Highlights:
-
•
Article have discussed need of liquid biopsy-based biomarker discovery for early diagnosis, treatment response, and disease prognosis.
-
•
Article have discussed about the types of liquid biopsies.
-
•
The article discusses the clinical utility of liquid biopsies for diagnostic, prognostic, and potential therapeutic purposes in solid tumors.
-
•
Article have discussed limitation and future prospects of liquid biopsies.
Keywords: Liquid biopsies, solid tumours, circulating tumour DNA, cell-free DNA, circulating tumour cells
Abstract
Late detection and lack of precision diagnostics are the major challenges in cancer prevention and management. Biomarker discovery in specific cancers, especially at the pre-invasive stage, is vital for early diagnosis, positive treatment response, and good disease prognosis. Traditional diagnostic measures require invasive procedures such as tissue excision using a needle, an endoscope, and/or surgical resection which can be unsafe, expensive, and painful. Additionally, the presence of comorbid conditions in individuals might render them ineligible for undertaking a tissue biopsy, and in some cases, it is difficult to access tumours depending on the site of occurrence. In this context, liquid biopsies are being explored for their clinical significance in solid malignancies management. These non-invasive or minimally invasive methods are being developed primarily for identification of biomarkers for early diagnosis and targeted therapeutics. In this review, we have summarised the use and importance of liquid biopsy as significant tool in diagnosis, prognosis prediction, and therapeutic development. We have also discussed the challenges that are encountered and future perspective.
Graphical abstract
Introduction
Technological advancements have led to significant improvement in areas of cancer diagnostics and therapeutics. Newer and highly sensitive and specific tools for tumour profiling and management are being commercialised or under development. However, the conventional biopsy methods available for tumour profiling are invasive, and also pose challenges in specific cases where tissue accessibility is a concern and also in patients with comorbidity. In this context, developing non-invasive liquid biopsy methods for early cancer detection has been the prime focus of researchers [1].
“Liquid biopsy" (LB) refers to a method for detecting the presence of specific molecular markers of a disease in liquid samples including blood, saliva, urine, and other minimally invasive biological samples [2]. LB is emerging as a major realm in the precision oncology approach [3]. LB has been established as an innovative diagnostic measure by analysing the Cell-free DNA (cfDNA), exosomes, circulating tumour cells (CTCs), proteins/ peptides, and circulating tumour DNA (ctDNA) [4] in biological fluids. A clear and comprehensive picture of these multiple components and molecules in the body fluids in specific cancers will lead to early and improved cancer detection, better cancer management, predicting prognosis, and effective monitoring of therapeutic response [5] (Fig. 1).
Fig. 1.
An overview of liquid biopsies and their applications. (Created with BioRender.com)
As of now, few LB based methods for cancer detection are available commercially. In lung and colorectal cancer (CRC), US-FDA has approved ctDNA-based diagnostic markers [6] and the CELLSEARCH® CTCs as a metastatic diagnostics tool for colon, breast, and prostate cancer (PC) [7,8]. Despite their clinical significance, utility of these methods is limited, as they are not globally recognized and are not part of a standard cancer diagnosis as well. Solid tumours are quite complex in their origin, either due to genetic aberrations or environmental insults [9]. LBs have been very advantageous in the detection and profiling of solid tumours in order to get an overall tumour progression scenario [7,10]. In this review, we have described the different types of liquid biopsies components and their clinical utility with respect to diagnostic, prognostic, and possible therapeutic uses in solid tumours.
Components of liquid biopsies and their use in solid tumours
Circulating tumour cells (CTC)
CTCs get detached from the site of primary tumour and disseminate into the bloodstream of cancer patients at different stages of cancer progression [11]. These CTCs can be easily isolated from human biological fluids (serum and plasma) and can be used for cancer diagnosis. Sample collection of bodily fluids offers twin advantages of being easily available and patient-friendly than conventional tissue biopsies. More importantly, it can detect tumour cells in the body fluid at an early stage, which allows for effective cancer management and therapeutic response [12,13]. Another significant advantage of these CTCs-based biomarkers in cancer detection is the sensitivity and specificity as chances of false positives are minimal [14].
CTCs were discovered by Thomas Ashworth. Detection and profiling of tumour through CTCs is more feasible than conventional biopsies and has led to the development of novel strategies not only in diagnosis, but also in the monitoring of metastasis and tumour recurrence of solid tumours [15]. Isolation of CTCs from from the primary solid tumours has been established and validated. Firstly, CTCs are collected and enriched based on their physical and biological characteristics. This is followed by detection of CTCs based on their size and electrical properties [13]. CELLSEARCH® was the first FDA-approved method that allowed scientists to enrich and culture CTCs from the peripheral blood of patients [15].
Several studies have reported the clinical significance of CTCs in detecting locally advanced cervical cancer [16,17]. Wen et al. [18] were also able to detect cervical cancer in serum samples of the patients using CTCs along with the Squamous cell carcinoma antigen (SCC-Ag), resulting in the development of an efficient predictive model for cervical cancer progression as compared to a single biomarker alone. Further, the significance of CTCs in Head and Neck cancer has been highlighted where a strong correlation between early clinical outcome and the presence of CTCs in patient samples has been observed, leading to a good prognosis [19].
Furthermore, a study has also compared CTC-based CELLSEARCH® and AdnaTest® tests to determine specificity and sensitivity variations, and observed that the combination of both the detection techniques was highly significant [20]. Because of the infrequent presence of CTCs in blood and their low number (very few in comparison to other blood cells), the development of more reliable CTCs detection approaches for diagnostics and therapeutic applications are under process (Table 1).
Table 1.
List of clinical trials conducted in liquid biopsies
| S. No. | Study title | Cancer | NCT number | Location |
|---|---|---|---|---|
| 1. | Study of Metabolites Markers in Adjuvant Breast Cancer | Breast cancer | NCT03367208 |
|
| 2. | Characterization & Comparison of Drugable Mutations in Primary and Metastatic Tumors, CTCs and cfDNA in MBCpatients | Metastatic Breast cancer | NCT02626039 | |
| 3. | Harvest of CTCs From MBC Patients Using the Parsortix™ PC1 System | Breast cancer | NCT03427450 |
|
| 4. | Determination of RAS Mutation Status in Liquid Biopsies in Subjects With RAS Wild-type.PERSEIDA Study | Colorectal cancer | NCT03957564 |
|
| 5. | Concordance of Inivata Liquid Biopsy with Standard Tissue Biopsy | Non-Small cell lung cancer | NCT03116633 |
|
| 6. | Concordance of Inivata Liquid Biopsy with Standard of Care Tissue Testing | Non-Small cell lung cancer | NCT02906852 |
|
| 7. | TAGRISSO (Osimertinib) in NSCLC Patients in Whom T790 Mutations Are Detected by Liquid Biopsy | Non-Small cell lung cancer | NCT03394118 |
|
| 8. | Therapeutic resistance and Clonal Evolution Assessed with Liquid Biopsy of NSCLC Patients in China |
|
NCT03059641 |
|
| 9. | Clinical Application of Liquid Biopsy for Precise Diagnosis and Prognosis in Lymphoma | Lymphomas | NCT04062877 |
|
| 10. | Study to Detect Biomarker Gradients in Coronary Arteries Using the Liquid Biopsy System | Coronary Heart Disease | NCT02119767 |
|
| 11. | A Study to Evaluate the Value of Circulating Tumour DNA in Follow-up of Patients with an Advanced Gastroenteropancreatic or Lung Neuroendocrine Tumour Under Everolimus +- SSA Treatment | Neuroendocrine Tumors | NCT05255133 |
|
| 12. | The Correlation of PD-L1 Expression in Non-small Lung Cancer Tissue and Peripheral Blood T Cell and Serum. | Non-small Cell Lung Cancer | NCT03073902 |
|
| 13. | Circulating Tumor DNA as Liquid Biopsy in Patients with Stage IV Solid Tumors, a Feasibility Study at MUSC HCC | Adenocarcinoma of the Colon Adenocarcinoma of the Rectum |
NCT03302325 |
|
| 14. | Detection of Circulating Tumor DNA Through Liquid Biopsies in Ovarian Cancer Patients and Evaluation of Prognostic and Predictive Values of Circulating Tumor DNA Assay. | Ovarian cancer | NCT05504174 |
|
| 15. | Liquid Biopsy Using Methylation Sequencing for Lung Cancer |
|
NCT04253509 |
|
| 16. | Olmutinib Trial in T790M (+) NSCLC Patients Detected by Liquid Biopsy Using BALF Extracellular Vesicular DNA |
|
NCT03228277 |
|
| 17. | Collection of Blood, Urine, and Stool to Monitor Metastatic Colorectal Cancers | Metastatic Colorectal cancer | NCT03563651 |
|
| 18. | Study of Circulating Tumor DNA (ctDNA) Kinetics in Immuno-oncology (IO-KIN) |
|
NCT04606940 |
|
| 19. | A Comprehensive Evaluation of Circulating Tumor DNA and Circulating Tumor Cells as a Predictive Marker in Lung Cancer |
|
NCT04254497 | |
| 20. | AI-EMERGE: Development and Validation of a Multi-analyte, Blood-based Colorectal Cancer Screening Test |
|
NCT03688906 |
|
| 21. | Noninvasive vs. Invasive Lung Evaluation |
|
NCT03615443 | |
| 22. | Mutational Landscape in Hepatocellular Carcinoma |
|
NCT03071458 |
|
| 23. | Use of Exome Sequence Analysis and Circulating Tumour in Assessing Tumour Heterogeneity in BRAF Mutant Melanoma |
|
NCT02251314 |
|
| 24. | Characterization & Comparison of Drugable Mutations in Primary and Metastatic Tumors, CTCs and cfDNA in MBCpatients |
|
NCT02626039 |
|
| 25. | Harvest of CTCs From MBC Patients Using the Parsortix™ PC1 System |
|
NCT03427450 |
|
| 26. | Multicenter Validation of the Sensitivity of Theranostic ALK Rearrangement Detection by FISH Analysis and Prevalence of Escaping Mutations in Circulating Tumor Cells for the Non-invasive Management of Lung Cancer Patients |
|
NCT02372448 |
|
| 27. | Circulating Tumor Cells and Tumor DNA in HCC and NET |
|
NCT02973204 |
|
| 28. | Circulating Tumour Cells in Somatuline Autogel Treated NeuroEndocrine Tumours Patients |
|
NCT02075606 |
|
| 29. | Diagnostic Accuracy of Circulating Tumor Cells (CTCs) and Onco-exosome Quantification in the Diagnosis of Pancreatic Cancer - PANC-CTC |
|
NCT03032913 |
|
| 30. | Liquid Biopsy in Lung Cancer |
|
NCT03479099 |
|
(Source: https://clinicaltrials.gov)
Circulating tumour DNA (ctDNA)
ctDNA-based liquid biopsy is an another tool for molecular diagnosis and surveillance of cancer [21]. Once released from tumour cells, ctDNA harboring original tumour mutations enters the bloodstream. Analysis of this ctDNA from blood samples has paved the way for determining the patients' genetic landscape, helping not only in tumour detection but also in saving time and cost as required for a genetic testing [22,23].
Several studies have established ctDNA as a diagnostic marker for molecular profiling of tumour e.g., in non-small cell lung cancer (NSCLC) and CRC: epidermal growth factor receptor (EGFR) and Vi-Ki-ras2 kirsten rat sarcoma viral oncogene homolog (KRAS) mutation, respectively [24,25]. Another study has shown its role in metastatic breast cancer for early diagnosis of disease and concluded that ctDNA might serve as a highly sensitive biomarker [26]. A study from John Hopkins screened 640 patient samples through the Digital Droplet PCR technique and found 48-73% of patients with colorectal, gastrointestinal, pancreatic, and breast cancer showing the presence of ctDNA [27]. Comparative analysis of BRCA1/2 and DNA repair gene mutation via liquid biopsy using ctDNA and tissue biopsy methods revealed concordance in the genetic landscape [28]. Preclinical and clinical research on ctDNA has shown its role in monitoring treatment response in real-time, selective treatment methods, and feasibility [29].
Cell-free DNA (cfDNA)
In 1948, Mandel and Metais discovered cfDNA, which has been observed to play a vital role in disease diagnosis. Apart from blood, cfDNA is also found in other biological fluids like saliva, urine, and cerebrospinal fluid [30,31]. It is released from cells mostly by apoptosis, necrosis, and active secretion [32]. Previous studies have shown cfDNA as an active biomarker for early cancer detection through digital polymerase chain reaction (dPCR), quantitative PCR (qPCR), and next-generation sequencing (NGS) based detection techniques [33].
A breast cancer study revealed the methylation dynamics of cfDNA during breast cancer progression [34]. An analysis of the genomic characterization of metastatic triple negative breast cancer (TNBC) showed that cfDNA is linked with poor survival in metastatic TNBC cohort [35]. cfDNA estimation and detection is favoured among liquid biopsies because it helps in monitoring the patient's real-time treatment outcome or response towards the targeted therapy [36]. However, cfDNA-based tools need to be validated on a large scale and in different tumours to increase its sensitivity for early diagnosis with high precision.
Exosomes
Exosomes are one of the emerging areas of biomarkers that are biologically active at various stages and also regulate multiple cell-cell interactions in carcinogenesis [37]. Exosomes are small cell-derived nanovesicles that help move the cargo from donor to the receiver cell. Exosomes derived from tumour serves as crucial biomarkers for early cancer detection and prognosis [38]. Exosomes are promising among the several liquid biopsy components that can be assessed due to their existence across all human body fluids and their possibilities for multi-component analysis. The concentration or amount of analytes in membrane-enclosed vesicles might provide greater precision and sensitivity than traditional liquid biopsies [39].
Several studies have shown a potential role of exosomes as diagnostics tools in solid tumours such as NSCLC [40], ovarian cancer [41], breast cancer [42], prostate cancer [43], hepatocellular carcinoma [44], CRC [45]. In a study on PC, exosomes in the blood and urine samples of invasive and metastatic cancer patients showed presence of PC-specific components that increase the detection specificity and sensitivity [46]. In a study on breast cancer, alterations in morphology and cargo of exosomes loaded with molecules like micro-RNAs and long noncoding RNAs were observed to be responsible for the activation of multiple aberrant signalling pathways [47]. Another study on TNBC breast cancer revealed the presence of specific miRNAs like miR246, 134, and 503 thought to be playing a significant role in tumour metastasis, and can be used as a metastatic biomarker [48]. However, cancer detection via exosomes also has its limitations, as some miRNAs encaged in these exosomes have been observed to be common in different cancers which make it difficult to differentiate among specific cancers. Nevertheless, exosomes need to be explored further for identification and development of specific biomarkers for different cancers [47].
Emerging clinical applications of liquid biopsy
Liquid biopsies have multiple benefits and have huge potential in improving multiple facets in cancer management, and tumour characterization, alongwith its implications in diagnosis, prognosis, treatment decisions, and long-term tracking. There are several aspects of cancer management where these liquid biopsies can be utilized, as discussed below in Table 2.
Table 2.
List of some liquid biopsy based available tests approved by FDA / CE-IVD
| S.no. | Test name (Manufacturer and approval) | Biomarker | Cancer Type | References |
|---|---|---|---|---|
| 1. | CellSearch® (Menarini Silocon Biosystems) FDA /CE-IVD | CTC with CD45-, EpCAM+ and (CK8, 18 and/or 19) |
|
[20] |
| 2. | Cobas® EGFR mutation test V2 (Roche) FDA/CE-IVD |
EGFR | NSCLC | [66] |
| 3. | Therascreen (Qiagen) FDA /CE-IVD |
PIK3CA | Breast | [67] |
| KRAS | CRC | [68] | ||
| BRAF | CRC | [68] | ||
| EGFR | NSCLC | [69] | ||
| FGFR | Urothelial | [70] | ||
| 4. | UroVysion (Abbott) FDA /CE-IVD |
Aneuploidy 3,7,17 and loss of 9p21 (p16) | Bladder (patients with haematuria) |
[71] |
| 5. | Epi proColon® (Epigenomics AG) FDA /CE-IVD |
SEPT9 methylation | CRC | [72] |
| 6. | Xpert® Bladder Cancer Detection Xpert® Bladder Cancer Monitor (Cepheid) CE-IVD |
UPK1B, IGF2, CRH, ANXA10, ABL1 |
Bladder (patients with haematuria) NMIBC |
[73] |
| 7. | Uromonitor (Uromonitor) CE-IVD |
FGFR3 and TERT PCR | NMIBC (patients with haematuria) |
[74] |
| 8. | OncoBEAM (Sysmex) CE-IVD |
KRAS & NRAS | mCRC | [75] |
| 9. | Idylla (Biocartis) CE-IVD |
KRAS NRAS, BRAF |
mCRC | [76] |
| 10. | HCCBlood Test (Epigenomics AG) CE-IVD |
SEPT9 methylation | HCC (patients with liver cirrhosis) |
[77] |
| 11. | COLOGUARD (ExactSciences) CE-IVD |
BMP3 & NDRG4 methylation, KRAS, ACTB Haemoglobin |
CRC or advanced adenoma |
[78] |
| 12. | IntPlex® (DiaDx) CE-IVD |
BRAF | mCRC | [79] |
| 13. | Target Selector™ (Biocept) CE-IVD |
EGFR | NSCLC | [80] |
| 14. | Epicheck (Nucleix) CE-IVD |
15 DNA methylation markers | Bladder | [81] |
(FDA: Food & Drug Administration; CE-IVD: In vitro Diagnostic device certification; CDx: Companion diagnostic; mCRC: metastatic colorectal carcinoma; CTC: Circulating tumor cells; NMIBC: Non-muscle invasive bladder cancer; NSCLC: Non-small cell lung carcinoma; HCC: Hepatocellular carcinoma;)
Diagnosis and tumour profiling
As discussed previously, the widely studied analytes of liquid biopsy are cfDNA, ctDNA, exoDNA and CTCs. It has been shown that quantity and quality [49,50] of cfDNA can differentiate between healthy persons and cancer patients. The total amount of cfDNA is usually found to be higher in cancer patients and it increases with disease progression and metastasis [51,52]. ctDNA is another tool which tells about the tumour genetic landscape including copy number variations (CNVs), methylation marks, insertions/ deletions, single nucleotide variants (SNVs) etc. These alterations revealed by LB using ctDNA are quite consistent with the conventional genetic testing through tissue biopsy in several solid tumours like lung [52], breast, colorectal, pancreatic, liver, esophageal, gastric, and ovarian cancers [50]. FDA has also approved tests for genetic testing for ovary, breast, lung and prostate cancer. EGFR Mutation Test v2 is currently being used for the NSCLC patients and is very beneficial for those who face difficulties in providing tumour biopsy samples [53]. Another approved test, Epi proColon® is commercially available which screens CRC via SEPTIN9 gene methylation [54].
Exosomes are released from the cells including tumour cells as membrane enclosed bodies, and reflects a more precise picture than ctDNA [55]. Contrary to exosomes, other membrane-derived vesicles originate from internal compartments known as tumour-associated microparticles (taMPs) that express markers on their cell surface based on their origin. The expression of cancer-specific markers such as CD147 in taMPs and epithelial cell adhesion molecules (EpCAM) are few examples that might be utilised for diagnosis. Further, double-positive taMPs are known to be linked strongly with CRC.
Lastly, CTCs from primary and metastatic tumour sites [55] appear to be augmented during metastasis and staging [56], indicating their diagnostic and prognostic significance. Although, the majority of systems used for CTC isolation, together with the CellSearch platform, are exclusively based on the identification of biomarkers such as EpCAM, and cytokeratins which are present on the cell surface, but an important subpopulation of CTCs may not be detected by these due to the absence of markers on cells that have undergone epithelial-to-mesenchymal transition (EMT) [57], [58], [59]. Efforts are currently being undertaken to combine the isolation/separation of such subsets of cells, e.g., by multiparametric analysis [59,60].
Treatment decision
US-FDA has approved six NGS-based tests for the treatment decision. A US-based company Foundation Medicine has three approvals: FoundationOne CDxTM, FoundationFocusTM CDxBRCA, and FoundationFocusTM CDxBRCA LOH. Among those three, the initial two tests detect somatic, and germline BRCA1/ BRCA2 mutations in ovarian cancer and FoundationFocus™ CDxBRCA LOH is being used for genomic loss of heterogenicity. FoundationOne CDxTM is approved for making treatment decisions in breast, colorectal melanoma, ovary, and NSCLC by identifying the insertions, substitutions, copy number changes, and deletions in 324 genes (FDA PMA P170019). Apart from this, FDA has authorised Illumina's PraxisTM Extended RAS panel to detect NRAS and KRAS mutations in CRC patients receiving Panitumumab therapy. In addition, ThermoFisher's Oncomine™ Dx Target Test is being used for detecting alterations among 23 genes for NSCLC, and MyChoice HRD CDx, (Myriad Genetic Laboratories) (FDA PMA P190014) for mutations in BRCA1 and BRCA2. All of these tests have been authorised for tissue samples. Although a similar test might be used to know the genetic landscape derived from ctDNA, validation studies must be conducted prior to clinical deployment [60].
Although, LB is a highly efficient and non-invasive method of testing compared to tumour tissue biopsy, it still needs to overcome the treatment decision issue, which is the prerequisite for large scale clinical trials demonstrating that using LB in treatment decisions can lead to better disease outcomes. Currently DYNAMIC-III clinical trial is going on to assess whether ctDNA-based chemotherapy decision is more effective than standard care treatment in stage III CRC patients. More clinical trial-based studies on LB are required concerning treatment decisions [60].
Follow-up of Cancer Patients
Apart from the above-mentioned uses, liquid biopsies are also used in monitoring cancer patients' disease progression and therapy response. Their non-invasive or less invasive character makes them an attractive option for disease monitoring for a longer period [49]. Contrarily, getting invasive tissue samples are one of the biggest challenges, and more significantly, these biopsies frequently overlook modifications observed in places apart from the primary tumour site that might impact and alter the therapeutic implications [49]. In addition, the imaging modalities frequently employed to monitor cancer patients undergoing therapy have numerous drawbacks. For instance, computed tomography (CT) scans has many disadvantages like radiation exposure to patients, expensive, less sensitive when detecting minor lesions, and delivering negligible information regarding genetic changes generated by treatment [61,62].
In addition, continuous collection of biofluids permits disease monitoring, therapy response evaluation, and detection of resistance mechanisms. Notably, because of the limited half-life of cfDNA, liquid biopsies permit the real-time monitoring of cancer patients. The utilisation of ctDNA analysis is currently being done to discover mutations in EGFR, ERBB2, PIK3CA, and RAS genes in NSCLC, CRC, and gastric cancer [63], [64], [65]. In addition, dynamics of ctDNA modifications can be monitored which can help in predicting response to treatment as well as clinical outcome and permitting prompt reorientation of treatment regimens. In fact, a decrease in ctDNA after therapy has been linked to a minimal risk of tumour progression and longer survival, whereas high levels of ctDNA have been linked to progression, relapse, and poor survival [60,66].
Compared to ctDNA, CTCs can better predict therapeutic response in comparison to imaging modalities. Identification of CTCc have also been done utilising a panel of genes which was instrumental in differentiating patients with metastatic colorectal cancer (mCRC) who showed no response to therapy, despite CT scans failing to detect them. Besides this, CTC analysis could also detect non-responders with decreased overall survival (OS) and progression-free survival (PFS) [50].
Limitations
Liquid biopsies have emerged as a highly promising tool in cancer diagnosis and monitoring. However, certain challenges related to sensitivity and specificity remains. The amount of ctDNA released into the bloodstream may vary, and tumours with low shedding of ctDNA may go undetected which leads to false-negatives or undefined results [67]. Besides, cancer with a highly complex pathophysiology, LBs may not be able to capture entirely a tumour's genetic or molecular landscape due to the limited amount of ctDNA present in the bloodstream or the release of of DNA from specific regions of the tumour. As a result, specific genetic alterations or mutations present in the tumour might be missed [67]. Further, it does not help localize the disease and does not identify the metastasized sites during tumour progression. Visual inspection is mandatory to detect solid tumours at higher stages, which is one of the major limitations of LB. Similarly, it is also difficult to detect particular cancers in the case of multiple cancers through liquid biopsy. In addition, isolating circulating tumour components is a challenge and requires advanced technologies. Lab to Lab variations, lack of standardized methods and quality control measures are significant issues that remain to be addressed.
Although, LBs hold promise as a non-invasive alternative to traditional tissue biopsies, their clinical utility is currently limited. Liquid biopsies are mainly used for specific cancer types and specific clinical scenarios, such as monitoring treatment response, detecting minimal residual disease, or identifying resistance mutations. Their utility as a primary diagnostic tool for cancer is still being explored [68]. Cost is another major limitation. LBs can be more expensive than traditional tissue biopsies, which limits their accessibility in specific healthcare settings or for patients without adequate insurance coverage [69]. Despite these limitations, LBs have shown great potential in certain applications and continue to be an active area of research and development in the field of cancer diagnostics and monitoring.
Conclusion and future prospective
Circulatory molecular markers are increasingly being researched for their use in precision diagnostics and targeted therapeutics in several diseases and disorders. Some of these markers carry specific disease signatures, and are termed as biomarkers. In this context, LBs present with vast benefits, such as being non-invasive, reproducible, rapid, and real-time. Although, it cannot be considered a replacement for tissue biopsy, but it has been shown to be an excellent alternative that needs to be tapped well. A comprehensive molecular understanding, development of innovative point of care strategies for isolation and precise detection of circulatory tumour components will definitely help in improving accessibility and affordability of LBs in the clinical settings.
Besides, advances in nanobiotechnologies including nanodiagnostics and nanomedicine will further help in tapping the full potential of LBs in solid tumours. As of now, detection of solid tumours using LBs along with visual inspection can help to precisely identify the severity of tumour progression and metastasis for better management. The area of liquid biopsy research has vast possibilities for cancer management, but it needs to be validated in such a manner so it can be used as a gold standard for early detection, disease progression, and treatment outcomes.
Funding
This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.
Ethical statement
Not applicable.
Conflicts of Interest
Authors declare no conflict of interest.
Acknowledgments
We would like to acknowledge the support of Indian Council of Medical Research, New Delhi.
CRediT authorship contribution statement
Sandeep Sisodiya: Writing – original draft, Data curation, Writing – review & editing. Vishakha Kasherwal: Writing – original draft, Data curation, Writing – review & editing. Asiya Khan: Writing – review & editing. Bishnudeo Roy: Writing – review & editing. Anjana Goel: Writing – review & editing. Sandeep Kumar: Writing – review & editing. Nazneen Arif: Writing – review & editing. Pranay Tanwar: Writing – review & editing. Showket Hussain: Conceptualization, Supervision.
Declaration of Competing Interest
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
References
- 1.Palmirotta R., Lovero D., Cafforio P., Felici C., Mannavola F., Pelle E., Quaresmini D., Tucci M., Silvestris F. Liquid biopsy of cancer: a multimodal diagnostic tool in clinical oncology. Ther Adv Med Oncol. 2018;10 doi: 10.1177/1758835918794630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Chen D., Xu T., Wang S., Chang H., Yu T., Zhu Y., Chen J. Liquid Biopsy Applications in the Clinic. Mol Diagn Ther. 2020;24:125–132. doi: 10.1007/s40291-019-00444-8. [DOI] [PubMed] [Google Scholar]
- 3.Nikanjam M., Kato S., Kurzrock R. Liquid biopsy: current technology and clinical applications. J Hematol Oncol. 2022;15:131. doi: 10.1186/s13045-022-01351-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Ijzerman M.J., de Boer J., Azad A., Degeling K., Geoghegan J., Hewitt C., Hollande F., Lee B., To Y.H., Tothill R.W., Wright G., Tie J., Dawson S.-J. Towards Routine Implementation of Liquid Biopsies in Cancer Management: It Is Always Too Early, until Suddenly It Is Too Late. Diagnostics. 2021 doi: 10.3390/diagnostics11010103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Ding Y., Li W., Wang K., Xu C., Hao M., Ding L. Perspectives of the Application of Liquid Biopsy in Colorectal Cancer. BioMed Research International. 2020;2020 doi: 10.1155/2020/6843180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Venook A.P. Colorectal Cancer Surveillance With Circulating Tumour DNA Assay. JAMA Network Open. 2022;5 doi: 10.1001/jamanetworkopen.2022.1100. e221100-e221100. [DOI] [PubMed] [Google Scholar]
- 7.De Rubis G., Krishnan S.Rajeev, Bebawy M. Liquid Biopsies in Cancer Diagnosis, Monitoring, and Prognosis. Trends in Pharmacological Sciences. 2019;40:172–186. doi: 10.1016/j.tips.2019.01.006. [DOI] [PubMed] [Google Scholar]
- 8.Mathai R.A., Vidya R.V., Reddy B.S., Thomas L., Udupa K., Kolesar J., Rao M. Potential Utility of Liquid Biopsy as a Diagnostic and Prognostic Tool for the Assessment of Solid Tumours: Implications in the Precision Oncology. Journal of Clinical Medicine. 2019 doi: 10.3390/jcm8030373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Chen Y., Song Y., Du W., Gong L., Chang H., Zou Z. Tumour-associated macrophages: an accomplice in solid tumour progression. Journal of Biomedical Science. 2019;26:78. doi: 10.1186/s12929-019-0568-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Zhang Y.-C., Zhou Q., Wu Y.-L. The emerging roles of NGS-based liquid biopsy in non-small cell lung cancer. Journal of Hematology & Oncology. 2017;10:167. doi: 10.1186/s13045-017-0536-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.P. Pinzani, V. D'Argenio, M. Del Re, C. Pellegrini, F. Cucchiara, F. Salvianti, S. Galbiati, Updates on liquid biopsy: current trends and future perspectives for clinical application in solid tumours, 59 (2021) 1181-1200, doi: 10.1515/cclm-2020-1685. [DOI] [PubMed]
- 12.Trujillo B., Wu A., Wetterskog D., Attard G. Blood-based liquid biopsies for prostate cancer: clinical opportunities and challenges. British Journal of Cancer. 2022;127:1394–1402. doi: 10.1038/s41416-022-01881-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Zhang H., Lin X., Huang Y., Wang M., Cen C., Tang S., Dique M.R., Cai L., Luis M.A., Smollar J., Wan Y., Cai F. Detection Methods and Clinical Applications of Circulating Tumour Cells in Breast Cancer. Frontiers in Oncology. 2021;11 doi: 10.3389/fonc.2021.652253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Brock G., Castellanos-Rizaldos E., Hu L., Coticchia C., Skog J. Liquid biopsy for cancer screening, patient stratification and monitoring. Translational Cancer Research. 2015;4:280–290. doi: 10.3978/j.issn.2218-676X.2015.06.05. [DOI] [Google Scholar]
- 15.Deng Z., Wu S., Wang Y., Shi D. Circulating tumour cell isolation for cancer diagnosis and prognosis. eBioMedicine. 2022;83 doi: 10.1016/j.ebiom.2022.104237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Pfitzner C., Schröder I., Scheungraber C., Dogan A., Runnebaum I.B., Dürst M., Häfner N. Digital-Direct-RT-PCR: a sensitive and specific method for quantification of CTC in patients with cervical carcinoma. Scientific reports. 2014;4:1–7. doi: 10.1038/srep03970. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Pao C.C., Hor J.J., Yang F.-P., Lin C.-Y., Tseng C.-J. Detection of human papillomavirus mRNA and cervical cancer cells in peripheral blood of cervical cancer patients with metastasis. Journal of clinical oncology. 1997;15:1008–1012. doi: 10.1200/JCO.1997.15.3.1008. [DOI] [PubMed] [Google Scholar]
- 18.Wen Y.-F., Cheng T.-T., Chen X.-L., Huang W.-J., Peng H.-H., Zhou T.-C., Lin X.-D., Zeng L.-S. Elevated circulating tumour cells and squamous cell carcinoma antigen levels predict poor survival for patients with locally advanced cervical cancer treated with radiotherapy. PLOS ONE. 2018;13 doi: 10.1371/journal.pone.0204334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.McMullen K.P., Chalmers J.J., Lang J.C., Kumar P., Jatana K.R. Circulating tumour cells in head and neck cancer: A review. World Journal of Otorhinolaryngology-Head and Neck Surgery. 2016;2:109–116. doi: 10.1016/j.wjorl.2016.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Gorges T.M., Stein A., Quidde J., Hauch S., Röck K., Riethdorf S., Joosse S.A., Pantel K. Improved Detection of Circulating Tumour Cells in Metastatic Colorectal Cancer by the Combination of the CellSearch® System and the AdnaTest®. PLOS ONE. 2016;11 doi: 10.1371/journal.pone.0155126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Lianidou E.S., Strati A., Markou A. Circulating tumour cells as promising novel biomarkers in solid cancers. Critical Reviews in Clinical Laboratory Sciences. 2014;51:160–171. doi: 10.3109/10408363.2014.896316. [DOI] [PubMed] [Google Scholar]
- 22.Heitzer E., Ulz P., Geigl J.B. Circulating Tumour DNA as a Liquid Biopsy for Cancer. Clinical Chemistry. 2015;61:112–123. doi: 10.1373/clinchem.2014.222679. [DOI] [PubMed] [Google Scholar]
- 23.Chen Q., Zhang Z.-H., Wang S., Lang J.-H. Circulating cell-free DNA or circulating tumour DNA in the management of ovarian and endometrial cancer. OncoTargets and therapy. 2019;12:11517. doi: 10.2147/OTT.S227156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Nacchio M., Sgariglia R., Gristina V., Pisapia P., Pepe F., De Luca C., Migliatico I., Clery E., Greco L., Vigliar E., Bellevicine C., Russo A., Troncone G., Malapelle U. KRAS mutations testing in non-small cell lung cancer: the role of Liquid biopsy in the basal setting. J Thorac Dis. 2020;12:3836–3843. doi: 10.21037/jtd.2020.01.19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Taly V., Pekin D., Benhaim L., Kotsopoulos S.K., Le Corre D., Li X., Atochin I., Link D.R., Griffiths A.D., Pallier K., Blons H., Bouché O., Landi B., Hutchison J.B., Laurent-Puig P. Multiplex Picodroplet Digital PCR to Detect KRAS Mutations in Circulating DNA from the Plasma of Colorectal Cancer Patients. Clinical Chemistry. 2013;59:1722–1731. doi: 10.1373/clinchem.2013.206359. [DOI] [PubMed] [Google Scholar]
- 26.Dawson S.-J., Tsui D.W.Y., Murtaza M., Biggs H., Rueda O.M., Chin S.-F., Dunning M.J., Gale D., Forshew T., Mahler-Araujo B., Rajan S., Humphray S., Becq J., Halsall D., Wallis M., Bentley D., Caldas C., Rosenfeld N. Analysis of Circulating Tumour DNA to Monitor Metastatic Breast Cancer. New England Journal of Medicine. 2013;368:1199–1209. doi: 10.1056/NEJMoa1213261. [DOI] [PubMed] [Google Scholar]
- 27.Alix-Panabières C., Pantel K. Clinical Applications of Circulating Tumour Cells and Circulating Tumour DNA as Liquid Biopsy. Cancer Discovery. 2016;6:479–491. doi: 10.1158/2159-8290.CD-15-1483. [DOI] [PubMed] [Google Scholar]
- 28.Tukachinsky H., Madison R.W., Chung J.H., Gjoerup O.V., Severson E.A., Dennis L., Fendler B.J., Morley S., Zhong L., Graf R.P., Ross J.S., Alexander B.M., Abida W., Chowdhury S., Ryan C.J., Fizazi K., Golsorkhi T., Watkins S.P., Simmons A., Loehr A., Venstrom J.M., Oxnard G.R. Genomic Analysis of Circulating Tumour DNA in 3,334 Patients with Advanced Prostate Cancer Identifies Targetable BRCA Alterations and AR Resistance Mechanisms. Clinical Cancer Research. 2021;27:3094–3105. doi: 10.1158/1078-0432.CCR-20-4805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Ignatiadis M., Sledge G.W., Jeffrey S.S. Liquid biopsy enters the clinic — implementation issues and future challenges. Nature Reviews Clinical Oncology. 2021;18:297–312. doi: 10.1038/s41571-020-00457-x. [DOI] [PubMed] [Google Scholar]
- 30.Bronkhorst A.J., Ungerer V., Holdenrieder S. The emerging role of cell-free DNA as a molecular marker for cancer management. Biomolecular Detection and Quantification. 2019;17 doi: 10.1016/j.bdq.2019.100087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Mithani S.K., Smith I.M., Zhou S., Gray A., Koch W.M., Maitra A., Califano J.A. Mitochondrial Resequencing Arrays Detect Tumour-Specific Mutations in Salivary Rinses of Patients with Head and Neck Cancer. Clinical Cancer Research. 2007;13:7335–7340. doi: 10.1158/1078-0432.CCR-07-0220. [DOI] [PubMed] [Google Scholar]
- 32.Jahr S., Hentze H., Englisch S., Hardt D., Fackelmayer F.O., Hesch R.-D., Knippers R. DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer research. 2001;61:1659–1665. [PubMed] [Google Scholar]
- 33.Liu S., Wang J. Current and Future Perspectives of Cell-Free DNA in Liquid Biopsy. Current Issues in Molecular Biology. 2022:2695–2709. doi: 10.3390/cimb44060184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Panagopoulou M., Esteller M., Chatzaki E. Circulating Cell-Free DNA in Breast Cancer: Searching for Hidden Information towards Precision Medicine. Cancers. 2021 doi: 10.3390/cancers13040728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Stover D.G., Parsons H.A., Ha G., Freeman S.S., Barry W.T., Guo H., Choudhury A.D., Gydush G., Reed S.C., Rhoades J., Rotem D., Hughes M.E., Dillon D.A., Partridge A.H., Wagle N., Krop I.E., Getz G., Golub T.R., Love J.C., Winer E.P., Tolaney S.M., Lin N.U., Adalsteinsson V.A. Association of Cell-Free DNA Tumour Fraction and Somatic Copy Number Alterations With Survival in Metastatic Triple-Negative Breast Cancer. Journal of Clinical Oncology. 2018;36:543–553. doi: 10.1200/JCO.2017.76.0033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Xu X., Yu Y., Shen M., Liu M., Wu S., Liang L., Huang F., Zhang C., Guo W., Liu T. Role of circulating free DNA in evaluating clinical tumour burden and predicting survival in Chinese metastatic colorectal cancer patients. BMC Cancer. 2020;20:1006. doi: 10.1186/s12885-020-07516-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Bondhopadhyay B., Sisodiya S., Alzahrani F.A., Bakhrebah M.A., Chikara A., Kasherwal V., Khan A., Rani J., Dar S.A., Akhter N., Tanwar P., Agrawal U., Hussain S. Exosomes: A Forthcoming Era of Breast Cancer Therapeutics. Cancers. 2021 doi: 10.3390/cancers13184672. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Fitts C.A., Ji N., Li Y., Tan C. Exploiting exosomes in cancer liquid biopsies and drug delivery. Advanced healthcare materials. 2019;8 doi: 10.1002/adhm.201801268. [DOI] [PubMed] [Google Scholar]
- 39.Kalluri R. The biology and function of exosomes in cancer. The Journal of clinical investigation. 2016;126:1208–1215. doi: 10.1172/JCI81135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Wang X., Jiang X., Li J., Wang J., Binang H., Shi S., Duan W., Zhao Y., Zhang Y. Serum exosomal miR-1269a serves as a diagnostic marker and plays an oncogenic role in non-small cell lung cancer. Thoracic cancer. 2020;11:3436–3447. doi: 10.1111/1759-7714.13644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Su Y.Y., Sun L., Guo Z.R., Li J.C., Bai T.T., Cai X.X., Li W.H., Zhu Y.F. Upregulated expression of serum exosomal miR-375 and miR-1307 enhance the diagnostic power of CA125 for ovarian cancer. Journal of ovarian research. 2019;12:1–9. doi: 10.1186/s13048-018-0477-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Li M., Zou X., Xia T., Wang T., Liu P., Zhou X., Wang S., Zhu W. A five-miRNA panel in plasma was identified for breast cancer diagnosis. Cancer medicine. 2019;8:7006–7017. doi: 10.1002/cam4.2572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Foroni C., Zarovni N., Bianciardi L., Bernardi S., Triggiani L., Zocco D., Venturella M., Chiesi A., Valcamonico F., Berruti A. When less is more: specific capture and analysis of tumour exosomes in plasma increases the sensitivity of liquid biopsy for comprehensive detection of multiple androgen receptor phenotypes in advanced prostate cancer patients. Biomedicines. 2020;8:131. doi: 10.3390/biomedicines8050131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Cui Y., Xu H.-F., Liu M.-Y., Xu Y.-J., He J.-C., Zhou Y., Cang S.-D. Mechanism of exosomal microRNA-224 in development of hepatocellular carcinoma and its diagnostic and prognostic value. World journal of gastroenterology. 2019;25:1890. doi: 10.3748/wjg.v25.i15.1890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Hao Y.X., Li Y.M., Ye M., Guo Y.Y., Li Q.W., Peng X.M., Wang Q., Zhang S.F., Zhao H.X., Zhang H. KRAS and BRAF mutations in serum exosomes from patients with colorectal cancer in a Chinese population. Oncology letters. 2017;13:3608–3616. doi: 10.3892/ol.2017.5889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Gao Z., Pang B., Li J., Gao N., Fan T., Li Y. Emerging role of exosomes in liquid biopsy for monitoring prostate cancer invasion and metastasis. Frontiers in Cell and Developmental Biology. 2021;9 doi: 10.3389/fcell.2021.679527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Shefer A., Yalovaya A., Tamkovich S. Exosomes in Breast Cancer: Involvement in Tumour Dissemination and Prospects for Liquid Biopsy. International Journal of Molecular Sciences. 2022 doi: 10.3390/ijms23168845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Halvaei S., Daryani S., Eslami-S Z., Samadi T., Jafarbeik-Iravani N., Bakhshayesh T.O., Majidzadeh-A K., Esmaeili R. Exosomes in Cancer Liquid Biopsy: A Focus on Breast Cancer. Molecular Therapy - Nucleic Acids. 2018;10:131–141. doi: 10.1016/j.omtn.2017.11.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Yu H., Han L., Yuan J., Sun Y. Circulating tumour cell free DNA from plasma and urine in the clinical management of colorectal cancer. Cancer Biomarkers. 2020;27:29–37. doi: 10.3233/CBM-182344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Martins I., Ribeiro I.P., Jorge J., Gonçalves A.C., Sarmento-Ribeiro A.B., Melo J.B., Carreira I.M. Liquid Biopsies: Applications for Cancer Diagnosis and Monitoring. Genes. 2021 doi: 10.3390/genes12030349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Herrmann S., Zhan T., Betge J., Rauscher B., Belle S., Gutting T., Schulte N., Jesenofsky R., Härtel N., Gaiser T., Hofheinz R.-D., Ebert M.P., Boutros M. Detection of mutational patterns in cell-free DNA of colorectal cancer by custom amplicon sequencing. Molecular Oncology. 2019;13:1669–1683. doi: 10.1002/1878-0261.12539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Balaji S.A., Shanmugam A., Chougule A., Sridharan S., Prabhash K., Arya A., Chaubey A., Hariharan A., Kolekar P., Sen M., Ravichandran A., Katragadda S., Sankaran S., Bhargava S., Kulkarni P., Rao S., Sunkavalli C., Banavali S., Joshi A., Noronha V., Dutt A., Bahadur U., Hariharan R., Veeramachaneni V., Gupta V. Analysis of solid tumour mutation profiles in liquid biopsy. Cancer Medicine. 2018;7:5439–5447. doi: 10.1002/cam4.1791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Oellerich M., Christenson R.H., Beck J., Walson P.D. Plasma EGFR mutation testing in non-small cell lung cancer: A value proposition. Clinica Chimica Acta. 2019;495:481–486. doi: 10.1016/j.cca.2019.05.019. [DOI] [PubMed] [Google Scholar]
- 54.Lamb Y.N., Dhillon S. Epi proColon® 2.0 CE: A Blood-Based Screening Test for Colorectal Cancer. Molecular Diagnosis & Therapy. 2017;21:225–232. doi: 10.1007/s40291-017-0259-y. [DOI] [PubMed] [Google Scholar]
- 55.Bernard V., Kim D.U., San Lucas F.A., Castillo J., Allenson K., Mulu F.C., Stephens B.M., Huang J., Semaan A., Guerrero P.A., Kamyabi N., Zhao J., Hurd M.W., Koay E.J., Taniguchi C.M., Herman J.M., Javle M., Wolff R., Katz M., Varadhachary G., Maitra A., Alvarez H.A. Circulating Nucleic Acids Are Associated With Outcomes of Patients With Pancreatic Cancer. Gastroenterology. 2019;156:108–118. doi: 10.1053/j.gastro.2018.09.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Gao W., Huang T., Yuan H., Yang J., Jin Q., Jia C., Mao G., Zhao J. Highly sensitive detection and mutational analysis of lung cancer circulating tumour cells using integrated combined immunomagnetic beads with a droplet digital PCR chip. Talanta. 2018;185:229–236. doi: 10.1016/j.talanta.2018.03.083. [DOI] [PubMed] [Google Scholar]
- 57.Ogle L.F., Orr J.G., Willoughby C.E., Hutton C., McPherson S., Plummer R., Boddy A.V., Curtin N.J., Jamieson D., Reeves H.L. Imagestream detection and characterisation of circulating tumour cells – A liquid biopsy for hepatocellular carcinoma? Journal of Hepatology. 2016;65:305–313. doi: 10.1016/j.jhep.2016.04.014. [DOI] [PubMed] [Google Scholar]
- 58.Aaltonen K.E., Novosadová V., Bendahl P.-O., Graffman C., Larsson A.-M., Rydén L. Molecular characterization of circulating tumour cells from patients with metastatic breast cancer reflects evolutionary changes in gene expression under the pressure of systemic therapy. Oncotarget. 2017;8(28) doi: 10.18632/oncotarget.17271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Qi L.-N., Xiang B.-D., Wu F.-X., Ye J.-Z., Zhong J.-H., Wang Y.-Y., Chen Y.-Y., Chen Z.-S., Ma L., Chen J., Gong W.-F., Han Z.-G., Lu Y., Shang J.-J., Li L.-Q. Circulating Tumour Cells Undergoing EMT Provide a Metric for Diagnosis and Prognosis of Patients with Hepatocellular Carcinoma. Cancer Research. 2018;78:4731–4744. doi: 10.1158/0008-5472.CAN-17-2459. [DOI] [PubMed] [Google Scholar]
- 60.Feng J., Li B., Ying J., Pan W., Liu C., Luo T., Lin H., Zheng L. Liquid Biopsy: Application in Early Diagnosis and Monitoring of Cancer. Small Structures. 2020;1 doi: 10.1002/sstr.202000063. [DOI] [Google Scholar]
- 61.Egyud M., Tejani M., Pennathur A., Luketich J., Sridhar P., Yamada E., Ståhlberg A., Filges S., Krzyzanowski P., Jackson J., Kalatskaya I., Jiao W., Nielsen G., Zhou Z., Litle V., Stein L., Godfrey T. Detection of Circulating Tumour DNA in Plasma: A Potential Biomarker for Esophageal Adenocarcinoma. The Annals of Thoracic Surgery. 2019;108:343–349. doi: 10.1016/j.athoracsur.2019.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Almodovar K., Iams W.T., Meador C.B., Zhao Z., York S., Horn L., Yan Y., Hernandez J., Chen H., Shyr Y., Lim L.P., Raymond C.K., Lovly C.M. Longitudinal Cell-Free DNA Analysis in Patients with Small Cell Lung Cancer Reveals Dynamic Insights into Treatment Efficacy and Disease Relapse. Journal of Thoracic Oncology. 2018;13:112–123. doi: 10.1016/j.jtho.2017.09.1951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Chen S., Zhao J., Cui L., Liu Y. Urinary circulating DNA detection for dynamic tracking of EGFR mutations for NSCLC patients treated with EGFR-TKIs. Clinical and Translational Oncology. 2017;19:332–340. doi: 10.1007/s12094-016-1534-9. [DOI] [PubMed] [Google Scholar]
- 64.Akamatsu H., Koh Y., Okamoto I., Fujimoto D., Bessho A., Azuma K., Morita S., Yamamoto N., Nakagawa K. Clinical significance of monitoring EGFR mutation in plasma using multiplexed digital PCR in EGFR mutated patients treated with afatinib (West Japan Oncology Group 8114LTR study) Lung Cancer. 2019;131:128–133. doi: 10.1016/j.lungcan.2019.03.021. [DOI] [PubMed] [Google Scholar]
- 65.Wang D.-S., Liu Z.-X., Lu Y.-X., Bao H., Wu X., Zeng Z.-L., Liu Z., Zhao Q., He C.-Y., Lu J.-H., Wang Z.-Q., Qiu M.-Z., Wang F., Wang F.-H., Li Y.-H., Wang X.-N., Xie D., Jia W.-H., Shao Y.W., Xu R.-H. Liquid biopsies to track trastuzumab resistance in metastatic HER2-positive gastric cancer. Gut. 2019;68:1152. doi: 10.1136/gutjnl-2018-316522. [DOI] [PubMed] [Google Scholar]
- 66.Khan K.H., Cunningham D., Werner B., Vlachogiannis G., Spiteri I., Heide T., Mateos J.F., Vatsiou A., Lampis A., Damavandi M.D., Lote H., Huntingford I.S., Hedayat S., Chau I., Tunariu N., Mentrasti G., Trevisani F., Rao S., Anandappa G., Watkins D., Starling N., Thomas J., Peckitt C., Khan N., Rugge M., Begum R., Hezelova B., Bryant A., Jones T., Proszek P., Fassan M., Hahne J.C., Hubank M., Braconi C., Sottoriva A., Valeri N. Longitudinal Liquid Biopsy and Mathematical Modeling of Clonal Evolution Forecast Time to Treatment Failure in the PROSPECT-C Phase II Colorectal Cancer Clinical Trial. Cancer Discovery. 2018;8:1270–1285. doi: 10.1158/2159-8290.CD-17-0891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Underwood J.J., Quadri R.S., Kalva S.P., Shah H., Sanjeevaiah A.R., Beg M.S., Sutphin P.D. Liquid Biopsy for Cancer: Review and Implications for the Radiologist. Radiology. 2019;294:5–17. doi: 10.1148/radiol.2019182584. [DOI] [PubMed] [Google Scholar]
- 68.Palmirotta R., Lovero D., Cafforio P., Felici C., Mannavola F., Pellè E., Quaresmini D., Tucci M., Silvestris F. Liquid biopsy of cancer: a multimodal diagnostic tool in clinical oncology. Therapeutic Advances in Medical Oncology. 2018;10 doi: 10.1177/1758835918794630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Bai Y., Zhao H. Liquid biopsy in tumours: opportunities and challenges. Annals of Translational Medicine. 2018;6(Supplement 1) doi: 10.21037/atm.2018.11.31. (November 29, 2018): Annals of Translational Medicine. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Major C., Wang S. Abstract 4010: Analytical concordance of 3 independent diagnostic assays for the detection of FGFR alterations in urothelial carcinoma tumor tissue. Cancer Research. 2022;82:4010. doi: 10.1158/1538-7445.AM2022-4010. [DOI] [Google Scholar]
- 71.Bhat A., Ritch C.R. Urinary biomarkers in bladder cancer: where do we stand? Current Opinion in Urology. 2019;29:203–209. doi: 10.1097/MOU.0000000000000605. [DOI] [PubMed] [Google Scholar]
- 72.J.D. Warren, W. Xiong, A.M. Bunker, C.P. Vaughn, L.V. Furtado, W.L. Roberts, J.C. Fang, W.S. Samowitz, K.A. Heichman, Septin 9 methylated DNA is a sensitive and specific blood test for colorectal cancer, BMC Medicine, 9 (2011) 133, doi: 10.1186/1741-7015-9-133. [DOI] [PMC free article] [PubMed]
- 73.Pichler R., Fritz J., Tulchiner G., Klinglmair G., Soleiman A., Horninger W., Klocker H., Heidegger I. Increased accuracy of a novel mRNA-based urine test for bladder cancer surveillance. BJU International. 2018;121:29–37. doi: 10.1111/bju.14019. [DOI] [PubMed] [Google Scholar]
- 74.Batista R., Vinagre J., Prazeres H., Sampaio C., Peralta P., Conceição P., Sismeiro A., Leão R., Gomes A., Furriel F., Oliveira C., Torres J.N., Eufrásio P., Azinhais P., Almeida F., Gonzalez E.R., Bidovanets B., Ecke T., Stinjs P., Pascual Á.S., Abdelmalek R., Villafruela A., Beardo-Villar P., Fidalgo N., Öztürk H., Gonzalez-Enguita C., Monzo J., Lopes T., Álvarez-Maestro M., Servan P.P., De La Cruz S.M.P., Perez M.P.S., Máximo V., Soares P. Validation of a Novel, Sensitive, and Specific Urine-Based Test for Recurrence Surveillance of Patients With Non-Muscle-Invasive Bladder Cancer in a Comprehensive Multicenter Study. Frontiers in Genetics. 2019;10 doi: 10.3389/fgene.2019.01237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.García-Foncillas J., Tabernero J., Élez E., Aranda E., Benavides M., Camps C., Jantus-Lewintre E., López R., Muinelo-Romay L., Montagut C., Antón A., López G., Díaz-Rubio E., Rojo F., Vivancos A. Prospective multicenter real-world RAS mutation comparison between OncoBEAM-based liquid biopsy and tissue analysis in metastatic colorectal cancer. British Journal of Cancer. 2018;119:1464–1470. doi: 10.1038/s41416-018-0293-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Makutani Y., Sakai K., Yamada M., Wada T., Chikugo T., Satou T., Iwasa Y., Yamamoto H., de Velasco M.A., Nishio K., Kawamura J. Performance of IdyllaTM RAS-BRAF mutation test for formalin-fixed paraffin-embedded tissues of colorectal cancer. International Journal of Clinical Oncology. 2022;27:1180–1187. doi: 10.1007/s10147-022-02167-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Li B., Huang H., Huang R., Zhang W., Zhou G., Wu Z., Lv C., Han X., Jiang L., Li Y., Li B., Zhang Z. <i>SEPT9</i> Gene Methylation as a Noninvasive Marker for Hepatocellular Carcinoma. Disease Markers. 2020;2020:6289063. doi: 10.1155/2020/6289063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Limburg P.J., Mahoney D.W., Ahlquist D.A., Allawi H.T., Johnson S.C., Kaiser M., Katerov V.E., Statz S., Graham R.P., Foote P.H., Doering K.A., Burger K.N., Lidgard G.P., Kisiel J.B. Comparison of Tissue-Based Molecular Markers in Younger versus Older Patients with Colorectal Neoplasia. Cancer Epidemiology, Biomarkers & Prevention. 2020;29:1570–1576. doi: 10.1158/1055-9965.EPI-19-1598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Pastor B., André T., Henriques J., Trouilloud I., Tournigand C., Jary M., Mazard T., Louvet C., Azan S., Bauer A., Roch B., Sanchez C., Vernerey D., Thierry A.R., Adenis A. Monitoring levels of circulating cell-free DNA in patients with metastatic colorectal cancer as a potential biomarker of responses to regorafenib treatment. Molecular Oncology. 2021;15:2401–2411. doi: 10.1002/1878-0261.12972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.J.C. Poole, S.-F. Wu, T.T. Lu, C.R.T. Vibat, A. Pham, E. Samuelsz, M. Patel, J. Chen, T. Daher, V.M. Singh, L.J. Arnold, Analytical validation of the Target Selector ctDNA platform featuring single copy detection sensitivity for clinically actionable EGFR, BRAF, and KRAS mutations, PLOS ONE, 14 (2019) e0223112, doi: 10.1371/journal.pone.0223112. [DOI] [PMC free article] [PubMed]
- 81.Witjes J.A., Morote J., Cornel E.B., Gakis G., van Valenberg F.J.P., Lozano F., Sternberg I.A., Willemsen E., Hegemann M.L., Paitan Y., Leibovitch I. Performance of the Bladder EpiCheckTM Methylation Test for Patients Under Surveillance for Non–muscle-invasive Bladder Cancer: Results of a Multicenter, Prospective. Blinded Clinical Trial, European Urology Oncology. 2018;1:307–313. doi: 10.1016/j.euo.2018.06.011. [DOI] [PubMed] [Google Scholar]


