Cell-free DNA-associated multi-feature applications in cancer diagnosis and treatment

Xiaolu Zhang, Jingwei Li, Xun Lan, Jie Li

PDF
Clinical and Translational Discovery ›› 2024, Vol. 4 ›› Issue (2) : e280. DOI: 10.1002/ctd2.280
REVIEW ARTICLE

Cell-free DNA-associated multi-feature applications in cancer diagnosis and treatment

Author information +
History +

Abstract

Malignant tumours pose significant challenges in terms of high morbidity and mortality rates, primarily due to the lack of large-scale applicable screening methods and efficient treatment strategies. However, the development of liquid biopsies, particularly circulating cell-free DNA (cfDNA), offers promising solutions characterised by their non-invasiveness and cost-effectiveness, providing comprehensive tumour information on a global scale. The release of cfDNA is predominantly associated with cell death and turnover, while its elimination occurs through nuclease digestion, renal excretion into the urine and uptake by the liver and spleen. Extensive research into the biological properties of cfDNA has led to the identification of novel applications, including non-invasive cancer screening, cancer subtype classification, tissue-of-origin detection and monitoring of treatment efficacy. Additionally, emerging fields such as methylation-omics, fragment-omics and nucleosome-omics show immense potential as tissue-and disease-specific markers. Therefore, this review aims to comprehensively introduce the latest detection techniques of cfDNA, along with detailed information on its characteristics and applications, providing valuable insights for cancer diagnosis and monitoring, which will assist us in purposefully enhancing relevant features for a more comprehensive application in clinical practice.

Keywords

cancer diagnosis / cancer treatment / cfDNA

Cite this article

Download citation ▾
Xiaolu Zhang, Jingwei Li, Xun Lan, Jie Li. Cell-free DNA-associated multi-feature applications in cancer diagnosis and treatment. Clinical and Translational Discovery, 2024, 4(2): e280 https://doi.org/10.1002/ctd2.280

References

[1]
SungH, FerlayJ, SiegelRL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209-249.
[2]
Arzuaga-MendezJ, Prieto-Fernández E, Lopez-LopezE, Martin-GuerreroI, García-Ruiz JC, García-OradA. Cell-free DNA as a biomarker in diffuse large B-cell lymphoma: a systematic review. Crit Rev Oncol Hematol. 2019;139:7-15.
[3]
MurtazaM, DawsonS-J, TsuiDWY, et al. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature. 2013;497(7447):108-112.
[4]
CristianoS, LealA, PhallenJ, et al. Genome-wide cell-free DNA fragmentation in patients with cancer. Nature. 2019;570(7761):385-389.
[5]
HeitzerE, HaqueIS, RobertsCES, Speicher MR. Current and future perspectives of liquid biopsies in genomics-driven oncology. Nat Rev Genet. 2019;20(2):71-88.
[6]
ZhuX, ChuC-J, PanW, LiY, HuangH, Zhao L. The correlation between periodontal parameters and cell-free DNA in the gingival crevicular fluid, saliva, and plasma in Chinese patients: a cross-sectional study. J Clin Med. 2022;11(23):6902.
[7]
IsolaG, Polizzi A, MascittiM, et al. Impact of periodontitis on circulating cell-free DNA levels as a measure of cardiovascular disease. Clin Oral Investig. 2023;27(11):6855-6863.
[8]
LuccheseA, Giovanni M, MaurizioM, ClaudioC. Reliability and efficacy of palifermin in prevention and management of oral mucositis in patients with acute lymphoblastic leukemia: a randomized, double-blind controlled clinical trial. Minerva Stomatol. 2016;65(1):43-53.
[9]
LenaertsL, Vandenberghe P, BrisonN, et al. Genomewide copy number alteration screening of circulating plasma DNA: potential for the detection of incipient tumors. Ann Oncol. 2019;30(1):85-95.
[10]
PantelK, Alix-Panabières C. Circulating tumour cells in cancer patients: challenges and perspectives. Trends Mol Med. 2010;16(9):398-406.
[11]
SchwarzenbachH, HoonDSB, PantelK. Cell-free nucleic acids as biomarkers in cancer patients. Nat Rev Cancer. 2011;11(6):426-437.
[12]
KalluriR, LeBleuVS. The biology, function, and biomedical applications of exosomes. Science. 2020;367(6478):eaau6977.
[13]
AnfossiS, Babayan A, PantelK, CalinGA. Clinical utility of circulating non-coding RNAs—an update. Nat Rev Clin Oncol. 2018;15(9):541-563.
[14]
HeitzerE, UlzP, GeiglJB. Circulating tumor DNA as a liquid biopsy for cancer. Clin Chem. 2015;61(1):112-123.
[15]
Santiago-WalkerA, Gagnon R, MazumdarJ, et al. Correlation of BRAF mutation status in circulating-free DNA and tumor and association with clinical outcome across four BRAFi and MEKi clinical trials. Clin Cancer Res. 2016;22(3):567-574.
[16]
Del ReM, Vivaldi C, RofiE, et al. Early changes in plasma DNA levels of mutant KRAS as a sensitive marker of response to chemotherapy in pancreatic cancer. Sci Rep. 2017;7(1):7931.
[17]
KrugerS, Heinemann V, RossC, et al. Repeated mutKRAS ctDNA measurements represent a novel and promising tool for early response prediction and therapy monitoring in advanced pancreatic cancer. Ann Oncol. 2018;29(12):2348-2355.
[18]
ShenSY, Singhania R, FehringerG, et al. Sensitive tumour detection and classification using plasma cell-free DNA methylomes. Nature. 2018;563(7732):579-583.
[19]
RussoM, MisaleS, WeiG, et al. Acquired resistance to the TRK inhibitor entrectinib in colorectal cancer. Cancer Discov. 2016;6(1):36-44.
[20]
WangD, LiuZ, LuY, et al. Liquid biopsies to track trastuzumab resistance in metastatic HER2-positive gastric cancer. Gut. 2019;68(7):1152-1161.
[21]
BullmanS, Pedamallu CS, SicinskaE, et al. Analysis of Fusobacterium persistence and antibiotic response in colorectal cancer. Science. 2017;358(6369):1443-1448.
[22]
DejeaCM, FathiP, CraigJM, et al. Patients with familial adenomatous polyposis harbor colonic biofilms containing tumorigenic bacteria. Science. 2018;359(6375):592-597.
[23]
GellerLT, Barzily-Rokni M, DaninoT, et al. Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine. Science. 2017;357(6356):1156-1160.
[24]
BuscarletM, Provost S, ZadaYF, et al. DNMT3A and TET2 dominate clonal hematopoiesis and demonstrate benign phenotypes and different genetic predispositions. Blood. 2017;130(6):753-762.
[25]
ShepherdMS, LiJ, WilsonNK, et al. Single-cell approaches identify the molecular network driving malignant hematopoietic stem cell self-renewal. Blood. 2018;132(8):791-803.
[26]
Acuna-HidalgoR, SengulH, SteehouwerM, et al. Ultra-sensitive sequencing identifies high prevalence of clonal hematopoiesis-associated mutations throughout adult life. Am Hum Genet. 2017;101(1):50-64.
[27]
CoombsCC, ZehirA, DevlinSM, et al. Therapy-related clonal hematopoiesis in patients with non-hematologic cancers is common and associated with adverse clinical outcomes. Cell Stem Cell. 2017;21(3):374-382.e4.
[28]
HuY, UlrichBC, SuppleeJ, et al. False-positive plasma genotyping due to clonal hematopoiesis. Clin Cancer Res. 2018;24(18):4437-4443.
[29]
NagaseR, InoueD, PastoreA, et al. Expression of mutant Asxl1 perturbs hematopoiesis and promotes susceptibility to leukemic transformation. J Exp Med. 2018;215(6):1729-1747.
[30]
PtashkinRN, Mandelker DL, CoombsCC, et al. Prevalence of clonal hematopoiesis mutations in tumor-only clinical genomic profiling of solid tumors. JAMA Oncol. 2018;4(11):1589.
[31]
RazaviP, LiBT, BrownDN, et al. High-intensity sequencing reveals the sources of plasma circulating cell-free DNA variants. Nat Med. 2019;25(12):1928-1937.
[32]
KrugAK, Enderle D, KarlovichC, et al. Improved EGFR mutation detection using combined exosomal RNA and circulating tumor DNA in NSCLC patient plasma. Ann Oncol. 2018;29(3):700-706.
[33]
MolpariaB, Nichani E, TorkamaniA. Assessment of circulating copy number variant detection for cancer screening. PLoS One. 2017;12(7):e0180647.
[34]
ChenX, ChangC-W, SpoerkeJM, et al. Low-pass whole-genome sequencing of circulating cell-free DNA demonstrates dynamic changes in genomic copy number in a squamous lung cancer clinical cohort. Clin Cancer Res. 2019;25(7):2254-2263.
[35]
XuJ-F, KangQ, MaX-Y, et al. A novel method to detect early colorectal cancer based on chromosome copy number variation in plasma. Cell Physiol Biochem. 2018;45(4):1444-1454.
[36]
ValpioneS, Galvani E, TweedyJ, et al. Immune awakening revealed by peripheral T cell dynamics after one cycle of immunotherapy. Nat Cancer. 2020;1(2):210-221.
[37]
AnagnostouV, FordePM, WhiteJR, et al. Dynamics of tumor and immune responses during immune checkpoint blockade in non-small cell lung cancer. Cancer Res. 2019;79(6):1214-1225.
[38]
GoldbergSB, Narayan A, KoleAJ, et al. Early assessment of lung cancer immunotherapy response via circulating tumor DNA. Clin Cancer Res. 2018;24(8):1872-1880.
[39]
HuangAC, PostowMA, OrlowskiRJ, et al. T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature. 2017;545(7652):60-65.
[40]
KriegC, Nowicka M, GugliettaS, et al. High-dimensional single-cell analysis predicts response to anti-PD-1 immunotherapy. Nat Med. 2018;24(2):144-153.
[41]
HuangAC, Orlowski RJ, XuX, et al. A single dose of neoadjuvant PD-1 blockade predicts clinical outcomes in resectable melanoma. Nat Med. 2019;25(3):454-461.
[42]
ThapaDR, Tonikian R, SunC, et al. Longitudinal analysis of peripheral blood T cell receptor diversity in patients with systemic lupus erythematosus by next-generation sequencing. Arthritis Res Ther. 2015;17(1):132.
[43]
GopalakrishnanV, Spencer CN, NeziL, et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science. 2018;359(6371):97-103.
[44]
JinC, Lagoudas GK, ZhaoC, et al. Commensal microbiota promote lung cancer development via γδ T cells. Cell. 2019;176(5):998-1013.e16.
[45]
MaC, HanM, HeinrichB, et al. Gut microbiome-mediated bile acid metabolism regulates liver cancer via NKT cells. Science. 2018;360(6391):eaan5931.
[46]
MatsonV, Fessler J, BaoR, et al. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science. 2018;359(6371):104-108.
[47]
MeiselM, Hinterleitner R, PacisA, et al. Microbial signals drive pre-leukaemic myeloproliferation in a Tet2-deficient host. Nature. 2018;557(7706):580-584.
[48]
PooreGD, Kopylova E, ZhuQ, et al. Microbiome analyses of blood and tissues suggest cancer diagnostic approach. Nature. 2020;579(7800):567-574.
[49]
LauJW, Lehnert E, SethiA, et al. The cancer genomics cloud: collaborative, reproducible, and democratized—a new paradigm in large-scale computational research. Cancer Res. 2017;77(21):e3-e6.
[50]
HillmannB, Al-Ghalith GA, Shields-CutlerRR. Evaluating the information content of shallow shotgun metagenomics. mSystems. 2018;3(6):e00069-18.
[51]
LawCW, ChenY, ShiW, SmythGK. voom: precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biol. 2014;15(2):R29.
[52]
MechamBH, NelsonPS, StoreyJD. Supervised normalization of microarrays. Bioinformatics. 2010;26(10):1308-1315.
[53]
RitchieME, Phipson B, WuD, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43(7):e47-e47.
[54]
GogginKP, Gonzalez-Pena V, InabaY, et al. Evaluation of plasma microbial cell-free DNA sequencing to predict bloodstream infection in pediatric patients with relapsed or refractory cancer. JAMA Oncol. 2020;6(4):552.
[55]
SerpasL, ChanRWY, JiangP, et al. Dnase1l3 deletion causes aberrations in length and end-motif frequencies in plasma DNA. Proc Natl Acad Sci U S A. 2019;116(2):641-649.
[56]
JiangP, SunK, PengW, et al. Plasma DNA end-motif profiling as a fragmentomic marker in cancer, pregnancy, and transplantation. Cancer Discov. 2020;10(5):664-673.
[57]
JiangP, XieT, DingSC, et al. Detection and characterization of jagged ends of double-stranded DNA in plasma. Genome Res. 2020;30(8):1144-1153.
[58]
HuangW. Cell-free DNA methylation profiling analysis—technologies and bioinformatics. Cancers. 2019;11(11):1741.
[59]
LiuL, ToungJM, JassowiczAF, et al. Targeted methylation sequencing of plasma cell-free DNA for cancer detection and classification. Ann Oncol. 2018;29(6):1445-1453.
[60]
LiuL, LiuH, ShaoD, et al. Development and clinical validation of a circulating tumor DNA test for the identification of clinically actionable mutations in nonsmall cell lung cancer. Genes Chromosomes Cancer. 2018;57(4):211-220.
[61]
ZhouX, ZhengH, FuH, Dillehay McKillip KL, PinneySM, LiuY. CRAG: de novo characterization of cell-free DNA fragmentation hotspots in plasma whole-genome sequencing. Genome Med. 2022;14(1):138.
[62]
RaoS, HanAL, ZukowskiA, et al. Transcription factor–nucleosome dynamics from plasma cfDNA identifies ER-driven states in breast cancer. Sci Adv. 2022;8(34):eabm4358.
[63]
DoebleyA-L, KoM, LiaoH, et al. A framework for clinical cancer subtyping from nucleosome profiling of cell-free DNA. Nat Commun. 2022;13(1):7475.
[64]
BarskiA, Cuddapah S, CuiK, et al. High-resolution profiling of histone methylations in the human genome. Cell. 2007;129(4):823-837.
[65]
SadehR, Sharkia I, FialkoffG, et al. ChIP-seq of plasma cell-free nucleosomes identifies gene expression programs of the cells of origin. Nat Biotechnol. 2021;39(5):586-598.
[66]
MossJ, Magenheim J, NeimanD, et al. Comprehensive human cell-type methylation atlas reveals origins of circulating cell-free DNA in health and disease. Nat Commun. 2018;9(1):5068.
[67]
MouliereF, Chandrananda D, PiskorzAM, et al. Enhanced detection of circulating tumor DNA by fragment size analysis. Sci Transl Med. 2018;10(466):eaat4921.
[68]
ChabonJJ, Hamilton EG, KurtzDM, et al. Integrating genomic features for non-invasive early lung cancer detection. Nature. 2020;580(7802):245-251.
[69]
LealA, Van Grieken NCT, PalsgroveDN, et al. White blood cell and cell-free DNA analyses for detection of residual disease in gastric cancer. Nat Commun. 2020;11(1):525.
[70]
SnyderMW, Kircher M, HillAJ, DazaRM, Shendure J. Cell-free DNA comprises an in vivo nucleosome footprint that informs its tissues-of-origin. Cell. 2016;164(1–2):57-68.
[71]
ScheininI, SieD, BengtssonH, et al. DNA copy number analysis of fresh and formalin-fixed specimens by shallow whole-genome sequencing with identification and exclusion of problematic regions in the genome assembly. Genome Res. 2014;24(12):2022-2032.
[72]
UlzP, Thallinger GG, AuerM, et al. Inferring expressed genes by whole-genome sequencing of plasma DNA. Nat Genet. 2016;48(10):1273-1278.
[73]
ZhuG, GuoYA, HoD, et al. Tissue-specific cell-free DNA degradation quantifies circulating tumor DNA burden. Nat Commun. 2021;12(1):2229.
[74]
BaeM, KimG, LeeT-R, et al. Integrative modeling of tumor genomes and epigenomes for enhanced cancer diagnosis by cell-free DNA. Nat Commun. 2023;14(1):2017.
[75]
LiJ, SunX, YangH, et al. Integrated analysis toolkit for dissecting whole-genome-wide features of cell-free DNA. Clin Transl Med. 2023;13(3):1212.
[76]
EsfahaniMS, Hamilton EG, MehrmohamadiM, et al. Inferring gene expression from cell-free DNA fragmentation profiles. Nat Biotechnol. 2022;40(4):585-597.
[77]
LiJ, LanX. Perspective on new cell-free DNA technologies for early cancer detection. Cancer Biol Med. 2023.
[78]
LiJ, XuM, PengJ, et al. Novel technologies in cfDNA analysis and potential utility in clinic. Chin J Cancer Res. 2021;33(6):708-718.

RIGHTS & PERMISSIONS

2024 2024 The Authors. Clinical and Translational Discovery published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.
PDF

Accesses

Citations

Detail

Sections
Recommended

/