Liquid biopsy for human cancer: cancer screening, monitoring, and treatment

Hao Wang1, Yi Zhang1, Hao Zhang1, Hui Cao2,3, Jinning Mao4, Xinxin Chen1, Liangchi Wang5, Nan Zhang6, Peng Luo7, Ji Xue8, Xiaoya Qi4, Xiancheng Dong9, Guodong Liu1(), Quan Cheng10,11()

PDF
MedComm ›› 2024, Vol. 5 ›› Issue (6) : e564. DOI: 10.1002/mco2.564
REVIEW

Liquid biopsy for human cancer: cancer screening, monitoring, and treatment

  • Hao Wang1, Yi Zhang1, Hao Zhang1, Hui Cao2,3, Jinning Mao4, Xinxin Chen1, Liangchi Wang5, Nan Zhang6, Peng Luo7, Ji Xue8, Xiaoya Qi4, Xiancheng Dong9, Guodong Liu1(), Quan Cheng10,11()
Author information +
History +

Abstract

Currently, tumor treatment modalities such as immunotherapy and targeted therapy have more stringent requirements for obtaining tumor growth information and require more accurate and easy-to-operate tumor information detection methods. Compared with traditional tissue biopsy, liquid biopsy is a novel, minimally invasive, real-time detection tool for detecting information directly or indirectly released by tumors in human body fluids, which is more suitable for the requirements of new tumor treatment modalities. Liquid biopsy has not been widely used in clinical practice, and there are fewer reviews of related clinical applications. This review summarizes the clinical applications of liquid biopsy components (e.g., circulating tumor cells, circulating tumor DNA, extracellular vesicles, etc.) in tumorigenesis and progression. This includes the development process and detection techniques of liquid biopsies, early screening of tumors, tumor growth detection, and guiding therapeutic strategies (liquid biopsy-based personalized medicine and prediction of treatment response). Finally, the current challenges and future directions for clinical applications of liquid biopsy are proposed. In sum, this review will inspire more researchers to use liquid biopsy technology to promote the realization of individualized therapy, improve the efficacy of tumor therapy, and provide better therapeutic options for tumor patients.

Keywords

Circulating tumor cell / Circulating tumor DNA / Exosomes / ICI / Immunotherapy / Liquid biopsy / Noninvasive tumor detection / PD-L1 / Tumor-related circulating cells

Cite this article

Download citation ▾
Hao Wang, Yi Zhang, Hao Zhang, Hui Cao, Jinning Mao, Xinxin Chen, Liangchi Wang, Nan Zhang, Peng Luo, Ji Xue, Xiaoya Qi, Xiancheng Dong, Guodong Liu, Quan Cheng. Liquid biopsy for human cancer: cancer screening, monitoring, and treatment. MedComm, 2024, 5(6): e564 https://doi.org/10.1002/mco2.564

References

1 RC Fitzgerald, AC Antoniou, L Fruk, N Rosenfeld. The future of early cancer detection. Nat Med. 2022;28(4):666-677.
2 S Connal, JM Cameron, A Sala, et al. Liquid biopsies: the future of cancer early detection. J Transl Med. 2023;21(1):118.
3 S Zhu, T Zhang, L Zheng, et al. Combination strategies to maximize the benefits of cancer immunotherapy. J Hematol Oncol. 2021;14(1):156.
4 J Larkin, V Chiarion-Sileni, R Gonzalez, et al. Five-year survival with combined nivolumab and ipilimumab in advanced melanoma. New Eng J Med. 2019;381(16):1535-1546.
5 RK Vaddepally, P Kharel, R Pandey, R Garje, AB Chandra. Review of indications of FDA-approved immune checkpoint inhibitors per NCCN guidelines with the level of evidence. Cancers. 2020;12(3):738.
6 EB Garon, MD Hellmann, NA Rizvi, et al. Five-year overall survival for patients with advanced non?small-cell lung cancer treated with pembrolizumab: results from the phase I KEYNOTE-001 study. J Clin Oncol. 2019;37(28):2518-2527.
7 O Hamid, C Robert, A Daud, et al. Five-year survival outcomes for patients with advanced melanoma treated with pembrolizumab in KEYNOTE-001. Ann Oncol. 2019;30(4):582-588.
8 JR Brahmer, R Govindan, RA Anders, et al. The Society for Immunotherapy of Cancer consensus statement on immunotherapy for the treatment of non-small cell lung cancer (NSCLC). J Immunother Cancer. 2018;6(1):75.
9 A Rizzo, AD Ricci. Biomarkers for breast cancer immunotherapy: pD-L1, TILs, and beyond. Expert Opin Investig Drugs. 2022;31(6):549-555.
10 M Mino-Kenudson, K Schalper, W Cooper, et al. Predictive biomarkers for immunotherapy in lung cancer: perspective from the International Association for the Study of Lung Cancer Pathology Committee. J Thorac Oncol. 2022;17(12):1335-1354.
11 AT Jacobs, D Martinez Castaneda-Cruz, MM Rose, et al. Targeted therapy for breast cancer: an overview of drug classes and outcomes. Biochem Pharmacol. 2022;204:115209.
12 FR Hirsch, GV Scagliotti, JL Mulshine, et al. Lung cancer: current therapies and new targeted treatments. Lancet. 2017;389(10066):299-311.
13 AN Geisler, GS Phillips, DM Barrios, et al. Immune checkpoint inhibitor-related dermatologic adverse events. J Am Acad Dermatol. 2020;83(5):1255-1268.
14 Shyam Sunder S, UC Sharma, S Pokharel. Adverse effects of tyrosine kinase inhibitors in cancer therapy: pathophysiology, mechanisms and clinical management. Signal Transduct Target Ther. 2023;8(1):262.
15 YL Tang, DD Li, JY Duan, LM Sheng, X Wang. Resistance to targeted therapy in metastatic colorectal cancer: current status and new developments. World J Gastroenterol. 2023;29(6):926-948.
16 F Pesapane, MB Suter, A Rotili, et al. Will traditional biopsy be substituted by radiomics and liquid biopsy for breast cancer diagnosis and characterisation? Med Oncol. 2020;37(4):29.
17 AM Tsimberidou, E Fountzilas, M Nikanjam, R Kurzrock. Review of precision cancer medicine: evolution of the treatment paradigm. Cancer Treat Rev. 2020;86:102019.
18 J Zugazagoitia, C Guedes, S Ponce, I Ferrer, S Molina-Pinelo, L Paz-Ares. Current challenges in cancer treatment. Clin Ther. 2016;38(7):1551-1566.
19 YMD Lo, N Corbetta, PF Chamberlain, et al. Presence of fetal DNA in maternal plasma and serum. Lancet. 1997;350(9076):485-487.
20 JA Canick, GE Palomaki, EM Kloza, GM Lambert-Messerlian, JE Haddow. The impact of maternal plasma DNA fetal fraction on next generation sequencing tests for common fetal aneuploidies. Prenat Diagn. 2013;33(7):667-674.
21 KL Reckamp, VO Melnikova, C Karlovich, et al. A highly sensitive and quantitative test platform for detection of NSCLC EGFR mutations in urine and plasma. J Thorac Oncol. 2016;11(10):1690-1700.
22 Y Wang, S Springer, CL Mulvey, et al. Detection of somatic mutations and HPV in the saliva and plasma of patients with head and neck squamous cell carcinomas. Sci Transl Med. 2015;7(293):293ra104.
23 H Kimura, Y Fujiwara, T Sone, et al. EGFR mutation status in tumour-derived DNA from pleural effusion fluid is a practical basis for predicting the response to gefitinib. Br J Cancer. 2006;95(10):1390-1395.
24 L De Mattos-Arruda, R Mayor, CKY Ng, et al. Cerebrospinal fluid-derived circulating tumour DNA better represents the genomic alterations of brain tumours than plasma. Nat Commun. 2015;6:8839.
25 F Diehl, K Schmidt, KH Durkee, et al. Analysis of mutations in DNA isolated from plasma and stool of colorectal cancer patients. Gastroenterology. 2008;135(2):489-498.
26 K Pantel, C Alix-Panabieres. Circulating tumour cells in cancer patients: challenges and perspectives. Trends Mol Med. 2010;16(9):398-406.
27 H Schwarzenbach, DS Hoon, K Pantel. Cell-free nucleic acids as biomarkers in cancer patients. Nat Rev Cancer. 2011;11(6):426-437.
28 S Anfossi, A Babayan, K Pantel, GA Calin. Clinical utility of circulating non-coding RNAs—an update. Nat Rev Clin Oncol. 2018;15(9):541-563.
29 R Kalluri, VS LeBleu. The biology, function, and biomedical applications of exosomes. Science. 2020;367(6478):eaau6977.
30 MG Best, N Sol, S In ’t Veld, et al. Swarm intelligence-enhanced detection of non-small-cell lung cancer using tumor-educated platelets. Cancer Cell. 2017;32(2):238-252. e9.
31 SK Min, SY Jung, HK Kang, SB Jo, MJ Kim, BM Min. MicroRNA-146a-5p limits elevated TGF-β signal during cell senescence. Mol Ther Nucleic Acids. 2017;7:335-338.
32 JCM Wan, C Massie, J Garcia-Corbacho, et al. Liquid biopsies come of age: towards implementation of circulating tumour DNA. Nat Rev Cancer. 2017;17(4):223-238.
33 DG Rothwell, M Ayub, N Cook, et al. Utility of ctDNA to support patient selection for early phase clinical trials: the TARGET study. Nat Med. 2019;25(5):738-743.
34 Z Zhang, H Wu, W Chong, L Shang, C Jing, L Li. Liquid biopsy in gastric cancer: predictive and prognostic biomarkers. Cell Death Dis. 2022;13(10):903.
35 K Pantel, C Alix-Panabieres. Liquid biopsy and minimal residual disease—latest advances and implications for cure. Nat Rev Clin Oncol. 2019;16(7):409-424.
36 N Aceto, A Bardia, DT Miyamoto, et al. Circulating tumor cell clusters are oligoclonal precursors of breast cancer metastasis. Cell. 2014;158(5):1110-1122.
37 Z Feng, J Wu, Y Lu, et al. Circulating tumor cells in the early detection of human cancers. Int J Biol Sci. 2022;18(8):3251-3265.
38 D Lin, L Shen, M Luo, et al. Circulating tumor cells: biology and clinical significance. Signal Transduct Target Ther. 2021;6(1):404.
39 H Chen, Y Li, Z Zhang, S Wang. Immunomagnetic separation of circulating tumor cells with microfluidic chips and their clinical applications. Biomicrofluidics. 2020;14(4):041502.
40 KS Law, CE Huang, SW Chen. Detection of circulating tumor cell-related markers in gynecologic cancer using microfluidic devices: a pilot study. Int J Mol Sci. 2023;24(3):2300.
41 VL Chen, Q Huang, R Harouaka, et al. A dual-filtration system for single-cell sequencing of circulating tumor cells and clusters in HCC. Hepatol Commun. 2022;6(6):1482-1491.
42 Y Liu, R Zhao, Z Xie, et al. Significance of circulating tumor cells detection in tumor diagnosis and monitoring. BMC Cancer. 2023;23(1):1195.
43 AA Javed, D Ding, A Hasanain, et al. Persistent circulating tumor cells at 1 year after oncologic resection predict late recurrence in pancreatic cancer. Ann Surg. 2023;277(6):859-865.
44 V Cappelletti, E Verzoni, R Ratta, et al. Analysis of single circulating tumor cells in renal cell carcinoma reveals phenotypic heterogeneity and genomic alterations related to progression. Int J Mol Sci. 2020;21(4):1475.
45 J Xu, K Liao, X Yang, C Wu, W Wu. Using single-cell sequencing technology to detect circulating tumor cells in solid tumors. Mol Cancer. 2021;20(1):104.
46 S Zhu, Z Xie, Y Chen, et al. Real-time detection of circulating tumor cells in bloodstream using plasmonic fiber sensors. Biosensors (Basel). 2022;12(11):968.
47 T Yu, C Wang, M Xie, et al. Heterogeneity of CTC contributes to the organotropism of breast cancer. Biomed Pharmacother. 2021;137:111314.
48 S Wu, L Gu, J Qin, et al. Rapid label-free isolation of circulating tumor cells from patients' peripheral blood using electrically charged Fe(3)O(4) nanoparticles. ACS Appl Mater Interfaces. 2020;12(4):4193-4203.
49 H Chen, Y Han, Q Li, Y Zou, S Wang, X Jiao. Clinical Microfluidic chip platform for the isolation of versatile circulating tumor cells. J Vis Exp. 2023(200).
50 IS Batth, A Mitra, S Rood, S Kopetz, D Menter, S Li. CTC analysis: an update on technological progress. Transl Res. 2019;212:14-25.
51 K Anitha, B Posinasetty, K Naveen Kumari, et al. Liquid biopsy for precision diagnostics and therapeutics. Clin Chim Acta. 2024;554:117746.
52 DK Dang, BH Park. Circulating tumor DNA: current challenges for clinical utility. J Clin Invest. 2022;132(12):e154941.
53 S Sawamura, T Mijiddorj Myangat, I Kajihara, et al. Genomic landscape of circulating tumour DNA in metastatic extramammary Paget's disease. Exp Dermatol. 2022;31(3):341-348.
54 F Mouliere, AR Thierry. The importance of examining the proportion of circulating DNA originating from tumor, microenvironment and normal cells in colorectal cancer patients. Expert Opin Biol Ther. 2012;12(1):S209-S215. Suppl.
55 V Candotto, F Pezzetti, A Scarano, A Agazzi, F Spadari, A Palmieri. Liquid biopsy. J Biol Regul Homeost Agents. 2019;33(1):1-6. 6 Suppl. Dental supplement.
56 MH Kim, GM Kim, JM Ahn, et al. Copy number aberrations in circulating tumor DNA enables prognosis prediction and molecular characterization of breast cancer. J Natl Cancer Inst. 2023;115(9):1036-1049.
57 I Garcia-Murillas, G Schiavon, B Weigelt, et al. Mutation tracking in circulating tumor DNA predicts relapse in early breast cancer. Sci Transl Med. 2015;7(302):302ra133.
58 L Zhang, R Parvin, Q Fan, F Ye. Emerging digital PCR technology in precision medicine. Biosens Bioelectron. 2022;211:114344.
59 M Postel, A Roosen, P Laurent-Puig, V Taly, SF Wang-Renault. Droplet-based digital PCR and next generation sequencing for monitoring circulating tumor DNA: a cancer diagnostic perspective. Expert Rev Mol Diagn. 2018;18(1):7-17.
60 L Meriranta, A Alkodsi, A Pasanen, et al. Molecular features encoded in the ctDNA reveal heterogeneity and predict outcome in high-risk aggressive B-cell lymphoma. Blood. 2022;139(12):1863-1877.
61 D Yu, Y Li, M Wang, et al. Exosomes as a new frontier of cancer liquid biopsy. Mol Cancer. 2022;21(1):56.
62 VS LeBleu, R Kalluri. Exosomes as a multicomponent biomarker platform in cancer. Trends Cancer. 2020;6(9):767-774.
63 X Yu, L Sha, L Dong, Y Cao, J Zhao. Recent advances in bio-sensing methods for the detection of tumor exosomes. Crit Rev Anal Chem. 2022;52(2):356-374.
64 G van Niel, G D'Angelo, G Raposo. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol. 2018;19(4):213-228.
65 N Ludwig, TL Whiteside, TE Reichert. Challenges in exosome isolation and analysis in health and disease. Int J Mol Sci. 2019;20(19):4684.
66 L Zhu, HT Sun, S Wang, et al. Isolation and characterization of exosomes for cancer research. J Hematol Oncol. 2020;13(1):152.
67 E Ococks, AM Frankell, N Masque Soler, et al. Longitudinal tracking of 97 esophageal adenocarcinomas using liquid biopsy sampling. Ann Oncol. 2021;32(4):522-532.
68 X Wang, J Xia, L Yang, J Dai, L He. Recent progress in exosome research: isolation, characterization and clinical applications. Cancer Gene Ther. 2023;30(8):1051-1065.
69 C Hu, W Jiang, M Lv, et al. Potentiality of exosomal proteins as novel cancer biomarkers for liquid biopsy. Front Immunol. 2022;13:792046.
70 TA Chan, M Yarchoan, E Jaffee, et al. Development of tumor mutation burden as an immunotherapy biomarker: utility for the oncology clinic. Ann Oncol. 2019;30(1):44-56.
71 P Hofman. PD-L1 immunohistochemistry for non-small cell lung carcinoma: which strategy should be adopted? Expert Rev Mol Diagn. 2017;17(12):1097-1108.
72 MS Tsao, KM Kerr, M Kockx, et al. PD-L1 immunohistochemistry comparability study in real-life clinical samples: results of blueprint phase 2 project. J Thorac Oncol. 2018;13(9):1302-1311.
73 M Ilie, J Benzaquen, V Hofman, et al. Immunotherapy in non-small cell lung cancer: biological principles and future opportunities. Curr Mol Med. 2017;17(8):527-540.
74 G Poulet, J Massias, V Taly. Liquid biopsy: general concepts. Acta Cytol. 2019;63(6):449-455.
75 J Marrugo-Ramírez, M Mir, J Samitier. Blood-based cancer biomarkers in liquid biopsy: a promising non-invasive alternative to tissue biopsy. Int J Mol Sci. 2018;19(10):2877.
76 L Licata, M Mariani, F Rossari, et al. Tissue- and liquid biopsy-based biomarkers for immunotherapy in breast cancer. Breast. 2023;69:330-341.
77 SM Batool, A Yekula, P Khanna, et al. The liquid biopsy consortium: challenges and opportunities for early cancer detection and monitoring. Cell Rep Med. 2023;4(10):101198.
78 BJ Mankidy, G Mohammad, K Trinh, et al. High risk lung nodule: a multidisciplinary approach to diagnosis and management. Respir Med. 2023;214:107277.
79 C Alix-Panabières, K Pantel. Liquid biopsy: from discovery to clinical application. Cancer Discov. 2021;11(4):858-873.
80 S Najafi, J Majidpoor, K Mortezaee. Liquid biopsy in colorectal cancer. Clin Chim Acta. 2024;553:117674.
81 U Malapelle, P Pisapia, F Pepe, et al. The evolving role of liquid biopsy in lung cancer. Lung Cancer. 2022;172:53-64.
82 H Ohmura, F Hanamura, Y Okumura, et al. Liquid biopsy for breast cancer and other solid tumors: a review of recent advances. Breast Cancer. 2024; Epub ahead of print.
83 A Raza, AQ Khan, VP Inchakalody, et al. Dynamic liquid biopsy components as predictive and prognostic biomarkers in colorectal cancer. J Exp Clin Cancer Res. 2022;41(1):99.
84 Y Suo, Z Gu, X Wei. Advances of in vivo flow cytometry on cancer studies. Cytometry A. 2020;97(1):15-23.
85 A Shaukat, TR Levin. Current and future colorectal cancer screening strategies. Nat Rev Gastroenterol Hepatol. 2022;19(8):521-531.
86 VHF de Jesus, ALA Dettino. Update on hepatocellular carcinoma from the 2018 Gastrointestinal Cancer Symposium (ASCO GI). J Hepatocell Carcinoma. 2018;5:87-90.
87 I Cima, SL Kong, D Sengupta, et al. Tumor-derived circulating endothelial cell clusters in colorectal cancer. Sci Transl Med. 2016;8(345):345ra89.
88 HM Kang, GH Kim, HK Jeon, et al. Circulating tumor cells detected by lab-on-a-disc: role in early diagnosis of gastric cancer. PLoS One. 2017;12(6):e0180251.
89 L Tang, S Zhao, W Liu, et al. Diagnostic accuracy of circulating tumor cells detection in gastric cancer: systematic review and meta-analysis. BMC Cancer. 2013;13:314.
90 C Rolfo, P Mack, GV Scagliotti, et al. Liquid biopsy for advanced NSCLC: a consensus statement from the International Association for the Study of Lung Cancer. J Thorac Oncol. 2021;16(10):1647-1662.
91 C Nicolazzo, L Barault, S Caponnetto, et al. True conversions from RAS mutant to RAS wild-type in circulating tumor DNA from metastatic colorectal cancer patients as assessed by methylation and mutational signature. Cancer Lett. 2021;507:89-96.
92 BM Szczerba, F Castro-Giner, M Vetter, et al. Neutrophils escort circulating tumour cells to enable cell cycle progression. Nature. 2019;566(7745):553-557.
93 JD Warren, W Xiong, AM Bunker, et al. Septin 9 methylated DNA is a sensitive and specific blood test for colorectal cancer. BMC Med. 2011;9:133.
94 A Loktionov. Biomarkers for detecting colorectal cancer non-invasively: dNA, RNA or proteins? World J Gastrointest Oncol. 2020;12(2):124-148.
95 J Hu, B Hu, YC Gui, ZB Tan, JW Xu. Diagnostic value and clinical significance of methylated SEPT9 for colorectal cancer: a meta-analysis. Med Sci Monit. 2019;25:5813-5822.
96 L Song, J Jia, X Peng, W Xiao, Y Li. The performance of the SEPT9 gene methylation assay and a comparison with other CRC screening tests: a meta-analysis. Sci Rep. 2017;7(1):3032.
97 H Luo, Q Zhao, W Wei, et al. Circulating tumor DNA methylation profiles enable early diagnosis, prognosis prediction, and screening for colorectal cancer. Sci Transl Med. 2020;12(524):eaax7533.
98 X Wu, Y Zhang, T Hu, et al. A novel cell-free DNA methylation-based model improves the early detection of colorectal cancer. Mol Oncol. 2021;15(10):2702-2714.
99 T Crook, R Leonard, K Mokbel, et al. Accurate screening for early-stage breast cancer by detection and profiling of circulating tumor cells. Cancers (Basel). 2022;14(14):3341.
100 L Konczalla, T Ghadban, KE Effenberger, et al. Prospective comparison of the prognostic relevance of circulating tumor cells in blood and disseminated tumor cells in bone marrow of a single patient's cohort with esophageal cancer. Ann Surg. 2021;273(2):299-305.
101 RJ Pan, HJ Hong, J Sun, et al. Detection and clinical value of circulating tumor cells as an assisted prognostic marker in colorectal cancer patients. Cancer Manag Res. 2021;13:4567-4578.
102 HJ Chalfin, T Pramparo, A Mortazavi, et al. Circulating tumor cell subtypes and T-cell populations as prognostic biomarkers to combination immunotherapy in patients with metastatic genitourinary cancer. Clin Cancer Res. 2021;27(5):1391-1398.
103 AM Miller, RH Shah, EI Pentsova, et al. Tracking tumour evolution in glioma through liquid biopsies of cerebrospinal fluid. Nature. 2019;565(7741):654-658.
104 M Tamminga, S de Wit, TJN Hiltermann, et al. Circulating tumor cells in advanced non-small cell lung cancer patients are associated with worse tumor response to checkpoint inhibitors. J Immunother Cancer. 2019;7(1):173.
105 S Camera, T Akin Telli, E Woff, et al. Prognostic value of the pace of tumor progression as assessed by serial (18)F-FDG PET/CT scan and liquid biopsy in refractory colorectal cancer: the CORIOLAN trial. Cancers (Basel). 2020;12(10):E2752.
106 M Bootsma, RR McKay, H Emamekhoo, et al. Longitudinal molecular profiling of circulating tumor cells in metastatic renal cell carcinoma. J Clin Oncol. 2022;40(31):3633-3641.
107 FC Bidard, N Kiavue, W Jacot, et al. Overall survival with circulating tumor cell count-driven choice of therapy in advanced breast cancer: a randomized trial. J Clin Oncol. 2024;42(4):383-389.
108 S Riethdorf, V Müller, S Loibl, et al. Prognostic impact of circulating tumor cells for breast cancer patients treated in the neoadjuvant “Geparquattro” trial. Clin Cancer Res. 2017;23(18):5384-5393.
109 FC Bidard, S Michiels, S Riethdorf, et al. Circulating tumor cells in breast cancer patients treated by neoadjuvant chemotherapy: a meta-analysis. J Natl Cancer Inst. 2018;110(6):560-567.
110 M Radovich, G Jiang, BA Hancock, et al. Association of circulating tumor DNA and circulating tumor cells after neoadjuvant chemotherapy with disease recurrence in patients with triple-negative breast cancer: preplanned secondary analysis of the BRE12-158 randomized clinical trial. JAMA Oncol. 2020;6(9):1410-1415.
111 NC Turner, C Swift, B Jenkins, et al. Results of the c-TRAK TN trial: a clinical trial utilising ctDNA mutation tracking to detect molecular residual disease and trigger intervention in patients with moderate- and high-risk early-stage triple-negative breast cancer. Ann Oncol. 2023;34(2):200-211.
112 J Tie, JD Cohen, Y Wang, et al. Circulating tumor DNA analyses as markers of recurrence risk and benefit of adjuvant therapy for stage III colon cancer. JAMA Oncol. 2019;5(12):1710-1717.
113 AU Rehman, P Khan, SK Maurya, et al. Liquid biopsies to occult brain metastasis. Mol Cancer. 2022;21(1):113.
114 L Sinoquet, W Jacot, L Gauthier, et al. Programmed cell death ligand 1-expressing circulating tumor cells: a new prognostic biomarker in non-small cell lung cancer. Clin Chem. 2021;67(11):1503-1512.
115 D Kong, W Zhang, Z Yang, et al. Correlation between PD-L1 expression ON CTCs and prognosis of patients with cancer: a systematic review and meta-analysis. Oncoimmunology. 2021;10(1):1938476.
116 P Winograd, S Hou, CM Court, et al. Hepatocellular carcinoma-circulating tumor cells expressing PD-L1 are prognostic and potentially associated with response to checkpoint inhibitors. Hepatol Commun. 2020;4(10):1527-1540.
117 MA Khattak, A Reid, J Freeman, et al. PD-L1 expression on circulating tumor cells may be predictive of response to pembrolizumab in advanced melanoma: results from a pilot study. Oncologist. 2020;25(3):e520-e527.
118 M Ikeda, Y Koh, S Teraoka, et al. Longitudinal evaluation of PD-L1 expression on circulating tumor cells in non-small cell lung cancer patients treated with nivolumab. Cancers (Basel). 2021;13(10):2290.
119 PJ Chiang, T Xu, TL Cha, et al. Programmed cell death ligand 1 expression in circulating tumor cells as a predictor of treatment response in patients with urothelial carcinoma. Biology (Basel). 2021;10(7):674.
120 S Bergmann, A Coym, L Ott, et al. Evaluation of PD-L1 expression on circulating tumor cells (CTCs) in patients with advanced urothelial carcinoma (UC). Oncoimmunology. 2020;9(1):1738798.
121 N Guibert, M Delaunay, A Lusque, et al. PD-L1 expression in circulating tumor cells of advanced non-small cell lung cancer patients treated with nivolumab. Lung Cancer. 2018;120:108-112.
122 W Kong, T Chen, Y Li. Diagnosis, monitoring, and prognosis of liquid biopsy in cancer immunotherapy. Methods Mol Biol. 2023;2695:127-143.
123 K Su, L Guo, K He, et al. PD-L1 expression on circulating tumor cells can be a predictive biomarker to PD-1 inhibitors combined with radiotherapy and antiangiogenic therapy in advanced hepatocellular carcinoma. Front Oncol. 2022;12:873830.
124 E Pantazaka, A Ntzifa, A Roumeliotou, et al. PD-L1/pS6 in circulating tumor cells (CTCs) during osimertinib treatment in patients with non-small cell lung cancer (NSCLC). Biomedicines. 2022;10(8):1893.
125 A Markou, E Tzanikou, E Lianidou. The potential of liquid biopsy in the management of cancer patients. Semin Cancer Biol. 2022;84:69-79.
126 C Yue, Y Jiang, P Li, et al. Dynamic change of PD-L1 expression on circulating tumor cells in advanced solid tumor patients undergoing PD-1 blockade therapy. Oncoimmunology. 2018;7(7):e1438111.
127 Z Tan, C Yue, S Ji, et al. Assessment of PD-L1 expression on circulating tumor cells for predicting clinical outcomes in patients with cancer receiving PD-1/PD-L1 blockade therapies. Oncologist. 2021;26(12):e2227-e2238.
128 JA Moran, DL Adams, MJ Edelman, et al. Monitoring PD-L1 expression on circulating tumor-associated cells in recurrent metastatic non-small-cell lung carcinoma predicts response to immunotherapy with radiation therapy. JCO Precis Oncol. 2022;6:e2200457.
129 Q Zhou, X Liu, J Li, et al. Circulating tumor cells PD-L1 expression detection and correlation of therapeutic efficacy of immune checkpoint inhibition in advanced non-small-cell lung cancer. Thorac Cancer. 2023;14(5):470-478.
130 RJ Hause, CC Pritchard, J Shendure, SJ Salipante. Classification and characterization of microsatellite instability across 18 cancer types. Nat Med. 2016;22(11):1342-1350.
131 B Duchemann, J Remon, M Naigeon, et al. Integrating circulating biomarkers in the immune checkpoint inhibitor treatment in lung cancer. Cancers (Basel). 2020;12(12):3625.
132 S Chakrabarti, L Bucheit, JS Starr, et al. Detection of microsatellite instability-high (MSI-H) by liquid biopsy predicts robust and durable response to immunotherapy in patients with pancreatic cancer. J Immunother Cancer. 2022;10(6):e004485.
133 P Barata, N Agarwal, R Nussenzveig, et al. Clinical activity of pembrolizumab in metastatic prostate cancer with microsatellite instability high (MSI-H) detected by circulating tumor DNA. J Immunother Cancer. 2020;8(2):e001065.
134 S Kamatham, F Shahjehan, PM Kasi. Circulating tumor DNA-based detection of microsatellite instability and response to immunotherapy in pancreatic cancer. Front Pharmacol. 2020;11:23.
135 OAM Saeed, SA Mann, C Luchini, et al. Evaluating mismatch repair deficiency for solid tumor immunotherapy eligibility: immunohistochemistry versus microsatellite molecular testing. Hum Pathol. 2021;115:10-18.
136 AB Silveira, FC Bidard, A Kasperek, et al. High-accuracy determination of microsatellite instability compatible with liquid biopsies. Clin Chem. 2020;66(4):606-613.
137 A Georgiadis, JN Durham, LA Keefer, et al. Noninvasive detection of microsatellite instability and high tumor mutation burden in cancer patients treated with PD-1 blockade. Clin Cancer Res. 2019;25(23):7024-7034.
138 M Del Re, F Cucchiara, E Rofi, et al. A multiparametric approach to improve the prediction of response to immunotherapy in patients with metastatic NSCLC. Cancer Immunol Immunother. 2021;70(6):1667-1678.
139 D de Miguel-Perez, A Russo, O Arrieta, et al. Extracellular vesicle PD-L1 dynamics predict durable response to immune-checkpoint inhibitors and survival in patients with non-small cell lung cancer. J Exp Clin Cancer Res. 2022;41(1):186.
140 SZ Rasihashemi, E Rezazadeh Gavgani, R Majidazar, et al. Tumor-derived exosomal PD-L1 in progression of cancer and immunotherapy. J Cell Physiol. 2022;237(3):1648-1660.
141 SM Morrissey, F Zhang, C Ding, et al. Tumor-derived exosomes drive immunosuppressive macrophages in a pre-metastatic niche through glycolytic dominant metabolic reprogramming. Cell Metab. 2021;33(10). 2040-2058.e10.
142 Y Wei, X Tang, Y Ren, et al. An RNA-RNA crosstalk network involving HMGB1 and RICTOR facilitates hepatocellular carcinoma tumorigenesis by promoting glutamine metabolism and impedes immunotherapy by PD-L1+ exosomes activity. Signal Transduct Target Ther. 2021;6(1):421.
143 K Zhou, S Guo, F Li, Q Sun, G Liang. Exosomal PD-L1: new insights into tumor immune escape mechanisms and therapeutic strategies. Front Cell Dev Biol. 2020;8:569219.
144 J Chen, Z Lin, L Liu, et al. GOLM1 exacerbates CD8(+) T cell suppression in hepatocellular carcinoma by promoting exosomal PD-L1 transport into tumor-associated macrophages. Signal Transduct Target Ther. 2021;6(1):397.
145 Kalantari Khandani N, A Ghahremanloo, SI Hashemy. Role of tumor microenvironment in the regulation of PD-L1: a novel role in resistance to cancer immunotherapy. J Cell Physiol. 2020;235(10):6496-6506.
146 L Guan, B Wu, T Li, et al. HRS phosphorylation drives immunosuppressive exosome secretion and restricts CD8(+) T-cell infiltration into tumors. Nat Commun. 2022;13(1):4078.
147 JC Lu, PF Zhang, XY Huang, et al. Amplification of spatially isolated adenosine pathway by tumor-macrophage interaction induces anti-PD1 resistance in hepatocellular carcinoma. J Hematol Oncol. 2021;14(1):200.
148 CH Lee, JH Bae, EJ Choe, et al. Macitentan improves antitumor immune responses by inhibiting the secretion of tumor-derived extracellular vesicle PD-L1. Theranostics. 2022;12(5):1971-1987.
149 DR Gandara, SM Paul, M Kowanetz, et al. Blood-based tumor mutational burden as a predictor of clinical benefit in non-small-cell lung cancer patients treated with atezolizumab. Nat Med. 2018;24(9):1441-1448.
150 BY Nabet, MS Esfahani, EJ Moding, et al. Noninvasive early identification of therapeutic benefit from immune checkpoint inhibition. Cell. 2020;183(2). 363-376.e13.
151 Y Khagi, AM Goodman, GA Daniels, et al. Hypermutated circulating tumor DNA: correlation with response to checkpoint inhibitor-based immunotherapy. Clin Cancer Res. 2017;23(19):5729-5736.
152 JC Stadler, Y Belloum, B Deitert, et al. Current and Future Clinical Applications of ctDNA in Immuno-Oncology. Cancer Res. 2022;82(3):349-358.
153 Z Wang, J Duan, S Cai, et al. Assessment of blood tumor mutational burden as a potential biomarker for immunotherapy in patients with non-small cell lung cancer with use of a next-generation sequencing cancer gene panel. JAMA Oncol. 2019;5(5):696-702.
154 R Kurzrock, C Aggarwal, C Weipert, et al. Prevalence of ARID1A mutations in cell-free circulating tumor DNA in a cohort of 71,301 patients and association with driver co-alterations. Cancers (Basel). 2022;14(17):4281.
155 DV Araujo, A Wang, D Torti, et al. Applications of circulating tumor DNA in a cohort of phase i solid tumor patients treated with immunotherapy. JNCI Cancer Spectr. 2021;5(3):pkaa122.
156 A Marabelle, M Fakih, J Lopez, et al. Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study. Lancet Oncol. 2020;21(10):1353-1365.
157 RM Samstein, CH Lee, AN Shoushtari, et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet. 2019;51(2):202-206.
158 M Poggio, T Hu, CC Pai, et al. Suppression of exosomal PD-L1 induces systemic anti-tumor immunity and memory. Cell. 2019;177(2). 414-427.e13.
159 Y Yang, CW Li, LC Chan, et al. Exosomal PD-L1 harbors active defense function to suppress T cell killing of breast cancer cells and promote tumor growth. Cell Res. 2018;28(8):862-864.
160 C Li, C Li, C Zhi, et al. Clinical significance of PD-L1 expression in serum-derived exosomes in NSCLC patients. J Transl Med. 2019;17(1):355.
161 W Yu, J Hurley, D Roberts, et al. Exosome-based liquid biopsies in cancer: opportunities and challenges. Ann Oncol. 2021;32(4):466-477.
162 D Daassi, KM Mahoney, GJ Freeman. The importance of exosomal PDL1 in tumour immune evasion. Nat Rev Immunol. 2020;20(4):209-215.
163 Y Lu, L Ye, X Jian, et al. Integrated microfluidic system for isolating exosome and analyzing protein marker PD-L1. Biosens Bioelectron. 2022;204:113879.
164 G Chen, AC Huang, W Zhang, et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature. 2018;560(7718):382-386.
165 C Zhang, Y Fan, X Che, et al. Anti-PD-1 therapy response predicted by the combination of exosomal PD-L1 and CD28. Front Oncol. 2020;10:760.
166 SM Morrissey, J Yan. Exosomal PD-L1: roles in tumor progression and immunotherapy. Trends Cancer. 2020;6(7):550-558.
167 MN Theodoraki, S Yerneni, WE Gooding, et al. Circulating exosomes measure responses to therapy in head and neck cancer patients treated with cetuximab, ipilimumab, and IMRT. Oncoimmunology. 2019;8(7):1593805.
168 M Cordonnier, C Nardin, G Chanteloup, et al. Tracking the evolution of circulating exosomal-PD-L1 to monitor melanoma patients. J Extracell Vesicles. 2020;9(1):1710899.
169 M Del Re, R Marconcini, G Pasquini, et al. PD-L1 mRNA expression in plasma-derived exosomes is associated with response to anti-PD-1 antibodies in melanoma and NSCLC. Br J Cancer. 2018;118(6):820-824.
170 C Zhang, X Chong, F Jiang, et al. Plasma extracellular vesicle derived protein profile predicting and monitoring immunotherapeutic outcomes of gastric cancer. J Extracell Vesicles. 2022;11(4):e12209.
171 J Sun, R Shi, X Zhang, et al. Characterization of immune landscape in papillary thyroid cancer reveals distinct tumor immunogenicity and implications for immunotherapy. Oncoimmunology. 2021;10(1):e1964189.
172 XX Peng, R Yu, X Wu, et al. Correlation of plasma exosomal microRNAs with the efficacy of immunotherapy in EGFR /ALK wild-type advanced non-small cell lung cancer. J Immunother Cancer. 2020;8(1):e000376.
173 SW Chen, SQ Zhu, X Pei, et al. Cancer cell-derived exosomal circUSP7 induces CD8(+) T cell dysfunction and anti-PD1 resistance by regulating the miR-934/SHP2 axis in NSCLC. Mol Cancer. 2021;20(1):144.
174 J Wu, D Zeng, S Zhi, et al. Single-cell analysis of a tumor-derived exosome signature correlates with prognosis and immunotherapy response. J Transl Med. 2021;19(1):381.
175 G Siravegna, S Marsoni, S Siena, A Bardelli. Integrating liquid biopsies into the management of cancer. Nat Rev Clin Oncol. 2017;14(9):531-548.
176 P Mondelo-Macia, J Garcia-Gonzalez, L Leon-Mateos, et al. Clinical potential of circulating free DNA and circulating tumour cells in patients with metastatic non-small-cell lung cancer treated with pembrolizumab. Mol Oncol. 2021;15(11):2923-2940.
177 MJM Magbanua, LB Swigart, HT Wu, et al. Circulating tumor DNA in neoadjuvant-treated breast cancer reflects response and survival. Ann Oncol. 2021;32(2):229-239.
178 I Garcia-Murillas, N Chopra, I Comino-Mendez, et al. Assessment of molecular relapse detection in early-stage breast cancer. JAMA Oncol. 2019;5(10):1473-1478.
179 PM Kasi, G Budde, M Krainock, et al. Circulating tumor DNA (ctDNA) serial analysis during progression on PD-1 blockade and later CTLA-4 rescue in patients with mismatch repair deficient metastatic colorectal cancer. J Immunother Cancer. 2022;10(1):e003312.
180 MD Hellmann, BY Nabet, H Rizvi, et al. Circulating tumor DNA analysis to assess risk of progression after long-term response to PD-(L)1 blockade in NSCLC. Clin Cancer Res. 2020;26(12):2849-2858.
181 T Powles, ZJ Assaf, N Davarpanah, et al. ctDNA guiding adjuvant immunotherapy in urothelial carcinoma. Nature. 2021;595(7867):432-437.
182 JH Lee, GV Long, AM Menzies, et al. Association between circulating tumor DNA and pseudoprogression in patients with metastatic melanoma treated with anti-programmed cell death 1 antibodies. JAMA Oncol. 2018;4(5):717-721.
183 E Takai, W Omata, Y Totoki, et al. Clonal dynamics of circulating tumor DNA during immune checkpoint blockade therapy for melanoma. Cancer Sci. 2021;112(11):4748-4757.
184 SN Lone, S Nisar, T Masoodi, et al. Liquid biopsy: a step closer to transform diagnosis, prognosis and future of cancer treatments. Mol Cancer. 2022;21(1):79.
185 G Torga, KJ Pienta. Patient-paired sample congruence between 2 commercial liquid biopsy tests. JAMA Oncol. 2018;4(6):868-870.
186 AE Revelo, A Martin, R Velasquez, et al. Liquid biopsy for lung cancers: an update on recent developments. Ann Transl Med. 2019;7(15):349.
187 D Chen, T Xu, S Wang, et al. Liquid biopsy applications in the clinic. Mol Diagn Ther. 2020;24(2):125-132.
188 L Raimondi, A De Luca, V Costa, et al. Circulating biomarkers in osteosarcoma: new translational tools for diagnosis and treatment. Oncotarget. 2017;8(59):100831-100851.
189 X Liu, J Yao, L Song, S Zhang, T Huang, Y Li. Local and abscopal responses in advanced intrahepatic cholangiocarcinoma with low TMB, MSS, pMMR and negative PD-L1 expression following combined therapy of SBRT with PD-1 blockade. J Immunother Cancer. 2019;7(1):204.
190 J Kong, D Ha, J Lee, et al. Network-based machine learning approach to predict immunotherapy response in cancer patients. Nat Commun. 2022;13(1):3703.
191 Z Lu, H Chen, X Jiao, et al. Prediction of immune checkpoint inhibition with immune oncology-related gene expression in gastrointestinal cancer using a machine learning classifier. J Immunother Cancer. 2020;8(2):e000631.
192 M Shehab, L Abualigah, Q Shambour, et al. Machine learning in medical applications: a review of state-of-the-art methods. Comput Biol Med. 2022;145:105458.
193 D Chowell, SK Yoo, C Valero, et al. Improved prediction of immune checkpoint blockade efficacy across multiple cancer types. Nat Biotechnol. 2022;40(4):499-506.
194 F Xie, J Zhang, J Wang, et al. Multifactorial deep learning reveals pan-cancer genomic tumor clusters with distinct immunogenomic landscape and response to immunotherapy. Clin Cancer Res. 2020;26(12):2908-2920.
195 M Zhou, Z Zhang, S Bao, et al. Computational recognition of lncRNA signature of tumor-infiltrating B lymphocytes with potential implications in prognosis and immunotherapy of bladder cancer. Brief Bioinform. 2021;22(3):bbaa047.
196 N Zhang, H Zhang, W Wu, et al. Machine learning-based identification of tumor-infiltrating immune cell-associated lncRNAs for improving outcomes and immunotherapy responses in patients with low-grade glioma. Theranostics. 2022;12(13):5931-5948.
197 Z Liu, G Lin, Z Yan, et al. Predictive mutation signature of immunotherapy benefits in NSCLC based on machine learning algorithms. Front Immunol. 2022;13:989275.
198 A Zaitsev, M Chelushkin, D Dyikanov, et al. Precise reconstruction of the TME using bulk RNA-seq and a machine learning algorithm trained on artificial transcriptomes. Cancer Cell. 2022;40(8):879-894. e16.
199 Y Park, MJ Kim, Y Choi, et al. Role of mass spectrometry-based serum proteomics signatures in predicting clinical outcomes and toxicity in patients with cancer treated with immunotherapy. J Immunother Cancer. 2022;10(3):e003566.
200 SM Liu, H Sun, JY Zhou, et al. Prediction of unfavourable response to checkpoint blockade in lung cancer patients through an integrated tumour-immune expression score. Transl Oncol. 2022;15(1):101254.
201 P Rich, RB Mitchell, E Schaefer, et al. Real-world performance of blood-based proteomic profiling in first-line immunotherapy treatment in advanced stage non-small cell lung cancer. J Immunother Cancer. 2021;9(10):e002989.
202 D Connors, J Allen, JD Alvarez, et al. International liquid biopsy standardization alliance white paper. Crit Rev Oncol Hematol. 2020;156:103112.
PDF

Accesses

Citations

Detail

Sections
Recommended

/