Although radiologic imaging and histologic assessment of tumor tissues are classic approaches for diagnosis and monitoring of treatment response, they have many limitations. These include challenges in distinguishing benign from malignant masses, difficult access to the tumor, high cost of the procedures, and tumor heterogeneity. In this setting, liquid biopsy has emerged as a potential alternative for both diagnostic and monitoring purposes. The approaches to liquid biopsy include cell-free DNA/circulating tumor DNA, long and micro noncoding RNAs, proteins/peptides, carbohydrates/lectins, lipids, and metabolites. Other approaches include detection and analysis of circulating tumor cells, extracellular vesicles, and tumor-activated platelets. Ultimately, reliable use of liquid biopsies requires bioinformatics and statistical integration of multiple datasets to achieve approval in a Clinical Laboratory Improvement Amendments setting. This review provides a balanced and critical assessment of recent discoveries regarding tumor-derived biomarkers in liquid biopsies along with the potential and pitfalls for cancer detection and longitudinal monitoring.

Routine clinical practice has relied on tissue sampling to diagnose tumors (1, 2). Despite the tremendous informative value of high-quality tumor biopsies, there are many limitations to their use. Tissue biopsies are invasive, costly, and a possible source of clinical complications. Adverse events such as air leaks in the chest, postprocedural hypotension, and hemorrhage occur in 1.6% to 17.1% of cases (3). Moreover, tissue preservation techniques (formalin fixation and paraffin embedding, snap freezing, and gel-preserved freezing; ref. 4) can distort the original quality of samples that were preserved for years. Liquid nitrogen–based snap freezing and more elaborate preservation methods are typically not available in a community hospital setting. Moreover, focal tissue sampling is not always able to capture the heterogeneity of the tumor microenvironment since biopsy cores cannot sample the entire tumor region. Finally, there are cases in which the primary site can be challenging to identify on imaging, or the tumor mass is localized in an area that is difficult to access, making it difficult to obtain a biopsy. Because of these and other limitations, the medical community has sought to produce and standardize noninvasive techniques that can be safely performed longitudinally and complement or substitute invasive approaches. Because liquid biopsies drawn from the entire tumor vascular drainage, this approach may be more reflective of the primary tumor and its collective metastases.

Because of their minimal invasiveness, liquid biopsies can be repeated and provide biological material that often requires minimal efforts for preservation (5), making this potentially one of the most useful techniques for early diagnosis, monitoring of therapy response, and prediction of prognosis. To date, several tumor-derived liquid biopsy biomarkers have been developed, including circulating tumor cells (CTC), extracellular vesicles (EV) or exosomes, cell-free (cfDNA) or circulating tumor DNA (ctDNA), epigenetically modified DNA, long and micro noncoding RNAs (ncRNA), and platelets (Fig. 1). Isolation of ctDNA and circulating cancer cells is typically influenced by disease burden and cancer type (6, 7). ctDNA is used to detect tumor-specific mutations or epigenetic modifications for genotyping metastasis (8), minimal residual disease (9), and staging for treatment decisions (10–12). The EVs from tumor cells can contain mRNAs, proteins, and other potential biomarkers (13). Platelets supply considerable amount of EVs to the circulation and also show robust uptake through their open canalicular system (14, 15). Moreover, platelets are among the easiest biological particles to isolate and can provide a complete molecular profile of their passage through a tumor (ref. 16; Fig. 1).

Figure 1.

The illustration represents the “Circulome” consisting of the different circulating factors derived from or influenced by cancer cells. MT: mitochondria; N: nucleus; ER: endoplasmic reticulum; MVE: multi-vesicular endosome; EE; early endosome; cf; circulating-free; tDNA: transfer DNA; miRNA: microRNA; mRNA: messenger RNA; lncRNA: long noncoding RNA.

Figure 1.

The illustration represents the “Circulome” consisting of the different circulating factors derived from or influenced by cancer cells. MT: mitochondria; N: nucleus; ER: endoplasmic reticulum; MVE: multi-vesicular endosome; EE; early endosome; cf; circulating-free; tDNA: transfer DNA; miRNA: microRNA; mRNA: messenger RNA; lncRNA: long noncoding RNA.

Close modal

Ultimately, effective liquid biopsies depend on the integration of multiple biomarkers, sampling approaches, and datasets. This results in a highly sensitive and specific representation of the disease status (17–19).

Tumor-derived factors

cf/ctDNA and CTCs

The presence of cfDNA in plasma and serum was first reported in 1948 by Mandel and Metais (20) but remained largely unexplored until the 1970s; since then it has drawn substantial attention for clinical use as a noninvasive biopsy (21). One of its most common applications in clinical practice is the analysis of circulating cfDNA to assess abnormal fetal conditions in high-risk pregnancies, such as trisomy of chromosomes 21, 18, or 13 (22). This technique, usually performed with high patient compliance, can be used as early as the 10th week of pregnancy until delivery and has almost completely replaced invasive diagnostic techniques such as amniocentesis.

DNA can be released into the bloodstream at any point from primary tumor cells, CTCs, or tumor-derived EVs. cfDNA typically consists of any circulating DNA molecule in the bloodstream, not necessarily cancer derived. In contrast, ctDNA is specifically tumor derived. Regardless of its source, cfDNA (or ctDNA) is actively secreted or passively released into the bloodstream by tumor cells undergoing apoptosis, necrosis (23), or immunotherapy-associated tumor killing (24–26). Some studies have shown concordance between ctDNA and DNA isolated from the original tumor tissue (27–29). Sequencing analyses have revealed similar mutations, copy-number variations, and gene amplification patterns between the ctDNA and DNA of tumor tissues (both primary and metastatic). However, other studies have found substantial rates of discordance between ctDNA and tumor DNA in identifying specific mutations. In a study on a cohort of patients with lung cancer (N = 38), a comparison between four platforms for mutation detection on ctDNA showed concordance between tumor tissue and ctDNA in detecting the EGFR T90M mutation ranging between 48% and 74% (30). In a heterogeneous cohort of 74 patients with different cancer types, concordance for detection of relevant alterations in 33 genes with microsatellite instability between cfDNA and matched tumor samples was tested. There was only 45% concordance when considering primary tumor samples (77% when considering DNA from metastatic disease; ref. 31). The discordant rates are often related to low variant allele frequency or intratumoral heterogeneity (32, 33).

Some studies have suggested that ctDNA analysis can complement DNA analysis from primary and metastatic tumor tissue biopsies when investigating TP53 mutations and other repair genes (34). A potential limitation of standard DNA-sequencing assays (e.g., next-generation sequencing) applied to ctDNA analyses (29) is characterized by the observation of a lower number of mutation reads, due to the small amount of tumor-specific DNA. To address this issue, more specific detection techniques have been developed, such as digital droplet PCR and several commercial assays for ctDNA amplification are being used in the clinical setting for various cancers (10, 11, 35). The short half-life in circulation, fragment size, and available volume are critical considerations when designing and evaluating ctDNA analysis approaches for plasma samples (8, 36, 37). Recently Natera’s Signatera (TM) MRD (molecular residual disease) test which uses ctDNA, has been granted Breakthrough Device Designation (38).

FDA-approved commercial assays for detection of mutations in circulating cancer-derived DNA include FoundationOne Liquid CDx [for non–small cell lung cancer (NSCLC) and prostate cancer] and Guardant360 CDx (for NSCLC and colorectal cancer), as companion diagnostic tests. While FoundationOne targets coding regions of 324 genes and among these, 72 genes with higher depth (39), the Guardant360 assay targets 54 genes, with complete exon coverage of 18 and coverage of critical regions of 36 genes (including ERBB2, EGFR, and MET; ref. 40).

Another opportunity for liquid biopsies involves CTCs that are cancer cells shed from the tumor into the bloodstream (41). Several studies have shown a high rate of concordance in gene mutations between circulating tumor cells and DNA from tumor tissue, making CTCs a potentially suitable candidate as a cancer biomarker (42–44). Clinically, the abundance of CTCs has been associated with a worse prognosis in prostate, lung, and other cancers. These clinical observations are corroborated by experimental findings that have demonstrated the potential of CTCs to form metastatic clusters (45). However, their role as a diagnostic marker remains unclear due, in part, to the lack of a specific marker for detection of all CTCs (46). A recent study in support of their diagnostic potential, including 112 patients with hepatocellular carcinoma, 20 healthy donors, and 12 patients with hepatitis B virus, reports detection of CTCs in 90% of the patients with hepatocellular carcinoma and in 17% of the patients with hepatitis B who further developed hepatocellular carcinoma (47). CTCs were proven to originate from the primary tumor, as suggested by identifying specific TP53 DNA mutations that matched the ones isolated in the primary tumor DNA. In addition, specific markers on CTCs (e.g., PD-L1) could be useful as a marker for response to therapy, as a recent pilot study on the efficacy of pembrolizumab in patients with advanced melanoma shows (48).

Despite their potential, CTCs are rare in the bloodstream [some studies report between one and six cells in 7.5 mL of whole blood (49)], and can be challenging to isolate. Up to now, the only FDA-approved method for CTC isolation has been the Cell Search System (Jenssen Diagnostics), which uses anti–EpCam-coated magnetic beads; this method has been used successfully in clinical trials for prognostic stratification. Several new techniques have since been tested to isolate higher quality material and avoid the bias of a priori knowledge of surface markers (needed for antibody-based isolation techniques). For example, acoustic separation uses the physical properties of CTCs to separate them from other circulating cells (50). Utilizing marker-directed antibodies for positive selection of cells has the apparent disadvantage of selecting just a specific subpopulation of circulating tumor-derived cells. Many tumor epithelial cells can express low levels of EpCAM or be entirely negative for it and positive for other cytokeratins (51). Besides that, many circulating cells have likely undergone epithelial–mesenchymal transition (EMT) with consequent loss of epithelial markers, which justifies the use of negative selection for CD45, without positive selection for cytokeratins (52). Although newer approaches, such as coupling CTC and ctDNA isolation to single-cell sequencing, may hold promise in the future (53, 54), the low specificity and high dilution represent the major drawbacks of using these currently in routine clinical practice. Finally, a specific phenomenon termed “clonal hematopoiesis of indeterminate potential” (55), may affect the reliability of DNA-sequencing data from cfDNA/ctDNA. Clonal hematopoiesis (CH) represents age-related acquisition of somatic mutations in hematopoietic cells, which leads to clonal expansion and potentially to hematopoietic diseases (56). Its incidence is quite high in the aging population, reaching 10% of healthy subjects after the age of 70 (57). This somatic mosaicism present in plasma could lead to misclassification of CH mutations as tumor derived and represents an important source of background noise which some have tried to control for using bioinformatics.

To overcome these limitations, scientists have investigated the presence of more specific tumor-derived circulating tumor biomarkers, including cancer-specific EVs and tumor-activated platelets.

EVs

EVs are membrane vesicles formed intracellularly and secreted in the extracellular space (58); they are generally distinguished on the basis of their size: small EVs (also called exosomes, 50 to 150 nm in diameter), medium EVs (150 to 300 nm in diameter), and large EVs (between 300 and 1,000 nm in diameter). EVs have different densities and biochemical compositions. Guidelines for classification and techniques for isolation and identification are described in detail on the International Society of Extracellular Vesicles (ISEV) website, https://www.isev.org/.

EVs are thought to originate from endosomes and intraluminal vesicles (ILV) located inside multivesicular bodies. When the early endosomal membrane folds on itself, it tends to sequester proteins, lipids, and cytosol. While part of this cargo is released by late endosomes in lysosomes, some of it can be incorporated into novel generated organelles. These organelles express tetraspanin (CD63) and the membrane proteins LAMP1 and LAMP2, and fuse with the plasma membrane to be released as EVs. The generation of EVs involves the endosomal sorting complex required for transport (ESCRT), which has three components (I, II, and III), and participate in the process of exosome assembly in different cell types. An ESCRT-independent mechanism for exosome generation is also present, as suggested by the evidence that selective inhibitors of ESCRT do not entirely suppress the biogenesis of EVs (59, 60). The heterogeneous cargos in EVs, including lipids, proteins, tumor, mitochondrial DNA (61), and coding along with ncRNAs (62, 63) can vary according to the producing cell and in response to external stimuli (64). These characteristics make them an attractive target for tumor profiling (65). Table 1 provides a schematic classification of EVs by size, together with a description of their cargo and cellular origin. EVs’ roles are not well established, but many studies show their clear biological impact on the recipient cell (66). For example, EVs derived from metastatic cancer cells are able to increase the invasive and migratory potential of the surrounding less aggressive cancer cells (67), while EVs from stromal cells can release ncRNAs into cancer cells and activate intracellular pathways favoring the expansion of therapy-resistant or tumor-initiating cell clones (68).

Table 1.

Comparison among three size-based categories of EVs, based on cargo, mechanisms of production, and detection markers.

EVs subtype (70, 71, 77, 78, 79, 80, 81, 82, 83)SizeCargoOriginMarker
Small EVs 50–150 nm diameter Lipids, proteins, RNAs, and DNA Folding of the endosomal membrane into ILVs. Fusion with plasma membrane and release CD63, CD9, CD81 
Medium EVs 150–300 nm diameter Cytosolic and plasma membrane-associated proteins, cytoskeletal proteins, integrins Outward budding of plasma membrane Tetraspanins 
Large EVs 300–1,000 nm diameter Cytosolic and plasma membrane-associated proteins, cytoskeletal proteins, integrins Outward budding of plasma membrane Tetraspanins 
EVs subtype (70, 71, 77, 78, 79, 80, 81, 82, 83)SizeCargoOriginMarker
Small EVs 50–150 nm diameter Lipids, proteins, RNAs, and DNA Folding of the endosomal membrane into ILVs. Fusion with plasma membrane and release CD63, CD9, CD81 
Medium EVs 150–300 nm diameter Cytosolic and plasma membrane-associated proteins, cytoskeletal proteins, integrins Outward budding of plasma membrane Tetraspanins 
Large EVs 300–1,000 nm diameter Cytosolic and plasma membrane-associated proteins, cytoskeletal proteins, integrins Outward budding of plasma membrane Tetraspanins 

Abbreviations: CD = cluster differentiation; EV = extracellular vesicle; ILVs = intraluminal vesicles.

Despite the growing evidence supporting their role in cancer metastasis and therapy evasion (61, 69), the effects of EVs during the earlier steps of tumorigenesis are less clear. To use EVs as cancer biomarkers, it is important to identify surface markers that could be used for isolation of cancer-derived EVs and standardize the isolation and enrichment techniques. For a better understanding of these issues, we reviewed standard and novel techniques for EV isolation.

EVs: standardized and novel techniques for collection, enrichment, and quantification

The ISEV defined a series of guidelines for isolation, separation concentration, and morphologic and functional characterization of EVs in 2014 and updated them in 2018 (70). Table 2 summarizes the principal EV isolation techniques, with advantages and disadvantages for each of them. Ultracentrifugation is the most used method and the first option suggested by the ISEV for isolation and concentration of EVs from cell culture, biological fluids, and tissues. It consists of a series of centrifugations at high speed for prolonged periods (relative centrifugal force of 100,000 × g for at least 2 hours), in sequential steps, that allow the isolation of large, medium, and small EVs. Differential ultracentrifugation, though, is time consuming and can result in a low-purity product. Van Deun and colleagues compared several exosome-isolation techniques from cell culture based on density gradient, ultracentrifugation, and precipitation (71). They found that density gradient–based methodologies resulted in a low yield of high-purity product, whereas the precipitation technique resulted in a high yield of low-purity product (primarily contaminated by serum proteins). Ultracentrifugation resulted in a medium purity and yield when compared with the previous two approaches (71). An example of a technique used to reach higher purity involves a density-gradient floatation approach together with ultracentrifugation (13). Complementary techniques can be adapted to concentrate the EVs further, such as filtration, size exclusion chromatography, and immune isolation (72). Currently, none of these methods is deemed the most efficient, and they should be chosen based on considering the final application of the isolated product.

Table 2.

Comparison among major isolation techniques for EVs based on throughput, purity, recovery rate, and needed equipment.

Technique of EVs isolation (58, 63, 64, 69, 70)ProsConsApplication
UC High sample throughput with relatively high product purity Time consuming (each centrifugation step is of at least 2 hours) (each cent Bulk EVs isolation from cell culture media 
Precipitation High sample throughput, and relatively low personnel specialization and machinery needed Very low product purity Bulk EVs isolation from cell culture media with subsequent product purification steps 
Density gradient based High product purity Low sample throughput and appropriate equipment required Isolation of EVs from body fluids 
Size-exclusion chromatography High product purity Low sample throughput and appropriate equipment required Isolation of EVs from body fluids 
Tangential flow filtration High product purity and recovery rate Low sample throughput and appropriate equipment required Isolation of EVs from body fluids 
Affinity based High product purity and recovery rate. Highly specificity Marker dependent Isolation of EVs from body fluids 
Technique of EVs isolation (58, 63, 64, 69, 70)ProsConsApplication
UC High sample throughput with relatively high product purity Time consuming (each centrifugation step is of at least 2 hours) (each cent Bulk EVs isolation from cell culture media 
Precipitation High sample throughput, and relatively low personnel specialization and machinery needed Very low product purity Bulk EVs isolation from cell culture media with subsequent product purification steps 
Density gradient based High product purity Low sample throughput and appropriate equipment required Isolation of EVs from body fluids 
Size-exclusion chromatography High product purity Low sample throughput and appropriate equipment required Isolation of EVs from body fluids 
Tangential flow filtration High product purity and recovery rate Low sample throughput and appropriate equipment required Isolation of EVs from body fluids 
Affinity based High product purity and recovery rate. Highly specificity Marker dependent Isolation of EVs from body fluids 

Abbreviations: EV = extracellular vesicle; SEC = size-exclusion chromatography; UC = ultracentrifugation.

New techniques, still needing further testing, include tangential-flow filtration (73), field-flow fractionation (74), field-free viscoelastic flow (75), alternating current electrodynamic (nanoshearing; ref. 76), acoustics (77), variations of size-exclusion chromatography (78), ion exchange chromatography (79), microfiltration (80), fluorescence-activated sorting (81), deterministic lateral displacement array (82), affinity-based isolation (83), fast protein/high-performance chromatography (84), high-resolution density gradient isolation (Jeppesen, Dennis K et al. “Reassessment of Exosome Composition.” Cell vol. 177,2 (2019): 428-445.e18. doi:10.1016/j.cell.2019.02.029), and other microfluidics techniques (85). While these techniques can help obtain purer EVs, obtaining a higher yield of cancer cell–derived EVs has been elusive. Surface marker-based detection techniques, such as fluorescence-activated sorting, could be used sequentially to achieve better results.

Multimarker kits use magnetic beads associated with a cocktail of antibodies against commonly recognized EV markers such as CD9, CD63, or CD81. However, even in this case, high specificity for cancer-derived vesicles is seldom reached. Most of the isolation techniques tend to yield a heterogeneous pellet of EVs, which is derived in great part from non–tumor-related cells. Therefore, a key point in establishing novel isolation techniques must include identifying cancer-specific markers for EVs; while there is likely to be variability according to tumor type, an ideal marker would be stable in an individual or patient cohort, to allow for diagnosis or longitudinal monitoring of the disease.

Non–tumor-derived factors

Tumor-activated platelets: origin and functions

Platelets are anucleate cell fragments produced by megakaryocytes (86), whose activation status and functions are altered as a result of the interactions with tumor cells (87). In human circulating blood, their count is between 150,000 and 400,000 per microliter (μL), making them a major sampling reservoir. Because of their small size (2–3 μmol/L in diameter; ref. 88), discoid structure, and biophysical characteristics, platelets circulate at the outermost shear fields of flowing blood, in close contact with the vascular endothelium and intravasating cancer cells. This, together with the leakiness of tumor vasculature, enhances tumor biomarker exchange between cancer cells and platelets, resulting in the biological “activation” of the latter. One of the most important properties acquired by platelets during this process is the capacity of active uptake and release of biomolecules through an open canalicular system (89–92). This activation process can occur at various stages of cancer development (93). Tumor-activated platelets that survive and recirculate without removal by the liver or spleen, can easily be isolated through blood biopsies. Once isolated, platelets can be analyzed to identify the adaptive changes they went through during cancer development; thus, representing a potentially useful circulating biomarker for cancer diagnosis, staging, and prognostication (94). Cancer cell–derived cytokines can increase platelet production. For example, IL6 enhances thrombopoietin synthesis in liver (95), which results in elevated platelet counts. Moreover, cancer cells recruit platelets to tumor niches and activate them via factors like TGFβ, ADP, and CD24; consequently, platelets trigger proangiogenic and proinflammatory stimuli in the tumor microenvironment (96). Platelet surface activation markers, such as activated α2b-β3, lysosomal-activated membrane protein (CD63), and P-selectin (CD62P) can be easily detected via flow cytometry together with platelet-specific antigens such as glycoprotein complex Ib-IX-V, GPIIb (CD41), and GPIIIa (CD61; ref. 97).

Tumor-activated platelet profiling

As for platelet-derived markers, researchers have lately focused greatly on RNA profiling. Despite being anucleate cell derivatives, platelets contain several types of RNAs, including messenger RNAs (precursor-mRNA and mature mRNA), structural and catalytic RNAs (rRNA, transfer RNA, and small nucleolar RNA), and regulatory RNAs (miRNA, long intergenic ncRNA, pseudogenes, and antisense RNA). Platelets can also translate mRNA into proteins (98) and splice mRNAs under external stimulation (99), to produce a unique array of mRNAs (100). Amplification and sequencing of platelet-derived mRNA in many patients with localized and metastatic cancer (NSCLC, colorectal, glioblastoma, pancreatic, hepatobiliary, and breast) have revealed the existence of a specific RNA profile present in patients with cancer (94). These mRNAs with increased expression in tumor-activated platelets were enriched for processes associated with vesicle-mediated transport and cytoskeletal protein binding. Diagnostic accuracy for this platelet-based pan-cancer classification reached an accuracy of 95% in distinguishing patients from healthy controls and 97% in predicting different tumor types. Two more recent studies focusing on breast cancer (101) and glioblastoma (102), confirmed the existence of different specific TEP-RNA-seq signatures in tumor patients versus healthy controls. Additional studies are needed to assess the stability of these mRNA signatures in several patient cohorts.

Tumor-activated platelet metabolism

Platelet metabolism is also influenced by interaction with cancer cells; in fact, platelet activation relies on metabolic switches that often involves lipid metabolism toward prostaglandins, prostacyclin, and thromboxane production. These components have a short half-life (seconds to a few minutes), and their synthesis can be influenced by external factors; proinflammatory stimuli tend to mobilize the membrane-bound arachidonic acid, which is used to synthesize bioactive lipids via the COX enzymes (103). Prostaglandins, in turn, favor tumorigenesis via several mechanisms, the most well-known being the modulation of the β-catenin pathway in colon cancer (104). Detection of metabolic profiles of tumor-activated platelets is therefore of great interest in liquid biopsy–based biomarker research. Platelet metabolomics has been extensively studied in the cardiovascular field, using nuclear magnetic resonance (105, 106). This technique could overcome some of the limitations of RNA sequencing, represented by the high cost and the complex interpretation of results.

When analyzing the content of platelets, it is extremely important to preemptively adopt an isolation technique that guarantees high purity of yield. White and red blood cells are common contaminants of the platelet-eluate and can significantly alter the quantity and quality of RNA and metabolites. For a better understanding of these issues, we review in the following paragraph, the major steps involved in isolation of platelets and some of the commonly used analytic techniques.

Tumor-activated platelets: standardized and novel techniques for isolation and morphologic analysis

The first well-established step for isolation of platelets consists of centrifugation of whole blood (6 minutes, 1,100 rpm) collected in a propylene tube containing acid citrate dextrose or sodium citrate (107). The product is a three-layered solution containing platelet-rich plasma (PRP) on top, a thin layer of white blood cells called the buffy coat, and red blood cells at the bottom. After PRP is collected, prostaglandin I2 (PGI2, prostacyclin) or EDTA is usually added [at a concentration of 0.5 μmol/L/L for the former (108) and 5 mmol/L for the latter], to avoid platelet activation. Subsequently, a series of differential centrifugation steps is performed to enrich the product for platelets. This step can be affected by the occurrence of white and red blood cell contamination and also by exposure to activating stimuli. Therefore, size-exclusion approaches such as elution on a sepharose column (109) are often adopted as an alternative to the sequential centrifugations. Microfluidics platforms exploit the small size of a single platelet (2–3 μmol/L in diameter) and can be combined with a second affinity-based technique such as acoustics (110) to achieve higher purity.

Isolated platelets can be analyzed by count, function, and morphology. Usually, functional tests assess platelet aggregation (in vitro platelet-to-platelet aggregate formation via GPIIb/IIIa), platelet adhesion (in vitro ability of platelets to adhere under shear conditions), and platelet activation (flow cytometry analyzes surface and intracellular activation markers). The study of tumor-activated platelets often requires both functional and morphologic analyses; while functional analyses are well standardized (111), the morphologic analyses are conducted mostly using experimental techniques.

Platelets are normally round or oval when in EDTA (Ethylenediaminetetraacetic acid) anticoagulant; upon activation, they develop a characteristic star-shaped appearance, where the central part is occupied by scattered granules, which appear azurophilic (112). The interaction with cancer cells results in an alteration in size, granule localization, mitochondria number and localization, and surface pseudopodia development. Wang and colleagues (113) reported a characteristic change in microtubule and granule distribution in circulating platelets from patients with ovarian cancer as compared with platelets from matched controls. This analysis relied on electron cryotomography, which allows for tridimensional imaging via the acquisition of multiple imaging stacks, resulting in a sharp image, avoiding sample fixation.

Platelet imaging should encompass high-resolution techniques such as structured illumination microscopy and single-molecule localization microscopy, which allow in depth analyses of the cytoskeleton and the actin nodules. Recent advances in multiplexing, with the development of multiplexed flow cytometry and mass cytometry able to target a high number of antigens at once, will more suitably serve platelet morphology studies (114, 115).

A single biomarker is not able to adequately detect and monitor tumors (116–120). The collection of large amounts of complex data represented by tumor tissue–based OMICS, liquid biopsies-derived DNA-sequencing, proteomics, and EVs and platelets will require artificial intelligence tools for integration and analysis (121–123). An attempt in this direction resulted in the generation of PlateletWeb, a platform that integrates literature data with available bioinformatics databases (such as the Uman Protein Database and Gene Ontology) on proteome and transcriptome, to provide a novel tool for the analysis of platelet signaling.

The increased number of targeted therapies available and the need to use them earlier during patients’ clinical course make diagnostic and prognostic protocols increasingly more important. Tumor biopsies are essential to diagnose cancer and provide helpful guidance to clinical practice. Nonetheless, they are limited in conveying all the necessary information due to biological and technical limitations. Liquid biopsies exploit the ability of cancer cells to release biological mediators that can influence the activity of other cells. These mediators can be detected and analyzed to provide information about tumor composition, metastatic behavior, and therapy sensitivity. Moreover, being minimally invasive, they can be repeated longitudinally with a higher level of compliance by patients.

CTCs and cfDNA have been used successfully in several prognostic and predictive clinical studies; moreover, novel strategies using EVs and tumor-activated platelets are being tested. Although these new technologies hold promise for clinical applications, they cannot fully substitute imaging and tumor tissue analysis; however, they can complement them. Because the biggest drawback of liquid biopsy is the low concentration of available cancer-derived material, it will be important to increase the specificity of isolation and enrichment techniques while maintaining their accessibility and cost-effectiveness.

The FDA-approved liquid biopsy–based tests primarily have two applications; these include as a companion diagnostic (FoundationOne and Guardant360) and predictor of outcome (Cell Search). The ct/DNA tests also provide insights regarding the genetic make-up of the tumor to guide targeted therapies. Nonetheless, these applications are biased on one side by the reduced specificity of the circulating cells captured by the assay and on the other by the potential confounding factor from hematopoietic precursors bearing clonal mutation (described above).

We, therefore, envision novel clinical applications for the newly developed liquid biopsy markers (circulating EVs and platelets), including potential for early diagnosis of individuals at risk for developing cancer (e.g., those with BRCA mutation). Clinical trials could benefit from the inclusion of liquid biopsy–based measurements, both at diagnosis and during treatment, ideally at multiple timepoints. Moreover, regular measurements for patients during follow-up or for healthy individuals at risk could be implemented. We recognize that the absence of a specific cancer-derived EV marker and the relatively short lifespan of platelets represent important limitations for their use in clinical practice; these and other assays will require further development and validation studies. Finally, merging multiple datasets to identify informative patterns is the ultimate step in identifying diagnostic, predictive, and prognostic biomarkers to be validated and approved for routine clinical use.

S. Corvigno reports grants from Amy Krouse Rosenthal Foundation during the conduct of the study. A.M. Johnson reports grants from Davidson Research Network during the conduct of the study. A.K. Sood reports grants from NCI and ACS during the conduct of the study; other support from Merck, GSK, Kiyatec, and BioPath outside the submitted work; in addition, A.K. Sood has a patent for EGFL6 antibodies issued. No disclosures were reported by the other authors.

Editorial support was provided by Tamara Locke in Editing Services, Research Medical Library, and the University of Texas MD Anderson Cancer Center. Figure 1 was produced using BioRender.

This work is supported, in part, by the Ovarian Cancer Moon Shot at the UT MD Anderson Cancer Center, R01CA177909, P50 CA098258, P50 CA217685, R35 CA209904, the American Cancer Society, the Ovarian Cancer Research Alliance, and the Frank McGraw Memorial Chair in Cancer Research at the UT MD Anderson Cancer Center.

S. Corvigno's research is supported by the Amy Krouse Rosenthal Foundation and Judy's Mission to End Ovarian Cancer Foundation Research Grant for Early Detection of Ovarian Cancer. AMJ's research is supported by the Davidson Research Network. V. Afshar-Kharghan is supported by NIH, NCI (CA231141, CA177909) grants. D.G. Menter is supported by Institutional Research Grant from UT MD Anderson Cancer Center; the Colorectal Cancer Moon Shot at the UT MD Anderson Cancer Center and 3P30CA016672, P30CA0166723, R01CA218230, and R01CA184843. Overall support was from the Cancer Center Core Grant, PI: Peter Pisters, P50CA221707. M.S. Cho is supported by “the Liz Tilberis Early Career Development Award from the Ovarian Cancer Research Alliance (OCRA) and Ovarian Cancer Research in honor of Liza Chance from the foundation of women cancer” (FWC).

K.-K. Wong is supported by DoD W81XWH-19–1-0169.

1.
Koljenović
S
,
Choo-Smith
L-P
,
Bakker Schut
TC
,
Kros
JM
,
van den Berge
HJ
,
Puppels
GJ
.
Discriminating vital tumor from necrotic tissue in human glioblastoma tissue samples by Raman spectroscopy
.
Lab Invest
2002
;
82
:
1265
77
.
2.
Rosenwald
S
,
Gilad
S
,
Benjamin
S
,
Lebanony
D
,
Dromi
N
,
Faerman
A
, et al
.
Validation of a microRNA-based qRT-PCR test for accurate identification of tumor tissue origin
.
Mod Pathol
2010
;
23
:
814
23
.
3.
Overman
MJ
,
Modak
J
,
Kopetz
S
,
Murthy
R
,
Yao
JC
,
Hicks
ME
, et al
.
Use of research biopsies in clinical trials: are risks and benefits adequately discussed?
J Clin Oncol
2013
;
31
:
17
22
.
4.
Ferrer
I
,
Armstrong
J
,
Capellari
S
,
Parchi
P
,
Arzberger
T
,
Bell
J
, et al
.
Effects of formalin fixation, paraffin embedding, and time of storage on DNA preservation in brain tissue: a BrainNet Europe study
.
Brain Pathol
2007
;
17
:
297
303
.
5.
Diaz
LA
,
Bardelli
A
.
Liquid biopsies: genotyping circulating tumor DNA
.
J Clin Oncol
2014
;
32
:
579
86
.
6.
Leal
A
,
van Grieken
NCT
,
Palsgrove
DN
,
Phallen
J
,
Medina
JE
,
Hruban
C
, et al
.
White blood cell and cell-free DNA analyses for detection of residual disease in gastric cancer
.
Nat Commun
2020
;
11
:
525
.
7.
Keller
L
,
Pantel
K
.
Unravelling tumour heterogeneity by single-cell profiling of circulating tumour cells
.
Nat Rev Cancer
2019
;
19
:
553
67
.
8.
Shi
X
,
Duose
DY
,
Mehrotra
M
,
Harmon
MA
,
Hu
P
,
Wistuba
II
, et al
.
Non-invasive genotyping of metastatic colorectal cancer using circulating cell free DNA
.
Cancer Genet
2019
;
237
:
82
9
.
9.
Morris
V
,
Dasari
A
,
Kopetz
S
.
Can circulating tumor DNA in early-stage colorectal cancer be more than a prognostic biomarker?
JAMA Oncol
2019
;
5
:
1101
3
.
10.
Parseghian
CM
,
Loree
JM
,
Morris
VK
,
Liu
X
,
Clifton
KK
,
Napolitano
S
, et al
.
Anti-EGFR-resistant clones decay exponentially after progression: implications for anti-EGFR re-challenge
.
Ann Oncol
2019
;
30
:
243
9
.
11.
Kopetz
S
,
Guthrie
KA
,
Morris
VK
,
Lenz
HJ
,
Magliocco
AM
,
Maru
D
, et al
.
Randomized trial of irinotecan and cetuximab with or without vemurafenib in BRAF-mutant metastatic colorectal cancer (SWOG S1406)
.
J Clin Oncol
2021
;
39
:
285
94
.
12.
Parseghian
CM
,
Napolitano
S
,
Loree
JM
,
Kopetz
S
.
Mechanisms of innate and acquired resistance to anti-EGFR therapy: a review of current knowledge with a focus on rechallenge therapies
.
Clin Cancer Res
2019
;
25
:
6899
908
.
13.
Fahrmann
JF
,
Mao
X
,
Irajizad
E
,
Katayama
H
,
Capello
M
,
Tanaka
I
, et al
.
Plasma-derived extracellular vesicles convey protein signatures that reflect pathophysiology in lung and pancreatic adenocarcinomas
.
Cancers
2020
;
12
:
1147
.
14.
Gajos
K
,
Kaminska
A
,
Awsiuk
K
,
Bajor
A
,
Gruszczynski
K
,
Pawlak
A
, et al
.
Immobilization and detection of platelet-derived extracellular vesicles on functionalized silicon substrate: cytometric and spectrometric approach
.
Anal Bioanal Chem
2017
;
409
:
1109
19
.
15.
Kanikarla-Marie
P
,
Lam
M
,
Menter
DG
,
Kopetz
S
.
Platelets, circulating tumor cells, and the circulome
.
Cancer Metastasis Rev
2017
;
36
:
235
48
.
16.
In 't Veld
S
,
Wurdinger
T
.
Tumor-educated platelets
.
Blood
2019
;
133
:
2359
64
.
17.
Bresalier
RS
,
Grady
WM
,
Markowitz
SD
,
Nielsen
HJ
,
Batra
SK
,
Lampe
PD
.
Biomarkers for early detection of colorectal cancer: the early detection research network, a framework for clinical translation
.
Cancer Epidemiol Biomarkers Prev
2020
;
29
:
2431
40
.
18.
Pepe
MS
,
Feng
Z
,
Janes
H
,
Bossuyt
PM
,
Potter
JD
.
Pivotal evaluation of the accuracy of a biomarker used for classification or prediction: standards for study design
.
J Natl Cancer Inst
2008
;
100
:
1432
8
.
19.
Low
SK
,
Zembutsu
H
,
Nakamura
Y
.
Breast cancer: the translation of big genomic data to cancer precision medicine
.
Cancer Sci
2018
;
109
:
497
506
.
20.
Mandel
P
,
Metais
P
.
Les acides nucléiques du plasma sanguin chez l'homme
.
C R Seances Soc Biol Fil
1948
;
142
:
241
3
.
21.
Mansueto
G
,
Benincasa
G
,
Della Mura
N
,
Nicoletti
GF
,
Napoli
C
.
Epigenetic-sensitive liquid biomarkers and personalised therapy in advanced heart failure: a focus on cell-free DNA and microRNAs
.
J Clin Pathol
2020
;
73
:
535
43
.
22.
Sentilhes
L
,
Salomon
LJ
,
Vayssiere
C
.
Cell-free DNA analysis for noninvasive examination of trisomy
.
N Engl J Med
2015
;
373
:
2581
2
.
23.
Heitzer
E
,
Ulz
P
,
Geigl
JB
.
Circulating tumor DNA as a liquid biopsy for cancer
.
Clin Chem
2015
;
61
:
112
23
.
24.
Nabet
BY
,
Esfahani
MS
,
Moding
EJ
,
Hamilton
EG
,
Chabon
JJ
,
Rizvi
H
, et al
.
Noninvasive early identification of therapeutic benefit from immune checkpoint inhibition
.
Cell
2020
;
183
:
363
76
.
25.
Cabel
L
,
Proudhon
C
,
Romano
E
,
Girard
N
,
Lantz
O
,
Stern
MH
, et al
.
Clinical potential of circulating tumour DNA in patients receiving anticancer immunotherapy
.
Nat Rev Clin Oncol
2018
;
15
:
639
50
.
26.
Abbosh
C
,
Birkbak
NJ
,
Wilson
GA
,
Jamal-Hanjani
M
,
Constantin
T
,
Salari
R
, et al
.
Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution
.
Nature
2017
;
545
:
446
51
.
27.
Wyatt
AW
,
Annala
M
,
Aggarwal
R
,
Beja
K
,
Feng
F
,
Youngren
J
, et al
.
Concordance of circulating tumor DNA and matched metastatic tissue biopsy in prostate cancer
.
J Natl Cancer Inst
2017
;
109
:
djx118
.
28.
Chae
YK
,
Davis
AA
,
Jain
S
,
Santa-Maria
C
,
Flaum
L
,
Beaubier
N
, et al
.
Concordance of genomic alterations by next-generation sequencing in tumor tissue versus circulating tumor DNA in breast cancer
.
Mol Cancer Ther
2017
;
16
:
1412
20
.
29.
Calapre
L
,
Giardina
T
,
Robinson
C
,
Reid
AL
,
Al-Ogaili
Z
,
Pereira
MR
, et al
.
Locus-specific concordance of genomic alterations between tissue and plasma circulating tumor DNA in metastatic melanoma
.
Mol Oncol
2019
;
13
:
171
84
.
30.
Thress
KS
,
Brant
R
,
Carr
TH
,
Dearden
S
,
Jenkins
S
,
Brown
H
, et al
.
EGFR mutation detection in ctDNA from NSCLC patient plasma: a cross-platform comparison of leading technologies to support the clinical development of AZD9291
.
Lung Cancer
2015
;
90
:
509
15
.
31.
Al Zoughbi
W
,
Fox
J
,
Beg
S
,
Papp
E
,
Hissong
E
,
Ohara
K
, et al
.
Validation of a circulating tumor DNA-based next-generation sequencing assay in a cohort of patients with solid tumors: a proposed solution for decentralized plasma testing
.
Oncologist
2021
;
26
:
e1971
e81
.
32.
Gupta
S
,
Hovelson
DH
,
Kemeny
G
,
Halabi
S
,
Foo
WC
,
Anand
M
, et al
.
Discordant and heterogeneous clinically relevant genomic alterations in circulating tumor cells vs plasma DNA from men with metastatic castration resistant prostate cancer
.
Genes Chromosomes Cancer
2020
;
59
:
225
39
.
33.
Wan
R
,
Wang
Z
,
Lee
JJ
,
Wang
S
,
Li
Q
,
Tang
F
, et al
.
Comprehensive analysis of the discordance of EGFR mutation status between tumor tissues and matched circulating tumor DNA in advanced non-small cell lung cancer
.
J Thorac Oncol
2017
;
12
:
1376
87
.
34.
Vandekerkhove
G
,
Struss
WJ
,
Annala
M
,
Kallio
HML
,
Khalaf
D
,
Warner
EW
, et al
.
Circulating tumor DNA abundance and potential utility in de novo metastatic prostate cancer
.
Eur Urol
2019
;
75
:
667
75
.
35.
Cheng
ML
,
Pectasides
E
,
Hanna
GJ
,
Parsons
HA
,
Choudhury
AD
,
Oxnard
GR
.
Circulating tumor DNA in advanced solid tumors: clinical relevance and future directions
.
CA Cancer J Clin
2021
;
71
:
176
90
.
36.
Shen
SY
,
Singhania
R
,
Fehringer
G
,
Chakravarthy
A
,
Roehrl
MHA
,
Chadwick
D
, et al
.
Sensitive tumour detection and classification using plasma cell-free DNA methylomes
.
Nature
2018
;
563
:
579
83
.
37.
Dasari
A
,
Morris
VK
,
Allegra
CJ
,
Atreya
C
,
Benson
AB
, 3rd,
Boland
P
, et al
.
ctDNA applications and integration in colorectal cancer: an NCI Colon and Rectal-Anal Task Forces whitepaper
.
Nat Rev Clin Oncol
2020
;
17
:
757
70
.
38.
Venook
AP
.
Colorectal cancer surveillance with circulating tumor DNA assay
.
JAMA Netw Open
2022;5(3):e221100.
.
39.
Woodhouse
R
,
Dennis
L
,
Li
MJ
,
Burns
C
,
Ma
P
,
Meng
W
, et al
.
Clinical and analytical validation of FoundationOne Liquid CDx, a novel 324-gene blood-based comprehensive genomic profiling assay
.
J Clin Oncol
38
:15s,
2020
(suppl; abstr e13685).
40.
Talasaz
A
,
Mortimer
S
,
Sebisanovic
D
,
Siew
L
,
Zapanta
A
,
Mei
GW
, et al
.
Use of the GUARDANT360 noninvasive tumor sequencing assay on 300 patients across colorectal, melanoma, bang, breast, and prostate cancers and its clinical utility
.
J Clin Oncol
2014
;
32
.
41.
Alix-Panabières
C
,
Pantel
K
.
Clinical applications of circulating tumor cells and circulating tumor DNA as liquid biopsy
.
Cancer Discov
2016
;
6
:
479
91
.
42.
Tan
CL
,
Lim
TH
,
Lim
TKH
,
Tan
DS-W
,
Chua
YW
,
Ang
MK
, et al
.
Concordance of anaplastic lymphoma kinase (ALK) gene rearrangements between circulating tumor cells and tumor in non-small cell lung cancer
.
Oncotarget
2016
;
7
:
23251
62
.
43.
Pailler
E
,
Adam
J
,
Barthélémy
A
,
Oulhen
M
,
Auger
N
,
Valent
A
, et al
.
Detection of circulating tumor cells harboring a unique ALK rearrangement in ALK-positive non–small-cell lung cancer
.
J Clin Oncol
2013
;
31
:
2273
81
.
44.
Ilie
M
,
Long
E
,
Butori
C
,
Hofman
V
,
Coelle
C
,
Mauro
V
, et al
.
ALK-gene rearrangement: a comparative analysis on circulating tumour cells and tumour tissue from patients with lung adenocarcinoma
.
Ann Oncol
2012
;
23
:
2907
13
.
45.
Aceto
N
,
Bardia
A
,
Miyamoto
DT
,
Donaldson
MC
,
Wittner
BS
,
Spencer
JA
, et al
.
Circulating tumor cell clusters are oligoclonal precursors of breast cancer metastasis
.
Cell
2014
;
158
:
1110
22
.
46.
Batth
IS
,
Mitra
A
,
Rood
S
,
Kopetz
S
,
Menter
D
,
Li
S
.
CTC analysis: an update on technological progress
.
Transl Res
2019
;
212
:
14
25
.
47.
Qi
LN
,
Xiang
BD
,
Wu
FX
,
Ye
JZ
,
Zhong
JH
,
Wang
YY
, et al
.
Circulating tumor cells undergoing EMT provide a metric for diagnosis and prognosis of patients with hepatocellular carcinoma
.
Cancer Res
2018
;
78
:
4731
44
.
48.
Khattak
MA
,
Reid
A
,
Freeman
J
,
Pereira
M
,
McEvoy
A
,
Lo
J
, et al
.
PD-L1 expression on circulating tumor cells may be predictive of response to pembrolizumab in advanced melanoma: results from a pilot study
.
Oncologist
2020
;
25
:
E520
E7
.
49.
Hashimoto
M
,
Tanaka
F
,
Yoneda
K
,
Takuwa
T
,
Kuroda
A
,
Matsumoto
S
, et al
.
The clinical value of circulating tumour cells (CTCs) in patients undergoing pulmonary metastasectomy for metastatic colorectal cancer
.
J Thorac Dis
2018
;
10
:
1569
77
.
50.
Li
P
,
Mao
Z
,
Peng
Z
,
Zhou
L
,
Chen
Y
,
Huang
PH
, et al
.
Acoustic separation of circulating tumor cells
.
Proc Natl Acad Sci U S A
2015
;
112
:
4970
5
.
51.
de Wit
S
,
van Dalum
G
,
Lenferink
AT
,
Tibbe
AG
,
Hiltermann
TJ
,
Groen
HJ
, et al
.
The detection of EpCAM(+) and EpCAM(-) circulating tumor cells
.
Sci Rep
2015
;
5
:
12270
.
52.
Pecot
CV
,
Bischoff
FZ
,
Mayer
JA
,
Wong
KL
,
Pham
T
,
Bottsford-Miller
J
, et al
.
A novel platform for detection of CK+ and CK- CTCs
.
Cancer Discov
2011
;
1
:
580
6
.
53.
Kim
O
,
Lee
D
,
Chungwon Lee
A
,
Lee
Y
,
Bae
HJ
,
Lee
HB
, et al
.
Whole genome sequencing of single circulating tumor cells isolated by applying a pulsed laser to cell-capturing microstructures
.
Small
2019
;
15
:
e1902607
.
54.
Cheng
YH
,
Chen
YC
,
Lin
E
,
Brien
R
,
Jung
S
,
Chen
YT
, et al
.
Hydro-Seq enables contamination-free high-throughput single-cell RNA-sequencing for circulating tumor cells
.
Nat Commun
2019
;
10
:
2163
.
55.
Steensma
DP
,
Bejar
R
,
Jaiswal
S
,
Lindsley
RC
,
Sekeres
MA
,
Hasserjian
RP
, et al
.
Clonal hematopoiesis of indeterminate potential and its distinction from myelodysplastic syndromes
.
Blood
2015
;
126
:
9
16
.
56.
Bowman
RL
,
Busque
L
,
Levine
RL
.
Clonal hematopoiesis and evolution to hematopoietic malignancies
.
Cell Stem Cell
2018
;
22
:
157
70
.
57.
Jaiswal
S
,
Fontanillas
P
,
Flannick
J
,
Manning
A
,
Grauman
PV
,
Mar
BG
, et al
.
Age-related clonal hematopoiesis associated with adverse outcomes
.
New Engl J Med
2014
;
371
:
2488
98
.
58.
Théry
C
,
Zitvogel
L
,
Amigorena
S
.
Exosomes: composition, biogenesis and function
.
Nat Rev Immunol
2002
;
2
:
569
79
.
59.
Colombo
M
,
Raposo
G
,
Thery
C
.
Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles
.
Annu Rev Cell Dev Biol
2014
;
30
:
255
89
.
60.
Henne
WM
,
Stenmark
H
,
Emr
SD
.
Molecular mechanisms of the membrane sculpting ESCRT pathway
.
Cold Spring Harb Perspect Biol
2013
;
5
:
a016766
.
61.
Sansone
P
,
Savini
C
,
Kurelac
I
,
Chang
Q
,
Amato
LB
,
Strillacci
A
, et al
.
Packaging and transfer of mitochondrial DNA via exosomes regulate escape from dormancy in hormonal therapy-resistant breast cancer
.
Proc Natl Acad Sci U S A
2017
;
114
:
E9066
E75
.
62.
Xu
R
,
Rai
A
,
Chen
M
,
Suwakulsiri
W
,
Greening
DW
,
Simpson
RJ
.
Extracellular vesicles in cancer - implications for future improvements in cancer care
.
Nat Rev Clin Oncol
2018
;
15
:
617
38
.
63.
Melo
SA
,
Sugimoto
H
,
O'Connell
JT
,
Kato
N
,
Villanueva
A
,
Vidal
A
, et al
.
Cancer exosomes perform cell-independent microRNA biogenesis and promote tumorigenesis
.
Cancer Cell
2014
;
26
:
707
21
.
64.
Burbidge
K
,
Zwikelmaier
V
,
Cook
B
,
Long
MM
,
Balva
B
,
Lonigro
M
, et al
.
Cargo and cell-specific differences in extracellular vesicle populations identified by multiplexed immunofluorescent analysis
.
J Extracell Vesicles
2020
;
9
:
1789326
.
65.
Iraci
N
,
Leonardi
T
,
Gessler
F
,
Vega
B
,
Pluchino
S
.
Focus on extracellular vesicles: physiological role and signalling properties of extracellular membrane vesicles
.
Int J Mol Sci
2016
;
17
:
171
.
66.
Yanez-Mo
M
,
Siljander
PR
,
Andreu
Z
,
Zavec
AB
,
Borras
FE
,
Buzas
EI
, et al
.
Biological properties of extracellular vesicles and their physiological functions
.
J Extracell Vesicles
2015
;
4
:
27066
.
67.
Zomer
A
,
Maynard
C
,
Verweij
FJ
,
Kamermans
A
,
Schafer
R
,
Beerling
E
, et al
.
In vivo imaging reveals extracellular vesicle-mediated phenocopying of metastatic behavior
.
Cell
2015
;
161
:
1046
57
.
68.
Boelens
MC
,
Wu
TJ
,
Nabet
BY
,
Xu
B
,
Qiu
Y
,
Yoon
T
, et al
.
Exosome transfer from stromal to breast cancer cells regulates therapy resistance pathways
.
Cell
2014
;
159
:
499
513
.
69.
Hoshino
A
,
Costa-Silva
B
,
Shen
TL
,
Rodrigues
G
,
Hashimoto
A
,
Tesic Mark
M
, et al
.
Tumour exosome integrins determine organotropic metastasis
.
Nature
2015
;
527
:
329
35
.
70.
Thery
C
,
Witwer
KW
,
Aikawa
E
,
Alcaraz
MJ
,
Anderson
JD
,
Andriantsitohaina
R
, et al
.
Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines
.
J Extracell Vesicles
2018
;
7
:
1535750
.
71.
Van Deun
J
,
Mestdagh
P
,
Sormunen
R
,
Cocquyt
V
,
Vermaelen
K
,
Vandesompele
J
, et al
.
The impact of disparate isolation methods for extracellular vesicles on downstream RNA profiling
.
J Extracell Vesicles
2014
;
3
.
72.
Mateescu
B
,
Kowal
EJ
,
van Balkom
BW
,
Bartel
S
,
Bhattacharyya
SN
,
Buzas
EI
, et al
.
Obstacles and opportunities in the functional analysis of extracellular vesicle RNA - an ISEV position paper
.
J Extracell Vesicles
2017
;
6
:
1286095
.
73.
Lamparski
HG
,
Metha-Damani
A
,
Yao
JY
,
Patel
S
,
Hsu
DH
,
Ruegg
C
, et al
.
Production and characterization of clinical grade exosomes derived from dendritic cells
.
J Immunol Methods
2002
;
270
:
211
26
.
74.
Zhang
H
,
Lyden
D
.
Asymmetric-flow field-flow fractionation technology for exomere and small extracellular vesicle separation and characterization
.
Nat Protoc
2019
;
14
:
1027
53
.
75.
Liu
C
,
Guo
J
,
Tian
F
,
Yang
N
,
Yan
F
,
Ding
Y
, et al
.
Field-free isolation of exosomes from extracellular vesicles by microfluidic viscoelastic flows
.
ACS Nano
2017
;
11
:
6968
76
.
76.
Vaidyanathan
R
,
Naghibosadat
M
,
Rauf
S
,
Korbie
D
,
Carrascosa
LG
,
Shiddiky
MJ
, et al
.
Detecting exosomes specifically: a multiplexed device based on alternating current electrohydrodynamic induced nanoshearing
.
Anal Chem
2014
;
86
:
11125
32
.
77.
Wu
M
,
Ouyang
Y
,
Wang
Z
,
Zhang
R
,
Huang
PH
,
Chen
C
, et al
.
Isolation of exosomes from whole blood by integrating acoustics and microfluidics
.
Proc Natl Acad Sci U S A
2017
;
114
:
10584
9
.
78.
Gamez-Valero
A
,
Monguio-Tortajada
M
,
Carreras-Planella
L
,
Franquesa
M
,
Beyer
K
,
Borras
FE
.
Size-exclusion chromatography-based isolation minimally alters extracellular vesicles' characteristics compared to precipitating agents
.
Sci Rep
2016
;
6
:
33641
.
79.
Heath
N
,
Grant
L
,
De Oliveira
TM
,
Rowlinson
R
,
Osteikoetxea
X
,
Dekker
N
, et al
.
Rapid isolation and enrichment of extracellular vesicle preparations using anion exchange chromatography
.
Sci Rep
2018
;
8
:
5730
.
80.
Merchant
ML
,
Powell
DW
,
Wilkey
DW
,
Cummins
TD
,
Deegens
JK
,
Rood
IM
, et al
.
Microfiltration isolation of human urinary exosomes for characterization by MS
.
Proteomics Clin Appl
2010
;
4
:
84
96
.
81.
Morales-Kastresana
A
,
Musich
TA
,
Welsh
JA
,
Telford
W
,
Demberg
T
,
Wood
JCS
, et al
.
High-fidelity detection and sorting of nanoscale vesicles in viral disease and cancer
.
J Extracell Vesicles
2019
;
8
:
1597603
.
82.
Wunsch
BH
,
Smith
JT
,
Gifford
SM
,
Wang
C
,
Brink
M
,
Bruce
RL
, et al
.
Nanoscale lateral displacement arrays for the separation of exosomes and colloids down to 20 nm
.
Nat Nanotechnol
2016
;
11
:
936
40
.
83.
Nakai
W
,
Yoshida
T
,
Diez
D
,
Miyatake
Y
,
Nishibu
T
,
Imawaka
N
, et al
.
A novel affinity-based method for the isolation of highly purified extracellular vesicles
.
Sci Rep
2016
;
6
:
33935
.
84.
Huang
T
,
He
J
.
Characterization of extracellular vesicles by size-exclusion high-performance liquid chromatography (HPLC)
.
Methods Mol Biol
2017
;
1660
:
191
9
.
85.
Zhang
P
,
Zhou
X
,
He
M
,
Shang
Y
,
Tetlow
AL
,
Godwin
AK
, et al
.
Ultrasensitive detection of circulating exosomes with a 3D-nanopatterned microfluidic chip
.
Nat Biomed Eng
2019
;
3
:
438
51
.
86.
Sim
X
,
Poncz
M
,
Gadue
P
,
French
DL
.
Understanding platelet generation from megakaryocytes: implications for in vitro-derived platelets
.
Blood
2016
;
127
:
1227
33
.
87.
Menter
DG
,
Kopetz
S
,
Hawk
E
,
Sood
AK
,
Loree
JM
,
Gresele
P
, et al
.
Platelet “first responders” in wound response, cancer, and metastasis
.
Cancer Metastasis Rev
2017
;
36
:
199
213
.
88.
Robier
C
.
Platelet morphology
.
J Lab Med
2020
;
44
:
231
9
.
89.
Ludhiadch
A
,
Muralidharan
A
,
Balyan
R
,
Munshi
A
.
The molecular basis of platelet biogenesis, activation, aggregation and implications in neurological disorders
.
Int J Neurosci
2020
;
130
:
1237
49
.
90.
Liu
T
,
Li
M
,
Tang
J
,
Li
J
,
Zhou
Y
,
Liu
Y
, et al
.
An acoustic strategy for gold nanoparticle loading in platelets as biomimetic multifunctional carriers
.
J Mater Chem B
2019
;
7
:
2138
44
.
91.
Selvadurai
MV
,
Hamilton
JR
.
Structure and function of the open canalicular system - the platelet's specialized internal membrane network
.
Platelets
2018
;
29
:
319
25
.
92.
Nguyen
TH
,
Schuster
N
,
Greinacher
A
,
Aurich
K
.
Uptake pathways of protein-coated magnetic nanoparticles in platelets
.
ACS Appl Mater Interfaces
2018
;
10
:
28314
21
.
93.
Haemmerle
M
,
Stone
RL
,
Menter
DG
,
Afshar-Kharghan
V
,
Sood
AK
.
The platelet lifeline to cancer: challenges and opportunities
.
Cancer Cell
2018
;
33
:
965
83
.
94.
Best Myron
G
,
Sol
N
,
Kooi
I
,
Tannous
J
,
Westerman Bart
A
,
Rustenburg
F
, et al
.
RNA-seq of tumor-educated platelets enables blood-based pan-cancer, multiclass, and molecular pathway cancer diagnostics
.
Cancer Cell
2015
;
28
:
666
76
.
95.
Stone
RL
,
Nick
AM
,
McNeish
IA
,
Balkwill
F
,
Han
HD
,
Bottsford-Miller
J
, et al
.
Paraneoplastic thrombocytosis in ovarian cancer
.
N Engl J Med
2012
;
366
:
610
8
.
96.
Kuznetsov
HS
,
Marsh
T
,
Markens
BA
,
Castano
Z
,
Greene-Colozzi
A
,
Hay
SA
, et al
.
Identification of luminal breast cancers that establish a tumor-supportive macroenvironment defined by proangiogenic platelets and bone marrow-derived cells
.
Cancer Discov
2012
;
2
:
1150
65
.
97.
van Velzen
JF
,
Laros-van Gorkom
BAP
,
Pop
GAM
,
van Heerde
WL
.
Multicolor flow cytometry for evaluation of platelet surface antigens and activation markers
.
Thromb Res
2012
;
130
:
92
8
.
98.
Schubert
S
,
Weyrich
AS
,
Rowley
JW
.
A tour through the transcriptional landscape of platelets
.
Blood
2014
;
124
:
493
502
.
99.
Schwertz
H
,
Tolley
ND
,
Foulks
JM
,
Denis
MM
,
Risenmay
BW
,
Buerke
M
, et al
.
Signal-dependent splicing of tissue factor pre-mRNA modulates the thrombogenecity of human platelets
.
J Exp Med
2006
;
203
:
2433
40
.
100.
Power
KA
,
McRedmond
JP
,
de
SA
,
Gallagher
WM
,
Gaora
PO
.
High-throughput proteomics detection of novel splice isoforms in human platelets
.
PLoS One
2009
;
4
:
e5001
.
101.
Liefaard
MC
,
Lips
E
,
Best
M
,
Sol
N
,
Veld
SI
,
Rookus
M
, et al
.
RNA signatures from tumor-educated platelets (TEP) enable detection of early-stage breast cancer
.
Ann Oncol
2019
;
30
.
102.
Sol
N
,
In 't Veld
S
,
Vancura
A
,
Tjerkstra
M
,
Leurs
C
,
Rustenburg
F
, et al
.
Tumor-educated platelet RNA for the detection and (Pseudo)progression monitoring of glioblastoma
.
Cell Rep Med
2020
;
1
:
100101
.
103.
Milde
M
,
Werthmann
RC
,
von Hayn
K
,
Bunemann
M
.
Dynamics of adenylate cyclase regulation via heterotrimeric G-proteins
.
Biochem Soc Trans
2014
;
42
:
239
43
.
104.
Castellone
MD
,
Teramoto
H
,
Williams
BO
,
Druey
KM
,
Gutkind
JS
.
Prostaglandin E-2 promotes colon cancer cell growth through a G(s)-axin-beta-catenin signaling axis
.
Science
2005
;
310
:
1504
10
.
105.
Chiang
JY
,
Lee
SH
,
Chen
YC
,
Wu
CK
,
Chuang
JY
,
Lo
SC
, et al
.
Metabolomic analysis of platelets of patients with aspirin non-response
.
Front Pharmacol
2019
;
10
:
1107
.
106.
Cenido
JF
,
Itin
B
,
Stark
RE
,
Huang
YY
,
Oquendo
MA
,
John Mann
J
, et al
.
Characterization of lipid rafts in human platelets using nuclear magnetic resonance: a pilot study
.
Biochem Biophys Rep
2017
;
10
:
132
6
.
107.
Haemmerle
M
,
Bottsford-Miller
J
,
Pradeep
S
,
Taylor
ML
,
Choi
HJ
,
Hansen
JM
, et al
.
FAK regulates platelet extravasation and tumor growth after antiangiogenic therapy withdrawal
.
J Clin Invest
2016
;
126
:
1885
96
.
108.
Hechler
B
,
Dupuis
A
,
Mangin
PH
,
Gachet
C
.
Platelet preparation for function testing in the laboratory and clinic: historical and practical aspects
.
Res Pract Thromb Haemost
2019
;
3
:
615
25
.
109.
Vollmar
B
,
Slotta
JE
,
Nickels
RM
,
Wenzel
E
,
Menger
MD
.
Comparative analysis of platelet isolation techniques for the in vivo study of the microcirculation
.
Microcirculation
2003
;
10
:
143
52
.
110.
Nam
J
,
Lim
H
,
Kim
D
,
Shin
S
.
Separation of platelets from whole blood using standing surface acoustic waves in a microchannel
.
Lab Chip
2011
;
11
:
3361
4
.
111.
Paniccia
R
,
Priora
R
,
Liotta
AA
,
Abbate
R
.
Platelet function tests: a comparative review
.
Vasc Health Risk Manag
2015
;
11
:
133
48
.
112.
Bain
B
.
Blood cells: an atlas of morphology (with clinical relevance)
.
Br J Haematol
2008
;
143
:
758
.
113.
Wang
R
,
Stone
RL
,
Kaelber
JT
,
Rochat
RH
,
Nick
AM
,
Vijayan
KV
, et al
.
Electron cryotomography reveals ultrastructure alterations in platelets from patients with ovarian cancer
.
Proc Natl Acad Sci U S A
2015
;
112
:
14266
71
.
114.
Spurgeon
BEJ
,
Aburima
A
,
Oberprieler
NG
,
Tasken
K
,
Naseem
KM
.
Multiplexed phosphospecific flow cytometry enables large-scale signaling profiling and drug screening in blood platelets
.
J Thromb Haemost
2014
;
12
:
1733
43
.
115.
Blair
TA
,
Michelson
AD
,
Frelinger
AL
.
Mass cytometry reveals distinct platelet subtypes in healthy subjects and novel alterations in surface glycoproteins in glanzmann thrombasthenia
.
Sci Rep
2018
;
8
:
10300
.
116.
Lamanna
J
,
Scott
EY
,
Edwards
HS
,
Chamberlain
MD
,
Dryden
MDM
,
Peng
J
, et al
.
Digital microfluidic isolation of single cells for -Omics
.
Nat Commun
2020
;
11
:
5632
.
117.
Zhu
Y
,
Orre
LM
,
Johansson
HJ
,
Huss
M
,
Boekel
J
,
Vesterlund
M
, et al
.
Discovery of coding regions in the human genome by integrated proteogenomics analysis workflow
.
Nat Commun
2018
;
9
:
903
.
118.
Weerts
MJA
,
Timmermans
EC
,
van de Stolpe
A
,
Vossen
R
,
Anvar
SY
,
Foekens
JA
, et al
.
Tumor-specific mitochondrial DNA variants are rarely detected in cell-free DNA
.
Neoplasia
2018
;
20
:
687
96
.
119.
Pisanic
TR
, 2nd,
Athamanolap
P
,
Poh
W
,
Chen
C
,
Hulbert
A
,
Brock
MV
, et al
.
DREAMing: a simple and ultrasensitive method for assessing intratumor epigenetic heterogeneity directly from liquid biopsies
.
Nucleic Acids Res
2015
;
43
:
e154
.
120.
Amorim
MG
,
Valieris
R
,
Drummond
RD
,
Pizzi
MP
,
Freitas
VM
,
Sinigaglia-Coimbra
R
, et al
.
A total transcriptome profiling method for plasma-derived extracellular vesicles: applications for liquid biopsies
.
Sci Rep
2017
;
7
:
14395
.
121.
Goldstein
E
,
Yeghiazaryan
K
,
Ahmad
A
,
Giordano
FA
,
Frohlich
H
,
Golubnitschaja
O
.
Optimal multiparametric set-up modelled for best survival outcomes in palliative treatment of liver malignancies: unsupervised machine learning and 3 PM recommendations
.
EPMA J
2020
;
11
:
505
15
.
122.
Ren
Y
,
Zhang
Y
,
Wang
D
,
Liu
F
,
Fu
Y
,
Xiang
S
, et al
.
SinoDuplex: an improved duplex sequencing approach to detect low-frequency variants in plasma cfDNA samples
.
Genomics Proteomics Bioinformatics
2020
;
18
:
81
90
.
123.
Chen
MX
,
Wang
SY
,
Kuo
CH
,
Tsai
IL
.
Metabolome analysis for investigating host-gut microbiota interactions
.
J Formos Med Assoc
2019
;
118
:
S10
22
.