Abstract
Circulating tumor cells (CTCs) are pivotal in the distant metastasis of tumors, serving as one of the primary materials for liquid biopsy. They hold significant clinical importance in assessing prognosis, predicting efficacy, evaluating therapeutic outcomes, and studying recurrence, metastasis, and resistance mechanisms in cancer patients. Nevertheless, the rareness and heterogeneity of CTC and the complexity of metastasis make the clinical application of CTC detection confront many challenges, which may need to be settled by some practical strategies. This article will review the content mentioned above.
Introduction
Malignant tumors are a significant cause of human mortality [1], responsible for over 90 % of deaths attributed to these tumors due to distant metastases [2]. The process of distant metastasis can be generally divided into several stages: Tumor cells detach from the primary tumor, breach the blood vessel barrier, and enter the bloodstream. Once in the circulatory system, these cells can survive, multiply, and eventually form metastatic foci. Among these, tumor cells that survive after breaching the blood vessel barrier in the circulatory system are known as Circulating Tumor Cells (CTCs), a heterogeneous group that may exhibit significant variations in cell size, morphology, molecular phenotype, activity level, metastatic potential, and proliferative capacity. CTCs can exist as single cells or aggregate with each other or blood-derived cells to form small multicellular aggregates (Circulating Tumor Microemboli, CTMs). Research has found that CTM is associated with poor clinical outcomes and is an independent prognostic marker [3]. Although the exact reasons for the formation of CTM are not fully understood, the aggregation of these cells might be a strategy for CTCs to cope with immune system attacks, and the microenvironment within CTM may be more conducive to maintaining their biological characteristics and helping them form metastatic foci at distant sites [4]. Therefore, CTM may hold more excellent research value than CTCs for studying metastasis mechanisms. Relevant research could potentially provide important insights into how metastasis initiates and develops, and it might expand the concept of “liquid biopsy” to other cell types beyond CTCs. CTCs are considered the “seeds” of tumor metastasis and an essential window for revealing the malignant behavior of tumors [5], 6]. This review will cover the clinical applications of CTC detection and the challenges and strategies encountered.
Clinical applications of CTC detection
As a critical step in the distant metastasis of malignant tumors, the presence or absence and quantity of CTCs in a patient’s bloodstream, on the one hand, represents the tumor’s ability to infiltrate into blood vessels, and on the other hand, indicates the possibility of forming metastatic foci in remote organs [7], 8]. Therefore, detecting and counting the number of CTCs can provide insights into the malignancy of the tumor and the risk of metastasis, carrying significant prognostic value. Moreover, CTCs are an essential component of “liquid biopsy,” compensating for the difficulty in dynamically acquiring tumor tissue. Performing real-time molecular and functional typing on them enables more precise guidance for the individualized treatment of patients [9], [10], [11]. The specific clinical applications of CTC detection include early screening of high-risk populations, accurate staging of confirmed patients, monitoring of recurrence and metastasis after early surgery, predicting prognosis before treatment initiation in advanced patients, and evaluating efficacy at the end of each treatment cycle, providing substantial assistance in the comprehensive management of patients.
Early screening of high-risk populations
Current clinical evidence suggests that cancer cells may have entered the bloodstream before patients exhibit symptoms. As a result, many anticipate using CTCs for screening high-risk populations [12], 13]. Unfortunately, sufficient evidence-based medicine currently does not support this application. Although individual case reports hint at the potential value of CTCs in this context [14], to truly implement them in clinical practice, apart from requiring long-term follow-up studies on large cohorts to obtain reliable, evidence-based medicine, there is an urgent need for more sensitive detection technologies to emerge.
Accurate staging of confirmed patients
The current clinical routine TNM staging system primarily evaluates the status of the primary tumor (Tumor, T), lymph nodes (Node, N), and distant metastasis (Metastasis, M) in cancer patients, providing significant guidance for treatment. However, according to reports in the literature, the detection rate of CTCs in early breast cancer, as determined by conventional pathology and imaging methods, ranges from 2 to 55 % [15]. This data suggests that even without evidence of distant metastasis, many patients already risk metastasis, making early treatment strategies potentially inappropriate. Considering this scenario, the American Joint Committee on Cancer (AJCC) introduced a new category, cM0 (i+), between MO (no distant metastasis) and Ml (distant metastasis) in its 2010 Cancer Staging Manual, marking the first time such a distinction was made in the TNM classification system, thus making the treatment more precise for patients [16].
Monitoring of postoperative recurrence and metastasis in early-stage patients
Even after undergoing adjuvant therapy post-surgery, 20–30 % of early-stage patients may experience recurrence and metastasis within years [17]. As the disease progresses in stages, if dynamic monitoring were initiated immediately following patients’ completion of their post-surgical adjuvant therapy, it might be possible to detect signs of recurrence and metastasis before they become visually apparent, allowing for earlier intervention and more control over treatment. Although there is currently a lack of evidence-based medicine supporting this approach, the potential application of CTCs in this phase is expected to expand significantly as future detection technologies improve.
Prognostic judgment of patients with advanced disease
For patients with advanced disease, the treatment strategy primarily focuses on palliative care, with greater emphasis on improving the quality of life for the patient. At this stage, CTC detection allows for more precise prognosis assessment, aiding personalized treatment, making it one of the most extensively studied areas in current CTC application research [18], [19], [20], [21], [22]. The most influential study was the prospective, multicenter registry clinical trial conducted by CellSearch system in the United States [22], which established a threshold of 5 CTCs per 7.5 mL in breast cancer and confirmed that CTCs are an independent predictor of both progression-free survival and overall survival. This work has been further validated in other breast cancer studies [17], 23] and has had a profound impact on CTC research in other cancer types, including colorectal cancer [24], [25], [26], prostate cancer [27], 28], non-small cell lung cancer [29], [30], [31], and small cell lung cancer [32], [33], [34], all of which have yielded similar findings.
Efficacy evaluation and efficacy prediction
Tumor markers and imaging tests are conventional methods for evaluating patient efficacy. However, the former has limited reference value due to individual patient differences. Moreover, the latter has a certain lag in time, typically requiring two treatment cycles (6–8 weeks) to reach a definitive conclusion. Previous literature reports have identified that the number of CTCs remains ≥5 after 1–2 treatment cycles as a standard for assessing whether a patient benefits from the treatment [35]. Compared to tumor markers, the evaluation of CTCs is more consistent with clinical outcomes, and compared to imaging, it provides more timely assessments, allowing for accurate evaluations by the end of the first course of treatment (3–4 weeks), which is particularly meaningful for patients undergoing expensive treatments [36], 37]. Although imaging methods remain the primary standard for evaluating patient efficacy, it is shown [38] that CTC can provide helpful corrections and supplements to traditional imaging evaluations [39], and the combination of the two can provide richer patient information, thus ensuring that patients can receive more rational treatment. Regarding the prognostic judgment and efficacy evaluation of advanced patients, a large amount of evidence-based medical evidence has been accumulated, which is the most classical clinical application of CTC [40], 41].
Late-stage patients undergo targeted therapy after the detection of relevant biomarkers to predict efficacy, such as EGFR mutations and ALK fusions in lung cancer [42], HER2 amplification in breast cancer [43], and KRAS mutations in colorectal cancer, among others [44]. In clinical practice, tumor tissue samples are the optimal source for these tests. However, late-stage patients may not always provide samples from the time of diagnosis or have enough material for testing, in which case CTCs can be a beneficial supplement. The aspect is that CTC reflects the patient’s real-time condition, and on the other hand, because CTC is the most dangerous cell in the patient’s body, treating these cells will bring more benefits to the patient. However, unlike histological testing, which has been repeatedly practiced and proven in clinical settings, the criteria for interpreting CTC tests must be better established and require validation and optimization through more clinical data.
In the realm of fundamental research, CTCs serve as a crucial material source for exploring pivotal issues such as the molecular mechanisms underlying epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET) [36], 45], 46], the phenotypic characteristics and functional identification of circulating tumor stem cells [47], the elucidation of resistance mechanisms and drug sensitivity analysis [48], distinguishing markers for CTC dormancy and fate determination [49], the causes and action mechanisms of CTMs [50], and so forth. Addressing these questions will aid in our understanding of the specific roles of CTCs in tumor progression, facilitate the optimization and improvement of CTC detection methods, and provide essential foundations for future drug development or prevention strategies.
Comparison of CTCs and circulating tumor DNA
CTCs and ctDNA (circulating tumor DNA) are frequently compared as essential components of liquid biopsies. Each has its advantages and can complement each other, reflecting the characteristics of tumors from different perspectives. The combined information from both aspects can provide a more comprehensive understanding of the patient’s condition [51], 52].
The advantage of ctDNA is that it is widely distributed and relatively homogeneous, and the corresponding digital PCR or second-generation sequencing technologies are well-established and sensitive enough to handle it. However, ctDNA is highly fragmented, so detection is limited to point mutations, amplifications and deletions, translocations, and methylation. In addition, due to the lack of cellular morphological support, it is challenging to distinguish tumor-derived ctDNA from normal Cell-free DNA (cfDNA), so it is necessary to exercise sufficient caution when the test results are negative [53], 54].
The advantage of CTCs lies in their complete cellular morphology, with the intracellular contents preserved mainly within the cell membrane. Even a single cell can possess a complete genome, transcriptome, proteome, and the potential for invasion, proliferation, and metastasis. As a result, the testing on CTCs is more comprehensive, with higher reliability of results. In addition to covering all tests of ctDNA, it also allows for related mRNA tests, detection of deletions, amplifications, and fusions at the chromosomal level, as well as counting, morphological observation, protein expression, and functional studies [55], 56]. However, the challenge with CTC testing is the lack of universally accepted reliable techniques. Obtaining a sufficient quantity and quality of CTCs that meet the requirements for downstream analysis is a prerequisite for advancing related work. For the foreseeable future, this remains a bottleneck affecting the development of the entire field.
The fundamental principles of CTC detection technology
The number of CTCs in peripheral blood is exceedingly scarce, with approximately 1 CTC found among 106−7 leukocytes. Consequently, CTC detection necessitates enrichment based on their biological or physical characteristics first, followed by identification of the enriched cells according to the CTC’s gene expression or functional attributes [57], 58] (Figure 1).

The fundamental principles of CTC detection technology.
The primary purpose of CTC enrichment is to optimize output and purity to maximize basic research and clinical applications. The main strategies can be divided into two categories: affinity-based and non-affinity-based. Affinity-based enrichment techniques, currently the most widely used methods for CTC enrichment, primarily rely on unique antigens expressed differently on CTCs compared to leukocytes to achieve this, including positive enrichment using antigens expressed on CTCs but not on leukocytes (such as epithelial cell adhesion molecule, EpCAM) and indirect negative enrichment using antigens expressed on leukocytes but not on CTCs (such as CD45). On the other hand, non-affinity-based enrichment exploits the differences in cell size, density, centrifugal force, and charge characteristics between CTCs and leukocytes to achieve enrichment, such as microfiltration methods, inertial chip technology, density gradient centrifugation, and dielectrophoresis, among others. While these methods may not have the specificity of affinity-based enrichment techniques, they yield more comprehensive cell populations, which will be beneficial for the analysis of CTC heterogeneity [55], [56], [57], [58].
CTC detection strategies primarily encompass phenotypic and functional assays. The most common phenotypic assays involve detecting specific protein expression using fluorescently labeled antibodies (e.g., for positive identification with cytokeratin, exclusion of leukocytes with CD45, and nuclear staining with DAPI), which offers the advantage of directly visualizing cells but is hindered by time-consuming and labor-intensive image scanning and interpretation, as well as susceptibility to subjective factors. Additionally, mRNA from epithelial-specific (such as cytokeratin 19) or tissue-specific sources (such as breast globin) can be quantitatively analyzed through reverse transcription polymerase chain reaction (RT-PCR) for phenotypic assessment, which is cost-effective, has established quality control systems, and allows for automated processing, providing relatively objective results. However, this method does not permit direct observation of cell morphology and cannot support subsequent functional analysis. Functional confirmation of CTCs is achieved through methods such as secretion of specific proteins, gel invasion assays, telomerase activity testing, and animal transplantation models, serving as definitive standards that can exclude false-positive results due to exceptional circumstances. However, these methodologies often necessitate extended periods of in vitro cell culture. They are also time-consuming, complex to execute, and challenging to scale up, making them less suitable for routine clinical diagnostics and more appropriate for functional basic research [55], [56], [57], [58].
Challenges and strategies for CTC detection technology
Dutzends of distinct commercial CTC detection systems have been developed relying on the enrichment and validation strategies introduced above [55], [56], [57], [58]. Unfortunately, these methods have limitations and have yet to be widely adopted in clinical practice. Future CTC detection technologies must address the following challenges and find appropriate solutions to meet clinical needs better.
Higher enrichment efficiency
The subsequent information will be more comprehensive and trustworthy only by capturing as many complete populations of CTCs as possible. However, a significant proportion of late-stage patients cannot detect CTCs or have insufficient numbers for downstream analysis, indicating room for improvement in the enrichment efficiency of detection methods. Researchers are also attempting to omit the enrichment step, which requires more sensitive digital RT-PCR techniques or high-resolution image scanning technologies, which currently need to be more widely applied [59]. The factors influencing enrichment efficiency can vary; thus, the corresponding solutions may differ.
Firstly, as the most commonly used enrichment method, positive sorting relying solely on the EpCAM leads to suboptimal) enrichment efficiency. A significant reason for this lies in the occurrence of epithelial-to-mesenchymal transition (EMT) during tumor metastasis, where cells lose their epithelial characteristics and gain mesenchymal traits, making them prone to be overlooked due to reduced or absent expression of EpCAM [60]. In response to this problem, some techniques have attempted to use combinations of antibodies or employ CD45-negative sorting. Others have opted for non-affinity-independent methods that do not rely on antigen expression. However, the heterogeneity of CTC populations may exceed our expectations, and each enrichment method has limitations. As a result, a recent trend has been the combination of multiple enrichment approaches to complement each other’s strengths and further enhance efficiency, albeit at the potential cost of prolonged processing steps that might affect CTC viability. Strategies for enriching CTCs in the future will need a more comprehensive understanding of their biological properties [59].
Another possible reason for the inefficiency of enrichment could be rough handling. For instance, in the classic CellSearch system, the enrichment and fluorescent labeling steps occur within a 15 mL centrifuge tube, with repeated washing steps that inevitably lead to unnecessary cell loss. A more advanced microfluidic chip offers a promising solution to this problem. Typically, these chips are about the size of a standard microscope slide and can process 1–2 mL of blood samples. They have a reduced risk of sample loss and significantly lower reagent costs [61], 62].
Detecting low blood volume can also impact detection effectiveness, primarily due to the limited number of CTCs. Additionally, the volume of blood sampled is usually only about 0.1 % of the total blood volume in an average human, leading to sampling errors that affect comparability between batches. Increasing the blood volume for detection is not feasible as it would be difficult for patients to accept and would add significant burdens to subsequent processing, making it hard to enhance detection throughput. In this context, in vivo detection might be a potential solution, such as the CellCollector technology, which involves inserting EpCAM-coated metal wires into a patient’s elbow vein for CTC collection, resulting in a processed blood volume of up to 1,500 mL after 30 min, significantly exceeding the volume used in external detection, with a higher detection rate than traditional Cellsearch systems [63]. Moreover, animal experiments suggest that the number of CTCs obtained through continuous external circulation enrichment is notably higher than that from single-sample results [64], 65]. However, the safety and reliability of this operation in humans still require further validation.
Specialized validation strategies
The significant presence of leukocytes in peripheral blood, which remains high even after CTC enrichment, requires specific methods for CTC identification. Among the existing methods, phenotypic identification is an immediate test but may yield false positives and negatives; functional identification offers better specificity but is time-consuming, making it unsuitable for routine clinical application. Recently, researchers have proposed using a glucose uptake experiment to identify CTCs, which operate on a principle similar to PET-CT, distinguishing between benign and malignant cells based on metabolic differences. This method combines the immediacy of conventional phenotypic identification with the ability to reflect the functional characteristics of the target cells, suggesting promising future applications if supported by more clinical data [66]. This approach also suggests that the development of specific identification strategies needs to fully account for the biological properties of CTCs, which is one of the essential responsibilities of basic CTC research.
Automation and standardization
Automation and standardization primarily encompass sample processing and image scanning analysis. In the realm of sample processing, this involves the development and operation of automated equipment, locking in operational procedures and technical parameters (such as centrifugation speed, incubation time, and volume of samples added), standardizing reagents and consumables (including volume, concentration, antibody clone numbers, material, manufacturer), implementing comprehensive quality control and troubleshooting strategies, among others. These steps are essential to mitigate artificial factors and ensure the comparability of test results. Numerous new technologies are making strides in these areas with promising outcomes. However, the automated image scanning analysis system still needs to improve for many testing methodologies. Relying solely on manual operations and analyses is time-consuming and labor-intensive, often leading to missed detections. A reliable image scanning analysis system necessitates high-performance microscopes to enhance image quality and requires meticulous optimization and locking of Huorescently labeled antibody concentrations along with corresponding exposure times. It demands the development of accompanying software and algorithms for automatic focus adjustment of microscopes, precise location of target cells, intelligent recognition, and interpretation, among other tasks [59]. This area represents a significant challenge in the future development of CTC detection technology, requiring deep interdisciplinary integration across fields such as biology, optics, mechanics, software development, and artificial intelligence to achieve breakthroughs.
Capturing single cells
We are increasingly aware that CTCs constitute a heterogeneous population, with only a fraction of cells exhibiting stem cell phenotypes or metastatic proliferation potential. If the entire population is analyzed in a lump, the information from these cells could be lost. To prevent this, single-cell analysis techniques that can identify each cell’s characteristics are essential [67], 68]. Currently, single-cell sequencing technologies used in tumor tissues, such as those from IOX genomics, assign a unique “barcode” to each cell on a slide, enabling high-throughput detection of thousands of cells [69]. However, due to the relatively low abundance of CTCs, these technologies are unsuitable, partly because they are expensive per cell and partly because of the limited number of cells affecting their differentiation. For single-cell analysis of CTCs, the first step involves capturing and recovering individual cells, which can be achieved through manual aspiration under a microscope, microdissection on a slide, determining coordinates for target cells using “traps” or “microhooks,” or sorting cells into liquid flows or single-cell microparticles based on fluorescence or charge signals. After obtaining the single cells, whole-genome amplification (WGA) is required, commonly performed using multiple displacement amplification or multiple annealing and looping-based amplification cycles [70]. In summary, the technical barriers for CTC single-cell analysis are high, typically requiring high-performance microscopes for cell confirmation, which is time-consuming and labor-intensive and does not quickly increase detection throughput. Currently, it is more commonly used in primary research fields.
Cultivation of CTCs in vitro
The in vitro culture of CTCs is a crucial approach for their functional analysis, which is essential for studying tumor metastasis and resistance mechanisms [48]. The outcomes of such studies can, in turn, drive improvements and enhancements to CTC detection technologies. However, establishing in vitro culture is complex. Even human primary tumors can only be propagated continuously in cell lines in rare instances. For CTCs, this challenge is even more pronounced because the establishment of in vitro culture for CTCs must meet two non-negotiable prerequisites simultaneously. Firstly, there needs to be appropriate technology capable of enriching a sufficient number of viable CTCs, with previous literature reporting successful in vitro cultures of CTCs at counts of 1,109 and 509 per 7.5 mL of peripheral blood [71]. Many existing CTC detection technologies fall short of achieving this. Secondly, a suitable culturing condition must be created that effectively stimulates and supports the growth of CTCs in vitro. To date, however, we lack the necessary understanding of the specific details of these culturing conditions.
For the first criterion, the most contradictory aspect is that the best methods for capturing CTCs either require cell fixation or expose cells to high shear forces, both of which reduce cell viability; conversely, the methods that best preserve cell viability often do not perform well in terms of efficiency and effectiveness when enriching for CTCs. Balancing cell viability with effective capture of CTCs is thus a challenging issue that detection technologies for CTCs must confront. The second criterion typically involves adding growth factors to stimulate epithelial cell growth, using methods such as adherent culture, serum-free suspension culture, and co-culture with feeder cells. While these methods are feasible, overall success rates are generally low and heavily dependent on the number and viability of enriched CTCs [71]. Recent reports indicate that organoid culture can be used to culture CTCs, which can maintain long-term cell survival under conventional culture conditions with the addition of growth factors by adding unique extracellular matrix materials. This method could be a promising direction for future CTC culture approaches [72].
Evidence-based medicine and regulatory approval
The primary reason for the limited clinical application of CTC detection is the need for evidence-based medical data, making it difficult to gain regulatory approval. It includes systematic evaluations, meta-analyses, animal studies, observational studies, diagnostic test accuracy studies, case reports, and expert opinions. The only system that US and Chinese regulatory bodies have approved is the CellSearch system, not just due to its standardized operational procedures and high comparability of results but, more importantly, because it facilitates multicenter clinical research. These studies are meticulously designed with well-structured follow-up data, providing high-level evidence that significantly guides clinical practice. However, many new CTC detection technologies are deficient in clinical research, merely demonstrating the essential performance of related technologies through testing on specific patients, which is far from accurate clinical application.
Furthermore, more universally accepted criteria for real-time companion diagnostics using CTCs need to be established, and future efforts will require large-scale clinical studies to establish and continuously refine these standards [71]. Conducting these multicenter clinical studies is challenging and lengthy, posing a significant burden on any CTC technology development company. However, this challenge is inevitable for the entire industry as it is necessary to accumulate sufficient and reliable evidence-based medical data to achieve clinical acceptance.
Summary
The close link between CTCs and tumor progression makes them a focal point in both clinical and basic research, providing significant guidance for patient clinical treatment and serving as a window into understanding the development of distant metastasis. However, despite a century and a half of relentless efforts, the detection technology for CTCs still needs improvement. On the one hand, this might be due to the complexity of the metastatic process, which we still need to learn more about, coupled with the rarity and heterogeneity of CTCs that pose significant challenges to their detection. Even if some CTCs can be captured, ensuring they will stay the same after leaving the body environment is challenging. On the other hand, the detection process for CTCs requires expertise across various fields, including materials, chemistry, biology, physics, electronics, optics, microfluidics, patient management, artificial intelligence, software development, and mechanical automation. It is unlikely that a single team with a specific knowledge background can handle its development process alone; instead, it necessitates the close collaboration of multiple specialized teams.
Currently, there are numerous commercialized detection methods both domestically and internationally. These methods have strengths and weaknesses, which are constantly being optimized and improved. It is not easy to pinpoint which one is the absolute best. As research on CTCs progresses, we are increasingly aware that each CTC detection technology has advantages and limitations. The application requirements for CTCs are diverse, and different needs may necessitate selecting targeted detection technologies. People often place unrealistic expectations on every specific detection technique, hoping that one method can solve all problems. However, a more practical approach is to ensure the standardization and normalization of the detection process while identifying clinical issues where this method can best leverage its strengths, continuously accumulating sufficient evidence-based medicine. Only in this way can CTC detection indeed contribute its value in clinical practice.
Funding source: National Natural Science Foundation of China
Award Identifier / Grant number: 82241230
Funding source: Municipal Natural Science Foundation of Shanghai
Award Identifier / Grant number: 23ZR1477400
Funding source: Basic Medicine Cultivation Program, Faculty of Medical Imaging, Naval Medical University
Award Identifier / Grant number: 2024PYA01
-
Research ethics: Not applicable.
-
Informed consent: Not applicable.
-
Author contributions: All authors have accepted responsibility for the entire content of this manuscript and approved its submission.
-
Use of Large Language Models, AI and Machine Learning Tools: None declared.
-
Conflict of interest: The authors state no conflict of interest.
-
Research funding: 1. National Natural Science Foundation of China (82241230). 2. Municipal Natural Science Foundation of Shanghai (23ZR1477400). 3. Basic Medicine Cultivation Program, Faculty of Medical Imaging, Naval Medical University (2024PYA01).
-
Data availability: Not applicable.
References
1. WHO. Cancer [online]. Available from: https://www.who.int/news-room/fact-sheets/detail/cancer [Accessed 4 Apr 2022].Search in Google Scholar
2. Shi, X, Wang, X, Yao, W, Shi, D, Shao, X, Lu, Z, et al.. Mechanism insights and therapeutic intervention of tumor metastasis: latest developments and perspectives. Signal Transduct Targeted Ther 2024;9:192. https://doi.org/10.1038/s41392-024-01885-2.Search in Google Scholar PubMed PubMed Central
3. Schuster, E, Taftaf, R, Reduzzi, C, Albert, MK, Romero-Calvo, I, Liu, H. Better together: circulating tumor cell clustering in metastatic cancer. Trends Cancer 2021;7:1020–32. https://doi.org/10.1016/j.trecan.2021.07.001.Search in Google Scholar PubMed PubMed Central
4. 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. https://doi.org/10.1016/j.cell.2014.07.013.Search in Google Scholar PubMed PubMed Central
5. Dianat-Moghadam, H, Azizi, M, Eslami-S, Z, Cortes-Hernandez, LE, Heidarifard, M, Nouri, M, et al.. The role of circulating tumor cells in the metastatic cascade: biology, technical challenges, and clinical relevance. Cancers 2020;12. https://doi.org/10.3390/cancers12040867.Search in Google Scholar PubMed PubMed Central
6. Chemi, F, Mohan, S, Guevara, T, Clipson, A, Rothwell, DG, Dive, C. Early dissemination of circulating tumor cells: biological and clinical insights. Front Oncol 2021;11:672195. https://doi.org/10.3389/fonc.2021.672195.Search in Google Scholar PubMed PubMed Central
7. Fabisiewicz, A, Grzybowska, E. CTC clusters in cancer progression and metastasis. Med Oncol 2017;34:12. https://doi.org/10.1007/s12032-016-0875-0.Search in Google Scholar PubMed
8. Pineiro, R, Martinez-Pena, I, Lopez-Lopez, R. Relevance of CTC clusters in breast cancer metastasis. Adv Exp Med Biol 2020;1220:93–115. https://doi.org/10.1007/978-3-030-35805-1_7.Search in Google Scholar PubMed
9. Wang, X, Wang, L, Lin, H, Zhu, Y, Huang, D, Lai, M, et al.. Research progress of CTC, ctDNA, and EVs in cancer liquid biopsy. Front Oncol 2024;14:1303335. https://doi.org/10.3389/fonc.2024.1303335.Search in Google Scholar PubMed PubMed Central
10. Alix-Panabieres, C, Pantel, K. Liquid biopsy: from discovery to clinical application. Cancer Discov 2021;11:858–73. https://doi.org/10.1158/2159-8290.cd-20-1311.Search in Google Scholar
11. Nikanjam, M, Kato, S, Kurzrock, R. Liquid biopsy: current technology and clinical applications. J Hematol Oncol 2022;15:131. https://doi.org/10.1186/s13045-022-01351-y.Search in Google Scholar PubMed PubMed Central
12. Lawrence, R, Watters, M, Davies, CR, Pantel, K, Lu, YJ. Circulating tumour cells for early detection of clinically relevant cancer. Nat Rev Clin Oncol 2023;20:487–500. https://doi.org/10.1038/s41571-023-00781-y.Search in Google Scholar PubMed PubMed Central
13. Feng, Z, Wu, J, Lu, Y, Chan, YT, Zhang, C, Wang, D, et al.. Circulating tumor cells in the early detection of human cancers. Int J Biol Sci 2022;18:3251–65. https://doi.org/10.7150/ijbs.71768.Search in Google Scholar PubMed PubMed Central
14. Poggiana, C, Rossi, E, Zamarchi, R. Possible role of circulating tumor cells in early detection of lung cancer. J Thorac Dis 2020;12:3821–35. https://doi.org/10.21037/jtd.2020.02.24.Search in Google Scholar PubMed PubMed Central
15. Banys, M, Muller, V, Melcher, C, Aktas, B, Kasimir-Bauer, S, Hagenbeck, C, et al.. Circulating tumor cells in breast cancer. Clin Chim Acta 2013;423:39–45. https://doi.org/10.1016/j.cca.2013.03.029.Search in Google Scholar PubMed
16. Edge, SB, Byrd, DR, Compton, CC, Fritz, AG, Greene, FL, Trotti, A, editors. AJCC cancer staging manual. New York: Springer; 2010.Search in Google Scholar
17. Jiang, ZF, Cristofanilli, M, Shao, ZM, Tong, ZS, Song, EW, Wang, XJ, et al.. Circulating tumor cells predict progression-free and overall survival in Chinese patients with metastatic breast cancer, HER2-positive or triple-negative (CBCSG004): a multicenter, double-blind, prospective trial. Ann Oncol 2013;24:2766–72. https://doi.org/10.1093/annonc/mdt246.Search in Google Scholar PubMed
18. Liu, J, Liu, Y, Gu, C, Zhang, L, Lu, X. Longitudinal change of circulating tumor cells during chemoradiation and its correlation with prognosis in advanced nonsmall-cell lung cancer patients. Cancer Biother Radiopharm 2023;38:305–12. https://doi.org/10.1089/cbr.2020.4096.Search in Google Scholar PubMed
19. Deng, J, Chen, W, Wu, X, Zhou, Y, Li, J. Evaluating the predictive power of circulating tumor cells for the prognosis of transarterial chemoembolization treatment on patients with advanced hepatocellular carcinoma. Medicine 2021;100:e24060. https://doi.org/10.1097/md.0000000000024060.Search in Google Scholar
20. Cai, D, Li, N, Jin, L, Qi, X, Hua, D, Wang, T. High CTC-TRPC5 expression significantly associated with poor prognosis in radical resected colorectal cancer patients. Front Mol Biosci 2021;8:727864. https://doi.org/10.3389/fmolb.2021.727864.Search in Google Scholar PubMed PubMed Central
21. Bidard, FC, Kiavue, N, Jacot, W, Bachelot, T, Dureau, S, Bourgeois, H, et al.. Overall survival with circulating tumor cell count-driven choice of therapy in advanced breast cancer: a randomized trial. J Clin Oncol 2024;42:383–9. https://doi.org/10.1200/jco.23.00456.Search in Google Scholar PubMed
22. Cristofanilli, M, Budd, GT, Ellis, MJ, Stopeck, A, Matera, J, Miller, MC, et al.. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N Engl J Med 2004;351:781–91. https://doi.org/10.1056/nejmoa040766.Search in Google Scholar
23. Bidard, FC, Peeters, DJ, Fehm, T, Nole, F, Gisbert-Criado, R, Mavroudis, D, et al.. Clinical validity of circulating tumour cells in patients with metastatic breast cancer: a pooled analysis of individual patient data. Lancet Oncol 2014;15:406–14. https://doi.org/10.1016/s1470-2045(14)70069-5.Search in Google Scholar PubMed
24. Cohen, SJ, Punt, CJ, Iannotti, N, Saidman, BH, Sabbath, KD, Gabrail, NY, et al.. Relationship of circulating tumor cells to tumor response, progression-free survival, and overall survival in patients with metastatic colorectal cancer. J Clin Oncol 2008;26:3213–21. https://doi.org/10.1200/jco.2007.15.8923.Search in Google Scholar
25. Shen, F, Zhu, Y, Wang, F, Cai, X, Ding, H, Zhou, F, et al.. Clinical significance of circulating tumour cells and tumour marker detection in the chemotherapeutic evaluation of advanced colorectal cancer. Colorectal Dis 2022;24:68–76. https://doi.org/10.1111/codi.15939.Search in Google Scholar PubMed PubMed Central
26. Yu, J, Zhang, J, Peng, T, Fei, Z, Jin, L, Yang, M. Predictive value of circulating tumor cells in prognosis of stage III/IV colorectal cancer after oxaliplatin-based first-line chemotherapy. Vivo 2022;36:806–13. https://doi.org/10.21873/invivo.12767.Search in Google Scholar PubMed PubMed Central
27. de Bono, JS, Scher, HI, Montgomery, RB, Parker, C, Miller, MC, Tissing, H, et al.. Circulating tumor cells predict survival benefit from treatment in metastatic castration-resistant prostate cancer. Clin Cancer Res 2008;14:6302–9. https://doi.org/10.1158/1078-0432.ccr-08-0872.Search in Google Scholar PubMed
28. Wu, YY, Ding, J, Bai, Q, Qi, J. Circulating tumor cells in the diagnosis and treatment of early and advanced prostate cancer. Zhonghua Nan ke Xue 2021;27:934–40.Search in Google Scholar
29. Krebs, MG, Sloane, R, Priest, L, Lancashire, L, Hou, JM, Greystoke, A, et al.. Evaluation and prognostic significance of circulating tumor cells in patients with non-small-cell lung cancer. J Clin Oncol 2011;29:1556–63. https://doi.org/10.1200/jco.2010.28.7045.Search in Google Scholar PubMed
30. Andrikou, K, Rossi, T, Verlicchi, A, Priano, I, Cravero, P, Burgio, MA, et al.. Circulating tumour cells: detection and application in advanced non-small cell lung cancer. Int J Mol Sci 2023;24. https://doi.org/10.3390/ijms242216085.Search in Google Scholar PubMed PubMed Central
31. Sun, Q, Li, W, Yang, D, Lin, PP, Zhang, L, Guo, H. The presence of small-size circulating tumor cells predicts worse prognosis in non-small cell lung cancer patients. Arch Pathol Lab Med 2024. https://doi.org/10.5858/arpa.2023-0455-OA.Search in Google Scholar PubMed
32. Su, Z, Wang, Z, Ni, X, Duan, J, Gao, Y, Zhuo, M, et al.. Inferring the evolution and progression of small-cell lung cancer by single-cell sequencing of circulating tumor cells. Clin Cancer Res 2019;25:5049–60. https://doi.org/10.1158/1078-0432.ccr-18-3571.Search in Google Scholar
33. Hou, JM, Krebs, MG, Lancashire, L, Sloane, R, Backen, A, Swain, RK, et al.. Clinical significance and molecular characteristics of circulating tumor cells and circulating tumor microemboli in patients with small-cell lung cancer. J Clin Oncol 2012;30:525–32. https://doi.org/10.1200/jco.2010.33.3716.Search in Google Scholar
34. Cheng, Y, Liu, XQ, Fan, Y, Liu, YP, Liu, Y, Liu, Y, et al.. Circulating tumor cell counts/change for outcome prediction in patients with extensive-stage small-cell lung cancer. Future Oncol 2016;12:789–99. https://doi.org/10.2217/fon.15.346.Search in Google Scholar PubMed
35. Alemar, J, Schuur, ER. Progress in using circulating tumor cell information to improve metastatic breast cancer therapy. JAMA Oncol 2013;2013:702732. https://doi.org/10.1155/2013/702732.Search in Google Scholar PubMed PubMed Central
36. Lin, D, Shen, L, Luo, M, Zhang, K, Li, J, Yang, Q, et al.. Circulating tumor cells: biology and clinical significance. Signal Transduct Targeted Ther 2021;6:404. https://doi.org/10.1038/s41392-021-00817-8.Search in Google Scholar PubMed PubMed Central
37. Messaritakis, I, Nikolaou, M, Koinis, F, Politaki, E, Koutsopoulos, A, Lagoudaki, E, et al.. Characterization of DLL3-positive circulating tumor cells (CTCs) in patients with small cell lung cancer (SCLC) and evaluation of their clinical relevance during front-line treatment. Lung Cancer 2019;135:33–9. https://doi.org/10.1016/j.lungcan.2019.06.025.Search in Google Scholar PubMed
38. Budd, GT, Cristofanilli, M, Ellis, MJ, Stopeck, A, Borden, E, Miller, MC, et al.. Circulating tumor cells versus imaging--predicting overall survival in metastatic breast cancer. Clin Cancer Res 2006;12:6403–9. https://doi.org/10.1158/1078-0432.ccr-05-1769.Search in Google Scholar PubMed
39. Shao, X, Jin, X, Chen, Z, Zhang, Z, Chen, W, Jiang, J, et al.. A comprehensive comparison of circulating tumor cells and breast imaging modalities as screening tools for breast cancer in Chinese women. Front Oncol 2022;12:890248. https://doi.org/10.3389/fonc.2022.890248.Search in Google Scholar PubMed PubMed Central
40. Yang, YP, Giret, TM, Cote, RJ. Circulating tumor cells from enumeration to analysis: current challenges and future opportunities. Cancers 2021;13. https://doi.org/10.3390/cancers13112723.Search in Google Scholar PubMed PubMed Central
41. Vasseur, A, Kiavue, N, Bidard, FC, Pierga, JY, Cabel, L. Clinical utility of circulating tumor cells: an update. Mol Oncol 2021;15:1647–66. https://doi.org/10.1002/1878-0261.12869.Search in Google Scholar PubMed PubMed Central
42. Liu, Q, Huang, Q, Yu, Z, Wu, H. Clinical characteristics of non-small cell lung cancer patients with EGFR mutations and ALK&ROS1 fusions. Clin Res J 2022;16:216–25. https://doi.org/10.1111/crj.13472.Search in Google Scholar PubMed PubMed Central
43. Swain, SM, Shastry, M, Hamilton, E. Targeting HER2-positive breast cancer: advances and future directions. Nat Rev Drug Discov 2023;22:101–26. https://doi.org/10.1038/s41573-022-00579-0.Search in Google Scholar PubMed PubMed Central
44. Zhu, G, Pei, L, Xia, H, Tang, Q, Bi, F. Role of oncogenic KRAS in the prognosis, diagnosis and treatment of colorectal cancer. Mol Cancer 2021;20:143. https://doi.org/10.1186/s12943-021-01441-4.Search in Google Scholar PubMed PubMed Central
45. Jie, XX, Zhang, XY, Xu, CJ. Epithelial-to-mesenchymal transition, circulating tumor cells and cancer metastasis: mechanisms and clinical applications. Oncotarget 2017;8:81558–71. https://doi.org/10.18632/oncotarget.18277.Search in Google Scholar PubMed PubMed Central
46. Hamilton, G, Rath, B. Mesenchymal-epithelial transition and circulating tumor cells in small cell lung cancer. Adv Exp Med Biol 2017;994:229–45. https://doi.org/10.1007/978-3-319-55947-6_12.Search in Google Scholar PubMed
47. Yang, MH, Imrali, A, Heeschen, C. Circulating cancer stem cells: the importance to select. Chin J Cancer Res 2015;27:437–49. https://doi.org/10.3978/j.issn.1000-9604.2015.04.08.Search in Google Scholar PubMed PubMed Central
48. Smit, DJ, Pantel, K, Jucker, M. Circulating tumor cells as a promising target for individualized drug susceptibility tests in cancer therapy. Biochem Pharmacol 2021;188:114589. https://doi.org/10.1016/j.bcp.2021.114589.Search in Google Scholar PubMed
49. Kang, Y, Pantel, K. Tumor cell dissemination: emerging biological insights from animal models and cancer patients. Cancer Cell 2013;23:573–81. https://doi.org/10.1016/j.ccr.2013.04.017.Search in Google Scholar PubMed PubMed Central
50. Umer, M, Vaidyanathan, R, Nguyen, NT, Shiddiky, M. Circulating tumor microemboli: progress in molecular understanding and enrichment technologies. Biotechnol Adv 2018;36:1367–89. https://doi.org/10.1016/j.biotechadv.2018.05.002.Search in Google Scholar PubMed
51. Heidrich, I, Ackar, L, Mossahebi, MP, Pantel, K. Liquid biopsies: potential and challenges. Int J Cancer 2021;148:528–45. https://doi.org/10.1002/ijc.33217.Search in Google Scholar PubMed
52. Martins, I, Ribeiro, IP, Jorge, J, Goncalves, AC, Sarmento-Ribeiro, AB, Melo, JB, et al.. Liquid biopsies: applications for cancer diagnosis and monitoring. Genes 2021;12. https://doi.org/10.3390/genes12030349.Search in Google Scholar PubMed PubMed Central
53. Calabuig-Farinas, S, Jantus-Lewintre, E, Herreros-Pomares, A, Camps, C. Circulating tumor cells versus circulating tumor DNA in lung cancer-which one will win? Transl Lung Cancer Res 2016;5:466–82. https://doi.org/10.21037/tlcr.2016.10.02.Search in Google Scholar PubMed PubMed Central
54. Wang, YH, Song, Z, Hu, XY, Wang, HS. Circulating tumor DNA analysis for tumor diagnosis. Talanta 2021;228:122220. https://doi.org/10.1016/j.talanta.2021.122220.Search in Google Scholar PubMed
55. Labib, M, Kelley, SO. Circulating tumor cell profiling for precision oncology. Mol Oncol 2021;15:1622–46. https://doi.org/10.1002/1878-0261.12901.Search in Google Scholar PubMed PubMed Central
56. Pantel, K, Alix-Panabieres, C. Liquid biopsy and minimal residual disease - latest advances and implications for cure. Nat Rev Clin Oncol 2019;16:409–24. https://doi.org/10.1038/s41571-019-0187-3.Search in Google Scholar PubMed
57. Habli, Z, AlChamaa, W, Saab, R, Kadara, H, Khraiche, ML. Circulating tumor cell detection technologies and clinical utility: challenges and opportunities. Cancers 2020;12. https://doi.org/10.3390/cancers12071930.Search in Google Scholar PubMed PubMed Central
58. Hu, X, Zang, X, Lv, Y. Detection of circulating tumor cells: advances and critical concerns. Oncol Lett 2021;21:422. https://doi.org/10.3892/ol.2021.12683.Search in Google Scholar PubMed PubMed Central
59. Schaffner, F, Merlin, JL, von Bubnoff, N, editors. Tumor Liquid Biopsies. Cham: Springer Cham; 2020.10.1007/978-3-030-26439-0Search in Google Scholar
60. Lowes, LE, Allan, AL. Circulating tumor cells and implications of the epithelial-to-mesenchymal transition. Adv Clin Chem 2018;83:121–81. https://doi.org/10.1016/bs.acc.2017.10.004.Search in Google Scholar PubMed
61. Cho, HY, Choi, JH, Lim, J, Lee, SN, Choi, JW. Microfluidic chip-based cancer diagnosis and prediction of relapse by detecting circulating tumor cells and circulating cancer stem cells. Cancers 2021;13. https://doi.org/10.3390/cancers13061385.Search in Google Scholar PubMed PubMed Central
62. Farshchi, F, Hasanzadeh, M. Microfluidic biosensing of circulating tumor cells (CTCs): recent progress and challenges in efficient diagnosis of cancer. Biomed Pharmacother 2021;134:111153. https://doi.org/10.1016/j.biopha.2020.111153.Search in Google Scholar PubMed
63. Theil, G, Boehm, C, Fischer, K, Bialek, J, Hoda, R, Weber, E, et al.. In vivo isolation of circulating tumor cells in patients with different stages of prostate cancer. Oncol Lett 2021;21:357. https://doi.org/10.3892/ol.2021.12618.Search in Google Scholar PubMed PubMed Central
64. Kim, TH, Wang, Y, Oliver, CR, Thamm, DH, Cooling, L, Paoletti, C, et al.. A temporary indwelling intravascular aphaeretic system for in vivo enrichment of circulating tumor cells. Nat Commun 2019;10:1478. https://doi.org/10.1038/s41467-019-09439-9.Search in Google Scholar PubMed PubMed Central
65. Tang, M, Xia, HF, Xu, CM, Feng, J, Ren, JG, Miao, F, et al.. Magnetic chip based extracorporeal circulation: a new tool for circulating tumor cell in vivo detection. Anal Chem 2019;91:15260–6. https://doi.org/10.1021/acs.analchem.9b04286.Search in Google Scholar PubMed
66. Li, Z, Wang, Z, Tang, Y, Lu, X, Chen, J, Dong, Y, et al.. Liquid biopsy-based single-cell metabolic phenotyping of lung cancer patients for informative diagnostics. Nat Commun 2019;10:3856. https://doi.org/10.1038/s41467-019-11808-3.Search in Google Scholar PubMed PubMed Central
67. Wu, CP, Wu, P, Zhao, HF, Liu, WL, Li, WP. Clinical applications of and challenges in single-cell analysis of circulating tumor cells. DNA Cell Biol 2018;37:78–89. https://doi.org/10.1089/dna.2017.3981.Search in Google Scholar PubMed
68. Dai, Z, Gu, XY, Xiang, SY, Gong, DD, Man, CF, Fan, Y. Research and application of single-cell sequencing in tumor heterogeneity and drug resistance of circulating tumor cells. Biomark Res 2020;8:60. https://doi.org/10.1186/s40364-020-00240-1.Search in Google Scholar PubMed PubMed Central
69. Ding, S, Chen, X, Shen, K. Single-cell RNA sequencing in breast cancer: understanding tumor heterogeneity and paving roads to individualized therapy. Cancer Commun 2020;40:329–44. https://doi.org/10.1002/cac2.12078.Search in Google Scholar PubMed PubMed Central
70. Gao, C, Zhang, M, Chen, L. The comparison of two single-cell sequencing platforms: BD rhapsody and 10x genomics chromium. Curr Genom 2020;21:602–9. https://doi.org/10.2174/1389202921999200625220812.Search in Google Scholar PubMed PubMed Central
71. Cote, RJ, Datar, RH, editors. Circulating Tumour Cells: Advances in Basic Science and Clinical Applications. Beijing: Beijing Science; 2018.Search in Google Scholar
72. Praharaj, PP, Bhutia, SK, Nagrath, S, Bitting, RL, Deep, G. Circulating tumor cell-derived organoids: current challenges and promises in medical research and precision medicine. Biochim Biophys Acta Rev Cancer 2018;1869:117–27. https://doi.org/10.1016/j.bbcan.2017.12.005.Search in Google Scholar PubMed PubMed Central
© 2024 Walter de Gruyter GmbH, Berlin/Boston
Articles in the same Issue
- Frontmatter
- Editorials
- The Friedewald formula strikes back
- Liquid biopsy in oncology: navigating technical hurdles and future transition for precision medicine
- The neglected issue of pyridoxal- 5′ phosphate
- Reviews
- Health literacy: a new challenge for laboratory medicine
- Clinical applications of circulating tumor cell detection: challenges and strategies
- Opinion Papers
- Pleural effusion as a sample matrix for laboratory analyses in cancer management: a perspective
- Interest of hair tests to discriminate a tail end of a doping regimen from a possible unpredictable source of a prohibited substance in case of challenging an anti-doping rule violation
- Perspectives
- Sigma Metrics misconceptions and limitations
- EN ISO 15189 revision: EFLM Committee Accreditation and ISO/CEN standards (C: A/ISO) analysis and general remarks on the changes
- General Clinical Chemistry and Laboratory Medicine
- Evaluation of current indirect methods for measuring LDL-cholesterol
- Verification of automated review, release and reporting of results with assessment of the risk of harm for patients: the procedure algorithm proposal for clinical laboratories
- Progranulin measurement with a new automated method: a step forward in the diagnostic approach to neurodegenerative disorders
- A comparative analysis of current С-peptide assays compared to a reference method: can we overcome inertia to standardization?
- Blood samples for ammonia analysis do not require transport to the laboratory on ice: a study of ammonia stability and cause of in vitro ammonia increase in samples from patients with hyperammonaemia
- A physio-chemical mathematical model of the effects of blood analysis delay on acid-base, metabolite and electrolyte status: evaluation in blood from critical care patients
- Evolution of autoimmune diagnostics over the past 10 years: lessons learned from the UK NEQAS external quality assessment EQA programs
- Comparison between monotest and traditional batch-based ELISA assays for therapeutic drug monitoring of infliximab and adalimumab levels and anti-drug antibodies
- Evaluation of pre-analytical factors impacting urine test strip and chemistry results
- Evaluation of AUTION EYE AI-4510 flow cell morphology analyzer for counting particles in urine
- Reference Values and Biological Variations
- Estimation of the allowable total error of the absolute CD34+ cell count by flow cytometry using data from UK NEQAS exercises 2004–2024
- Establishment of gender– and age–related reference intervals for serum uric acid in adults based on big data from Zhejiang Province in China
- Cancer Diagnostics
- Tumor specific protein 70 targeted tumor cell isolation technology can improve the accuracy of cytopathological examination
- Cardiovascular Diseases
- Diagnostic performance of Mindray CL1200i high sensitivity cardiac troponin I assay compared to Abbott Alinity cardiac troponin I assay for the diagnosis of type 1 and 2 acute myocardial infarction in females and males: MERITnI study
- Infectious Diseases
- Evidence-based assessment of the application of Six Sigma to infectious disease serology quality control
- Letters to the Editor
- Evaluating the accuracy of ChatGPT in classifying normal and abnormal blood cell morphology
- Refining within-subject biological variation estimation using routine laboratory data: practical applications of the refineR algorithm
- Early rule-out high-sensitivity troponin protocols require continuous analytical robustness: a caution regarding the potential for troponin assay down-calibration
- Biochemical evidence of vitamin B12 deficiency: a crucial issue to address supplementation in pregnant women
- Plasmacytoid dendritic cell proliferation and acute myeloid leukemia with minimal differentiation (AML-M0)
- Failing methemoglobin blood gas analyses in a sodium nitrite intoxication
Articles in the same Issue
- Frontmatter
- Editorials
- The Friedewald formula strikes back
- Liquid biopsy in oncology: navigating technical hurdles and future transition for precision medicine
- The neglected issue of pyridoxal- 5′ phosphate
- Reviews
- Health literacy: a new challenge for laboratory medicine
- Clinical applications of circulating tumor cell detection: challenges and strategies
- Opinion Papers
- Pleural effusion as a sample matrix for laboratory analyses in cancer management: a perspective
- Interest of hair tests to discriminate a tail end of a doping regimen from a possible unpredictable source of a prohibited substance in case of challenging an anti-doping rule violation
- Perspectives
- Sigma Metrics misconceptions and limitations
- EN ISO 15189 revision: EFLM Committee Accreditation and ISO/CEN standards (C: A/ISO) analysis and general remarks on the changes
- General Clinical Chemistry and Laboratory Medicine
- Evaluation of current indirect methods for measuring LDL-cholesterol
- Verification of automated review, release and reporting of results with assessment of the risk of harm for patients: the procedure algorithm proposal for clinical laboratories
- Progranulin measurement with a new automated method: a step forward in the diagnostic approach to neurodegenerative disorders
- A comparative analysis of current С-peptide assays compared to a reference method: can we overcome inertia to standardization?
- Blood samples for ammonia analysis do not require transport to the laboratory on ice: a study of ammonia stability and cause of in vitro ammonia increase in samples from patients with hyperammonaemia
- A physio-chemical mathematical model of the effects of blood analysis delay on acid-base, metabolite and electrolyte status: evaluation in blood from critical care patients
- Evolution of autoimmune diagnostics over the past 10 years: lessons learned from the UK NEQAS external quality assessment EQA programs
- Comparison between monotest and traditional batch-based ELISA assays for therapeutic drug monitoring of infliximab and adalimumab levels and anti-drug antibodies
- Evaluation of pre-analytical factors impacting urine test strip and chemistry results
- Evaluation of AUTION EYE AI-4510 flow cell morphology analyzer for counting particles in urine
- Reference Values and Biological Variations
- Estimation of the allowable total error of the absolute CD34+ cell count by flow cytometry using data from UK NEQAS exercises 2004–2024
- Establishment of gender– and age–related reference intervals for serum uric acid in adults based on big data from Zhejiang Province in China
- Cancer Diagnostics
- Tumor specific protein 70 targeted tumor cell isolation technology can improve the accuracy of cytopathological examination
- Cardiovascular Diseases
- Diagnostic performance of Mindray CL1200i high sensitivity cardiac troponin I assay compared to Abbott Alinity cardiac troponin I assay for the diagnosis of type 1 and 2 acute myocardial infarction in females and males: MERITnI study
- Infectious Diseases
- Evidence-based assessment of the application of Six Sigma to infectious disease serology quality control
- Letters to the Editor
- Evaluating the accuracy of ChatGPT in classifying normal and abnormal blood cell morphology
- Refining within-subject biological variation estimation using routine laboratory data: practical applications of the refineR algorithm
- Early rule-out high-sensitivity troponin protocols require continuous analytical robustness: a caution regarding the potential for troponin assay down-calibration
- Biochemical evidence of vitamin B12 deficiency: a crucial issue to address supplementation in pregnant women
- Plasmacytoid dendritic cell proliferation and acute myeloid leukemia with minimal differentiation (AML-M0)
- Failing methemoglobin blood gas analyses in a sodium nitrite intoxication