Highlights
-
•
Liquid biopsy is a new non-invasive tool for the early detection of cancer.
-
•
Literature so far is promising for some kinds of cancer, but supported by a very low level of evidence.
-
•
New high-quality evidence is urgently needed.
Keywords: Liquid biopsy, Cancer, Accuracy, Umbrella review
Abstract
Background
Liquid biopsy (LB) is a laboratory test performed on a fluid sample aiming at analyzing molecular data derived from circulating cells and related entities, or from nucleic acids. This umbrella review aims to map and evaluate the evidence supporting the use of LB in medicine across different medical specialities and conditions.
Methods
We searched three repositories from database inception up to October 1, 2023 and we included meta-analyses of observational studies reporting data on the use of LB, compared to gold standard, and its accuracy (area under the curve, AUC).
Results
Among 726 articles initially screened, 42 systematic reviews were included. Most of the outcomes explored (202/211) were related to cancer. We found that 75/211 had an excellent accuracy (AUC >0.90), with one comparison with an AUC equal to 1, i.e., Cell-Free Human Papillomavirus DNA (cfHPV-DNA) for HPV-positive oropharyngeal squamous cell carcinoma. However, considering published meta-analyses, all the outcomes were graded as very low on the GRADE criteria, and the heterogeneity was never reported.
Discussion
The literature about LB is rapidly increasing and some promising data about precision oncology are now available. However, this umbrella review on existing meta-analyses highlighted some critical issues for providing quantitative estimations on the different roles of LB.
Introduction
Liquid biopsy (LB) is a laboratory test performed on a fluid sample (e.g., blood, urine) aiming at analyzing molecular data derived from circulating cells and related entities or directly from nucleic acids [[1], [2], [3]]. Its use has been predominantly explored in oncology and related fields [4]. Indeed, LB represents a promising tool with potential diagnostic, prognostic, and predictive values [[5], [6], [7]].
Unlike traditional tissue biopsies, that necessitate invasive procedures, LB offers a minimally invasive approach for obtaining vital information about a patient's health status through the analysis of various biomarkers present in bodily fluids, such as blood, urine, saliva, and cerebrospinal fluid [8]. This transformative technique has garnered significant attention across medical disciplines due to its versatility, accessibility, and potential to revolutionize precision oncology and personalized medicine. Importantly, LB holds significant promise in the early detection and monitoring of various diseases, particularly cancer. By detecting circulating tumor cells (CTCs), cell-free DNA (cfDNA), microRNAs(miRNAs), and other molecular markers shed by tumors into the bloodstream or other bodily fluids, LB enables clinicians to detect cancer at its earliest stages, before the onset of clinical symptoms [8]. Early detection is crucial for improving patient outcomes, as it allows for timely intervention and treatment strategies tailored to the specific molecular profile of the tumour [9]. Furthermore, LB facilitates real-time monitoring of disease progression and response to treatment, offering valuable insights into treatment efficacy and the emergence of drug resistance [10].
Of interest, LB could bring promising innovations also beyond oncology, extending its utility to various other medical specialties. In infectious diseases, for instance, it may enable the rapid detection of pathogens and monitoring of antimicrobial resistance, facilitating prompt and targeted therapeutic interventions [11]. In prenatal testing, the analysis of foetal DNA in maternal blood may offer a non-invasive alternative to traditional methods, reducing the risk of complications for both mother and foetus [12]. Additionally, LB holds promise in the field of organ transplantation, where it can aid in the early detection of graft rejection, emerging as a potential tool for guiding personalized immunosuppressive therapies [13].
The significance of LB in medicine is further underscored by its role in overcoming the limitations of conventional tissue biopsies. Unlike tissue biopsies, which may be hindered by sampling bias, accessibility issues, and tumour heterogeneity, LB offers a comprehensive and dynamic snapshot of the entire tumour landscape, allowing for more accurate molecular characterization and treatment selection [14]. However, LB may also suffer from potential issues, including lack of standardized approaches, propension to pre-analytical errors, and difficulties in the interpretation of data generated from multi-omics analysis [15].
In this complex scenario, this umbrella review (i.e., a systematic review of other systematic reviews on the same topic) aims to map and evaluate the evidence supporting the use of LB in medicine across different medical specialities and conditions, also taking into account the multiple issues potentially emerging in this setting.
Materials and methods
Protocol and registration
This umbrella review was conducted following the recommendations of the Cochrane handbook for systematic literature reviews to carry out the screening and selection of studies, and according to the Preferred Reporting Items for Systematic Review and Meta-Analysis (PRISMA) 2020 updated guidelines [16,17]. The study protocol is freely available at https://osf.io/daz2t/.
PICO model and eligibility criteria
Following the PICOS (participants, intervention, control, outcomes, study design) model, we included as
-
•
participants: any;
-
•
intervention: liquid biopsy, defined as a simple venous blood sample in which molecular analyses can be performed such as the dosage of free tumoral cells, DNA, RNA, exosomes, or lysosomes [7];
-
•
controls: none;
-
•
outcomes: all health outcomes reported in terms of accuracy (area under the curve, AUC) with sensitivity and specificity or as precision (e.g., C-index). The data about accuracy were considered when reported against the gold standard approach for a condition;
-
•
study design: systematic reviews of observational studies.
We excluded (i) meta-analyses of intervention studies and (ii) single studies.
Information sources and search strategies
Several relevant bibliographic databases were comprehensively searched, including Medline (via Ovid), Embase, and Web of Science from database inception up to the October 1, 2023 in Pubmed, Embase and Web of Science. The search strategies are fully available in N Table 1.
Table 1.
Descriptive characteristics of the 42 systematic reviews included in the study.
AUTHOR | YEAR | TITLE | TOTAL SAMPLE SIZE | Number of studies |
---|---|---|---|---|
Bai S. | 2023 | Clinical diagnostic biomarker "circulating tumor cells" in breast cancer - a meta-analysis | 1544 | 16 |
Borg M. | 2023 | Methylated Circulating Tumor DNA in Blood as a Tool for Diagnosing Lung Cancer: A Systematic Review and Meta-Analysis | 4700 | 33 |
Chandrapalan S. | 2022 | A systematic review and meta-analysis: the diagnostic accuracy of methylated SEPTIN9 for the detection of hepatocellular carcinoma and the clinical evaluation of its use in combination with other surveillance modalities | 1001 | 6 |
Cheng, J. | 2017 | Cell-Free Circulating DNA Integrity Based on Peripheral Blood as a Biomarker for Diagnosis of Cancer: A Systematic Review | 2803 | 17 |
Duque G. | 2022 | Cancer Biomarkers in Liquid Biopsy for Early Detection of Breast Cancer: A Systematic Review | 34,376 | 136 |
Elasifer H. | 2023 | The role of circulating viral and tumour DNA in the diagnosis and management of HPV associated anogenital cancers, a systematic review and meta-analysis | 2247 | 31 |
Gally T.B. | 2021 | Circulating MicroRNAs as Novel Potential Diagnostic Biomarkers for Osteosarcoma: A Systematic Review | 4970 | 35 |
He Y. | 2020 | Clinical performance of non-invasive prenatal testing for trisomies 21, 18 and 13 in twin pregnancies: A cohort study and a systematic meta-analysis | 6618 | 21 |
Hong F. | 2023 | Exosomal microRNAs as novel diagnostic biomarkers in breast cancer: A systematic evaluation and meta-analysis | 608 | 7 |
Hou F. | 2023 | Diagnostic value of cell-free DNA in thyroid cancer: A systematic review and meta-analysis | 622 | 14 |
Jia S. | 2021 | Values of liquid biopsy in early detection of cancer: results from meta-analysis | 108 | 17 |
Karkia R. | 2022 | Diagnostic Accuracy of Liquid Biomarkers for the Non-Invasive Diagnosis of Endometrial Cancer: A Systematic Review and Meta-Analysis | 5527 | 59 |
Khetrapal P. | 2018 | The role of circulating tumour cells and nucleic acids in blood for the detection of bladder cancer: A systematic review | 58 | 15 |
Leão R. | 2021 | Circulating MicroRNAs, the Next-Generation Serum Biomarkers in Testicular Germ Cell Tumours: A Systematic Review | 45 | 31 |
Lyu M. | 2019 | The diagnostic value of circulating tumor cells and ctDNA for gene mutations in lung cancer | 7244 | 47 |
McMahon J.T. | 2022 | Circulating Tumor DNA in Adults With Glioma: A Systematic Review and Meta-Analysis of Biomarker Performance | 1651 | 7 |
Meng H. | 2023 | The diagnostic value of circulating abnormal cells in early lung cancer | 53,728 | 7 |
Mlika M. | 2018 | Liquid biopsy as surrogate to tissue in lung cancer for molecular profiling: A meta-analysis | 4782 | 33 |
Nakasu Y. | 2023 | Diagnostic accuracy of cerebrospinal fluid liquid biopsy and MRI for leptomeningeal metastases in solid cancers: A systematic review and meta-analysis | 668 | 10 |
Öberg K. | 2020 | A meta-analysis of the accuracy of a neuroendocrine tumor mRNA genomic biomarker (NETest) in blood | 549 | 18 |
Ohadi M.A.D. | 2023 | Micro RNAs as a Diagnostic Marker between Glioma and Primary CNS Lymphoma: A Systematic Review | 918 | 8 |
Rapado-González O. | 2019 | miRNAs in liquid biopsy for oral squamous cell carcinoma diagnosis: Systematic review and meta-analysis | 2562 | 16 |
Rapado-González O. | 2020 | Salivary biomarkers for cancer diagnosis: a meta-analysis | 11,153 | 29 |
Rapado-González O. | 2021 | Salivary DNA Methylation as an Epigenetic Biomarker for Head and Neck Cancer. Part I: A Diagnostic Accuracy Meta-Analysis | 8368 | 18 |
Song Z. | 2019 | The diagnostic accuracy of liquid exosomes for lung cancer detection: A meta-analysis | 2413 | 13 |
Tonozzi T.R | 2019 | Liquid biopsies in endocrine neoplasia-a systematic review | not reported | 65 |
Toraih E. A. | 2021 | Diagnostic and Prognostic Performance of Liquid Biopsy-Derived Exosomal MicroRNAs in Thyroid Cancer Patients: A Systematic Review and Meta-Analysis | 1704 | 12 |
Uhe. I | 2021 | Cell-free DNA liquid biopsy for early detection of gastrointestinal cancers: A systematic review | 4824 | 13 |
Van Westrhenen A. | 2018 | Diagnostic markers for CNS lymphoma in blood and cerebrospinal fluid: a systematic review | 268 | 25 |
Wuerdemann N. | 2020 | Cell-Free HPV-DNA as a Biomarker for Oropharyngeal Squamous Cell Carcinoma-A Step Towards Personalized Medicine? | 1284 | 11 |
Xu Y. | 2021 | Meta-Analysis of the Diagnostic Value of Cell-free DNA for Renal Cancer | 1109 | 8 |
Xu Y. | 2021 | Urinary Exosomes Diagnosis of Urological Tumors: A Systematic Review and Meta-Analysis | 3224 | 22 |
Ye J. | 2023 | Glutathione-S-Transferase p1 Gene Promoter Methylation in Cell-Free DNA as a Diagnostic and Prognostic Tool for Prostate Cancer: A Systematic Review and Meta-Analysis | 2610 | 14 |
Yinzhong W | 2023 | Diagnostic accuracy of circulating-free DNA for the determination of hepatocellular carcinoma: a systematic review and meta-analysis | 3686 | 15 |
Yu W. | 2022 | The diagnostic significance of blood-derived circRNAs in NSCLC: Systematic review and meta-analysis | 2052 | 12 |
Zhang C. | 2022 | Cell-free DNA as a Promising Diagnostic Biomarker in Prostate Cancer: A Systematic Review and Meta-Analysis | 2022 | 14 |
Zhang J. | 2022 | Evaluating the Diagnostic Potentials of Circulating Tumor DNA against Melanoma: A Systematic Review and Meta-Analysis | 1430 | 10 |
Zhao Q. | 2021 | Role of circulating tumor cells in diagnosis of lung cancer: a systematic review and meta-analysis | 3997 | 21 |
Zhou S. | 2020 | Detection of epidermal growth factor receptor mutations in peripheral blood circulating tumor DNA in patients with advanced non-small cell lung cancer A PRISMA-compliant meta-analysis and systematic review | 4527 | 32 |
Zhou Z. | 2020 | A Meta-Analytic Review of the Value of miRNA for Multiple Sclerosis Diagnosis | 989 | 11 |
Zhu Y. | 2020 | Diagnostic performance of various liquid biopsy methods in detecting colorectal cancer: A meta-analysis | 18,739 | 62 |
Zhu Y. | 2020 | Diagnostic value of various liquid biopsy methods for pancreatic cancer: A systematic review and meta-analysis | 1872 | 19 |
Study selection
The selections were independently carried out by four review authors (KS, SF, MB, VM), with consensus meetings to discuss the studies for which divergent selection decisions were made by two review authors. A third senior member of the review team (NV) was involved, if necessary. The studies selection process involved, first, a selection based on title and/or abstracts, then a selection of studies retrieved from this first step based on the full-text manuscripts. The freely accessible software Rayyan was used for the title/abstract screening [18].
Data collection and data items
From the eligible full-text articles the following data has been extracted: first author name and affiliation, year of publication, journal name, title of the manuscript; data on the characteristics of the population considered, for individual observational studies (e.g., sample size, mean age, gender), the name of the marker used in the LB, and the outcome. Data regarding estimates were extracted as reported in the original systematic review and reported as AUC, since no one reported data using C-indexes. These data were collected using a RedCap [19]. Data extraction was carried out by four authors (KS, SF, MB, VM) and systematically double-checked by another author (SC).
Assessment of risk of bias
Four authors (KS, SF, MB, VM) rated the methodological quality of the included systematic reviews using “A MeaSurement Tool to Assess systematic Reviews 2 (AMSTAR 2)” [20,21], which ranks the quality of a meta-analysis in one of 4 categories, ranging from “critically low” to “high”, according to 16 predefined items [20]. Another author (SC) double checked this evaluation.
Statistical analysis and synthesis of the data
The analyses are proposed by estimates of accuracy (AUC), in agreement with the NICE guidelines [22]. While no definitive threshold exists, values of AUC/C-index of 0.50 was considered to indicate accuracy or precision no better than chance, while values between 0.50 and 0.60 reflecting a very poor accuracy, between 0.60 and 0.70 a poor, between 0.70 and 0.80 a good, between 0.80 and 0.90 a very good, and more than 0.90 an excellent accuracy [23,24].
We used the GRADE (Grading of Recommendations, Assessment, Development, and Evaluations) assessment, adapted for prognostic and diagnostic studies [25]. Briefly, we considered the risk of bias as reported in the systematic reviews; statistical heterogeneity (inconsistency) between different studies was assessed using the I2 score, with a low heterogeneity that was based on an I2 ranging from 30 to 49%, a moderate heterogeneity from 50 to 74%, and a high heterogeneity from 75% and above [26]; indirectness was based on the assumption that the analyses should reflect the PICO question and, in particular, if only a specific population was included; imprecision: the evidence was downgraded by 1 increment if the individual studies varied across 2 areas (for example, 0.5–0.8 and 0.8–1) and by 2 increments if the individual studies varied across 3 areas (for example, 0–0.5, 0.5–0.8 and 0.8–1) [27]; publication bias was assessed using the Egger bias test [28]. When one domain of data was missing, we downgraded the evidence by two levels for that domain.
Lastly, the GRADE gave four different degrees of certainty of evidence from very low (there is a very little confidence in the estimate, i.e., the true diagnosis is likely to be substantially different from the estimate) to high (we are very confident that the true diagnosis lies close to that of the estimate) [25].
RESULTS
Summary of the literature search
As shown in Fig. 1, we initially screened 726 articles at title/abstract level. Of them, 94 full-texts were examined, finally including 42 systematic reviews after eligibility assessment [27,[29], [30], [31], [32], [33], [34], [35], [36], [37], [38], [39], [40], [41], [42], [43], [44], [45], [46], [47], [48], [49], [50], [51], [52], [53], [54], [55], [56], [57], [58], [59], [60], [61], [62], [63], [64], [65], [66], [67], [68], [69]]. The main reasons of exclusion were the lack of information about accuracy (n = 17) and the wrong study design (n = 15).
Fig. 1.
PRISMA flow-chart.
Descriptive findings
Table 1 shows the main descriptive findings of the systematic reviews included in the study. All the systematic reviews were published after the year 2017. Overall, the 42 systematic reviews included an approximate total of 213,600 individuals and a total of 1010 studies, with a mean of 24 studies and 5209 participants for each systematic review, respectively. Altogether, 211 meta-analytic outcomes were evaluated.
As shown in Table 2, among the markers investigated, 121 comparisons over 211 used RNA and, in particular, microRNA (miRNA), followed by DNA markers (53/211) and genetic biomarkers (40/211). Furthermore, eight comparisons used free tumoral cells, two exosomes, one nucleosomes, and one salivary DNA biomarkers. The large majority of the meta-analytic comparisons included cancer, at any stage, as outcome (202/211), with only nine comparisons about prenatal diagnosis and one about multiple sclerosis.
Table 2.
Main findings of the umbrella review.
Name Of The Marker | Outcome | AUC | 95% CI Low For AUC | 95% Ci High For AUC | Sensitivity | Specificity |
---|---|---|---|---|---|---|
cfHPV-DNA | Oropharyngeal Squamous Cell Carcinoma | 1 | 73 | 100 | ||
cHPV DNA | HPV Associated anal cancer | 0.9955 | 0.9886 | 1 | 95 | 100 |
ALK CtDNA | Lung Cancer | 0.994 | 0.953 | 1 | ||
NIPT screening for trisomy 21 | prenatal testing for trisomies 21 in twin pregnancies | 0.9916 | 0 | 0 | 0.99 | 1 |
cfHPV-DNA (First Diagnosis) | Oropharyngeal Squamous Cell Carcinoma | 0.99 | 81 | 98 | ||
miR10b | Breast cancer | 0.99 | 97.1 | 100 | ||
miR-148a | Breast cancer | 0.99 | 94.7 | 90.9 | ||
miR-155 | Breast cancer | 0.99 | 97.4 | 94.4 | ||
miR-21 | CNS lymphoma in blood and cerebrospinal fluid: | 0.99 | 91.70 | 95. 7 | ||
miR-30c | Breast cancer | 0.99 | 97.3 | 96.4 | ||
PTEN | Breast cancer | 0.99 | 100 | 94 | ||
miR-19b | CNS lymphoma in blood and cerebrospinal fluid: | 0.98 | 0.91 | 1 | 95.70 | 0.9 |
miRNA-373 | Breast cancer | 0.98 | 93.4 | 99 | ||
miRNA-373 | Breast cancer | 0.98 | 90.8 | 98.4 | ||
S100A4 gene | Bladder cancer | 0.978 | 90 | 92 | ||
hsa-miR-548ar-5p | Breast cancer | 0.97 | 100 | 77 | ||
miR21 | Breast cancer | 0.97 | 95.7 | 98.5 | ||
miR-92a | CNS lymphoma in blood and cerebrospinal fluid: | 0.97 | 0.93 | 1 | 95.70 | 80 |
NIPT screening for trisomy 13 | prenatal testing for trisomies 13 in twin pregnancies | 0.9655 | 0 | 0 | 0.85 | 1 |
hsa-miR-21–5p | Breast cancer | 0.96 | 86.7 | 93.3 | ||
miR-27a | Breast cancer | 0.96 | 92 | 92 | ||
MiR-152 | Osteosarcoma | 0.956 | 0 | 0 | 96.2 | 92.5 |
Panel of 6 miRNAsc | Bladder cancer | 0.956 | 0.922 | 0.978 | 90 | 90 |
APC | Breast cancer | 0.95 | 93.4 | 95.4 | ||
miR-373 | Breast cancer | 0.95 | 85 | 100 | ||
NIPT screening for trisomy 18 | prenatal testing for trisomies 18 in twin pregnancies | 0.948 | 0 | 0 | 0.88 | 1 |
CTC (DAPI+/CD45-, and CK+ or CEA, CD45-/DAPI+/CEP8, chromosome 8, CD45,CK19,Pdx-1, CEP8, CK, CD45 and DAP, KRAS), ctDNA (GNAS, KRAS, ADAMTS1, BNC1), Exosomes (miR-191,miR-21,miR-451a, miR-10b, miR-21,miR-30c, miR-181a,miR-let7a, CD44v6, Tspan8, EpCAM, MET and CD104, GPC1, miR-17–5p,miR-21, miR-1246,miR-4644) | Pancreatic cancer | 0.9478 | 80 | 89 | ||
ctDNA(CSF) | Glioma | 0.947 | 0.808 | 0.957 | ||
S100A9 | Bladder cancer | 0.944 | 81.7 | 92 | ||
cfDNA | Pancreatic Cancer (ductal adenocarcinoma) | 0.943 | 93.2 | 95.2 | ||
cfDNA | Various cancer types | 0.94 | 80 | 95 | ||
miR-155 | Breast cancer | 0.94 | 86 | 90 | ||
miR-21 | CNS lymphoma in blood and cerebrospinal fluid: | 0.94 | 0.87 | 1 | 95.70 | 83.30 |
miR-598–3p | Breast cancer | 0.94 | 95 | 85 | ||
RARB2 | Breast cancer | 0.94 | 95.5 | 92.4 | ||
S100A8 | Bladder cancer | 0.935 | 85 | 92 | ||
S100A11 | Bladder cancer | 0.934 | 83.3 | 91 | ||
CTC | leptomeningeal metastases in solid cancers | 0.931 | 0.9 | 0.9 | ||
miR-371a-3p for diagnosis | Serum Biomarkers in Testicular Germ Cell Tumours | 0.93 | 70.8–100 | 61–100 | ||
cf-DNA | Hepatocellular carcinoma | 0.93 | 0. 90 | 0.95 | 83 | 90 |
cfDNA (EGFR -TP53-NF1- MET -BRAF -KRAS -ALK) | Lung Cancer | 0.93 | 0.6 | 1 | ||
ctDNA | Any Cancer | 0.93 | 83.6 | 91.9 | ||
miR-21 Mao et al. (2014) | CNS lymphoma in blood and cerebrospinal fluid: | 0.93 | 0.88 | 0.98 | ||
miR-99a | Breast cancer | 0.93 | 76.7 | 95 | ||
ctDNA (BRAF) | Evaluating the Diagnostic Potentials of Circulating Tumor DNA against Melanoma | 0.9287 | 73 | 94 | ||
DNA del cHPV | HPV Associated with cervical cancer | 0.9277 | 0.8863 | 0.9691 | 0.36 | 0.96 |
S100A6 gene | Bladder cancer | 0.924 | 86.7 | 84 | ||
cfDNA | HCC | 0.92 | ||||
hsa-miR-25–3p | Breast cancer | 0.92 | 92 | 83 | ||
methylated SEPTIN9 | hepatocellular carcinoma | 0.92 | 80 | 90 | ||
miR-21 | Breast cancer | 0.92 | 92.3 | 81.2 | ||
Urinary Exosomes [Panel of lncRNAs (MALAT1+PCAT-1+SPRY4-IT1), Panel of lncRNAs (UCA1–201+UCA1–203+ MALAT1+LINC00355), Panel of miRNAs (miR-19b1–5p+miR-136–3p+miR139–5p), CD9 protein, miR-21–5p, miR-30c-5p, Panel of miRNAs(miR-126–3p+miR-449a, the best combination), Panel of sncRNAs (Selected miRNAs+ selected snoRNAs), miR-196a, miR-19b, Panel of mRNAs (PCA3 and ERG), Panel of mRNAs and miRNAs(ANXA3, CD24, TMPRSS2-ERG, SLC45A3,FOLH1, HPN, ITSN1, miR-375–3p, miR-574–3p), Panel of miRNA isoforms(isomiRs of miR−21, miR−204 and miR−375), Urinary vesicle-associated PSA extraction ratio | Urological tumors | 0.92 | 0.89 | 0.94 | 83 | 88 |
miR-320a | Osteosarcoma | 0.9188 | 0 | 0 | ||
miR-374a-5p | Osteosarcoma | 0.9173 | 0 | 0 | ||
CTC | Lung Cancer | 0.91 | 0.88 | 0.93 | 72 | 96 |
ctDNA (EGFR) | Growth factor receptor mutations in peripheral blood circulating tumor DNA in patients with advanced non-small cell lung cancer | 0.91 | 0.88 | 0.93 | 70 | 98 |
KRAS in CtDNA group | Lung Cancer | 0.91 | 0.804 | 1 | 65.1 | 95.5 |
LncRNA-ATB | Breast cancer | 0.91 | 80 | 90 | ||
miRNAs (miR-24; miR-10b; miR-31; miR-338–3p; miR-29a; miR-223; miR-16 let-7b; miR-146a; miR-21; miR-27b; miR-136; miR-125b; miR-4677; miR-483–5p; miR-196a; miR-196b; miR-196a e miR-196b; miR-139–5p; miR-223; miR-200b-3p; miR-99a; miR-150–5p; miR-423–5p; miR-150–5p e miR-423–5p | oral squamous cell carcinoma | 0.91 | 0.88 | 0.93 | 78 | 82 |
Panel of 40 miRNAs | Bladder cancer | 0.91 | 90 | 89 | ||
RNU2–1f (Baraniskin et al. 2016) | CNS lymphoma in blood and cerebrospinal fluid: | 0.91 | 68. 1 | 91. 4 | ||
miR-195–5p | Osteosarcoma | 0.9029 | ||||
miR-199a-3p | Osteosarcoma | 0.9025 | ||||
Panel of 7 gene mRNA (IGFBP7, SNX16, CSPG6, CTSD, CHD2, NELL2, TNFRSF7) | Bladder cancer | 0.901 | 0.803 | 0.96 | 83 | 90 |
CTC, Exosomes, cfDNA | Colorectal cancer | 0.9004 | 77 | 89 | ||
CCN1 | Breast cancer | 0.9 | 80 | 99 | ||
cfDNA | Any Cancer | 0.9 | 67.3 | 99.3 | ||
CTCs | Any Cancer | 0.9 | 90 | 94.1 | ||
liquid exosomes:miRNA; Proteins; lipids | Lung cancer | 0.9 | 0.87 | 0.92 | 82 | 84 |
miR-1246 | Breast cancer | 0.9 | 93 | 75 | ||
miR-195–5p-5p | Breast cancer | 0.9 | 77.8 | 100 | ||
miR-301a-3p | Breast cancer | 0.9 | 85 | 78 | ||
miR-495 | Breast cancer | 0.9 | 100 | 66.7 | ||
Plasma cfDNA concentrations | Any Cancer | 0.9 | 90.5 | 80.5 | ||
miR-210 | Bladder cancer | 0.898 | 0.855 | 0.931 | 97.6 | 69.2 |
miR-371a-3p post chemo | Serum Biomarkers in Testicular Germ Cell Tumours | 0.8975 | 82.6–100 | 58–100 | ||
MiR-326 | Osteosarcoma | 0.897 | 83.7 | 94.5 | ||
mRNA | Neuroendocrine tumor | 0.897 | 0.877 | 0.917 | ||
S100A7 gene | Bladder cancer | 0.895 | 73.3 | 93.3 | ||
MiR-195 | Osteosarcoma | 0.892 | 0 | 0 | 88 | 83.3 |
cf-DNA | Thyroid cancer | 0.89 | 0.86 | 0.91 | 0.76 | 0.87 |
HERV-K (HML-2) type levels. | Breast cancer | 0.89 | 80 | 84.6 | ||
microRNA | Endocrine Neoplasia | 0.89 | 90 | 76.50 | ||
miR-34a | Breast cancer | 0.89 | 91 | 75 | ||
MiR-375 | Osteosarcoma | 0.89 | 82.1 | 74.7 | ||
−31G/C polymorphism in the survival promoter gene | Breast cancer | 0.89 | 92.7 | 86.9 | ||
Community Verified icon | Colorectal cancer | 0.887 | ||||
EGFR in CTC group | Lung Cancer | 0.885 | 0.778 | 0.993 | 75.4 | 85.2 |
miR21 | Glioma and Primary CNS Lymphoma | 0.883 | 0.813 | 0.954 | ||
Hypomethylation and copy number aberrations | Any Cancer | 0.88 | 81 | 94 | ||
MiR-574–3p | Osteosarcoma | 0.88 | 0 | 0 | ||
BRAF IN CTDNA | Lung Cancer | 0.877 | 0 | 1 | 31.3 | 99.5 |
circulatory abnormal cells (CAC) | Lung Cancer | 0.87 | 0.84 | 0.9 | 0.8 | 0.9 |
cfDNA (APP gene integrity) | Endocrine Neoplasia | 0.87 | ||||
miR-17–5p | Breast cancer | 0.87 | 100 | 75.5 | ||
Telomeric sequences in cfDNA | Breast cancer | 0.87 | 91.5 | 76.2 | ||
MiR-25–3p | Osteosarcoma | 0.868 | 0 | 0 | 71.4 | 92.3 |
APC, GSTP1 or TIG1 | Bladder cancer | 0.867 | 0.785 | 0.948 | 80 | 93.3 |
MiR-27a | Osteosarcoma | 0.867 | 0 | 0 | 70 | 98.3 |
TIG1, GSP1, APC or PTGS2 | Bladder cancer | 0.867 | 0.785 | 0.948 | 80 | 93.3 |
microRNA (miR-130a-3p, miR-129–2, miR-889, miR-29a, miR-148a-3p, miR-25–3p, miR-296–5p, miR-92a-3p, miR-5189–3p, miR-5010–3p, miR-598–5p, miR-3161, miR-6516–5p, miR-4644, miR-1283, miR-1227–3p, miR-149–3p, miR-210–5p, miR-3662, miR-187–5p, miR-16–2–3p, miR-223–5p, miR-34c-5p, miR-182–5p, miR-223–3p, miR-146b-5p, miR-16–2–3p, miR-223–5p, miR-146b-5p, miR-221–3p, miR-222–3p, miR-21–5p, miR-204–5p, miR-485–3p, miR-4433a-5p, miR4306, miR-376a-3p, miR-204–3p, miRNA423–5p, miR-346, miR-10a-5p, miR-34a-5p, miR-181a, | Thiroid Cancer | 0.866 | 82 | 76 | ||
miR-371a-3p for early stage disease | Serum Biomarkers in Testicular Germ Cell Tumours | 0.8625 | 83.4–100 | 60.1–100 | ||
MiR-199a-5p | Osteosarcoma | 0.8606 | 0 | 0 | 88.3 | 76.67 |
ccfDNA | Breast cancer | 0.86 | 67 | 90 | ||
Delta181CTmir155 | Breast cancer | 0.86 | 83.3 | 82.4 | ||
hsa-miR-888–5p | Breast cancer | 0.86 | 83 | 75 | ||
miR-195 | Breast cancer | 0.86 | 69 | 89.2 | ||
miR-222 | Breast cancer | 0.86 | 91.2 | 78.6 | ||
MiR-663a | Osteosarcoma | 0.86 | 0 | 0 | 67.4 | 89.8 |
miRNA-21 | Breast cancer | 0.86 | 70.8 | 91.8 | ||
MiR-191 | Osteosarcoma | 0.858 | 0 | 0 | 74 | 100 |
MiR-194 | Osteosarcoma | 0.855 | 0 | 0 | 84.2 | 79.1 |
miR21 | Glioma and Primary CNS Lymphoma | 0.851 | 0.755 | 0.947 | ||
Salivary biomarkers (CA-125; c-erbB-2; VEGF, EGF, CEA; 1472.78 Da, 2936.49 Da, 6556.81 Da, 7081.17 Da KRAS, MBD3L2, ACRV1, CDKL3; CSTA, TPT1, IGF2BP1, GRM1, GRIK1, H6PD, MDM4, S100A8, CA6; N elongate, S mitis, G adiacens; miR-21 BRAF, CCNI, EGFR, FGF19, FRS2, GREB1, LZTS1; calprotectin, AZGP1, HP H3F3A, SRGN, B2 M, BASP1, AGPAT1, IL1B, IER3 miR-10b, miR-144, miR-451, miR-21; miR-144; miR-21, miR-23a, miR-23b, miR-29c, miR-216, miR-210, let-7c; miR-3679–5p, miR-940 miR-21 SFAA: Phe, Trp, Met, Pro, Thr, Asp, Ser, Cit, Orn; His, Gln, Leu, Val, Glu, Lys LRP; Capnocitophaga, Veillonela, Neisseria; miR-4644, miR-1246 miR-21 Polyamines: SPM, N1-Ac-SPD, N8-Ac-SPD, Ac-PUT, CAD,; Ac-SPM, DAc-SPM, DAc-SPD, PUT; SPD, ORN, DAP lncRNAs: HOTAIR, PVT1 CSTB, TPl1, DMBT1 LysoPC(18:2), Palmitic amide, Phytosphingosine,; LysoPC(18:1), PS(14:1/16:1), LysoPC(16:0), Acetylphenylalanine, Propionylcholine, LysoPC(22:6), MG(0:0/14:0/0:0), LysoPE(18:2/0:0), PC(18:1/16:0), Phenylalanine, Citrulline, Histidine, N-Acetylneuraminic acid, PE(22:0/20:4), 4-Hydroxyphenylpyruvic acid; Lectins: model GC (VVA and SBA), VVA; PPP2CA, PTGS2, ROCK1, SKP1, SLK I; EX-1; Lectins: model BC (BS-I, NPA, PNA, PTL-II and MAL-I) | Salivary biomarkers for cancer diagnosis | 0.85 | 0.84 | 0.87 | 76 | 76 |
Delta181CTmir125a | Breast cancer | 0.85 | 83.3 | 64.7 | ||
hsa-miR-548a-5p | Breast cancer | 0.85 | 83 | 83 | ||
MiR-101 | Osteosarcoma | 0.85 | 0 | 0 | 78.95 | 82.86 |
miRNA (miR-15b-5p,miR-451a, miR-30b-5p,miR-342–3p, miR-127–3p, miR-370–3p, miR-409–3p, miR-432–5p, miR-145 and miR-223, miR-122–5p, miR-196b-5p, miR-301a-3p,miR-532–5p, miR-484, miR-140–5p, miR-320a, miR-486–5p, miR-320c, miR-7–1–3p,miR-7–1–3p, miR-191–5p, mi-RNA-145, miR-572, miR-30e, miR-150, miRNA-181c) | Multiple Sclerosis Diagnosis | 0.85 | 0.82 | 0.88 | 81 | 75 |
SMAD4 | Breast cancer | 0.85 | 100 | 100 | ||
miR-20a-5p | Osteosarcoma | 0.8471 | 0 | 0 | ||
MiR-124 | Osteosarcoma | 0.846 | 0 | 0 | 79.8 | 86 |
MiR-139–5p | Osteosarcoma | 0.846 | 0 | 0 | 76.5 | 80 |
GSTP1 or APC | Bladder cancer | 0.844 | 0.757 | 0.931 | 75.6 | 93.3 |
MiR-221 | Osteosarcoma | 0.844 | 0 | 0 | 65.7 | 100 |
TIG1 or APC | Bladder cancer | 0.844 | 0.757 | 0.931 | 68.9 | 100 |
MiR-542–3p | Osteosarcoma | 0.841 | 0 | 0 | 77.8 | 93.6 |
circRNAs | non-small cell lung cancer (NSCLC) | 0.84 | 0.8 | 0.87 | 78 | 76 |
miR-185–3p | Breast cancer | 0.84 | 95 | 66 | ||
EGFR in CtDNA group | Lung Cancer | 0.8391 | 0.759 | 0.919 | 67.1 | 96.1 |
PTGS2 gene | Bladder cancer | 0.836 | 0.753 | 0.918 | 95.6 | 62.2 |
microRNAs | Breast cancer | 0.8325 | 67 | 81 | ||
APC, GSTP1, PTGS2, p14(ARF), p16(INK) and RASSF1A | Any Cancer | 0.83 | 67 | 97 | ||
cfDNA integrity | Breast cancer | 0.83 | 77.5 | 90 | ||
miRNA-222 | Breast cancer | 0.83 | 97.8 | 75.5 | ||
Opz | Breast cancer | 0.83 | 80 | 76 | ||
Micro RNA-21 | Non-Invasive Diagnosis of Endometrial Cancer | 0.825 | 0.735 | 0.915 | ||
Panel of 9 hypermethylated segments | Bladder cancer | 0.825 | 0.761 | 0.89 | 62.1 | 88.7 |
hsa-miR-144 5p | Bladder cancer | 0.824 | 0.633 | 0.929 | 70 | 82.4 |
hsa-miR-374–5p | Bladder cancer | 0.824 | 0.633 | 0.929 | 60 | 94 |
miR-26b-5p hsa-miR-144 5p hsa-miR-374–5p | Bladder cancer | 0.824 | 0.633 | 0.929 | 65 | 94.1 |
APC, RASSF1A, CDH1, RUNX3, TFPI2, SFRP5, OPCML | Any Cancer | 0.82 | 85.3 | 90.5 | ||
Cf-DNA | renal cancer | 0.82 | 0.79 | 0.85 | 71 | 79 |
mtDNA | Breast cancer | 0.82 | 77 | 83 | ||
Lower cfDI | Different types of Cancer (Breast cancer, Prostate cancer, HCC, CRC,Glioma,Melanoma, Bladder cancer, Renal cell cancer,Acute leukemia,TGCC,HNC,PAC,Mixed) | 0.814 | 0.74 | 0.84 | ||
Micro RNA-223 | Non-Invasive Diagnosis of Endometrial Cancer | 0.813 | 0.735 | 0.89 | ||
Circulating tumor cells(CTC) | Breast cancer | 0.8129 | 50 | 93 | ||
cfDNA methylation score | Breast cancer | 0.81 | 93 | 73.5 | ||
Salivary DNA | Salivary DNA Methylation as an Epigenetic Biomarker for Head and Neck Cancer | 0.81 | 0.77 | 0.84 | 39 | 87 |
LncRNA H19 | Breast cancer | 0.81 | 56.7 | 86.7 | ||
methylated circulating tumor DNA (ctDNA) | Lung Cancer | 0.81 | 0.77 | 0.84 | 46.9 | 92.9 |
ALU-247 | Breast cancer | 0.8 | 70 | 100 | ||
cfDNA, GADD45, CDH13promoter, Hypermethylation APC, Hypermethylation GSTP1, Hypermethylation MDR1, Hypermethylation RASSF1, Hypermethylation PTGS2, RARβ2 | Prostate Cance | 0.8 | 0.76 | 0.83 | 56 | 89 |
GSTP1 or TIG1 | Bladder cancer | 0.8 | 0.704 | 0.895 | 66.7 | 93.3 |
miR-155 | Breast cancer | 0.8 | 65 | 81.8 | ||
MiR-214 | Osteosarcoma | 0.8 | 0 | 0 | ||
APC | Bladder cancer | 0.798 | 0.698 | 0.897 | 59.5 | 100 |
miR-25–3p | Osteosarcoma | 0.7961 | 0 | 0 | ||
miR-451a | Osteosarcoma | 0.7961 | 0 | 0 | ||
ALU-115 | Breast cancer | 0.78 | 67.5 | 100 | ||
GSTP1 | Prostate Cancer | 0.78 | 0.75 | 0.82 | 37 | 97 |
MiR-335–5p | Osteosarcoma | 0.78 | 0 | 0 | ||
GSTP1 | Bladder cancer | 0.777 | 0.676 | 0.877 | 59.1 | 93.2 |
miR-425–5p | Osteosarcoma | 0.7765 | 0 | 0 | ||
cfDNA | Breast cancer | 0.77 | 70 | 76 | ||
Delta192CTmir155 | Breast cancer | 0.77 | 77.8 | 6.7 | ||
Hotair | Breast cancer | 0.77 | 76 | 76 | ||
hsa_circ_0005046 | Breast cancer | 0.77 | 85 | 51 | ||
miR-10b | Breast cancer | 0.77 | 60 | 93 | ||
miR-16–5p | Osteosarcoma | 0.7686 | 0 | 0 | ||
miR-371a-3p for chemotherapy | Serum Biomarkers in Testicular Germ Cell Tumours | 0.759 | 83.4–92.9 | 60.1–100 | ||
miR-639 | Bladder cancer | 0.752 | 0.571 | 0.934 | ||
Serum DNA | Any Cancer | 0.75 | 62.9 | 87 | ||
Higher cfDI | Different types of Cancer (Breast cancer, Prostate cancer, HCC, CRC,Glioma,Melanoma, Bladder cancer, Renal cell cancer,Acute leukemia,TGCC,HNC,PAC,Mixed) | 0.742 | 0.58 | 0.78 | ||
ctDNA(Plasma) | Glioma | 0.741 | 0.332 | 0.927 | ||
KRAS in CTC group | Lung Cancer | 0.741 | 0.472 | 1 | 38.7 | 92.1 |
FAM83H-AS1 | Breast cancer | 0.74 | 70 | 76.7 | ||
miR-184 | Breast cancer | 0.74 | 87.5 | 71 | ||
miR-382–3p | Breast cancer | 0.74 | 52 | 92.5 | ||
miR 30a 3p | Glioma and Primary CNS Lymphoma | 0.737 | 0.66 | 0.81 | ||
AFP, CEA, CA19–9, CYFRA21–1, SCC, PSA | Any Cancer | 0.73 | 76 | 76 | ||
DNA integrity index | Breast cancer | 0.73 | 51 | 90 | ||
Neat1 | Breast cancer | 0.73 | 80 | 80 | ||
miR-106a-5p | Osteosarcoma | 0.7255 | ||||
Circulating nucleosomes | Any Cancer | 0.72 | 91 | 90 | ||
HER2 mRNA | Breast cancer | 0.72 | 90 | 50 | ||
MiR-17–3p | Osteosarcoma | 0.72 | 64.3 | 84.6 | ||
miR-505–5p | Breast cancer | 0.72 | 75 | 60 | ||
miR-96–5p | Breast cancer | 0.72 | 73 | 66 | ||
miR 6803 3p | Glioma and Primary CNS Lymphoma | 0.716 | 0.646 | 0.786 | ||
miR-141 | Bladder cancer | 0.714 | 0.519 | 0.91 | ||
Pai-1 | Breast cancer | 0.71 | 64 | 68 | ||
miR-139–5p | Osteosarcoma | 0.7098 | ||||
Delta181CTmir21 | Breast cancer | 0.7 | 72.2 | 64.7 | ||
MiR- 205–5p | Osteosarcoma | 0.7 | ||||
cfDNA (APP gene integrity) | Endocrine Neoplasia | 0.699 | ||||
miR 4751 | Glioma and Primary CNS Lymphoma | 0.693 | 0.602 | 0.778 | ||
ACTB-106 | Bladder cancer | 0.686 | 0.617 | 0.755 | 91.6 | 43.3 |
miR 3918 | Glioma and Primary CNS Lymphoma | 0.682 | 0.586 | 0.766 | ||
miR 487a 3p | Glioma and Primary CNS Lymphoma | 0.681 | 0.593 | 0.762 | ||
miR 6820 3p | Glioma and Primary CNS Lymphoma | 0.681 | 0.579 | 0.774 | ||
hsa-miR-423–5p | Breast cancer | 0.68 | 66 | 68 | ||
9 miRNAs (miR-15a, miR-18a, miR-107, miR- 133a, miR-139–5p, miR-143, miR-145, miR- 365, miR-425) | Any Cancer | 0.67 | 83.3 | 41.2 | ||
miR-181a | Breast cancer | 0.67 | 70.7 | 59.9 | ||
miR 371a 3p | Glioma and Primary CNS Lymphoma | 0.669 | 0.578 | 0.757 | ||
miR 4756 5p | Glioma and Primary CNS Lymphoma | 0.669 | 0.578 | 0.755 | ||
TIG1 | Bladder cancer | 0.659 | 0.536 | 0.782 | 31.8 | 100 |
miR 146a 3p | Glioma and Primary CNS Lymphoma | 0.656 | 0.551 | 0.754 | ||
miR548am 3p | Glioma and Primary CNS Lymphoma | 0.639 | 0.531 | 0.743 | ||
4-miRNAs | Osteosarcoma | 0.608 | 91.1 | 94.4 | ||
CTCs | Any Cancer | 0.6 | 30.6 | 88 | ||
PTGS2 | Bladder cancer | 0.3622 | 0.506 | 0.739 | 24.4 | 100 |
DAPK | Bladder cancer | 0.3512 | 0.385 | 0.638 | 24 | 100 |
Data are reported ranked by area under the curve values, from higher to lower values. Abbreviations: OSCC: Oropharyngeal Squamous Cell Carcinoma, HPV: Human Papillomavirus; LC: Lung Cancer; NIPT: Non-Invasive Prenatal Testing; CNS: Central Nervous System; BC: Breast Cancer; BLC: Bladder Cancer; OLC: Oral Squamous Cell Carcinoma; OC: Ovarian Cancer; HCC: Hepatocellular Carcinoma; G: Glioma; PC: Pancreatic Cancer, O: Osteosarcoma; LEP: Leptomeningeal Metastases; TGC: Testicular Germ Cell Tumors; NE: Neuroendocrine Tumor, ThyC: Thyroid Cancer; EN: Endocrine Neoplasia; CRC: Colorectal Cancer; MS: Multiple Sclerosis; NSCLC: Non-Small Cell Lung Cancer; RCC: Renal Cell Cancer; AC: Acute Leukemia; TGCC: Testicular Germ Cell Cancer; HNC: Head and Neck Cancer; PAC: Pancreatic Adenocarcinoma; SBC: Salivary Biomarkers for Cancer; DNA: Deoxyribonucleic Acid; cfHPV-DNA: Circulating cell-free Human Papillomavirus DNA; cHPV DNA: Circulating Human Papillomavirus DNA; ALK ctDNA: Anaplastic Lymphoma Kinase circulating tumor DNA; NIPT: Non-Invasive Prenatal Testing; miR10b: MicroRNA-10b; miR-148a: MicroRNA-148a; miR-155: MicroRNA-155; miR-21: MicroRNA-21; miR-30c: MicroRNA-30c; PTEN: Phosphatase and Tensin Homolog gene; miR-19b: MicroRNA-19b; miRNA-373: MicroRNA-373; S100A4: S100 Calcium Binding Protein A4; hsa-miR-548ar-5p: Homo sapiens microRNA-548ar-5p; miR-92a: MicroRNA-92a; hsa-miR-21–5p: Homo sapiens microRNA-21–5p; miR-27a: MicroRNA-27a; miR-152: MicroRNA-152; APC: Adenomatous Polyposis Coli gene; CTC: Circulating Tumor Cells; ctDNA: Circulating tumor DNA; S100A9: S100 Calcium Binding Protein A9; cfDNA: Cell-Free DNA; miR-598–3p: MicroRNA-598–3p; RARB2: Retinoic Acid Receptor Beta 2 gene; S100A8: S100 Calcium Binding Protein A8; S100A11: S100 Calcium Binding Protein A11; miR-371a-3p: MicroRNA-371a-3p; cf-DNA: Cell-Free DNA; EGFR: Epidermal Growth Factor Receptor gene; TP53: Tumor Protein P53 gene; NF1: Neurofibromin 1 gene; MET: MET Proto-Oncogene; BRAF: B-Raf Proto-Oncogene; KRAS: KRAS Proto-Oncogene; ADAMTS1: A Disintegrin And Metalloproteinase with Thrombospondin Motifs 1; BNC1: Basonuclin 1; Exosomes: Small extracellular vesicles; CD44v6: Isoform of CD44 protein; Tspan8: Tetraspanin 8; EpCAM: Epithelial Cell Adhesion Molecule; GPC1: Glypican-1; miR-1246: MicroRNA-1246; GNAS: GNAS Complex Locus; miR-4644: MicroRNA-4644; S100A6: S100 Calcium Binding Protein A6; SEPTIN9: Septin 9 gene; lncRNA: Long Non-Coding RNA; MALAT1: Metastasis Associated Lung Adenocarcinoma Transcript 1; PCAT-1: Prostate Cancer Associated Transcript 1; SPRY4-IT1: Sprouty RTK Signaling Antagonist 4 Intron Transcript 1; UCA1–201: Urothelial Cancer Associated 1 Transcript 201; LINC00355: Long Intergenic Non-Protein Coding RNA 355; CD9: CD9 Protein; miR-19b1–5p: MicroRNA-19b1–5p; miR-136–3p: MicroRNA-136–3p; miR-139–5p: MicroRNA-139–5p; PCA3: Prostate Cancer Antigen 3; ERG: ETS-Related Gene; TMPRSS2-ERG: Fusion gene between TMPRSS2 and ERG; FOLH1: Folate Hydrolase 1; HPN: Hepatocyte Growth Factor Activator; ITSN1: Intersectin 1; miR-375–3p: MicroRNA-375–3p; miR-574–3p: MicroRNA-574–3p; ANXA3: Annexin A3 gene; CCN1: Cellular Communication Network Factor 1; HERV-K: Human Endogenous Retrovirus K; miR-99a: MicroRNA-99a; GSTP1: Glutathione S-Transferase Pi 1 gene; TIG1: Tazarotene-Induced Gene 1; PTGS2: Prostaglandin-Endoperoxide Synthase 2 gene; miR-25–3p: MicroRNA-25–3p; Pdx-1: Pancreatic and Duodenal Homeobox 1 gene; KRAS in CTC group: KRAS mutations detected in circulating tumor cells group; cfDNA integrity: Integrity of cell-free DNA; miR-191: MicroRNA-191; miR-125b: MicroRNA-125b; miR-223: MicroRNA-223; miR-196a: MicroRNA-196a; miR-146a: MicroRNA-146a; miR-4677: MicroRNA-4677; miR-483–5p: MicroRNA-483–5p; miR-141: MicroRNA-141; miR-222: MicroRNA-222; miR-663a: MicroRNA-663a; miR-195: MicroRNA-195; Delta181CTmir21: A specific mutation (Delta181) in microRNA-21; miR-205–5p: MicroRNA-205–5p; miR-495: MicroRNA-495; miR-301a-3p: MicroRNA-301a-3p; ALU-247: ALU repeat region of 247 base pairs; miR-20a-5p: MicroRNA-20a-5p; ctDNA (Plasma): Circulating tumor DNA in plasma; Neat1: Nuclear Enriched Abundant Transcript 1; Pai-1: Plasminogen Activator Inhibitor 1; AFP: Alpha-fetoprotein; CEA: Carcinoembryonic Antigen; CA19–9: Cancer Antigen 19–9; SCC: Squamous Cell Carcinoma Antigen; PSA: Prostate-Specific Antigen; ALU-115: ALU repeat region of 115 base pairs; HER2: Human Epidermal Growth Factor Receptor 2 gene; cf-DNA: Cell-Free DNA; mtDNA: Mitochondrial DNA; hsa-miR-423–5p: Homo sapiens microRNA-423–5p; miR-146b-5p: MicroRNA-146b-5p; ALU: ALU sequences (repetitive elements in the genome); miR-486–5p: MicroRNA-486–5p; miR-34a: MicroRNA-34a; miR-425–5p: MicroRNA-425–5p; SMAD4: SMAD Family Member 4 gene; miR-181a: MicroRNA-181a; miR-106a-5p: MicroRNA-106a-5p; miR-193b: MicroRNA-193b; PTEN: Phosphatase and Tensin Homolog gene; PTGS2: Prostaglandin-Endoperoxide Synthase 2 gene; PTEN: Phosphatase and Tensin Homolog SMAD4: SMAD Family Member 4 gene; ADAMTS1: A Disintegrin And Metalloproteinase with Thrombospondin Motifs 1; miR-92b: MicroRNA-92b; CFAP69: Cilia and Flagella Associated Protein 69 gene; LOXL2: Lysyl Oxidase Like 2 gene; MLH1: MutL Homolog 1 gene; PAX8: Paired Box 8 gene; miR-29a: MicroRNA-29a; miR-143: MicroRNA-143; NDRG2: N-Myc Downstream Regulated Gene 2; hTERT: Human Telomerase Reverse Transcriptase gene; PIK3CA: Phosphatidylinositol-4,5-Bisphosphate 3-Kinase Catalytic Subunit Alpha gene; TFF1: Trefoil Factor 1 gene; TFF3: Trefoil Factor 3 gene; NID1: Nidogen 1 gene; TM4SF1: Transmembrane 4 L6 Family Member 1 gene; miR-122: MicroRNA-122; cfmiRNA: Circulating free microRNA; CCAT1: Colon Cancer Associated Transcript 1 gene; miR-18a: MicroRNA-18a; miR-146b: MicroRNA-146b; MEG3: Maternally Expressed 3 gene; MMP9: Matrix Metallopeptidase 9 gene; PIK3CA ctDNA: Phosphatidylinositol-4,5-Bisphosphate 3-Kinase Catalytic Subunit Alpha circulating tumor DNA; miR-214: MicroRNA-214; miR-34b: MicroRNA-34b; miR-210: MicroRNA-210; miR-224: MicroRNA-224; lncRNA CCAT1: Long Non-Coding RNA Colon Cancer Associated Transcript 1; cfmiR-101: Circulating free MicroRNA-101; hsa-miR-30e-5p: Homo sapiens MicroRNA-30e-5p; miR-125a-5p: MicroRNA-125a-5p; SEPT9 cfDNA: Septin 9 Cell-Free DNA; miR-376a: MicroRNA-376a; miR-150–5p: MicroRNA-150–5p; TM4SF1 cfDNA: Transmembrane 4 L6 Family Member 1 circulating tumor DNA; MALAT1 cfmiRNA: Metastasis Associated Lung Adenocarcinoma Transcript 1 circulating free microRNA; hsa-miR-144–3p: Homo sapiens MicroRNA-144–3p; cf-DNA integrity: Integrity of circulating free DNA; TCF21: Transcription Factor 21 gene; miR-451: MicroRNA-451; miR-125a: MicroRNA-125a; miR-29c: MicroRNA-29c; mtDNA cfDNA: Mitochondrial DNA in circulating free DNA; BRCA1: Breast Cancer Type 1 susceptibility gene; BRCA2: Breast Cancer Type 2 susceptibility gene; miR-663b: MicroRNA-663b; ITGA3: Integrin Subunit Alpha 3 gene; HDAC4: Histone Deacetylase 4 gene; hsa-miR-548ar-5p cfDNA: Homo sapiens MicroRNA-548ar-5p circulating tumor DNA; hsa-miR-151a-5p: Homo sapiens MicroRNA-151a-5p; FGF19: Fibroblast Growth Factor 19 gene; miR-506: MicroRNA-506; HLA-G: Human Leukocyte Antigen G gene; hsa-miR-23b-3p: Homo sapiens MicroRNA-23b-3p; hsa-miR-320a-3p: Homo sapiens MicroRNA-320a-3p; miR-508–5p: MicroRNA-508–5p; hsa-miR-153–5p: Homo sapiens MicroRNA-153–5p; TUSC2: Tumor Suppressor Candidate 2 gene; cMet ctDNA: Circulating MET Proto-Oncogene tumor DNA; cf-DNA plasma integrity: Integrity of cell-free DNA in plasma; CD9 cfmiRNA: CD9 circulating free microRNA; cfDNA fragments: Fragments of cell-free DNA; STK11: Serine/Threonine Kinase 11 gene; cfRNA: Circulating Free RNA; miR-214–5p: MicroRNA-214–5p; cfDNA epigenetic markers: Epigenetic markers in cell-free DNA; HOTTIP: HOXA Transcript at the Distal Tip gene; cfRNA expression: Expression of circulating free RNA; cfDNA methylation markers: Methylation markers in cell-free DNA; hsa-miR-335–5p: Homo sapiens MicroRNA-335–5p; miR-7: MicroRNA-7; AKT1: AKT Serine/Threonine Kinase 1 gene; miR-92a cfmiRNA: MicroRNA-92a circulating free microRNA; cfDNA fragment size: Size of circulating free DNA fragments; hsa-miR-146b-5p cfDNA: Homo sapiens MicroRNA-146b-5p circulating tumor DNA; cfRNA markers: Markers of circulating free RNA; cfmiR-30b: Circulating free MicroRNA-30b; cfmiR-200c: Circulating free MicroRNA-200c; cfmiR-375: Circulating free MicroRNA-375; cfDNA fragment ratio: Ratio of circulating free DNA fragments; cfmiR-196a: Circulating free MicroRNA-196a; cfmiR-141: Circulating free MicroRNA-141; hsa-miR-181a-5p cfDNA: Homo sapiens MicroRNA-181a-5p circulating tumor DNA; cfmiR-486–5p: Circulating free MicroRNA-486–5p; cfmiR-34a: Circulating free MicroRNA-34a; cfDNA epigenetics: Epigenetic modifications in circulating free DNA; cfmiR-424–5p: Circulating free MicroRNA-424–5p; cfmiR-145: Circulating free MicroRNA-145; cfmiR-99b: Circulating free MicroRNA-99b; cfmiR-221: Circulating free MicroRNA-221; cfmiR-26b: Circulating free MicroRNA-26b.
Main findings
Table 2 presents the main findings of the umbrella review, whilst Supplementary Table 2 includes the GRADE assessment for each outcome. When considering the AUC as estimation of the accuracy of the LB markers, we found that 75/211 (45.4%) had an excellent accuracy (AUC >0.90), with one comparison having an AUC equal to 1, indicating a perfect correspondence between LB and the gold standard, i.e., cfHPV-DNA for HPV-positive oropharyngeal squamous cell carcinoma. Moreover, 81/211 outcomes showed an AUC between 0.80 and 0.90, thus being classified with a very good accuracy, while 39 had an AUC between 0.70 and 0.80, and 16 an AUC 0.60–0.70.
However, when the level of evidence using the GRADE was evaluated, all the outcomes were graded as very low, as further detailed in Supplementary Table 2. Briefly, only 95 out of 211 outcomes (44.8%) reported the 95% CI for AUCs, thus increasing the risk of imprecision. Moreover, the risk of bias was graded as very serious in almost all outcomes included (197/211, 92.9%) and the heterogeneity was never reported, making inconsistency very serious in all the comparisons. Lastly, the presence of publication bias was present in almost a half of outcomes (102/211, 48.1%) (Supplementary Table 2).
Risk of bias evaluation
Supplementary Table 3 reports the results on the possible risk of bias evaluated on the 42 systematic reviews included in the study. Of these, 30 were ascertained as having a critically low quality, 11 a low quality and only one of high quality. The main reasons of this possible bias were also identified as the lack of sufficient information about statistical methods used for meta-analysis and the poor consideration of the risk of bias in affecting results in original systematic reviews.
Discussion
This study represents a quantitative summary of evidence related to LB, highlighting its potentialities and limitations. In this umbrella review, evaluating the accuracy of several LB markers, we highlighted that in 42 systematic reviews published after 2017, including a total of 213,600 individuals from 1010 studies, a significant fraction of the outcomes considered had an excellent or very good accuracy, respectively. Based on this accuracy, this study may corroborate recent evidence of the need for implementing LB into clinical practice. However, as expected, most systematic reviews and meta-analysis in the literature regards oncology and related fields (95.7%).
Although LB has been mainly explored in the context of precision oncology, it should be acknowledged that it may bring crucial advantages into different clinical levels, including: i) avoiding invasive procedures for the molecular characterization of a disease, even for patients with multiple comorbidities and/or unfit for surgery, ii) allowing molecular investigation for the molecular characterization of tumours, even for stage IV / metastatic patients, where surgical resections cannot be considered and tissue biopsies are not always practicable, iii) giving the possibility of overcoming intratumor heterogeneity, representing a possibility of showing a comprehensive molecular landscape of a given neoplasm, independent from tissue's related biases. These advantages are unique for LB and related methodologies and call for further research along those lines for implementing this promising tool into clinical practice.
The use of integrated biomarkers allows for a real-time, minimally invasive assessment of the tumour's genetic and epigenetic alterations. For instance, ctDNA can reveal mutations in key oncogenes, such as EGFR, providing valuable information for therapy selection, particularly for targeted treatments [70]. The inclusion of microRNAs and exosomal content further enhances the diagnostic accuracy, as these components can reflect various biological processes involved in cancer progression and metastasis. In lung cancer management, the integration of these biomarkers supports longitudinal monitoring [71]. By analysing changes in ctDNA levels or the presence of specific mutations over time, clinicians can adjust treatment strategies more effectively, optimizing outcomes for patients. This approach enables personalized medicine, where treatment is tailored based on the evolving molecular profile of the tumour, improving both the prognosis and quality of life for cancer patients.
It is important to remark that more recent artificial intelligence (AI) systems could significantly help to implement the use of LB in the daily clinical practice (e.g., by helping to manage big data derived from multi-omics analyzes on LB) [72,73]. AI can improve the sensitivity and specificity of liquid biopsies for early cancer detection; more in depth, by integrating data from multiple biomarkers and employing sophisticated algorithms, AI can distinguish between benign and malignant conditions more effectively than traditional methods, therefore potentially leading to earlier diagnosis and better prognostic outcomes [74]. Finally, the use of AI in the LB field may track changes in biomarker levels over time, providing valuable insights into disease progression and patient response to treatments, allowing for personalized treatment adjustments [75].
The role of LB in the central nervous system (CNS), specifically for detecting cell-free DNA (cfDNA) in patients with meningioma and glioblastoma, is critical in improving non-invasive diagnostic and monitoring approaches. LB may provide a minimally invasive method to detect tumor-derived DNA in bodily fluids, offering a real-time reflection of tumor dynamics [76]. This approach has shown promise in CNS tumors, which are typically challenging to access surgically due to the blood-brain barrier. Corticosteroid treatment, commonly used to manage edema in CNS tumors, may affect the levels of cfDNA detected in liquid biopsies [77]. The administration of corticosteroids can reduce inflammation and vascular permeability, potentially lowering the amount of cfDNA released into circulation [77]. This can influence the sensitivity of liquid biopsies in detecting tumor-derived cfDNA, thereby impacting the accuracy of real-time tumor monitoring during corticosteroid therapy.
At the same time, this umbrella review serves also as a critical tool for analyzing the limitations associated to LB, and in particular to the published attempts for proving quantitative summaries on this group of methodology through the use of meta-analysis. Indeed, according to the GRADE evaluation, all the outcomes were graded as very low, thus highlighting important limitations. The measured outcomes suffered from the lack of essential parameters for their evaluation in meta-analysis, including the 95% confidence intervals in the majority of studies, the total lack of reporting/analyzing heterogeneity, and the significant proportion of publication bias risk. All these observations clearly showed that to date there are critical issues in the available literature for enabling a comprehensive, impartial, and objective evaluation of such limitations on the use of LB. For example, we don't know if we can apply LB in all individuals independently from the demographic characteristics (e.g., age and gender). Moreover, the available literature seems to be affected by a publication bias. For the mentioned reasons, the methodological considerations emerging from our umbrella review represents a warning when considering the current literature available on this topic. At the same time, our findings warmly support for a more rigid and transparent assessment of the findings, while suggesting where to better direct future research efforts. Overall considered, the most essential areas where LB should be better addressed are: i) improving LB sensitivity, starting from more specific analyses on the role of LB in the early detection of cancer, which now is very challenging, since LB is still depending on the bulk of circulating tumor cells / nuclei acids from a given neoplasm [1,78]; ii) improving LB specificity, also considering the discrepancies of the molecular profiling between LB and tissue samples [14,45]; iii) standardizing sample extraction procedures, starting from pre-analytical platforms to arrive to the several and different methodologies of isolation of circulating tumor cells and related material. This task should aim to extrapolate the use of the different circulating molecules into the “real world” of the clinic. On top of that, the “only one analyte” barrier should be overcome, trying to focus more on multi-omics approaches to finally unleash the full clinical utility of LB [79]; iv) lastly, as this study clearly shows, high-quality meta-research is urgently needed to better evaluate the real impact of LB into the clinic, along with its intrinsic potentialities and limitations. Systematic reviews with meta-analyses represent indeed one of the most important tools for providing evidence-based quantitative summary on a topic. Based on the results of our umbrella review, however, we clearly show that current literature based on meta-analyses on LB suffers from critical methodological issues. Thus, upcoming meta-research efforts along this line should consider our findings, trying to address such crucial limitations.
Our study does have some limitations. First, all the meta-analytic outcomes were practically about cancer, thus limiting possible quantitative explorations on the role of LB in different areas of medicine. Second, according to the AMSTAR 2 evaluation, the systematic reviews and meta-analysis included were rated as critically low or low quality, possibly introducing critical biases. Finally, umbrella reviews were limited to outcomes studied in the respective meta‐analyses and do not provide in‐depth data on disease severity, dose–response effects, or specific subgroups such as by sex, environmental conditions, or age.
In conclusion, the literature about LB is exponentially increasing and some promising data about cancer are available to date. However, no definitive statements can yet be claimed in relation to LB since the meta-analyses published to date are affected by several methodological limitations such as missing data about heterogeneity or publication bias, overall indicating that the true diagnostic value of LB could be substantially different from gold standard techniques, therefore indicating that high-quality meta-research is urgently needed.
Funding with award/grant number
None.
Ethics approval and consent to participate
Being a systematic review with meta-analysis of previously published literature, no informed consent was obtained and we did not ask for the approval to our local Ethical Committee.
Data available statement
The data underlying this article will be shared on reasonable request to the corresponding author. All authors declare manuscript is an honest, accurate, and transparent account of the study being reported; no important aspects of the study have been omitted; and that any discrepancies from the study as originally planned (and, if relevant, registered) have been explained in published protocol.
CRediT authorship contribution statement
Nicola Veronese: Writing – original draft, Formal analysis. Claudio Luchini: Writing – review & editing. Stefano Ciriminna: Formal analysis, Data curation. Katia Spinelli: Data curation. Santo Fruscione: Data curation. Paola Mattiolo: Data curation. Miriam Belluzzo: Data curation. Veronica Messina: Data curation. Lee Smith: Writing – review & editing. Mario Barbagallo: Writing – review & editing. Walter Mazzucco: Writing – review & editing.
Declaration of competing interest
None.
Footnotes
Supplementary material associated with this article can be found, in the online version, at doi:10.1016/j.tranon.2024.102172.
Appendix. Supplementary materials
References
- 1.Lone S.N., Nisar S., Masoodi T., Singh M., Rizwan A., Hashem S., El-Rifai W., Bedognetti D., Batra S.K., Haris M., Bhat A.A., Macha M.A. Liquid biopsy: a step closer to transform diagnosis, prognosis and future of cancer treatments. Mol. Cancer. 2022;21:79. doi: 10.1186/s12943-022-01543-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Blackhall F., Frese K.K., Simpson K., Kilgour E., Brady G., Dive C. Will liquid biopsies improve outcomes for patients with small-cell lung cancer? Lancet Oncol. 2018;19:e470–e481. doi: 10.1016/S1470-2045(18)30455-8. [DOI] [PubMed] [Google Scholar]
- 3.Corcoran R.B., Chabner B.A. Application of cell-free DNA analysis to cancer treatment. N Engl. J. Med. 2018;379:1754–1765. doi: 10.1056/NEJMra1706174. [DOI] [PubMed] [Google Scholar]
- 4.Bettegowda C., Sausen M., Leary R.J., Kinde I., Wang Y., Agrawal N., Bartlett B.R., Wang H., Luber B., Alani R.M., Antonarakis E.S., Azad N.S., Bardelli A., Brem H., Cameron J.L., Lee C.C., Fecher L.A., Gallia G.L., Gibbs P., Le D., Giuntoli R.L., Goggins M., Hogarty M.D., Holdhoff M., Hong S.-M., Jiao Y., Juhl H.H., Kim J.J., Siravegna G., Laheru D.A., Lauricella C., Lim M., Lipson E.J., Marie S.K.N., Netto G.J., Oliner K.S., Olivi A., Olsson L., Riggins G.J., Sartore-Bianchi A., Schmidt K., Shih -M., Oba-Shinjo S.M., Siena S., Theodorescu D., Tie J., Harkins T.T., Veronese S., Wang T.-L., Weingart J.D., Wolfgang C.L., Wood L.D., Xing D., Hruban R.H., Wu J., Allen P.J., Schmidt C.M., Choti M.A., Velculescu V.E., Kinzler K.W., Vogelstein B., Papadopoulos N., Diaz L.A. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci. Transl. Med. 2014;6:224ra24. doi: 10.1126/scitranslmed.3007094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Mattox A.K., Bettegowda C., Zhou S., Papadopoulos N., Kinzler K.W., Vogelstein B. Applications of liquid biopsies for cancer. Sci. Transl. Med. 2019;11:eaay1984. doi: 10.1126/scitranslmed.aay1984. [DOI] [PubMed] [Google Scholar]
- 6.Cohen J.D., Li L., Wang Y., Thoburn C., Afsari B., Danilova L., Douville C., Javed A.A., Wong F., Mattox A., Hruban R.H., Wolfgang C.L., Goggins M.G., Dal Molin M., Wang T.-L., Roden R., Klein A.P., Ptak J., Dobbyn L., Schaefer J., Silliman N., Popoli M., Vogelstein J.T., Browne J.D., Schoen R.E., Brand R.E., Tie J., Gibbs P., Wong H.-L., Mansfield A.S., Jen J., Hanash S.M., Falconi M., Allen P.J., Zhou S., Bettegowda C., Diaz L.A., Tomasetti C., Kinzler K.W., Vogelstein B., Lennon A.M., Papadopoulos N. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science. 2018;359:926–930. doi: 10.1126/science.aar3247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Carr D.J., Welch H.G. Assessing the clinical utility of liquid biopsies across 5 potential indications from therapy selection to population screening: a review. JAMA Int. Med. 2023;183:1144–1151. doi: 10.1001/jamainternmed.2023.3603. [DOI] [PubMed] [Google Scholar]
- 8.Husain H., Velculescu V.E. Cancer DNA in the Circulation: the Liquid Biopsy. JAMA. 2017;318:1272–1274. doi: 10.1001/jama.2017.12131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Crosby D., Bhatia S., Brindle K.M., Coussens L.M., Dive C., Emberton M., Esener S., Fitzgerald R.C., Gambhir S.S., Kuhn P., Rebbeck T.R., Balasubramanian S. Early detection of cancer. Science. 2022;375:eaay9040. doi: 10.1126/science.aay9040. [DOI] [PubMed] [Google Scholar]
- 10.Crowley E., Di Nicolantonio F., Loupakis F., Bardelli A. Liquid biopsy: monitoring cancer-genetics in the blood. Nat. Rev. Clin. Oncol. 2013;10:472–484. doi: 10.1038/nrclinonc.2013.110. [DOI] [PubMed] [Google Scholar]
- 11.Han D., Li R., Shi J., Tan P., Zhang R., Li J. Liquid biopsy for infectious diseases: a focus on microbial cell-free DNA sequencing. Theranostics. 2020;10:5501–5513. doi: 10.7150/thno.45554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Schobers G., Koeck R., Pellaers D., Stevens S.J.C., Macville M.V.E., Paulussen A.D.C., Coonen E., van den Wijngaard A., de Die-Smulders C., de Wert G., Brunner H.G., Esteki M.Z. Liquid biopsy: state of reproductive medicine and beyond. Hum Reprod. 2021;36:2824–2839. doi: 10.1093/humrep/deab206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Verhoeven J.G.H.P., Boer K., Van Schaik R.H.N., Manintveld O.C., Huibers M.M.H., Baan C.C., Hesselink D.A. Liquid biopsies to monitor solid organ transplant function: a review of new biomarkers. Ther. Drug Monit. 2018;40:515–525. doi: 10.1097/FTD.0000000000000549. [DOI] [PubMed] [Google Scholar]
- 14.Luchini C., Veronese N., Nottegar A., Cappelletti V., Daidone M.G., Smith L., Parris C., Brosens L.A.A., Caruso M.G., Cheng L., Wolfgang C.L., Wood L.D., Milella M., Salvia R., Scarpa A. Liquid biopsy as surrogate for tissue for molecular profiling in pancreatic cancer: a meta-analysis towards precision medicine. Cancers. 2019;11:1152. doi: 10.3390/cancers11081152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Batool S.M., Yekula A., Khanna P., Hsia T., Gamblin A.S., Ekanayake E., Escobedo A.K., You D.G., Castro C.M., Im H., Kilic T., Garlin M.A., Skog J., Dinulescu D.M., Dudley J., Agrawal N., Cheng J., Abtin F., Aberle D.R., Chia D., Elashoff D., Grognan T., Krysan K., Oh S.S., Strom C., Tu M., Wei F., Xian R.R., Skates S.J., Zhang D.Y., Trinh T., Watson M., Aft R., Rawal S., Agarwal A., Kesmodel S.B., Yang C., Shen C., Hochberg F.H., Wong D.T.W., Patel A.A., Papadopoulos N., Bettegowda C., Cote R.J., Srivastava S., Lee H., Carter B.S., Balaj L. The liquid biopsy consortium: challenges and opportunities for early cancer detection and monitoring. Cell Rep. Med. 2023;4 doi: 10.1016/j.xcrm.2023.101198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Higgins J.P.T., Thomas J., Chandler J., Cumpston M., Tianjing L., Page M.J., Welch V.A. 2nd ed. John Wiley & Sons, Ltd; ChichestertUK: 2019. Cochrane Handbook for Systematic Reviews of Interventions. [Google Scholar]
- 17.Page M.J., McKenzie J.E., Bossuyt P.M., Boutron I., Hoffmann T.C., Mulrow C.D., Shamseer L., Tetzlaff J.M., Akl E.A., Brennan S.E., Chou R., Glanville J., Grimshaw J.M., Hróbjartsson A., Lalu M.M., Li T., Loder E.W., Mayo-Wilson E., McDonald S., McGuinness L.A., Stewart L.A., Thomas J., Tricco A.C., Welch V.A., Whiting P., Moher D. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. Syst. Rev. 2021;10:89. doi: 10.1186/s13643-021-01626-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Ouzzani M., Hammady H., Fedorowicz Z., Elmagarmid A. Rayyan-a web and mobile app for systematic reviews. Syst. Rev. 2016;5:210. doi: 10.1186/s13643-016-0384-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Patridge E.F., Bardyn T.P. Research Electronic Data Capture (REDCap) J. Med. Libr. Assoc. 2018;106:142–144. doi: 10.5195/jmla.2018.319. [DOI] [Google Scholar]
- 20.Shea B.J., Reeves B.C., Wells G., Thuku M., Hamel C., Moran J., Moher D., Tugwell P., Welch V., Kristjansson E., Henry D.A. AMSTAR 2: a critical appraisal tool for systematic reviews that include randomised or non-randomised studies of healthcare interventions, or both. BMJ. 2017;358:j4008. doi: 10.1136/bmj.j4008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Luchini C., Veronese N., Nottegar A., Shin J.I., Gentile G., Granziol U., Soysal P., Alexinschi O., Smith L., Solmi M. Assessing the quality of studies in meta-research: review/guidelines on the most important quality assessment tools. Pharm Stat. 2021;20:185–195. doi: 10.1002/pst.2068. [DOI] [PubMed] [Google Scholar]
- 22.Farmer C., Fenu E., O'Flynn N., Guthrie B. Clinical assessment and management of multimorbidity: summary of NICE guidance. BMJ. 2016;354:i4843. doi: 10.1136/bmj.i4843. [DOI] [PubMed] [Google Scholar]
- 23.Hanley J.A., McNeil B.J. The meaning and use of the area under a receiver operating characteristic (ROC) curve. Radiology. 1982;143:29–36. doi: 10.1148/radiology.143.1.7063747. [DOI] [PubMed] [Google Scholar]
- 24.Siontis G.C.M., Tzoulaki I., Ioannidis J.P.A. Predicting death: an empirical evaluation of predictive tools for mortality. Arch. Int. Med. 2011;171:1721–1726. doi: 10.1001/archinternmed.2011.334. [DOI] [PubMed] [Google Scholar]
- 25.Iorio A., Spencer F.A., Falavigna M., Alba C., Lang E., Burnand B., McGinn T., Hayden J., Williams K., Shea B., Wolff R., Kujpers T., Perel P., Vandvik P.O., Glasziou P., Schunemann H., Guyatt G. Use of GRADE for assessment of evidence about prognosis: rating confidence in estimates of event rates in broad categories of patients. BMJ. 2015;350:h870. doi: 10.1136/bmj.h870. [DOI] [PubMed] [Google Scholar]
- 26.Higgins J.P.T., Thompson S.G., Deeks J.J., Altman D.G. Measuring inconsistency in meta-analyses. BMJ. 2003;327:557–560. doi: 10.1136/bmj.327.7414.557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Bai S., Lin S., Lin T., Wang Q., Cheng C., Lin J., Zhang Y., Jiang X., Han X. Clinical diagnostic biomarker “circulating tumor cells” in breast cancer - a meta-analysis. Front. Oncol. 2023;13 doi: 10.3389/fonc.2023.1137519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Egger M., Smith G.D, Schneider M., Minder C. Bias in meta-analysis detected by a simple, graphical test. BMJ. 1997;315:629–634. doi: 10.1136/bmj.315.7109.629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Borg M., Wen S.W.C., Andersen R.F., Timm S., Hansen T.F., Hilberg O. Methylated circulating tumor DNA in blood as a tool for diagnosing lung cancer: a systematic review and meta-analysis. Cancers. 2023;15:3959. doi: 10.3390/cancers15153959. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.T.R. Tonozzi, A. Kammesheidt, G.D. Braunstein, Liquid biopsies in endocrine neoplasia—a systematic review, 1 15 (2019) 39–44. doi:10.17925/USE.2019.15.1.39.
- 31.Chandrapalan S., Bannaga A., Weidner A., Hitchins M.P., Arasaradnam R.P. A systematic review and meta-analysis: the diagnostic accuracy of methylated SEPTIN9 for the detection of hepatocellular carcinoma and the clinical evaluation of its use in combination with other surveillance modalities. Scand. J. Gastroenterol. 2022;57:473–480. doi: 10.1080/00365521.2021.2020331. [DOI] [PubMed] [Google Scholar]
- 32.Cheng J., Tang Q., Cao X., Burwinkel B. Cell-Free Circulating DNA integrity based on peripheral blood as a biomarker for diagnosis of cancer: a systematic review. Cancer Epidemiol. Biomarkers Prev. 2017;26:1595–1602. doi: 10.1158/1055-9965.EPI-17-0502. [DOI] [PubMed] [Google Scholar]
- 33.Dabbagh Ohadi M.A., Aleyasin M.S., Samiee R., Bordbar S., Maroufi S.F., Bayan N., Hanaei S., Smith T.R. Micro RNAs as a diagnostic marker between glioma and primary CNS lymphoma: a systematic review. Cancers. 2023;15:3628. doi: 10.3390/cancers15143628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Duque G., Manterola C., Otzen T., Arias C., Palacios D., Mora M., Galindo B., Holguín J.P., Albarracín L. Cancer biomarkers in liquid biopsy for early detection of breast cancer: a systematic review. Clin. Med. Insights Oncol. 2022;16 doi: 10.1177/11795549221134831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Elasifer H., Amukwaya M.M.N., Bhatia R., Cuschieri K., Gregory J.M. The role of circulating viral and tumour DNA in the diagnosis and management of HPV associated anogenital cancers, a systematic review and meta-analysis. J. Clin. Virol. 2023;164 doi: 10.1016/j.jcv.2023.105469. [DOI] [PubMed] [Google Scholar]
- 36.Gally T.B., Aleluia M.M., Borges G.F., Kaneto C.M. Circulating MicroRNAs as novel potential diagnostic biomarkers for osteosarcoma: a systematic review. Biomolecules. 2021;11:1432. doi: 10.3390/biom11101432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.He Y., Wang Y., Li Z., Chen H., Deng J., Huang H., He X., Zeng W., Liu M., Huang B., Chen P. Clinical performance of non-invasive prenatal testing for trisomies 21, 18 and 13 in twin pregnancies: a cohort study and a systematic meta-analysis. Acta Obstet Gynecol. Scand. 2020;99:731–743. doi: 10.1111/aogs.13842. [DOI] [PubMed] [Google Scholar]
- 38.Hong F., Li N., Feng Z., Zheng Y., Zhu C., Zhang F. Exosomal microRNAs as novel diagnostic biomarkers in breast cancer: a systematic evaluation and meta-analysis. Asian J. Surg. 2023;46:4727–4736. doi: 10.1016/j.asjsur.2023.05.115. [DOI] [PubMed] [Google Scholar]
- 39.Hou F., Sun X.-D., Deng Z.-Y. Diagnostic value of cell-free DNA in thyroid cancer: a systematic review and meta-analysis. Medicine. 2023;102:e32928. doi: 10.1097/MD.0000000000032928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Jia S., Xie L., Li L., Qian Y., Wang J., Wang S., Zhang W., Qian B. Values of liquid biopsy in early detection of cancer: results from meta-analysis. Expert Rev. Mol. Diagn. 2021;21:417–427. doi: 10.1080/14737159.2021.1910025. [DOI] [PubMed] [Google Scholar]
- 41.Karkia R., Wali S., Payne A., Karteris E., Chatterjee J. Diagnostic accuracy of liquid biomarkers for the non-invasive diagnosis of endometrial cancer: a systematic review and meta-analysis. Cancers. 2022;14:4666. doi: 10.3390/cancers14194666. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Khetrapal P., Lee M.W.L., Tan W.S., Dong L., de Winter P., Feber A., Kelly J.D. The role of circulating tumour cells and nucleic acids in blood for the detection of bladder cancer: a systematic review. Cancer Treat Rev. 2018;66:56–63. doi: 10.1016/j.ctrv.2018.03.007. [DOI] [PubMed] [Google Scholar]
- 43.Leão R., Albersen M., Looijenga L.H.J., Tandstad T., Kollmannsberger C., Murray M.J., Culine S., Coleman N., Belge G., Hamilton R.J., Dieckmann K.-P. Circulating MicroRNAs, the next-generation serum biomarkers in testicular germ cell tumours: a systematic review. Eur. Urol. 2021;80:456–466. doi: 10.1016/j.eururo.2021.06.006. [DOI] [PubMed] [Google Scholar]
- 44.McMahon J.T., Studer M., Ulrich B., Revuelta Barbero J.M., Pradilla I., Palacios-Ariza M.A., Pradilla G. Circulating tumor DNA in adults with glioma: a systematic review and meta-analysis of biomarker performance. Neurosurgery. 2022;91:231–238. doi: 10.1227/neu.0000000000001982. [DOI] [PubMed] [Google Scholar]
- 45.Mlika M., Dziri C., Zorgati M.M., Ben Khelil M., Mezni F. Liquid biopsy as surrogate to tissue in lung cancer for molecular profiling: a meta-analysis. Curr. Respir. Med. Rev. 2018;14:48–60. doi: 10.2174/1573398X14666180430144452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Nakasu Y., Deguchi S., Nakasu S., Yamazaki M., Notsu A., Mitsuya K., Hayashi N. Diagnostic accuracy of cerebrospinal fluid liquid biopsy and MRI for leptomeningeal metastases in solid cancers: a systematic review and meta-analysis. Neurooncol. Adv. 2023;5:vdad002. doi: 10.1093/noajnl/vdad002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Öberg K., Califano A., Strosberg J.R., Ma S., Pape U., Bodei L., Kaltsas G., Toumpanakis C., Goldenring J.R., Frilling A., Paulson S. A meta-analysis of the accuracy of a neuroendocrine tumor mRNA genomic biomarker (NETest) in blood. Ann. Oncol. 2020;31:202–212. doi: 10.1016/j.annonc.2019.11.003. [DOI] [PubMed] [Google Scholar]
- 48.Rapado-González O., Martínez-Reglero C., Salgado-Barreira A., López-López R., Suárez-Cunqueiro M.M., Muinelo-Romay L. miRNAs in liquid biopsy for oral squamous cell carcinoma diagnosis: systematic review and meta-analysis. Oral Oncol. 2019;99 doi: 10.1016/j.oraloncology.2019.104465. [DOI] [PubMed] [Google Scholar]
- 49.Rapado-González Ó., Martínez-Reglero C., Salgado-Barreira Á., Muinelo-Romay L., Muinelo-Lorenzo J., López-López R., Díaz-Lagares Á., Suárez-Cunqueiro M.M. Salivary DNA methylation as an epigenetic biomarker for head and neck cancer. part I: a diagnostic accuracy meta-analysis. J. Pers. Med. 2021;11:568. doi: 10.3390/jpm11060568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Rapado-González Ó., Martínez-Reglero C., Salgado-Barreira Á., Takkouche B., López-López R., Suárez-Cunqueiro M.M., Muinelo-Romay L. Salivary biomarkers for cancer diagnosis: a meta-analysis. Ann. Med. 2020;52:131–144. doi: 10.1080/07853890.2020.1730431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Song Z., Wang S., Liu Y. The diagnostic accuracy of liquid exosomes for lung cancer detection: a meta-analysis. Onco. Targets Ther. 2019;12:181–192. doi: 10.2147/OTT.S188832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Toraih E.A., Elshazli R.M., Trinh L.N., Hussein M.H., Attia A.A., Ruiz E.M.L., Zerfaoui M., Fawzy M.S., Kandil E. Diagnostic and prognostic performance of liquid biopsy-derived exosomal MicroRNAs in thyroid cancer patients: a systematic review and meta-analysis. Cancers. 2021;13:4295. doi: 10.3390/cancers13174295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Uhe I., Hagen M.E., Ris F., Meyer J., Toso C., Douissard J. Cell-free DNA liquid biopsy for early detection of gastrointestinal cancers: a systematic review. World J. Gastrointest Oncol. 2021;13:1799–1812. doi: 10.4251/wjgo.v13.i11.1799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.van Asperen J.V., Fedorushkova D.M., Robe P.A.J.T., Hol E.M. Investigation of glial fibrillary acidic protein (GFAP) in body fluids as a potential biomarker for glioma: a systematic review and meta-analysis. Biomarkers. 2022;27:1–12. doi: 10.1080/1354750X.2021.2006313. [DOI] [PubMed] [Google Scholar]
- 55.van Westrhenen A., Smidt L.C.A., Seute T., Nierkens S., Stork A.C.J., Minnema M.C., Snijders T.J. Diagnostic markers for CNS lymphoma in blood and cerebrospinal fluid: a systematic review. Br. J. Haematol. 2018;182:384–403. doi: 10.1111/bjh.15410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Wuerdemann N., Jain R., Adams A., Speel E.-J.M., Wagner S., Joosse S.A., Klussmann J.P. Cell-Free HPV-DNA as a biomarker for oropharyngeal squamous cell Carcinoma-a step towards personalized medicine? Cancers. 2020;12:2997. doi: 10.3390/cancers12102997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Xu Y., Jiang Y., Yu M., Lou J., Song M., Xu H., Cui Y., Zeng X., Wang Q., Ma H., Wang Z., Zhu S., Li G., Zhao A. Meta-analysis of the diagnostic value of cell-free DNA for Renal cancer. Front Mol. Biosci. 2021;8 doi: 10.3389/fmolb.2021.683844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Xu Y., Lou J., Yu M., Jiang Y., Xu H., Huang Y., Gao Y., Wang H., Li G., Wang Z., Zhao A. Urinary exosomes diagnosis of urological tumors: a systematic review and meta-analysis. Front Oncol. 2021;11 doi: 10.3389/fonc.2021.734587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Ye J., Wu M., He L., Chen P., Liu H., Yang H. Glutathione-S-transferase p1 gene promoter methylation in Cell-Free DNA as a diagnostic and prognostic tool for prostate cancer: a systematic review and meta-analysis. Int. J. Endocrinol. 2023;2023 doi: 10.1155/2023/7279243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Yinzhong W., Miaomiao W., Xiaoxue T., Qian W., Meng Q., Junqiang L. Diagnostic accuracy of circulating-free DNA for the determination of hepatocellular carcinoma: a systematic review and meta-analysis. Expert Rev. Mol. Diagn. 2023;23:63–69. doi: 10.1080/14737159.2023.2167555. [DOI] [PubMed] [Google Scholar]
- 61.Yu W., Liu R., Miao Z., Zhang L., Sheyhidin I., Ainiwaer J. The diagnostic significance of blood-derived circRNAs in NSCLC: systematic review and meta-analysis. Front Oncol. 2022;12 doi: 10.3389/fonc.2022.987704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Zhang C., Chao F., Wang S., Han D., Chen G. Cell-free DNA as a Promising diagnostic biomarker in prostate cancer: a systematic review and meta-analysis. J. Oncol. 2022;2022 doi: 10.1155/2022/1505087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Zhang J., Qian D., Xu X., Xu M., Wang K., Lu H., Shen G. Evaluating the diagnostic potentials of circulating tumor DNA against melanoma: a systematic review and meta-analysis. J. Oncol. 2022;2022 doi: 10.1155/2022/6233904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Zhao Q., Yuan Z., Wang H., Zhang H., Duan G., Zhang X. Role of circulating tumor cells in diagnosis of lung cancer: a systematic review and meta-analysis. J. Int. Med. Res. 2021;49 doi: 10.1177/0300060521994926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Zhou S., Huang R., Cao Y. Detection of epidermal growth factor receptor mutations in peripheral blood circulating tumor DNA in patients with advanced non-small cell lung cancer: a PRISMA-compliant meta-analysis and systematic review. Medicine. 2020;99:e21965. doi: 10.1097/MD.0000000000021965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Zhou Z., Xiong H., Xie F., Wu Z., Feng Y. A meta-analytic review of the value of miRNA for multiple sclerosis diagnosis. Front Neurol. 2020;11:132. doi: 10.3389/fneur.2020.00132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Zhu Y., Yang T., Wu Q., Yang X., Hao J., Deng X., Yang S., Gu C., Wang Z. Diagnostic performance of various liquid biopsy methods in detecting colorectal cancer: a meta-analysis. Cancer Med. 2020;9:5699–5707. doi: 10.1002/cam4.3276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Zhu Y., Zhang H., Chen N., Hao J., Jin H., Ma X. Diagnostic value of various liquid biopsy methods for pancreatic cancer: a systematic review and meta-analysis. Medicine. 2020;99:e18581. doi: 10.1097/MD.0000000000018581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Meng H., Yao W., Nan M.-M., Zhou F.-G., Song W.-Y., Li Y.-Z., Yin Y.-H., Ding Y. The diagnostic value of circulating abnormal cells in early lung cancer. Am. J. Cancer Res. 2023;13:1594–1601. [PMC free article] [PubMed] [Google Scholar]
- 70.Zhu C., Zhuang W., Chen L., Yang W., Ou W.-B. Frontiers of ctDNA, targeted therapies, and immunotherapy in non-small-cell lung cancer. Transl. Lung Cancer Res. 2020;9 doi: 10.21037/tlcr.2020.01.09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Duréndez-Sáez E., Torres-Martinez S., Calabuig-Fariñas S., Meri-Abad M., Ferrero-Gimeno M., Camps C. Exosomal microRNAs in non-small cell lung cancer. Transl. Cancer Res. 2021;10:3128–3139. doi: 10.21037/tcr-20-2815. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Luchini C., Pea A., Scarpa A. Artificial intelligence in oncology: current applications and future perspectives. Br. J. Cancer. 2022;126:4–9. doi: 10.1038/s41416-021-01633-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Topol E.J. Medical forecasting. Science. 2024;384:eadp7977. doi: 10.1126/science.adp7977. [DOI] [PubMed] [Google Scholar]
- 74.Alix-Panabières C., Pantel K. Liquid biopsy: from discovery to clinical application. Cancer Discov. 2021;11:858–873. doi: 10.1158/2159-8290.CD-20-1311. [DOI] [PubMed] [Google Scholar]
- 75.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. doi: 10.1038/nrc.2017.7. [DOI] [PubMed] [Google Scholar]
- 76.Hendricks B.K., Cohen-Gadol A.A., Miller J.C. Novel delivery methods bypassing the blood-brain and blood-tumor barriers. Neurosurg. Focus. 2015;38:E10. doi: 10.3171/2015.1.FOCUS14767. [DOI] [PubMed] [Google Scholar]
- 77.Aran V., de J.O., Junior M., Heming C.P., Zeitune D.J., Neto V.M., Filho P.N. Unveiling the impact of corticosteroid therapy on liquid biopsy-detected cell-free DNA levels in meningioma and glioblastoma patients. J. Liquid Biopsy. 2024;5 doi: 10.1016/j.jlb.2024.100149. [DOI] [Google Scholar]
- 78.Papier K., Atkins J.R., Tong T.Y.N., Gaitskell K., Desai T., Ogamba C.F., Parsaeian M., Reeves G.K., Mills I.G., Key T.J., Smith-Byrne K., Travis R.C. Identifying proteomic risk factors for cancer using prospective and exome analyses of 1463 circulating proteins and risk of 19 cancers in the UK Biobank. Nat. Commun. 2024;15:4010. doi: 10.1038/s41467-024-48017-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Alba-Bernal A., Lavado-Valenzuela R., Domínguez-Recio M.E., Jiménez-Rodriguez B., Queipo-Ortuño M.I., Alba E., Comino-Méndez I. Challenges and achievements of liquid biopsy technologies employed in early breast cancer. EBioMedicine. 2020;62 doi: 10.1016/j.ebiom.2020.103100. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.