossRef] [PubMed]
- Shen, S.Y.; Singhania, R.; Fehringer, G.; Chakravarthy, A.; Roehrl, M.H.A.; Chadwick, D.; Zuzarte, P.C.; Borgida, A.; Wang, T.T.; Li, T.; et al. Sensitive tumour detection and classification using plasma cell-free DNA methylomes. Nature 2018, 563, 579–583. [Google Scholar] [CrossRef] [PubMed]
- Liu, M.C.; Oxnard, G.R.; Klein, E.A.; Swanton, C.; Seiden, M.V. Sensitive and specific multi-cancer detection and localization using methylation signatures in cell-free DNA. Ann. Oncol. 2020, 31, 745–759. [Google Scholar] [CrossRef]
- Flores, B.C.T.; Correia, M.P.; Rodríguez, J.G.; Henrique, R.; Jerónimo, C. Bridging the Gaps between Circulating Tumor Cells and DNA Methylation in Prostate Cancer. Cancers 2021, 13, 4209. [Google Scholar] [CrossRef] [PubMed]
- Nassar, F.J.; Msheik, Z.S.; Nasr, R.R.; Temraz, S.N. Methylated circulating tumor DNA as a biomarker for colorectal cancer diagnosis, prognosis, and prediction. Clin. Epigenet. 2021, 13, 111. [Google Scholar] [CrossRef] [PubMed]
- Guo, S.; Diep, D.; Plongthongkum, N.; Fung, H.L.; Zhang, K.; Zhang, K. Identification of methylation haplotype blocks aids in deconvolution of heterogeneous tissue samples and tumor tissue-of-origin mapping from plasma DNA. Nat. Genet. 2017, 49, 635–642. [Google Scholar] [CrossRef] [PubMed]
- Hao, X.; Luo, H.; Krawczyk, M.; Wei, W.; Wang, W.; Wang, J.; Flagg, K.; Hou, J.; Zhang, H.; Yi, S.; et al. DNA methylation markers for diagnosis and prognosis of common cancers. Proc. Natl. Acad. Sci. USA 2017, 114, 7414–7419. [Google Scholar] [CrossRef]
- Lorente, D.; Omlin, A.; Zafeiriou, Z.; Nava-Rodrigues, D.; Pérez-López, R.; Pezaro, C.; Mehra, N.; Sheridan, E.; Figueiredo, I.; Riisnaes, R.; et al. Castration-Resistant Prostate Cancer Tissue Acquisition From Bone Metastases for Molecular Analyses. Clin. Genitourin. Cancer 2016, 14, 485–493. [Google Scholar] [CrossRef] [PubMed]
- Gerlinger, M.; Rowan, A.J.; Horswell, S.; Larkin, J.; Endesfelder, D.; Gronroos, E.; Martinez, P.; Matthews, N.; Stewart, A.; Tarpey, P.; et al. Intratumor Heterogeneity and Branched Evolution Revealed by Multiregion Sequencing. N. Engl. J. Med. 2012, 366, 883–892. [Google Scholar] [CrossRef] [PubMed]
- Bardelli, A.; Pantel, K. Liquid Biopsies, What We Do Not Know (Yet). Cancer Cell 2017, 31, 172–179. [Google Scholar] [CrossRef] [PubMed]
- Clinton, T.N.; Chen, Z.; Wise, H.; Lenis, A.T.; Chavan, S.; Donoghue, M.T.A.; Almassi, N.; Chu, C.E.; Dason, S.; Rao, P.; et al. Genomic heterogeneity as a barrier to precision oncology in urothelial cancer. Cell Rep. 2022, 41, 111859. [Google Scholar] [CrossRef] [PubMed]
- Herberts, C.; Annala, M.; Sipola, J.; Ng, S.W.S.; Chen, X.E.; Nurminen, A.; Korhonen, O.V.; Munzur, A.D.; Beja, K.; Schönlau, E.; et al. Deep whole-genome ctDNA chronology of treatment-resistant prostate cancer. Nature 2022, 608, 199–208. [Google Scholar] [CrossRef] [PubMed]
- Wyatt, A.W.; Annala, M.; Aggarwal, R.; Beja, K.; Feng, F.; Youngren, J.; Foye, A.; Lloyd, P.; Nykter, M.; Beer, T.M.; et al. Concordance of Circulating Tumor DNA and Matched Metastatic Tissue Biopsy in Prostate Cancer. J. Natl. Cancer Inst. 2017, 109, djx118. [Google Scholar] [CrossRef] [PubMed]
- Wolf, A.M.; Wender, R.C.; Etzioni, R.B.; Thompson, I.M.; D’Amico, A.V.; Volk, R.J.; Brooks, D.D.; Dash, C.; Guessous, I.; Andrews, K.; et al. American Cancer Society guideline for the early detection of prostate cancer: Update 2010. CA Cancer J Clin 2010, 60, 70–98. [Google Scholar] [CrossRef]
- Kilpeläinen, T.P.; Tammela, T.L.; Roobol, M.; Hugosson, J.; Ciatto, S.; Nelen, V.; Moss, S.; Määttänen, L.; Auvinen, A. False-positive screening results in the European randomized study of screening for prostate cancer. Eur. J. Cancer 2011, 47, 2698–2705. [Google Scholar] [CrossRef] [PubMed]
- Loeb, S.; Vellekoop, A.; Ahmed, H.U.; Catto, J.; Emberton, M.; Nam, R.; Rosario, D.J.; Scattoni, V.; Lotan, Y. Systematic review of complications of prostate biopsy. Eur. Urol. 2013, 64, 876–892. [Google Scholar] [CrossRef] [PubMed]
- Ahmed, H.U.; El-Shater Bosaily, A.; Brown, L.C.; Gabe, R.; Kaplan, R.; Parmar, M.K.; Collaco-Moraes, Y.; Ward, K.; Hindley, R.G.; Freeman, A.; et al. Diagnostic accuracy of multi-parametric MRI and TRUS biopsy in prostate cancer (PROMIS): A paired validating confirmatory study. Lancet 2017, 389, 815–822. [Google Scholar] [CrossRef]
- Aravanis, A.M.; Lee, M.; Klausner, R.D. Next-Generation Sequencing of Circulating Tumor DNA for Early Cancer Detection. Cell 2017, 168, 571–574. [Google Scholar] [CrossRef] [PubMed]
- Hennigan, S.T.; Trostel, S.Y.; Terrigino, N.T.; Voznesensky, O.S.; Schaefer, R.J.; Whitlock, N.C.; Wilkinson, S.; Carrabba, N.V.; Atway, R.; Shema, S.; et al. Low Abundance of Circulating Tumor DNA in Localized Prostate Cancer. JCO Precis. Oncol. 2019, 3, 1–13. [Google Scholar] [CrossRef] [PubMed]
- Berman, B.P.; Weisenberger, D.J.; Aman, J.F.; Hinoue, T.; Ramjan, Z.; Liu, Y.; Noushmehr, H.; Lange, C.P.E.; van Dijk, C.M.; Tollenaar, R.A.E.M.; et al. Regions of focal DNA hypermethylation and long-range hypomethylation in colorectal cancer coincide with nuclear lamina–associated domains. Nat. Genet. 2012, 44, 40–46. [Google Scholar] [CrossRef] [PubMed]
- Guo, H.; Vuille, J.A.; Wittner, B.S.; Lachtara, E.M.; Hou, Y.; Lin, M.; Zhao, T.; Raman, A.T.; Russell, H.C.; Reeves, B.A.; et al. DNA hypomethylation silences anti-tumor immune genes in early prostate cancer and CTCs. Cell 2023, 186, 2765–2782.e28. [Google Scholar] [CrossRef] [PubMed]
- Klein, E.A.; Richards, D.; Cohn, A.; Tummala, M.; Lapham, R.; Cosgrove, D.; Chung, G.; Clement, J.; Gao, J.; Hunkapiller, N.; et al. Clinical validation of a targeted methylation-based multi-cancer early detection test using an independent validation set. Ann. Oncol. 2021, 32, 1167–1177. [Google Scholar] [CrossRef] [PubMed]
- Hubbell, E.; Clarke, C.A.; Aravanis, A.M.; Berg, C.D. Modeled Reductions in Late-stage Cancer with a Multi-Cancer Early Detection Test. Cancer Epidemiol. Biomark. Prev. 2021, 30, 460–468. [Google Scholar] [CrossRef] [PubMed]
- Ellinger, J.; Haan, K.; Heukamp, L.C.; Kahl, P.; Büttner, R.; Müller, S.C.; von Ruecker, A.; Bastian, P.J. CpG island hypermethylation in cell-free serum DNA identifies patients with localized prostate cancer. Prostate 2008, 68, 42–49. [Google Scholar] [CrossRef] [PubMed]
- Brikun, I.; Nusskern, D.; Decatus, A.; Harvey, E.; Li, L.; Freije, D. A panel of DNA methylation markers for the detection of prostate cancer from FV and DRE urine DNA. Clin. Epigenet. 2018, 10, 91. [Google Scholar] [CrossRef] [PubMed]
- Haldrup, C.; Pedersen, A.L.; Øgaard, N.; Strand, S.H.; Høyer, S.; Borre, M.; Ørntoft, T.F.; Sørensen, K.D. Biomarker potential of ST6GALNAC3 and ZNF660 promoter hypermethylation in prostate cancer tissue and liquid biopsies. Mol. Oncol. 2018, 12, 545–560. [Google Scholar] [CrossRef] [PubMed]
- Constâncio, V.; Nunes, S.P.; Moreira-Barbosa, C.; Freitas, R.; Oliveira, J.; Pousa, I.; Oliveira, J.; Soares, M.; Dias, C.G.; Dias, T.; et al. Early detection of the major male cancer types in blood-based liquid biopsies using a DNA methylation panel. Clin. Epigenet. 2019, 11, 175. [Google Scholar] [CrossRef]
- Reis, I.M.; Ramachandran, K.; Speer, C.; Gordian, E.; Singal, R. Serum GADD45a methylation is a useful biomarker to distinguish benign vs malignant prostate disease. Br. J. Cancer 2015, 113, 460–468. [Google Scholar] [CrossRef] [PubMed]
- O’Reilly, E.; Tuzova, A.V.; Walsh, A.L.; Russell, N.M.; O’Brien, O.; Kelly, S.; Dhomhnallain, O.N.; DeBarra, L.; Dale, C.M.; Brugman, R.; et al. epiCaPture: A Urine DNA Methylation Test for Early Detection of Aggressive Prostate Cancer. JCO Precis. Oncol. 2019, 3, 1–18. [Google Scholar] [CrossRef]
- Henrique, R.; Jerónimo, C. Molecular detection of prostate cancer: A role for GSTP1 hypermethylation. Eur. Urol. 2004, 46, 660–669; discussion 669. [Google Scholar] [CrossRef]
- Wu, T.; Giovannucci, E.; Welge, J.; Mallick, P.; Tang, W.Y.; Ho, S.M. Measurement of GSTP1 promoter methylation in body fluids may complement PSA screening: A meta-analysis. Br. J. Cancer 2011, 105, 65–73. [Google Scholar] [CrossRef]
- Helzer, K.T.; Sharifi, M.N.; Sperger, J.M.; Shi, Y.; Annala, M.; Bootsma, M.L.; Reese, S.R.; Taylor, A.; Kaufmann, K.R.; Krause, H.K.; et al. Fragmentomic analysis of circulating tumor DNA-targeted cancer panels. Ann. Oncol. 2023, 34, 813–825. [Google Scholar] [CrossRef] [PubMed]
- Spratt, D.E.; Yousefi, K.; Deheshi, S.; Ross, A.E.; Den, R.B.; Schaeffer, E.M.; Trock, B.J.; Zhang, J.; Glass, A.G.; Dicker, A.P.; et al. Individual Patient-Level Meta-Analysis of the Performance of the Decipher Genomic Classifier in High-Risk Men after Prostatectomy to Predict Development of Metastatic Disease. J. Clin. Oncol. 2017, 35, 1991–1998. [Google Scholar] [CrossRef]
- Spratt, D.E.; Huang, H.-C.; Michalski, J.M.; Davicioni, E.; Berlin, A.; Simko, J.; Efstathiou, J.A.; Tran, P.T.; Thompson, D.; Parliament, M.; et al. Validation of the performance of the Decipher biopsy genomic classifier in intermediate-risk prostate cancer on the phase III randomized trial NRG Oncology/RTOG 0126. J. Clin. Oncol. 2022, 40, 269. [Google Scholar] [CrossRef]
- Robinson, D.; Van Allen, E.M.; Wu, Y.M.; Schultz, N.; Lonigro, R.J.; Mosquera, J.M.; Montgomery, B.; Taplin, M.E.; Pritchard, C.C.; Attard, G.; et al. Integrative clinical genomics of advanced prostate cancer. Cell 2015, 161, 1215–1228. [Google Scholar] [CrossRef] [PubMed]
- van Dessel, L.F.; van Riet, J.; Smits, M.; Zhu, Y.; Hamberg, P.; van der Heijden, M.S.; Bergman, A.M.; van Oort, I.M.; de Wit, R.; Voest, E.E.; et al. The genomic landscape of metastatic castration-resistant prostate cancers reveals multiple distinct genotypes with potential clinical impact. Nat. Commun. 2019, 10, 5251. [Google Scholar] [CrossRef]
- Sonpavde, G.; Agarwal, N.; Pond, G.R.; Nagy, R.J.; Nussenzveig, R.H.; Hahn, A.W.; Sartor, O.; Gourdin, T.S.; Nandagopal, L.; Ledet, E.M.; et al. Circulating tumor DNA alterations in patients with metastatic castration-resistant prostate cancer. Cancer 2019, 125, 1459–1469. [Google Scholar] [CrossRef]
- Tukachinsky, H.; Madison, R.W.; Chung, J.H.; Gjoerup, O.V.; Severson, E.A.; Dennis, L.; Fendler, B.J.; Morley, S.; Zhong, L.; Graf, R.P.; et al. Genomic Analysis of Circulating Tumor DNA in 3,334 Patients with Advanced Prostate Cancer Identifies Targetable BRCA Alterations and AR Resistance Mechanisms. Clin. Cancer Res. 2021, 27, 3094–3105. [Google Scholar] [CrossRef]
- Annala, M.; Vandekerkhove, G.; Khalaf, D.; Taavitsainen, S.; Beja, K.; Warner, E.W.; Sunderland, K.; Kollmannsberger, C.; Eigl, B.J.; Finch, D.; et al. Circulating Tumor DNA Genomics Correlate with Resistance to Abiraterone and Enzalutamide in Prostate Cancer. Cancer Discov. 2018, 8, 444–457. [Google Scholar] [CrossRef] [PubMed]
- Annala, M.; Taavitsainen, S.; Khalaf, D.J.; Vandekerkhove, G.; Beja, K.; Sipola, J.; Warner, E.W.; Herberts, C.; Wong, A.; Fu, S.; et al. Evolution of Castration-Resistant Prostate Cancer in ctDNA during Sequential Androgen Receptor Pathway Inhibition. Clin. Cancer Res. 2021, 27, 4610–4623. [Google Scholar] [CrossRef]
- Romanel, A.; Gasi Tandefelt, D.; Conteduca, V.; Jayaram, A.; Casiraghi, N.; Wetterskog, D.; Salvi, S.; Amadori, D.; Zafeiriou, Z.; Rescigno, P.; et al. Plasma AR and abiraterone-resistant prostate cancer. Sci. Transl. Med. 2015, 7, 312re310. [Google Scholar] [CrossRef]
- Chi, K.N.; Annala, M.; Sunderland, K.; Khalaf, D.; Finch, D.; Oja, C.D.; Vergidis, J.; Zulfiqar, M.; Beja, K.; Vandekerkhove, G.; et al. A randomized phase II cross-over study of abiraterone + prednisone (ABI) vs enzalutamide (ENZ) for patients (pts) with metastatic, castration-resistant prostate cancer (mCRPC). J. Clin. Oncol. 2017, 35, 5002. [Google Scholar] [CrossRef]
- Rebello, R.J.; Oing, C.; Knudsen, K.E.; Loeb, S.; Johnson, D.C.; Reiter, R.E.; Gillessen, S.; Van der Kwast, T.; Bristow, R.G. Prostate cancer. Nat. Rev. Dis. Primers 2021, 7, 9. [Google Scholar] [CrossRef]
- Sato, Y. Clinical utility of liquid biopsy-based companion diagnostics in the non-small-cell lung cancer treatment. Explor. Target. Antitumor Ther. 2022, 3, 630–642. [Google Scholar] [CrossRef]
- Antonarakis, E.S.; Lu, C.; Wang, H.; Luber, B.; Nakazawa, M.; Roeser, J.C.; Chen, Y.; Mohammad, T.A.; Chen, Y.; Fedor, H.L.; et al. AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer. N. Engl. J. Med. 2014, 371, 1028–1038. [Google Scholar] [CrossRef]
- Ryan, C.J.; Smith, M.R.; de Bono, J.S.; Molina, A.; Logothetis, C.J.; de Souza, P.; Fizazi, K.; Mainwaring, P.; Piulats, J.M.; Ng, S.; et al. Abiraterone in metastatic prostate cancer without previous chemotherapy. N. Engl. J. Med. 2013, 368, 138–148. [Google Scholar] [CrossRef] [PubMed]
- Scher, H.I.; Fizazi, K.; Saad, F.; Taplin, M.E.; Sternberg, C.N.; Miller, K.; de Wit, R.; Mulders, P.; Chi, K.N.; Shore, N.D.; et al. Increased survival with enzalutamide in prostate cancer after chemotherapy. N. Engl. J. Med. 2012, 367, 1187–1197. [Google Scholar] [CrossRef]
- O’Donnell, A.; Judson, I.; Dowsett, M.; Raynaud, F.; Dearnaley, D.; Mason, M.; Harland, S.; Robbins, A.; Halbert, G.; Nutley, B.; et al. Hormonal impact of the 17alpha-hydroxylase/C(17,20)-lyase inhibitor abiraterone acetate (CB7630) in patients with prostate cancer. Br. J. Cancer 2004, 90, 2317–2325. [Google Scholar] [CrossRef] [PubMed]
- Scher, H.I.; Beer, T.M.; Higano, C.S.; Anand, A.; Taplin, M.E.; Efstathiou, E.; Rathkopf, D.; Shelkey, J.; Yu, E.Y.; Alumkal, J.; et al. Antitumour activity of MDV3100 in castration-resistant prostate cancer: A phase 1-2 study. Lancet 2010, 375, 1437–1446. [Google Scholar] [CrossRef] [PubMed]
- Quigley, D.A.; Dang, H.X.; Zhao, S.G.; Lloyd, P.; Aggarwal, R.; Alumkal, J.J.; Foye, A.; Kothari, V.; Perry, M.D.; Bailey, A.M.; et al. Genomic Hallmarks and Structural Variation in Metastatic Prostate Cancer. Cell 2018, 174, 758–769.e9. [Google Scholar] [CrossRef] [PubMed]
- Lorente, D.; Mateo, J.; Zafeiriou, Z.; Smith, A.D.; Sandhu, S.; Ferraldeschi, R.; de Bono, J.S. Switching and withdrawing hormonal agents for castration-resistant prostate cancer. Nat. Rev. Urol. 2015, 12, 37–47. [Google Scholar] [CrossRef] [PubMed]
- Armstrong, A.J.; Halabi, S.; Luo, J.; Nanus, D.M.; Giannakakou, P.; Szmulewitz, R.Z.; Danila, D.C.; Healy, P.; Anand, M.; Rothwell, C.J.; et al. Prospective Multicenter Validation of Androgen Receptor Splice Variant 7 and Hormone Therapy Resistance in High-Risk Castration-Resistant Prostate Cancer: The PROPHECY Study. J. Clin. Oncol. 2019, 37, 1120–1129. [Google Scholar] [CrossRef] [PubMed]
- Conteduca, V.; Wetterskog, D.; Sharabiani, M.T.A.; Grande, E.; Fernandez-Perez, M.P.; Jayaram, A.; Salvi, S.; Castellano, D.; Romanel, A.; Lolli, C.; 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. 2017, 28, 1508–1516. [Google Scholar] [CrossRef]
- De Laere, B.; van Dam, P.J.; Whitington, T.; Mayrhofer, M.; Diaz, E.H.; Van den Eynden, G.; Vandebroek, J.; Del-Favero, J.; Van Laere, S.; Dirix, L.; 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. [Google Scholar] [CrossRef] [PubMed]
- Swami, U.; Zimmerman, R.M.; Nussenzveig, R.H.; Hernandez, E.J.; Jo, Y.; Sayegh, N.; Wesolowski, S.; Kiedrowski, L.A.; Barata, P.C.; Lemmon, G.H.; et al. Genomic landscape of advanced prostate cancer patients with BRCA1 versus BRCA2 mutations as detected by comprehensive genomic profiling of cell-free DNA. Front. Oncol. 2022, 12, 966534. [Google Scholar] [CrossRef] [PubMed]
- Pritchard, C.C.; Mateo, J.; Walsh, M.F.; De Sarkar, N.; Abida, W.; Beltran, H.; Garofalo, A.; Gulati, R.; Carreira, S.; Eeles, R.; et al. Inherited DNA-Repair Gene Mutations in Men with Metastatic Prostate Cancer. N. Engl. J. Med. 2016, 375, 443–453. [Google Scholar] [CrossRef]
- Abida, W.; Patnaik, A.; Campbell, D.; Shapiro, J.; Bryce, A.H.; McDermott, R.; Sautois, B.; Vogelzang, N.J.; Bambury, R.M.; Voog, E.; et al. Rucaparib in Men with Metastatic Castration-Resistant Prostate Cancer Harboring a BRCA1 or BRCA2 Gene Alteration. J. Clin. Oncol. 2020, 38, 3763–3772. [Google Scholar] [CrossRef]
- Hussain, M.; Mateo, J.; Fizazi, K.; Saad, F.; Shore, N.; Sandhu, S.; Chi, K.N.; Sartor, O.; Agarwal, N.; Olmos, D.; et al. Survival with Olaparib in Metastatic Castration-Resistant Prostate Cancer. N. Engl. J. Med. 2020, 383, 2345–2357. [Google Scholar] [CrossRef] [PubMed]
- 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. Engl. J. Med. 2015, 373, 1697–1708. [Google Scholar] [CrossRef] [PubMed]
- Chi, K.N.; Barnicle, A.; Sibilla, C.; Lai, Z.; Corcoran, C.; Barrett, J.C.; Adelman, C.A.; Qiu, P.; Easter, A.; Dearden, S.; et al. Detection of BRCA1, BRCA2, and ATM Alterations in Matched Tumor Tissue and Circulating Tumor DNA in Patients with Prostate Cancer Screened in PROfound. Clin. Cancer Res. 2023, 29, 81–91. [Google Scholar] [CrossRef] [PubMed]
- Goodall, J.; Mateo, J.; Yuan, W.; Mossop, H.; Porta, N.; Miranda, S.; Perez-Lopez, R.; Dolling, D.; Robinson, D.R.; Sandhu, S.; et al. Circulating Cell-Free DNA to Guide Prostate Cancer Treatment with PARP Inhibition. Cancer Discov. 2017, 7, 1006–1017. [Google Scholar] [CrossRef]
- Quigley, D.; Alumkal, J.J.; Wyatt, A.W.; Kothari, V.; Foye, A.; Lloyd, P.; Aggarwal, R.; Kim, W.; Lu, E.; Schwartzman, J.; et al. Analysis of Circulating Cell-Free DNA Identifies Multiclonal Heterogeneity of BRCA2 Reversion Mutations Associated with Resistance to PARP Inhibitors. Cancer Discov. 2017, 7, 999–1005. [Google Scholar] [CrossRef] [PubMed]
- Anscher, M.S.; Chang, E.; Gao, X.; Gong, Y.; Weinstock, C.; Bloomquist, E.; Adeniyi, O.; Charlab, R.; Zimmerman, S.; Serlemitsos-Day, M.; et al. FDA Approval Summary: Rucaparib for the Treatment of Patients with Deleterious BRCA-Mutated Metastatic Castrate-Resistant Prostate Cancer. Oncologist 2021, 26, 139–146. [Google Scholar] [CrossRef] [PubMed]
- Woodhouse, R.; Li, M.; Hughes, J.; Delfosse, D.; Skoletsky, J.; Ma, P.; Meng, W.; Dewal, N.; Milbury, C.; Clark, T.; et al. Clinical and analytical validation of FoundationOne Liquid CDx, a novel 324-Gene cfDNA-based comprehensive genomic profiling assay for cancers of solid tumor origin. PLoS ONE 2020, 15, e0237802. [Google Scholar] [CrossRef] [PubMed]
- Sweeney, C.; Bracarda, S.; Sternberg, C.N.; Chi, K.N.; Olmos, D.; Sandhu, S.; Massard, C.; Matsubara, N.; Alekseev, B.; Parnis, F.; et al. Ipatasertib plus abiraterone and prednisolone in metastatic castration-resistant prostate cancer (IPATential150): A multicentre, randomised, double-blind, phase 3 trial. Lancet 2021, 398, 131–142. [Google Scholar] [CrossRef] [PubMed]
- Bang, S.; Won, D.; Shin, S.; Cho, K.S.; Park, J.W.; Lee, J.; Choi, Y.D.; Kang, S.; Lee, S.T.; Choi, J.R.; et al. Circulating Tumor DNA Analysis on Metastatic Prostate Cancer with Disease Progression. Cancers 2023, 15, 3998. [Google Scholar] [CrossRef] [PubMed]
- Dong, X.; Zheng, T.; Zhang, M.; Dai, C.; Wang, L.; Wang, L.; Zhang, R.; Long, Y.; Wen, D.; Xie, F.; et al. Circulating Cell-Free DNA-Based Detection of Tumor Suppressor Gene Copy Number Loss and Its Clinical Implication in Metastatic Prostate Cancer. Front. Oncol. 2021, 11, 720727. [Google Scholar] [CrossRef]
- Lanka, S.M.; Zorko, N.A.; Antonarakis, E.S.; Barata, P.C. Metastatic Castration-Resistant Prostate Cancer, Immune Checkpoint Inhibitors, and Beyond. Curr. Oncol. 2023, 30, 4246–4256. [Google Scholar] [CrossRef] [PubMed]
- Marabelle, A.; Le, D.T.; Ascierto, P.A.; Giacomo, A.M.D.; Jesus-Acosta, A.D.; Delord, J.-P.; Geva, R.; Gottfried, M.; Penel, N.; Hansen, A.R.; et al. Efficacy of Pembrolizumab in Patients with Noncolorectal High Microsatellite Instability/Mismatch Repair–Deficient Cancer: Results From the Phase II KEYNOTE-158 Study. J. Clin. Oncol. 2020, 38, 1–10. [Google Scholar] [CrossRef]
- Muzny, D.M.; Bainbridge, M.N.; Chang, K.; Dinh, H.H.; Drummond, J.A.; Fowler, G.; Kovar, C.L.; Lewis, L.R.; Morgan, M.B.; Newsham, I.F.; et al. Comprehensive molecular characterization of human colon and rectal cancer. Nature 2012, 487, 330–337. [Google Scholar] [CrossRef]
- Dudley, J.C.; Lin, M.T.; Le, D.T.; Eshleman, J.R. Microsatellite Instability as a Biomarker for PD-1 Blockade. Clin. Cancer Res. 2016, 22, 813–820. [Google Scholar] [CrossRef] [PubMed]
- Abida, W.; Cheng, M.L.; Armenia, J.; Middha, S.; Autio, K.A.; Vargas, H.A.; Rathkopf, D.; Morris, M.J.; Danila, D.C.; Slovin, S.F.; et al. Analysis of the Prevalence of Microsatellite Instability in Prostate Cancer and Response to Immune Checkpoint Blockade. JAMA Oncol. 2019, 5, 471–478. [Google Scholar] [CrossRef]
- Black, J.R.M.; McGranahan, N. Genetic and non-genetic clonal diversity in cancer evolution. Nat. Rev. Cancer 2021, 21, 379–392. [Google Scholar] [CrossRef] [PubMed]
- Wyatt, A.W.; Azad, A.A.; Volik, S.V.; Annala, M.; Beja, K.; McConeghy, B.; Haegert, A.; Warner, E.W.; Mo, F.; Brahmbhatt, S.; et al. Genomic Alterations in Cell-Free DNA and Enzalutamide Resistance in Castration-Resistant Prostate Cancer. JAMA Oncol. 2016, 2, 1598–1606. [Google Scholar] [CrossRef]
- Lallous, N.; Volik, S.V.; Awrey, S.; Leblanc, E.; Tse, R.; Murillo, J.; Singh, K.; Azad, A.A.; Wyatt, A.W.; LeBihan, S.; 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. [Google Scholar] [CrossRef]
- Tolmeijer, S.H.; Boerrigter, E.; Sumiyoshi, T.; Kwan, E.M.; Ng, S.W.S.; Annala, M.; Donnellan, G.; Herberts, C.; Benoist, G.E.; Hamberg, P.; et al. Early On-treatment Changes in Circulating Tumor DNA Fraction and Response to Enzalutamide or Abiraterone in Metastatic Castration-Resistant Prostate Cancer. Clin. Cancer Res. 2023, 29, 2835–2844. [Google Scholar] [CrossRef]
- Conteduca, V.; Wetterskog, D.; Scarpi, E.; Romanel, A.; Gurioli, G.; Jayaram, A.; Lolli, C.; Tandefelt, D.G.; Schepisi, G.; Casadei, C.; et al. Plasma tumour DNA as an early indicator of treatment response in metastatic castration-resistant prostate cancer. Br. J. Cancer 2020, 123, 982–987. [Google Scholar] [CrossRef]
- Jayaram, A.; Wingate, A.; Wetterskog, D.; Wheeler, G.; Sternberg, C.N.; Jones, R.; Berruti, A.; Lefresne, F.; Lahaye, M.; Thomas, S.; et al. Plasma tumor gene conversions after one cycle abiraterone acetate for metastatic castration-resistant prostate cancer: A biomarker analysis of a multicenter international trial. Ann. Oncol. 2021, 32, 726–735. [Google Scholar] [CrossRef]
- Silva, R.; Moran, B.; Baird, A.M.; O’Rourke, C.J.; Finn, S.P.; McDermott, R.; Watson, W.; Gallagher, W.M.; Brennan, D.J.; Perry, A.S. Longitudinal analysis of individual cfDNA methylome patterns in metastatic prostate cancer. Clin. Epigenet. 2021, 13, 168. [Google Scholar] [CrossRef]
- Tessneer, K.L.; Pasula, S.; Cai, X.; Dong, Y.; Liu, X.; Yu, L.; Hahn, S.; McManus, J.; Chen, Y.; Chang, B.; et al. Endocytic adaptor protein epsin is elevated in prostate cancer and required for cancer progression. ISRN Oncol. 2013, 2013, 420597. [Google Scholar] [CrossRef]
- Mahon, K.L.; Qu, W.; Lin, H.M.; Spielman, C.; Cain, D.; Jacobs, C.; Stockler, M.R.; Higano, C.S.; de Bono, J.S.; Chi, K.N.; et al. Serum Free Methylated Glutathione S-transferase 1 DNA Levels, Survival, and Response to Docetaxel in Metastatic, Castration-resistant Prostate Cancer: Post Hoc Analyses of Data from a Phase 3 Trial. Eur. Urol. 2019, 76, 306–312. [Google Scholar] [CrossRef]
- Birkenkamp-Demtröder, K.; Nordentoft, I.; Christensen, E.; Høyer, S.; Reinert, T.; Vang, S.; Borre, M.; Agerbæk, M.; Jensen, J.B.; Ørntoft, T.F.; et al. Genomic Alterations in Liquid Biopsies from Patients with Bladder Cancer. Eur. Urol. 2016, 70, 75–82. [Google Scholar] [CrossRef]
- Agarwal, N.; Pal, S.K.; Hahn, A.W.; Nussenzveig, R.H.; Pond, G.R.; Gupta, S.V.; Wang, J.; Bilen, M.A.; Naik, G.; Ghatalia, P.; et al. Characterization of metastatic urothelial carcinoma via comprehensive genomic profiling of circulating tumor DNA. Cancer 2018, 124, 2115–2124. [Google Scholar] [CrossRef]
- Jain, M.; Kamalov, D.; Tivtikyan, A.; Balatsky, A.; Samokhodskaya, L.; Okhobotov, D.; Kozlova, P.; Pisarev, E.; Zvereva, M.; Kamalov, A. Urine TERT promoter mutations-based tumor DNA detection in patients with bladder cancer: A pilot study. Mol. Clin. Oncol. 2021, 15, 253. [Google Scholar] [CrossRef]
- Dudley, J.C.; Schroers-Martin, J.; Lazzareschi, D.V.; Shi, W.Y.; Chen, S.B.; Esfahani, M.S.; Trivedi, D.; Chabon, J.J.; Chaudhuri, A.A.; Stehr, H.; et al. Detection and Surveillance of Bladder Cancer Using Urine Tumor DNA. Cancer Discov. 2019, 9, 500–509. [Google Scholar] [CrossRef]
- Xu, Y.; Kim, Y.H.; Jeong, P.; Piao, X.M.; Byun, Y.J.; Kang, H.W.; Kim, W.T.; Lee, J.Y.; Kim, I.Y.; Moon, S.K.; et al. Diagnostic value of combined IQGAP3/BMP4 and IQGAP3/FAM107A expression ratios in urinary cell-free DNA for discriminating bladder cancer from hematuria. Urol. Oncol. 2019, 37, 86–96. [Google Scholar] [CrossRef]
- Cheng, T.H.T.; Jiang, P.; Teoh, J.Y.C.; Heung, M.M.S.; Tam, J.C.W.; Sun, X.; Lee, W.S.; Ni, M.; Chan, R.C.K.; Ng, C.F.; et al. Noninvasive Detection of Bladder Cancer by Shallow-Depth Genome-Wide Bisulfite Sequencing of Urinary Cell-Free DNA for Methylation and Copy Number Profiling. Clin. Chem. 2019, 65, 927–936. [Google Scholar] [CrossRef]
- Xu, Y.; Ma, X.; Ai, X.; Gao, J.; Liang, Y.; Zhang, Q.; Ma, T.; Mao, K.; Zheng, Q.; Wang, S.; et al. A Urine-Based Liquid Biopsy Method for Detection of Upper Tract Urinary Carcinoma. Front. Oncol. 2020, 10, 597486. [Google Scholar] [CrossRef]
- Zeng, S.; Ying, Y.; Xing, N.; Wang, B.; Qian, Z.; Zhou, Z.; Zhang, Z.; Xu, W.; Wang, H.; Dai, L.; et al. Noninvasive Detection of Urothelial Carcinoma by Cost-effective Low-coverage Whole-genome Sequencing from Urine-Exfoliated Cell DNA. Clin. Cancer Res. 2020, 26, 5646–5654. [Google Scholar] [CrossRef]
- Christensen, E.; Birkenkamp-Demtröder, K.; Sethi, H.; Shchegrova, S.; Salari, R.; Nordentoft, I.; Wu, H.-T.; Knudsen, M.; Lamy, P.; Lindskrog, S.V.; et al. Early Detection of Metastatic Relapse and Monitoring of Therapeutic Efficacy by Ultra-Deep Sequencing of Plasma Cell-Free DNA in Patients with Urothelial Bladder Carcinoma. J. Clin. Oncol. 2019, 37, 1547–1557. [Google Scholar] [CrossRef]
- Christensen, E.; Birkenkamp-Demtröder, K.; Nordentoft, I.; Høyer, S.; van der Keur, K.; van Kessel, K.; Zwarthoff, E.; Agerbæk, M.; Ørntoft, T.F.; Jensen, J.B.; et al. Liquid Biopsy Analysis of FGFR3 and PIK3CA Hotspot Mutations for Disease Surveillance in Bladder Cancer. Eur. Urol. 2017, 71, 961–969. [Google Scholar] [CrossRef]
- Shohdy, K.S.; Villamar, D.M.; Cao, Y.; Trieu, J.; Price, K.S.; Nagy, R.; Tagawa, S.T.; Molina, A.M.; Sternberg, C.N.; Nanus, D.M.; et al. Serial ctDNA analysis predicts clinical progression in patients with advanced urothelial carcinoma. Br. J. Cancer 2022, 126, 430–439. [Google Scholar] [CrossRef]
- Grivas, P.; Lalani, A.A.; Pond, G.R.; Nagy, R.J.; Faltas, B.; Agarwal, N.; Gupta, S.V.; Drakaki, A.; Vaishampayan, U.N.; Wang, J.; et al. Circulating Tumor DNA Alterations in Advanced Urothelial Carcinoma and Association with Clinical Outcomes: A Pilot Study. Eur. Urol. Oncol. 2020, 3, 695–699. [Google Scholar] [CrossRef]
- Vandekerkhove, G.; Todenhöfer, T.; Annala, M.; Struss, W.J.; Wong, A.; Beja, K.; Ritch, E.; Brahmbhatt, S.; Volik, S.V.; Hennenlotter, J.; et al. Circulating Tumor DNA Reveals Clinically Actionable Somatic Genome of Metastatic Bladder Cancer. Clin. Cancer Res. 2017, 23, 6487–6497. [Google Scholar] [CrossRef]
- Puntoni, M.; Petrera, M.; Campora, S.; Garrone, E.; Defferrari, C.; Torrisi, R.; Johansson, H.; Bruno, S.; Curotto, A.; DeCensi, A. Prognostic Significance of VEGF after Twenty-Year Follow-up in a Randomized Trial of Fenretinide in Non–Muscle-Invasive Bladder Cancer. Cancer Prev. Res. 2016, 9, 437–444. [Google Scholar] [CrossRef]
- Christensen, E.; Nordentoft, I.; Birkenkamp-Demtröder, K.; Elbæk, S.K.; Lindskrog, S.V.; Taber, A.; Andreasen, T.G.; Strandgaard, T.; Knudsen, M.; Lamy, P.; et al. Cell-Free Urine and Plasma DNA Mutational Analysis Predicts Neoadjuvant Chemotherapy Response and Outcome in Patients with Muscle-Invasive Bladder Cancer. Clin. Cancer Res. 2023, 29, 1582–1591. [Google Scholar] [CrossRef]
- Powles, T.; Assaf, Z.J.; Davarpanah, N.; Banchereau, R.; Szabados, B.E.; Yuen, K.C.; Grivas, P.; Hussain, M.; Oudard, S.; Gschwend, J.E.; et al. ctDNA guiding adjuvant immunotherapy in urothelial carcinoma. Nature 2021, 595, 432–437. [Google Scholar] [CrossRef]
- Robertson, A.G.; Kim, J.; Al-Ahmadie, H.; Bellmunt, J.; Guo, G.; Cherniack, A.D.; Hinoue, T.; Laird, P.W.; Hoadley, K.A.; Akbani, R.; et al. Comprehensive Molecular Characterization of Muscle-Invasive Bladder Cancer. Cell 2017, 171, 540–556.e525. [Google Scholar] [CrossRef]
- Ross, J.S.; Wang, K.; Khaira, D.; Ali, S.M.; Fisher, H.A.; Mian, B.; Nazeer, T.; Elvin, J.A.; Palma, N.; Yelensky, R.; et al. Comprehensive genomic profiling of 295 cases of clinically advanced urothelial carcinoma of the urinary bladder reveals a high frequency of clinically relevant genomic alterations. Cancer 2016, 122, 702–711. [Google Scholar] [CrossRef]
- Vandekerkhove, G.; Lavoie, J.-M.; Annala, M.; Murtha, A.J.; Sundahl, N.; Walz, S.; Sano, T.; Taavitsainen, S.; Ritch, E.; Fazli, L.; et al. Plasma ctDNA is a tumor tissue surrogate and enables clinical-genomic stratification of metastatic bladder cancer. Nat. Commun. 2021, 12, 184. [Google Scholar] [CrossRef]
- Barata, P.C.; Koshkin, V.S.; Funchain, P.; Sohal, D.; Pritchard, A.; Klek, S.; Adamowicz, T.; Gopalakrishnan, D.; Garcia, J.; Rini, B.; et al. Next-generation sequencing (NGS) of cell-free circulating tumor DNA and tumor tissue in patients with advanced urothelial cancer: A pilot assessment of concordance. Ann. Oncol. 2017, 28, 2458–2463. [Google Scholar] [CrossRef]
- Klempner, S.J.; Fabrizio, D.; Bane, S.; Reinhart, M.; Peoples, T.; Ali, S.M.; Sokol, E.S.; Frampton, G.; Schrock, A.B.; Anhorn, R.; et al. Tumor Mutational Burden as a Predictive Biomarker for Response to Immune Checkpoint Inhibitors: A Review of Current Evidence. Oncologist 2020, 25, e147–e159. [Google Scholar] [CrossRef]
- Alexandrov, L.B.; Nik-Zainal, S.; Wedge, D.C.; Aparicio, S.A.J.R.; Behjati, S.; Biankin, A.V.; Bignell, G.R.; Bolli, N.; Borg, A.; Børresen-Dale, A.-L.; et al. Signatures of mutational processes in human cancer. Nature 2013, 500, 415–421. [Google Scholar] [CrossRef]
- Maia, M.C.; Salgia, M.; Pal, S.K. Harnessing cell-free DNA: Plasma circulating tumour DNA for liquid biopsy in genitourinary cancers. Nat. Rev. Urol. 2020, 17, 271–291. [Google Scholar] [CrossRef]
- Rosenberg, J.E.; Hoffman-Censits, J.; Powles, T.; van der Heijden, M.S.; Balar, A.V.; Necchi, A.; Dawson, N.; O’Donnell, P.H.; Balmanoukian, A.; Loriot, Y.; et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: A single-arm, multicentre, phase 2 trial. Lancet 2016, 387, 1909–1920. [Google Scholar] [CrossRef]
- Necchi, A.; Anichini, A.; Raggi, D.; Briganti, A.; Massa, S.; Lucianò, R.; Colecchia, M.; Giannatempo, P.; Mortarini, R.; Bianchi, M.; et al. Pembrolizumab as Neoadjuvant Therapy before Radical Cystectomy in Patients with Muscle-Invasive Urothelial Bladder Carcinoma (PURE-01): An Open-Label, Single-Arm, Phase II Study. J. Clin. Oncol. 2018, 36, 3353–3360. [Google Scholar] [CrossRef]
- Sheng, X.; Chen, H.; Hu, B.; Yao, X.; Liu, Z.; Yao, X.; Guo, H.; Hu, Y.; Ji, Z.; Luo, H.; et al. Safety, Efficacy, and Biomarker Analysis of Toripalimab in Patients with Previously Treated Advanced Urothelial Carcinoma: Results from a Multicenter Phase II Trial POLARIS-03. Clin. Cancer Res. 2022, 28, 489–497. [Google Scholar] [CrossRef]
- Zang, J.; Zhang, R.; Jin, D.; Xie, F.; Shahatiaili, A.; Wu, G.; Zhang, Y.; Zhao, Z.; Du, P.; Jia, S.; et al. Integrated longitudinal circulating tumor DNA profiling predicts immunotherapy response of metastatic urothelial carcinoma in the POLARIS-03 trial. J. Pathol. 2023, 261, 198–209. [Google Scholar] [CrossRef]
- Szabados, B.; Kockx, M.; Assaf, Z.J.; van Dam, P.J.; Rodriguez-Vida, A.; Duran, I.; Crabb, S.J.; Van Der Heijden, M.S.; Pous, A.F.; Gravis, G.; et al. Final Results of Neoadjuvant Atezolizumab in Cisplatin-ineligible Patients with Muscle-invasive Urothelial Cancer of the Bladder. Eur. Urol. 2022, 82, 212–222. [Google Scholar] [CrossRef]
- Raja, R.; Kuziora, M.; Brohawn, P.Z.; Higgs, B.W.; Gupta, A.; Dennis, P.A.; Ranade, K. Early Reduction in ctDNA Predicts Survival in Patients with Lung and Bladder Cancer Treated with Durvalumab. Clin. Cancer Res. 2018, 24, 6212–6222. [Google Scholar] [CrossRef]
- van Dorp, J.; Pipinikas, C.; Suelmann, B.B.M.; Mehra, N.; van Dijk, N.; Marsico, G.; van Montfoort, M.L.; Hackinger, S.; Braaf, L.M.; Amarante, T.; et al. High- or low-dose preoperative ipilimumab plus nivolumab in stage III urothelial cancer: The phase 1B NABUCCO trial. Nat. Med. 2023, 29, 588–592. [Google Scholar] [CrossRef]
- Nassar, A.H.; Umeton, R.; Kim, J.; Lundgren, K.; Harshman, L.; Van Allen, E.M.; Preston, M.; Dong, F.; Bellmunt, J.; Mouw, K.W.; et al. Mutational Analysis of 472 Urothelial Carcinoma Across Grades and Anatomic Sites. Clin. Cancer Res. 2019, 25, 2458–2470. [Google Scholar] [CrossRef]
- Guercio, B.J.; Sarfaty, M.; Teo, M.Y.; Ratna, N.; Duzgol, C.; Funt, S.A.; Lee, C.-H.; Aggen, D.H.; Regazzi, A.M.; Chen, Z.; et al. Clinical and Genomic Landscape of FGFR3-Altered Urothelial Carcinoma and Treatment Outcomes with Erdafitinib: A Real-World Experience. Clin. Cancer Res. 2023, 29, 4586–4595. [Google Scholar] [CrossRef]
- Sternberg, C.N.; Petrylak, D.P.; Bellmunt, J.; Nishiyama, H.; Necchi, A.; Gurney, H.; Lee, J.L.; van der Heijden, M.S.; Rosenbaum, E.; Penel, N.; et al. FORT-1: Phase II/III Study of Rogaratinib Versus Chemotherapy in Patients with Locally Advanced or Metastatic Urothelial Carcinoma Selected Based on FGFR1/3 mRNA Expression. J. Clin. Oncol. 2023, 41, 629–639. [Google Scholar] [CrossRef]
- Facchinetti, F.; Hollebecque, A.; Braye, F.; Vasseur, D.; Pradat, Y.; Bahleda, R.; Pobel, C.; Bigot, L.; Déas, O.; Florez Arango, J.D.; et al. Resistance to Selective FGFR Inhibitors in FGFR-Driven Urothelial Cancer. Cancer Discov. 2023, 13, 1998–2011. [Google Scholar] [CrossRef]
- Plimack, E.R.; Dunbrack, R.L.; Brennan, T.A.; Andrake, M.D.; Zhou, Y.; Serebriiskii, I.G.; Slifker, M.; Alpaugh, K.; Dulaimi, E.; Palma, N.; et al. Defects in DNA Repair Genes Predict Response to Neoadjuvant Cisplatin-based Chemotherapy in Muscle-invasive Bladder Cancer. Eur. Urol. 2015, 68, 959–967. [Google Scholar] [CrossRef]
- Galsky, M.D.; Wang, H.; Hahn, N.M.; Twardowski, P.; Pal, S.K.; Albany, C.; Fleming, M.T.; Starodub, A.; Hauke, R.J.; Yu, M.; et al. Phase 2 Trial of Gemcitabine, Cisplatin, plus Ipilimumab in Patients with Metastatic Urothelial Cancer and Impact of DNA Damage Response Gene Mutations on Outcomes. Eur. Urol. 2018, 73, 751–759. [Google Scholar] [CrossRef] [PubMed]
- Powles, T.; Carroll, D.; Chowdhury, S.; Gravis, G.; Joly, F.; Carles, J.; Fléchon, A.; Maroto, P.; Petrylak, D.; Rolland, F.; et al. An adaptive, biomarker-directed platform study of durvalumab in combination with targeted therapies in advanced urothelial cancer. Nat. Med. 2021, 27, 793–801. [Google Scholar] [CrossRef]
- Witjes, J.A.; Bruins, H.M.; Cathomas, R.; Compérat, E.M.; Cowan, N.C.; Gakis, G.; Hernández, V.; Linares Espinós, E.; Lorch, A.; Neuzillet, Y.; et al. European Association of Urology Guidelines on Muscle-invasive and Metastatic Bladder Cancer: Summary of the 2020 Guidelines. Eur. Urol. 2021, 79, 82–104. [Google Scholar] [CrossRef] [PubMed]
- Nawaf, C.; Shiang, A.; Chauhan, P.S.; Chaudhuri, A.A.; Agarwal, G.; Smith, Z.L. Circulating tumor DNA based minimal residual disease detection and adjuvant treatment decision-making for muscle-invasive bladder cancer guided by modern clinical trials. Transl. Oncol. 2023, 37, 101763. [Google Scholar] [CrossRef]
- Carrasco, R.; Ingelmo-Torres, M.; Trullas, R.; Roldán, F.L.; Rodríguez-Carunchio, L.; Juez, L.; Sureda, J.; Alcaraz, A.; Mengual, L.; Izquierdo, L. Tumor-Agnostic Circulating Tumor DNA Testing for Monitoring Muscle-Invasive Bladder Cancer. Int. J. Mol. Sci. 2023, 24, 16578. [Google Scholar] [CrossRef]
- Birkenkamp-Demtröder, K.; Christensen, E.; Nordentoft, I.; Knudsen, M.; Taber, A.; Høyer, S.; Lamy, P.; Agerbæk, M.; Jensen, J.B.; Dyrskjøt, L. Monitoring Treatment Response and Metastatic Relapse in Advanced Bladder Cancer by Liquid Biopsy Analysis. Eur. Urol. 2018, 73, 535–540. [Google Scholar] [CrossRef]
- Kovacs, G.; Akhtar, M.; Beckwith, B.J.; Bugert, P.; Cooper, C.S.; Delahunt, B.; Eble, J.N.; Fleming, S.; Ljungberg, B.; Medeiros, L.J.; et al. The Heidelberg classification of renal cell tumours. J. Pathol. 1997, 183, 131–133. [Google Scholar] [CrossRef]
- Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
- National Cancer Institute: Surveillance, E., and End Results Program. Cancer Stat Facts: Kidney and Renal Pelvis Cancer. Available online: https://seer.cancer.gov/statfacts/html/kidrp.html (accessed on 18 June 2024).
- Choueiri, T.K.; Motzer, R.J. Systemic Therapy for Metastatic Renal-Cell Carcinoma. N. Engl. J. Med. 2017, 376, 354–366. [Google Scholar] [CrossRef]
- Nuzzo, P.V.; Berchuck, J.E.; Korthauer, K.; Spisak, S.; Nassar, A.H.; Abou Alaiwi, S.; Chakravarthy, A.; Shen, S.Y.; Bakouny, Z.; Boccardo, F.; et al. Detection of renal cell carcinoma using plasma and urine cell-free DNA methylomes. Nat. Med. 2020, 26, 1041–1043. [Google Scholar] [CrossRef] [PubMed]
- Zill, O.A.; Banks, K.C.; Fairclough, S.R.; Mortimer, S.A.; Vowles, J.V.; Mokhtari, R.; Gandara, D.R.; Mack, P.C.; Odegaard, J.I.; Nagy, R.J.; et al. The Landscape of Actionable Genomic Alterations in Cell-Free Circulating Tumor DNA from 21,807 Advanced Cancer Patients. Clin. Cancer Res. 2018, 24, 3528–3538. [Google Scholar] [CrossRef]
- Kubiliute, R.; Jarmalaite, S. Epigenetic Biomarkers of Renal Cell Carcinoma for Liquid Biopsy Tests. Int. J. Mol. Sci. 2021, 22, 8846. [Google Scholar] [CrossRef] [PubMed]
- Outeiro-Pinho, G.; Barros-Silva, D.; Aznar, E.; Sousa, A.I.; Vieira-Coimbra, M.; Oliveira, J.; Gonçalves, C.S.; Costa, B.M.; Junker, K.; Henrique, R.; et al. MicroRNA-30a-5p(me): A novel diagnostic and prognostic biomarker for clear cell renal cell carcinoma in tissue and urine samples. J. Exp. Clin. Cancer Res. 2020, 39, 98. [Google Scholar] [CrossRef] [PubMed]
- Lin, Y.L.; Wang, Y.P.; Li, H.Z.; Zhang, X. Aberrant Promoter Methylation of PCDH17 (Protocadherin 17) in Serum and its Clinical Significance in Renal Cell Carcinoma. Med. Sci. Monit. 2017, 23, 3318–3323. [Google Scholar] [CrossRef]
- Skrypkina, I.; Tsyba, L.; Onyshchenko, K.; Morderer, D.; Kashparova, O.; Nikolaienko, O.; Panasenko, G.; Vozianov, S.; Romanenko, A.; Rynditch, A. Concentration and Methylation of Cell-Free DNA from Blood Plasma as Diagnostic Markers of Renal Cancer. Dis. Markers 2016, 2016, 3693096. [Google Scholar] [CrossRef] [PubMed]
- Hoque, M.O.; Begum, S.; Topaloglu, O.; Jeronimo, C.; Mambo, E.; Westra, W.H.; Califano, J.A.; Sidransky, D. Quantitative detection of promoter hypermethylation of multiple genes in the tumor, urine, and serum DNA of patients with renal cancer. Cancer Res. 2004, 64, 5511–5517. [Google Scholar] [CrossRef] [PubMed]
- Perego, R.A.; Corizzato, M.; Brambilla, P.; Ferrero, S.; Bianchi, C.; Fasoli, E.; Signorini, S.; Torsello, B.; Invernizzi, L.; Bombelli, S.; et al. Concentration and microsatellite status of plasma DNA for monitoring patients with renal carcinoma. Eur. J. Cancer 2008, 44, 1039–1047. [Google Scholar] [CrossRef] [PubMed]
- Mouliere, F.; Chandrananda, D.; Piskorz, A.M.; Moore, E.K.; Morris, J.; Ahlborn, L.B.; Mair, R.; Goranova, T.; Marass, F.; Heider, K.; et al. Enhanced detection of circulating tumor DNA by fragment size analysis. Sci. Transl. Med. 2018, 10, eaat4921. [Google Scholar] [CrossRef] [PubMed]
- Bacon, J.V.W.; Annala, M.; Soleimani, M.; Lavoie, J.M.; So, A.; Gleave, M.E.; Fazli, L.; Wang, G.; Chi, K.N.; Kollmannsberger, C.K.; et al. Plasma Circulating Tumor DNA and Clonal Hematopoiesis in Metastatic Renal Cell Carcinoma. Clin. Genitourin. Cancer 2020, 18, 322–331.e2. [Google Scholar] [CrossRef] [PubMed]
- Yamamoto, Y.; Uemura, M.; Fujita, M.; Maejima, K.; Koh, Y.; Matsushita, M.; Nakano, K.; Hayashi, Y.; Wang, C.; Ishizuya, Y.; et al. Clinical significance of the mutational landscape and fragmentation of circulating tumor DNA in renal cell carcinoma. Cancer Sci. 2019, 110, 617–628. [Google Scholar] [CrossRef]
- Maia, M.C.; Bergerot, P.G.; Dizman, N.; Hsu, J.; Jones, J.; Lanman, R.B.; Banks, K.C.; Pal, S.K. Association of Circulating Tumor DNA (ctDNA) Detection in Metastatic Renal Cell Carcinoma (mRCC) with Tumor Burden. Kidney Cancer 2017, 1, 65–70. [Google Scholar] [CrossRef]
- Chen, W.; Zhuang, J.; Wang, P.P.; Jiang, J.; Lin, C.; Zeng, P.; Liang, Y.; Zhang, X.; Dai, Y.; Diao, H. DNA methylation-based classification and identification of renal cell carcinoma prognosis-subgroups. Cancer Cell Int. 2019, 19, 185. [Google Scholar] [CrossRef] [PubMed]
- de Martino, M.; Klatte, T.; Haitel, A.; Marberger, M. Serum cell-free DNA in renal cell carcinoma: A diagnostic and prognostic marker. Cancer 2012, 118, 82–90. [Google Scholar] [CrossRef] [PubMed]
- Jung, M.; Ellinger, J.; Gevensleben, H.; Syring, I.; Lüders, C.; de Vos, L.; Pützer, S.; Bootz, F.; Landsberg, J.; Kristiansen, G.; et al. Cell-Free SHOX2 DNA Methylation in Blood as a Molecular Staging Parameter for Risk Stratification in Renal Cell Carcinoma Patients: A Prospective Observational Cohort Study. Clin. Chem. 2019, 65, 559–568. [Google Scholar] [CrossRef] [PubMed]
- Hahn, A.W.; Gill, D.M.; Maughan, B.; Agarwal, A.; Arjyal, L.; Gupta, S.; Streeter, J.; Bailey, E.; Pal, S.K.; Agarwal, N. Correlation of genomic alterations assessed by next-generation sequencing (NGS) of tumor tissue DNA and circulating tumor DNA (ctDNA) in metastatic renal cell carcinoma (mRCC): Potential clinical implications. Oncotarget 2017, 8, 33614–33620. [Google Scholar] [CrossRef] [PubMed]
- Khagi, Y.; Goodman, A.M.; Daniels, G.A.; Patel, S.P.; Sacco, A.G.; Randall, J.M.; Bazhenova, L.A.; Kurzrock, R. Hypermutated Circulating Tumor DNA: Correlation with Response to Checkpoint Inhibitor-Based Immunotherapy. Clin. Cancer Res. 2017, 23, 5729–5736. [Google Scholar] [CrossRef]
- Feng, G.; Ye, X.; Fang, F.; Pu, C.; Huang, H.; Li, G. Quantification of plasma cell-free DNA in predicting therapeutic efficacy of sorafenib on metastatic clear cell renal cell carcinoma. Dis. Markers 2013, 34, 105–111. [Google Scholar] [CrossRef] [PubMed]
- Pal, S.K.; Sonpavde, G.; Agarwal, N.; Vogelzang, N.J.; Srinivas, S.; Haas, N.B.; Signoretti, S.; McGregor, B.A.; Jones, J.; Lanman, R.B.; et al. Evolution of Circulating Tumor DNA Profile from First-line to Subsequent Therapy in Metastatic Renal Cell Carcinoma. Eur. Urol. 2017, 72, 557–564. [Google Scholar] [CrossRef]
- Gonzalgo, M.L.; Eisenberger, C.F.; Lee, S.M.; Trock, B.J.; Marshall, F.F.; Hortopan, S.; Sidransky, D.; Schoenberg, M.P. Prognostic Significance of Preoperative Molecular Serum Analysis in Renal Cancer. Clin. Cancer Res. 2002, 8, 1878–1881. [Google Scholar]
- Lasseter, K.; Nassar, A.H.; Hamieh, L.; Berchuck, J.E.; Nuzzo, P.V.; Korthauer, K.; Shinagare, A.B.; Ogorek, B.; McKay, R.; Thorner, A.R.; et al. Plasma cell-free DNA variant analysis compared with methylated DNA analysis in renal cell carcinoma. Genet. Med. 2020, 22, 1366–1373. [Google Scholar] [CrossRef]
- Wan, J.; Zhu, L.; Jiang, Z.; Cheng, K. Monitoring of plasma cell-free DNA in predicting postoperative recurrence of clear cell renal cell carcinoma. Urol. Int. 2013, 91, 273–278. [Google Scholar] [CrossRef]
- Chera, B.S.; Kumar, S.; Shen, C.; Amdur, R.; Dagan, R.; Green, R.; Goldman, E.; Weiss, J.; Grilley-Olson, J.; Patel, S.; et al. Plasma Circulating Tumor HPV DNA for the Surveillance of Cancer Recurrence in HPV-Associated Oropharyngeal Cancer. J. Clin. Oncol. 2020, 38, 1050–1058. [Google Scholar] [CrossRef] [PubMed]
- Naegele, S.; Efthymiou, V.; Das, D.; Sadow, P.M.; Richmon, J.D.; Iafrate, A.J.; Faden, D.L. Detection and Monitoring of Circulating Tumor HPV DNA in HPV-Associated Sinonasal and Nasopharyngeal Cancers. JAMA Otolaryngol. Head Neck Surg. 2023, 149, 179–181. [Google Scholar] [CrossRef] [PubMed]
- Balachandra, S.; Kusin, S.B.; Lee, R.; Blackwell, J.M.; Tiro, J.A.; Cowell, L.G.; Chiang, C.M.; Wu, S.Y.; Varma, S.; Rivera, E.L.; et al. Blood-based biomarkers of human papillomavirus-associated cancers: A systematic review and meta-analysis. Cancer 2021, 127, 850–864. [Google Scholar] [CrossRef] [PubMed]
- Olesen, T.B.; Sand, F.L.; Rasmussen, C.L.; Albieri, V.; Toft, B.G.; Norrild, B.; Munk, C.; Kjær, S.K. Prevalence of human papillomavirus DNA and p16(INK4a) in penile cancer and penile intraepithelial neoplasia: A systematic review and meta-analysis. Lancet Oncol. 2019, 20, 145–158. [Google Scholar] [CrossRef] [PubMed]
- Ellinger, J.; Wittkamp, V.; Albers, P.; Perabo, F.G.E.; Mueller, S.C.; Ruecker, A.v.; Bastian, P.J. Cell-Free Circulating DNA: Diagnostic Value in Patients with Testicular Germ Cell Cancer. J. Urol. 2009, 181, 363–371. [Google Scholar] [CrossRef] [PubMed]
- Ellinger, J.; Albers, P.; Müller, S.C.; von Ruecker, A.; Bastian, P.J. Circulating mitochondrial DNA in the serum of patients with testicular germ cell cancer as a novel noninvasive diagnostic biomarker. BJU Int. 2009, 104, 48–52. [Google Scholar] [CrossRef] [PubMed]
- Ellinger, J.; Albers, P.; Perabo, F.G.; Müller, S.C.; von Ruecker, A.; Bastian, P.J. CpG island hypermethylation of cell-free circulating serum DNA in patients with testicular cancer. J. Urol. 2009, 182, 324–329. [Google Scholar] [CrossRef] [PubMed]
- Raos, D.; Oršolić, D.; Mašić, S.; Tomić, M.; Krasić, J.; Tomašković, I.; Gabaj, N.N.; Gelo, N.; Kaštelan, Ž.; Kuliš, T.; et al. cfDNA methylation in liquid biopsies as potential testicular seminoma biomarker. Epigenomics 2022, 14, 1493–1507. [Google Scholar] [CrossRef] [PubMed]
- Nazha, B.; Zhuang, T.Z.; Dada, H.I.; Drusbosky, L.M.; Brown, J.T.; Ravindranathan, D.; Carthon, B.C.; Kucuk, O.; Goldman, J.; Master, V.A.; et al. Blood-Based Next-Generation Sequencing in Adrenocortical Carcinoma. Oncologist 2022, 27, 462–468. [Google Scholar] [CrossRef]
- Raj, N.; Zheng, Y.; Kelly, V.; Katz, S.S.; Chou, J.; Do, R.K.G.; Capanu, M.; Zamarin, D.; Saltz, L.B.; Ariyan, C.E.; et al. PD-1 Blockade in Advanced Adrenocortical Carcinoma. J. Clin. Oncol. 2020, 38, 71–80. [Google Scholar] [CrossRef]
- Mota, J.M.; Sousa, L.G.; Braghiroli, M.I.; Siqueira, L.T.; Neto, J.E.B.; Chapchap, P.; Hoff, A.A.O.; Hoff, P.M. Pembrolizumab for metastatic adrenocortical carcinoma with high mutational burden: Two case reports. Medicine 2018, 97, e13517. [Google Scholar] [CrossRef] [PubMed]
- Garinet, S.; Nectoux, J.; Neou, M.; Pasmant, E.; Jouinot, A.; Sibony, M.; Orhant, L.; Pipoli da Fonseca, J.; Perlemoine, K.; Bricaire, L.; et al. Detection and monitoring of circulating tumor DNA in adrenocortical carcinoma. Endocr.-Relat. Cancer 2018, 25, L13–L17. [Google Scholar] [CrossRef] [PubMed]
- Fiala, C.; Diamandis, E.P. Utility of circulating tumor DNA in cancer diagnostics with emphasis on early detection. BMC Med. 2018, 16, 166. [Google Scholar] [CrossRef] [PubMed]
- Esfahani, M.S.; Hamilton, E.G.; Mehrmohamadi, M.; Nabet, B.Y.; Alig, S.K.; King, D.A.; Steen, C.B.; Macaulay, C.W.; Schultz, A.; Nesselbush, M.C.; et al. Inferring gene expression from cell-free DNA fragmentation profiles. Nat. Biotechnol. 2022, 40, 585–597. [Google Scholar] [CrossRef] [PubMed]
- Cristiano, S.; Leal, A.; Phallen, J.; Fiksel, J.; Adleff, V.; Bruhm, D.C.; Jensen, S.; Medina, J.E.; Hruban, C.; White, J.R.; et al. Genome-wide cell-free DNA fragmentation in patients with cancer. Nature 2019, 570, 385–389. [Google Scholar] [CrossRef] [PubMed]
- Abbosh, C.; Birkbak, N.J.; Wilson, G.A.; Jamal-Hanjani, M.; Constantin, T.; Salari, R.; Le Quesne, J.; Moore, D.A.; Veeriah, S.; Rosenthal, R.; et al. Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution. Nature 2017, 545, 446–451. [Google Scholar] [CrossRef]
- Merker, J.D.; Oxnard, G.R.; Compton, C.; Diehn, M.; Hurley, P.; Lazar, A.J.; Lindeman, N.; Lockwood, C.M.; Rai, A.J.; Schilsky, R.L.; et al. Circulating Tumor DNA Analysis in Patients with Cancer: American Society of Clinical Oncology and College of American Pathologists Joint Review. J. Clin. Oncol. 2018, 36, 1631–1641. [Google Scholar] [CrossRef]
CtDNA Detection Method | Approach | Sensitivity | Potential Applications | Limitations |
---|---|---|---|---|
Droplet Digtal PCR (ddPCR) | Quantification of targeted genetic alterations [7,9] | High for pre-specified mutations [7,9] | Detecting targeted mutations, tracking treatment response, relapse, and recurrence | Omits genetic alterations or subclonal populations not pre-targeted |
Beads, Emulsion, Amplification, and Magnetics (BEAMing) | Quantification of targeted genetic alterations [9] | High for pre-specified mutations [9] | Detecting targeted mutations, tracking treatment response, relapse, and recurrence | Omits genetic alterations or subclonal populations not targeted |
Next-generation Sequencing (NGS) | Detection of a range of genetic alterations, including variant allele frequency, insertions, deletions, and structural variants [11] | High for a range of genetic alterations [11] | Identification of actionable or high-risk alterations, detection of alterations associated with mechanisms of treatment resistance, detection of tumor genetic evolution, detection of subclonal populations | High costs, multi-week turnaround time, data which may be difficult to interpret or ambiguous |
Immunoprecipitation-based Epigenetic Targeted Assays | Detection of cancer-specific epigenetic patterns including DNA methylation, nucleosome positioning, and histone modification [14] | Variable [17,18] | Early cancer diagnosis and prognosis | Potentially lower sensitivity, challenges in interpreting data, greater technical challenges in interpretation of results |
Agent | ctDNA Detectable Target | Drug Target | Reference |
---|---|---|---|
Androgen Receptor Signaling Inhibitors (Enzalutamide and Abiraterone) | Androgen receptor alterations, including LBD mutations and structural alterations (such as AR-V7), AR amplifications, and AR copy number alterations | Ligand binding domain of androgen receptor and downstream androgen receptor signaling | [53,58,59,60,61,62,63,64,65,66,67] |
PARP Inhibitors (Olaparib, Rucaparib) | Homologous recombination repair deficits including BRCA1, BRCA2, ATM, CHEK2, RAD51D, and PALB2 | Poly adenosine diphosphate (ADP)–ribose polymerase (PARP) inhibitor (single-strand DNA repair) | [69,70,71,72,73,74,75,76,77] |
AKT inhibitors (Ipatasertib) | PTEN loss | PTEN-PI3K-AKT pathway | [78,79,80] |
Immune Checkpoint Inhibitors (Pembrolizumab) | Microsatellite instability, mismatch repair deficiency, and tumor mutational burden | Anti-programmed cell death protein 1 (Anti-PD-1) | [81,82,84,85] |
Agent | Genetic Target | Drug Target | Reference |
---|---|---|---|
Immune Modulatory Therapy (Pembrolizumab, Atezolizumab, Toripalimab, Ipilimumab, and Durvalumab) | Microsatellite instability, mismatch repair deficiency, tumor mutational burden | Anti-PD-1, Anti-PD-L1, Anti-CTLA-4 | [118,119,120,121,122,124,156,157] |
FGFR3 Inhibitors (Rogaratinib, Edafitinib, Furtibatinib, and Pemigatinib) | FGFR3 | FGFR | [126,127,128] |
Cisplatin-based Therapies | DNA damage response genes including ATM, RBI, and FANCC, nucleotide excision repair genes like ERCC2 | Cross-links DNA | [129,130,131] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Gerke, M.B.; Jansen, C.S.; Bilen, M.A. Circulating Tumor DNA in Genitourinary Cancers: Detection, Prognostics, and Therapeutic Implications. Cancers 2024, 16, 2280. https://doi.org/10.3390/cancers16122280
Gerke MB, Jansen CS, Bilen MA. Circulating Tumor DNA in Genitourinary Cancers: Detection, Prognostics, and Therapeutic Implications. Cancers. 2024; 16(12):2280. https://doi.org/10.3390/cancers16122280
Chicago/Turabian StyleGerke, Margo B., Caroline S. Jansen, and Mehmet A. Bilen. 2024. "Circulating Tumor DNA in Genitourinary Cancers: Detection, Prognostics, and Therapeutic Implications" Cancers 16, no. 12: 2280. https://doi.org/10.3390/cancers16122280
APA StyleGerke, M. B., Jansen, C. S., & Bilen, M. A. (2024). Circulating Tumor DNA in Genitourinary Cancers: Detection, Prognostics, and Therapeutic Implications. Cancers, 16(12), 2280. https://doi.org/10.3390/cancers16122280