Letter to the Editor Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Apr 21, 2024; 30(15): 2175-2178
Published online Apr 21, 2024. doi: 10.3748/wjg.v30.i15.2175
Circulating tumor DNA in liquid biopsy: Current diagnostic limitation
Shi-Cai Liu, School of Medical Information, Wannan Medical College, Wuhu 241002, Anhui Province, China
ORCID number: Shi-Cai Liu (0000-0003-1270-4729).
Author contributions: Liu SC conceived the theme, and prepared the manuscript. All authors have read and approved the version to be published.
Supported by Talent Scientific Research Start-up Foundation of Wannan Medical College, No. WYRCQD2023045.
Conflict-of-interest statement: The authors declare no competing interests.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Shi-Cai Liu, PhD, Academic Research, School of Medical Information, Wannan Medical College, No. 22 Wenchang West Road, Wuhu 241002, Anhui Province, China. liushicainj@163.com
Received: February 2, 2024
Peer-review started: February 2, 2024
First decision: February 29, 2024
Revised: March 7, 2024
Accepted: April 2, 2024
Article in press: April 2, 2024
Published online: April 21, 2024

Abstract

With the rapid development of science and technology, cell-free DNA (cfDNA) is rapidly becoming an important biomarker for tumor diagnosis, monitoring and prognosis, and this cfDNA-based liquid biopsy technology has great potential to become an important part of precision medicine. cfDNA is the total amount of free DNA in the systemic circulation, including DNA fragments derived from tumor cells and all other somatic cells. Tumor cells release fragments of DNA into the bloodstream, and this source of cfDNA is called circulating tumor DNA (ctDNA). cfDNA detection has become a major focus in the field of tumor research in recent years, which provides a new opportunity for non-invasive diagnosis and prognosis of cancer. In this paper, we discuss the limitations of the study on the origin and dynamics analysis of ctDNA, and how to solve these problems in the future. Although the future faces major challenges, it also contains great potential.

Key Words: Cell-free DNA, Circulating tumor DNA, Liquid biopsy, Cancer, Diagnosis, Prognosis

Core Tip: Tumor liquid biopsy based on cell-free DNA detection has become a major hotspot in the field of tumor research in recent years. Circulating tumor DNA (ctDNA) is a DNA fragment that breaks down from cells in primary tumors or even new tumors formed by metastasis, and enters the peripheral circulation. ctDNA analysis provides a non-invasive method for cancer detection and monitoring, which is important for the management of clinical patients.



TO THE EDITOR

We read with interest a basic study by Terasawa et al[1], who assessed the origin of circulating tumor DNA (ctDNA), elucidated the dynamics of ctDNA levels, assessed ctDNA levels using xenografted mice after treatment, and determined whether tumor size and tumor invasion correlated with ctDNA levels. ctDNA is the cell-free DNA (cfDNA) of tumor origin. We congratulate the authors on their work and contributions in this field. At present, it is not clear what factors (e.g., tumor size and tumor invasion) affect the levels of ctDNA, and there are always questions about the origin of ctDNA. It is crucial to address these issues in order to make ctDNA an effective and practical biomarker for liquid biopsy in clinical practice, and to fully tap into its potential. In this regard, Terasawa et al[1] explored the origin and dynamics of ctDNA with potential contributions to this issue.

Using BALB/c-nu/nu mice inoculated with the TE11 cell line, the authors established tumor xenotransplantation. In order to perform ctDNA analysis, several groups of mice were killed at appropriate time points after xenotransplantation. The findings shed light on the origin and dynamics of ctDNA, suggesting that tumor size is an important factor. Moreover, the results of this study showed that when the tumor was completely removed, the ctDNA disappeared after ≥ 1 d. Although the results were satisfactory, in future studies, researchers need to further understand the biological characteristics of ctDNA (such as ctDNA release and clearance mechanisms) and improve the sensitivity of ctDNA detection. The authors also pointed out in their discussion that, with respect to residual tumors, although all mice underwent pathological autopsies in this study, it was not possible to completely identify residual tumors, which is essentially a sensitivity issue of ctDNA detection.

In the early stage of cancer, the content of ctDNA in plasma is low, and the number of somatic mutations based on a certain mutation is even lower[2]. The detection and analysis of ctDNA brings many obstacles to clinical cancer detection, especially early detection. Previous research has shown that only when the ctDNA content in cfDNA is ≥ 10%, accurate information of tumor can be obtained[3]. However, with the exception of some patients with advanced tumors who have high amounts of ctDNA in plasma, the ctDNA content in most patients with tumors does not meet this standard[4]. This makes detecting ctDNA difficult, especially in the early stages of cancer. At present, increasing the depth of sequencing is mainly used to improve the sensitivity and accuracy of ctDNA detection, but increasing the depth of sequencing may bring false positive results, because cfDNA of non-tumor origin may also carry various tumor-associated mutations[5]. These problems have always limited the clinical application of ctDNA liquid biopsy.

Due to these shortcomings, in addition to detecting tumor mutations, the field of liquid cancer biopsy is actively exploring non-invasive detection methods based on other characteristics of plasma cfDNA, such as fragment size[6,7], methylation[8-11], end coordinates[12], and chromatin accessibility of cfDNA[13], which are currently being studied. Renaud et al[14] used fragment length characteristics of cfDNA to diagnose metastatic castration-resistant prostate cancer. Heeke et al[15] used cfDNA methylation data to classify small cell lung cancer (SCLC) and discovered SCLC subgroups based on DNA methylation data, which can serve as potential biomarkers to guide patient classification and clinical precision treatment. The United States Food and Drug Administration has approved the first blood-based colorectal cancer (CRC) screening product, SEPT9 gene testing[16,17]. The study by Jiang et al[12] showed that cancer-related end coordinates in plasma cfDNA can be applied to liver cancer early detection. Using plasma cfDNA and protein markers, Cohen et al[18] developed CancerSEEK, which can detect eight common cancers of the lung, colorectum, liver, stomach, esophagus, pancreas, breast, or ovary. In my previous studies, cfDNA chromatin open state was used to distinguish patients with esophageal cancer from individuals without cancer[13]. Li et al[19] developed a multimodal epigenetic sequencing analysis (MESA) method based on cfDNA, MESA, for the detection of CRC, which can capture and integrate various epigenetic features in cfDNA, such as cfDNA methylation and nucleosome occupancy. These studies open up new ideas for cfDNA-based liquid biopsy in non-invasive diagnosis. Many studies have shown that the total level of cfDNA is correlated with tumor staging[20,21], suggesting that cfDNA has prognostic potential. In addition, cfDNA can serve as a real-time indicator of therapeutic efficacy, allowing for earlier observation of therapeutic effects than clinical trials, thanks to its short half-life[22-24].

At present, research on improving the sensitivity of ctDNA detection mainly focuses on in vitro sequencing and analysis, such as detecting multi somatic mutations and integrating DNA methylation or fragmentation patterns[25,26]. An inherent challenge faced by all these methods is the low amount of ctDNA in the collected blood samples, which limits sensitivity. Increasing the volume of the blood sample can improve sensitivity of the detection. However, for patients who are weak or ill, it is impractical. In addition, some have proposed methods that are closer to tumor sampling or increase tumor DNA loss. These methods also have certain limitations, such as requiring prior knowledge of tumor location, being limited to specific primary tumors, requiring invasive surgery, and being costly. Recently, a research team has developed two intravenous inducers to improve the recovery of ctDNA in blood collection, which can temporarily delay the clearance of cfDNA in the body[27]. Uptake by liver-resident macrophages and degradation by circulating nucleases are two natural mechanisms by which cfDNA is cleared. In this study, the authors developed two intravenous inducers that can act on these mechanisms and improve the recovery rate of ctDNA when used 1-2 h before blood withdrawal. Although this strategy can significantly improve the sensitivity of ctDNA detection in preclinical models, the safety and tolerability of the formulation, as well as whether this effect can be translated into human patients, remain to be determined.

With the rapid development of science and technology, cfDNA is rapidly becoming an important biomarker for tumor diagnosis, monitoring and prognosis, and this cfDNA-based liquid biopsy technology has great potential to become an important tool of precision medicine. Despite its enormous potential, there is still a long way to go. Research on blood sample collection, cfDNA isolation, and Next-Generation Sequencing data analysis is currently limited and needs to be focused on in the future. In addition, the biological characteristics of ctDNA are also of great research value. Meanwhile, it is necessary to confirm the effectiveness and practicability of cfDNA as a diagnosis/prognosis marker to further promote the clinical application of cfDNA-based liquid biopsy.

Footnotes

Provenance and peer review: Unsolicited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country/Territory of origin: China

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): 0

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Huang CT, Taiwan S-Editor: Qu XL L-Editor: A P-Editor: Cai YX

References
1.  Terasawa H, Kinugasa H, Nouso K, Yamamoto S, Hirai M, Tanaka T, Takaki A, Okada H. Circulating tumor DNA dynamics analysis in a xenograft mouse model with esophageal squamous cell carcinoma. World J Gastroenterol. 2021;27:7134-7143.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
2.  Siravegna G, Marsoni S, Siena S, Bardelli A. Integrating liquid biopsies into the management of cancer. Nat Rev Clin Oncol. 2017;14:531-548.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1017]  [Cited by in F6Publishing: 1163]  [Article Influence: 166.1]  [Reference Citation Analysis (0)]
3.  Murtaza M, Dawson SJ, Tsui DW, Gale D, Forshew T, Piskorz AM, Parkinson C, Chin SF, Kingsbury Z, Wong AS, Marass F, Humphray S, Hadfield J, Bentley D, Chin TM, Brenton JD, Caldas C, Rosenfeld N. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature. 2013;497:108-112.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1288]  [Cited by in F6Publishing: 1252]  [Article Influence: 113.8]  [Reference Citation Analysis (0)]
4.  Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y, Agrawal N, Bartlett BR, Wang H, Luber B, Alani RM, Antonarakis ES, Azad NS, Bardelli A, Brem H, Cameron JL, Lee CC, Fecher LA, Gallia GL, Gibbs P, Le D, Giuntoli RL, Goggins M, Hogarty MD, Holdhoff M, Hong SM, Jiao Y, Juhl HH, Kim JJ, Siravegna G, Laheru DA, Lauricella C, Lim M, Lipson EJ, Marie SK, Netto GJ, Oliner KS, Olivi A, Olsson L, Riggins GJ, Sartore-Bianchi A, Schmidt K, Shih lM, Oba-Shinjo SM, Siena S, Theodorescu D, Tie J, Harkins TT, Veronese S, Wang TL, Weingart JD, Wolfgang CL, Wood LD, Xing D, Hruban RH, Wu J, Allen PJ, Schmidt CM, Choti MA, Velculescu VE, Kinzler KW, Vogelstein B, Papadopoulos N, Diaz LA Jr. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med. 2014;6:224ra24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2770]  [Cited by in F6Publishing: 3117]  [Article Influence: 311.7]  [Reference Citation Analysis (0)]
5.  Aravanis AM, Lee M, Klausner RD. Next-Generation Sequencing of Circulating Tumor DNA for Early Cancer Detection. Cell. 2017;168:571-574.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 232]  [Cited by in F6Publishing: 236]  [Article Influence: 33.7]  [Reference Citation Analysis (0)]
6.  Mouliere F, Chandrananda D, Piskorz AM, Moore EK, Morris J, Ahlborn LB, Mair R, Goranova T, Marass F, Heider K, Wan JCM, Supernat A, Hudecova I, Gounaris I, Ros S, Jimenez-Linan M, Garcia-Corbacho J, Patel K, Østrup O, Murphy S, Eldridge MD, Gale D, Stewart GD, Burge J, Cooper WN, van der Heijden MS, Massie CE, Watts C, Corrie P, Pacey S, Brindle KM, Baird RD, Mau-Sørensen M, Parkinson CA, Smith CG, Brenton JD, Rosenfeld N. Enhanced detection of circulating tumor DNA by fragment size analysis. Sci Transl Med. 2018;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 622]  [Cited by in F6Publishing: 555]  [Article Influence: 92.5]  [Reference Citation Analysis (0)]
7.  Underhill HR, Kitzman JO, Hellwig S, Welker NC, Daza R, Baker DN, Gligorich KM, Rostomily RC, Bronner MP, Shendure J. Fragment Length of Circulating Tumor DNA. PLoS Genet. 2016;12:e1006162.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 374]  [Cited by in F6Publishing: 424]  [Article Influence: 53.0]  [Reference Citation Analysis (0)]
8.  Gao Q, Lin YP, Li BS, Wang GQ, Dong LQ, Shen BY, Lou WH, Wu WC, Ge D, Zhu QL, Xu Y, Xu JM, Chang WJ, Lan P, Zhou PH, He MJ, Qiao GB, Chuai SK, Zang RY, Shi TY, Tan LJ, Yin J, Zeng Q, Su XF, Wang ZD, Zhao XQ, Nian WQ, Zhang S, Zhou J, Cai SL, Zhang ZH, Fan J. Unintrusive multi-cancer detection by circulating cell-free DNA methylation sequencing (THUNDER): development and independent validation studies. Ann Oncol. 2023;34:486-495.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 10]  [Reference Citation Analysis (0)]
9.  Zhou X, Cheng Z, Dong M, Liu Q, Yang W, Liu M, Tian J, Cheng W. Tumor fractions deciphered from circulating cell-free DNA methylation for cancer early diagnosis. Nat Commun. 2022;13:7694.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 8]  [Reference Citation Analysis (0)]
10.  Luo H, Wei W, Ye Z, Zheng J, Xu RH. Liquid Biopsy of Methylation Biomarkers in Cell-Free DNA. Trends Mol Med. 2021;27:482-500.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 111]  [Article Influence: 37.0]  [Reference Citation Analysis (0)]
11.  Luo H, Zhao Q, Wei W, Zheng L, Yi S, Li G, Wang W, Sheng H, Pu H, Mo H, Zuo Z, Liu Z, Li C, Xie C, Zeng Z, Li W, Hao X, Liu Y, Cao S, Liu W, Gibson S, Zhang K, Xu G, Xu RH. Circulating tumor DNA methylation profiles enable early diagnosis, prognosis prediction, and screening for colorectal cancer. Sci Transl Med. 2020;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 186]  [Cited by in F6Publishing: 203]  [Article Influence: 50.8]  [Reference Citation Analysis (0)]
12.  Jiang P, Sun K, Tong YK, Cheng SH, Cheng THT, Heung MMS, Wong J, Wong VWS, Chan HLY, Chan KCA, Lo YMD, Chiu RWK. Preferred end coordinates and somatic variants as signatures of circulating tumor DNA associated with hepatocellular carcinoma. Proc Natl Acad Sci U S A. 2018;115:E10925-E10933.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 112]  [Article Influence: 18.7]  [Reference Citation Analysis (0)]
13.  Liu S, Wu J, Xia Q, Liu H, Li W, Xia X, Wang J. Finding new cancer epigenetic and genetic biomarkers from cell-free DNA by combining SALP-seq and machine learning. Comput Struct Biotechnol J. 2020;18:1891-1903.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 10]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
14.  Renaud G, Nørgaard M, Lindberg J, Grönberg H, De Laere B, Jensen JB, Borre M, Andersen CL, Sørensen KD, Maretty L, Besenbacher S. Unsupervised detection of fragment length signatures of circulating tumor DNA using non-negative matrix factorization. Elife. 2022;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
15.  Heeke S, Gay CM, Estecio MR, Tran H, Morris BB, Zhang B, Tang X, Raso MG, Rocha P, Lai S, Arriola E, Hofman P, Hofman V, Kopparapu P, Lovly CM, Concannon K, De Sousa LG, Lewis WE, Kondo K, Hu X, Tanimoto A, Vokes NI, Nilsson MB, Stewart A, Jansen M, Horváth I, Gaga M, Panagoulias V, Raviv Y, Frumkin D, Wasserstrom A, Shuali A, Schnabel CA, Xi Y, Diao L, Wang Q, Zhang J, Van Loo P, Wang J, Wistuba II, Byers LA, Heymach JV. Tumor- and circulating-free DNA methylation identifies clinically relevant small cell lung cancer subtypes. Cancer Cell. 2024;42:225-237.e5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
16.  Song L, Li Y. SEPT9: A Specific Circulating Biomarker for Colorectal Cancer. Adv Clin Chem. 2015;72:171-204.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 54]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
17.  Church TR, Wandell M, Lofton-Day C, Mongin SJ, Burger M, Payne SR, Castaños-Vélez E, Blumenstein BA, Rösch T, Osborn N, Snover D, Day RW, Ransohoff DF; PRESEPT Clinical Study Steering Committee, Investigators and Study Team. Prospective evaluation of methylated SEPT9 in plasma for detection of asymptomatic colorectal cancer. Gut. 2014;63:317-325.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 486]  [Cited by in F6Publishing: 509]  [Article Influence: 50.9]  [Reference Citation Analysis (0)]
18.  Cohen JD, Li L, Wang Y, Thoburn C, Afsari B, Danilova L, Douville C, Javed AA, Wong F, Mattox A, Hruban RH, Wolfgang CL, Goggins MG, Dal Molin M, Wang TL, Roden R, Klein AP, Ptak J, Dobbyn L, Schaefer J, Silliman N, Popoli M, Vogelstein JT, Browne JD, Schoen RE, Brand RE, Tie J, Gibbs P, Wong HL, Mansfield AS, Jen J, Hanash SM, Falconi M, Allen PJ, Zhou S, Bettegowda C, Diaz LA Jr, Tomasetti C, Kinzler KW, Vogelstein B, Lennon AM, Papadopoulos N. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science. 2018;359:926-930.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1752]  [Cited by in F6Publishing: 1552]  [Article Influence: 258.7]  [Reference Citation Analysis (0)]
19.  Li Y, Xu J, Chen C, Lu Z, Wan D, Li D, Li JS, Sorg AJ, Roberts CC, Mahajan S, Gallant MA, Pinkoviezky I, Cui Y, Taggart DJ, Li W. Multimodal epigenetic sequencing analysis (MESA) of cell-free DNA for non-invasive colorectal cancer detection. Genome Med. 2024;16:9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Reference Citation Analysis (0)]
20.  Picardo F, Romanelli A, Muinelo-Romay L, Mazza T, Fusilli C, Parrella P, Barbazán J, Lopez-López R, Barbano R, De Robertis M, Taffon C, Bordoni V, Agrati C, Costantini M, Ricci F, Graziano P, Maiello E, Muscarella LA, Fazio VM, Poeta ML. Diagnostic and Prognostic Value of B4GALT1 Hypermethylation and Its Clinical Significance as a Novel Circulating Cell-Free DNA Biomarker in Colorectal Cancer. Cancers (Basel). 2019;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 26]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
21.  Fu R, Huang J, Tian X, Liang C, Xiong Y, Zhang JT, Jiang B, Dong S, Gong Y, Gao W, Li F, Shi Y, Liu Z, Gao X, Chen R, Zhong W, Zhang Y. Postoperative circulating tumor DNA can refine risk stratification in resectable lung cancer: results from a multicenter study. Mol Oncol. 2023;17:825-838.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
22.  Stejskal P, Goodarzi H, Srovnal J, Hajdúch M, van 't Veer LJ, Magbanua MJM. Circulating tumor nucleic acids: biology, release mechanisms, and clinical relevance. Mol Cancer. 2023;22:15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 29]  [Article Influence: 29.0]  [Reference Citation Analysis (0)]
23.  Ko JMY, Ng HY, Lam KO, Chiu KWH, Kwong DLW, Lo AWI, Wong JC, Lin RCW, Fong HCH, Li JYK, Dai W, Law S, Lung ML. Liquid Biopsy Serial Monitoring of Treatment Responses and Relapse in Advanced Esophageal Squamous Cell Carcinoma. Cancers (Basel). 2020;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
24.  Boonstra PA, Wind TT, van Kruchten M, Schuuring E, Hospers GAP, van der Wekken AJ, de Groot DJ, Schröder CP, Fehrmann RSN, Reyners AKL. Clinical utility of circulating tumor DNA as a response and follow-up marker in cancer therapy. Cancer Metastasis Rev. 2020;39:999-1013.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 30]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
25.  Liu S, Wang J. Current and Future Perspectives of Cell-Free DNA in Liquid Biopsy. Curr Issues Mol Biol. 2022;44:2695-2709.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
26.  Roviello G, Lavacchi D, Antonuzzo L, Catalano M, Mini E. Liquid biopsy in colorectal cancer: No longer young, but not yet old. World J Gastroenterol. 2022;28:1503-1507.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
27.  Martin-Alonso C, Tabrizi S, Xiong K, Blewett T, Sridhar S, Crnjac A, Patel S, An Z, Bekdemir A, Shea D, Wang ST, Rodriguez-Aponte S, Naranjo CA, Rhoades J, Kirkpatrick JD, Fleming HE, Amini AP, Golub TR, Love JC, Bhatia SN, Adalsteinsson VA. Priming agents transiently reduce the clearance of cell-free DNA to improve liquid biopsies. Science. 2024;383:eadf2341.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]