Appearance
This report is written by MaltSci based on the latest literature and research findings
How does liquid biopsy enhance cancer diagnosis?
Abstract
The diagnosis and management of cancer have traditionally relied on invasive tissue biopsies, which pose risks and complications. Liquid biopsy, a non-invasive method that analyzes tumor-derived materials in bodily fluids, presents a revolutionary alternative. This review explores how liquid biopsy improves cancer diagnosis by detecting circulating tumor cells (CTCs), cell-free DNA (cfDNA), and exosomes. These biomarkers offer critical insights into tumor biology and dynamics, enabling early cancer detection, real-time monitoring of treatment responses, and identification of minimal residual disease, thus supporting personalized treatment strategies. Current advancements in technologies such as next-generation sequencing and microfluidics have enhanced the sensitivity and specificity of liquid biopsy, though challenges remain, including the need for standardized protocols and cost considerations. This review provides a comprehensive overview of the mechanisms of liquid biopsy, its clinical applications, advantages over traditional biopsies, and the challenges it faces. The potential of liquid biopsy to transform cancer care through technological advancements and integration into clinical practice is significant, making it a pivotal tool in the future of oncology.
Outline
This report will discuss the following questions.
- 1 Introduction
- 2 Mechanisms of Liquid Biopsy
- 2.1 Circulating Tumor Cells (CTCs)
- 2.2 Cell-Free DNA (cfDNA)
- 2.3 Exosomes and Other Biomarkers
- 3 Clinical Applications of Liquid Biopsy
- 3.1 Early Detection of Cancer
- 3.2 Monitoring Treatment Response
- 3.3 Detecting Minimal Residual Disease
- 4 Advantages Over Traditional Biopsies
- 4.1 Non-Invasiveness
- 4.2 Real-Time Monitoring
- 4.3 Comprehensive Tumor Profiling
- 5 Challenges and Limitations
- 5.1 Technical Challenges
- 5.2 Standardization and Validation
- 5.3 Ethical Considerations
- 6 Future Directions
- 6.1 Advances in Technology
- 6.2 Integration into Clinical Practice
- 6.3 Potential for Personalized Medicine
- 7 Summary
1 Introduction
The diagnosis and management of cancer have long relied on traditional tissue biopsies, which, despite their efficacy, are often invasive and carry significant risks of complications. Recent advancements in diagnostic methodologies have led to the emergence of liquid biopsy, a non-invasive approach that analyzes tumor-derived materials found in bodily fluids, such as blood, urine, and saliva. Liquid biopsy offers a revolutionary alternative, allowing for the detection of circulating tumor cells (CTCs), cell-free DNA (cfDNA), and exosomes, thus providing critical insights into tumor biology and dynamics without the need for invasive procedures [1][2]. This innovative technique not only facilitates early detection of cancer but also enables real-time monitoring of treatment responses and the identification of minimal residual disease, ultimately supporting personalized treatment strategies [3][4].
The significance of liquid biopsy in oncology cannot be overstated. With the potential to improve diagnostic accuracy and enhance patient outcomes, liquid biopsy represents a paradigm shift in how cancer is diagnosed and managed [5][6]. The ability to obtain real-time molecular information about tumors aids in tailoring therapies based on individual patient needs, thus aligning with the principles of precision medicine [2]. Moreover, liquid biopsy techniques can identify genetic mutations and tumor heterogeneity, providing a more comprehensive understanding of the disease and its progression [1][7].
Current research indicates that liquid biopsy is gaining traction as a crucial tool in early cancer detection and monitoring. Numerous studies have highlighted its application in various cancer types, including non-small cell lung carcinoma (NSCLC), where early detection significantly improves patient outcomes [3]. The integration of advanced technologies, such as next-generation sequencing and microfluidics, has further enhanced the sensitivity and specificity of liquid biopsy [7][8]. However, challenges remain, including the need for standardized protocols and the high costs associated with advanced diagnostic technologies [4][9].
This review will explore the mechanisms underlying liquid biopsy technology, detailing the roles of CTCs, cfDNA, and exosomes as key biomarkers in cancer diagnostics. We will examine the clinical applications of liquid biopsy, focusing on its use in early cancer detection, monitoring treatment responses, and detecting minimal residual disease. Additionally, we will discuss the advantages of liquid biopsy over traditional biopsies, including its non-invasive nature, ability for real-time monitoring, and comprehensive tumor profiling capabilities. We will also address the challenges and limitations faced by liquid biopsy, including technical hurdles, standardization issues, and ethical considerations. Finally, we will consider future directions for research and implementation in oncology, highlighting the potential for liquid biopsy to revolutionize cancer care through advances in technology and integration into clinical practice.
In summary, this review aims to provide a comprehensive overview of how liquid biopsy is transforming cancer diagnosis and management, synthesizing current literature and emerging findings to illuminate its critical role in the future of oncology. As we continue to advance our understanding of cancer biology and improve diagnostic methodologies, liquid biopsy stands at the forefront of efforts to enhance patient care and outcomes in the realm of cancer treatment [10][11].
2 Mechanisms of Liquid Biopsy
2.1 Circulating Tumor Cells (CTCs)
Liquid biopsy represents a transformative approach in cancer diagnosis, primarily through the analysis of circulating tumor cells (CTCs). CTCs are malignant cells that detach from primary tumors or metastases and enter the bloodstream, providing crucial insights into the tumor's biology and dynamics. The mechanisms by which liquid biopsy, particularly through CTC analysis, enhances cancer diagnosis can be outlined as follows:
Minimally Invasive Sampling: Liquid biopsy allows for the collection of blood samples, which is significantly less invasive compared to traditional tissue biopsies. This ease of access facilitates repeated sampling over time, enabling continuous monitoring of tumor evolution and patient response to treatment [12][13].
Real-Time Monitoring: The analysis of CTCs enables real-time assessment of cancer progression, treatment response, and the detection of minimal residual disease (MRD). This is particularly beneficial for adjusting therapeutic strategies promptly based on the tumor's evolving characteristics [14][15].
Characterization of Tumor Heterogeneity: CTCs provide a snapshot of the tumor's molecular characteristics, reflecting intratumoral heterogeneity. Advanced technologies now allow for the digital genomic and next-generation sequencing of CTCs, which can differentiate molecular subtypes of cancer and track genetic variations over time. This capability is essential for personalized medicine, as it helps identify actionable mutations and therapeutic targets [13][16].
Predictive Biomarkers: The enumeration and molecular analysis of CTCs have been shown to correlate with prognosis, therapy response, and disease progression. High CTC counts are often associated with lower survival rates, while reductions in CTC levels during treatment may indicate positive therapeutic responses [17][18].
Functional Studies and Drug Testing: CTC-derived cell lines and explants provide valuable platforms for functional studies that elucidate the metastatic process and therapeutic responses. These models can also facilitate personalized drug testing, allowing clinicians to tailor treatments based on the specific biology of a patient's cancer [12][13].
Comprehensive Biomarker Analysis: Beyond CTCs, liquid biopsy can also analyze circulating tumor DNA (ctDNA), circulating microRNAs (cfmiRNAs), and extracellular vesicles (EVs). This multifaceted approach enhances the diagnostic accuracy and provides a broader understanding of the tumor's biology [13][14].
In summary, liquid biopsy, particularly through the analysis of CTCs, improves cancer diagnosis by offering a non-invasive, real-time, and comprehensive assessment of tumor biology. It allows for continuous monitoring, enhances the understanding of tumor heterogeneity, and provides predictive biomarkers that facilitate personalized treatment strategies. This innovative approach holds significant promise for advancing cancer care and improving patient outcomes.
2.2 Cell-Free DNA (cfDNA)
Liquid biopsy represents a significant advancement in cancer diagnostics, primarily through the analysis of cell-free DNA (cfDNA) found in blood or other body fluids. This minimally invasive technique allows for the collection of tumor-derived genetic material without the need for traditional tissue biopsies, which can be invasive and may not accurately represent tumor heterogeneity. The mechanisms through which liquid biopsy improves cancer diagnosis include enhanced sensitivity, comprehensive assessment of tumor characteristics, and the ability to monitor disease progression dynamically.
One of the primary advantages of cfDNA analysis in liquid biopsies is its ability to provide a comprehensive view of tumor heterogeneity. As cfDNA is released from all tumor sites into the bloodstream, it offers a more reliable representation of the tumor's genetic landscape compared to a single tissue biopsy, which may only capture a localized area of the tumor. This comprehensive approach is particularly beneficial for assessing tumor evolution and therapy response, as it reflects the dynamic changes in the tumor's genetic profile over time [19].
The sensitivity of cfDNA detection has been significantly enhanced by advances in next-generation sequencing (NGS) technologies. These methods allow for the detection of low-frequency mutations and other genetic alterations that might be missed in traditional tissue biopsies. The ability to identify such alterations can lead to more accurate diagnoses and tailored treatment strategies. For instance, cfDNA analysis can facilitate the early detection of minimal residual disease, improving the chances of successful treatment outcomes [20].
Furthermore, cfDNA analysis extends beyond the identification of mutations. Emerging research has highlighted the importance of genomic and epigenomic features, such as methylation patterns and fragmentomics, which can provide deeper insights into tumor biology. This multi-modal assessment enables clinicians to monitor disease progression and treatment response more effectively, thus improving overall patient management [21].
In specific contexts, such as hepatocellular carcinoma (HCC), liquid biopsy has shown promise in enhancing early detection and monitoring treatment responses. Traditional diagnostic methods, like abdominal ultrasonography combined with alpha-fetoprotein (AFP) analysis, have limitations in sensitivity. In contrast, cfDNA analysis offers real-time insights into tumor burden and genetic profiles, which can significantly improve diagnostic accuracy and treatment monitoring [22].
In summary, liquid biopsy, particularly through the analysis of cfDNA, enhances cancer diagnosis by providing a non-invasive, sensitive, and comprehensive method for understanding tumor biology. It allows for dynamic monitoring of disease progression and treatment response, ultimately leading to improved patient outcomes and more personalized therapeutic strategies [23][24][25].
2.3 Exosomes and Other Biomarkers
Liquid biopsy represents a transformative approach in cancer diagnosis, offering significant advantages over traditional tissue biopsies. This minimally invasive technique allows for the analysis of various biomarkers present in biofluids, such as blood, urine, and saliva, providing insights into the tumor's molecular characteristics and its microenvironment.
Exosomes, which are nanosized extracellular vesicles secreted by all cell types, including tumor cells, play a crucial role in liquid biopsy. They carry a diverse array of biomolecules, including proteins, nucleic acids (DNA and RNA), and lipids, that reflect the physiological and pathological states of their parent cells. The presence of these biomolecules in exosomes facilitates intercellular communication and can significantly influence tumor progression, metastasis, and therapeutic responses [26][27][28].
One of the key mechanisms through which liquid biopsy enhances cancer diagnosis is by enabling the detection of tumor-derived exosomes and their specific cargo. For instance, exosomal microRNAs (miRNAs) have emerged as promising biomarkers due to their stability in biofluids and their ability to provide real-time information about tumor dynamics [29][30]. The detection of these exosomal miRNAs can aid in the early diagnosis of various cancers, including breast cancer, by revealing alterations in the tumor's genetic landscape long before clinical symptoms manifest [28][30].
Furthermore, liquid biopsy technologies, particularly those utilizing exosomes, allow for the simultaneous detection of multiple biomarkers, thereby improving diagnostic accuracy. For example, a novel microfluidic approach has been developed that utilizes optical transparent antibody-conjugated microbeads to capture and enrich exosomes, allowing for the detection of multiple cancer biomarkers from a single plasma sample. This method demonstrated a high diagnostic potential, with area under curve (AUC) values indicating excellent discrimination between cancer patients and healthy individuals [28].
In addition to their role in diagnosis, exosomes are also instrumental in monitoring treatment responses and disease progression. The analysis of exosomal contents can provide valuable information on how tumors respond to therapies, enabling personalized treatment strategies. For instance, changes in the protein and RNA profiles of exosomes can indicate therapeutic efficacy or resistance, guiding clinicians in optimizing treatment regimens [26][31].
Moreover, liquid biopsy minimizes the need for invasive procedures, reducing patient discomfort and risk while allowing for frequent monitoring of cancer progression and treatment responses. This capability is particularly advantageous in cases where tumors are difficult to access or when rapid changes in tumor dynamics need to be assessed [27][28].
In summary, liquid biopsy enhances cancer diagnosis through the utilization of exosomes and other biomarkers by providing a non-invasive, real-time, and comprehensive analysis of tumor characteristics. This approach not only facilitates early detection but also supports ongoing monitoring and personalized treatment strategies, representing a significant advancement in oncology.
3 Clinical Applications of Liquid Biopsy
3.1 Early Detection of Cancer
Liquid biopsy has emerged as a transformative approach in oncology, significantly enhancing cancer diagnosis, particularly in the realm of early detection. This innovative method analyzes circulating tumor components, such as circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), extracellular vesicles (EVs), and tumor-educated platelets (TEPs), found in bodily fluids. The advantages of liquid biopsy over traditional tissue biopsy are manifold, particularly in its non-invasive nature, allowing for repeated sampling without the associated risks of invasive procedures.
One of the primary benefits of liquid biopsy is its ability to provide real-time insights into tumor dynamics, which is crucial for early detection and monitoring of cancer progression. This is particularly important for cancers like non-small cell lung carcinoma (NSCLC), where early detection is vital for improving patient outcomes. Studies indicate that liquid biopsy can detect molecular alterations indicative of malignancy before the clinical manifestation of the disease, thereby facilitating timely intervention (Chang et al., 2022; Coppola et al., 2025) [1][3].
Furthermore, liquid biopsy technologies have advanced significantly, incorporating methods such as next-generation sequencing, polymerase chain reaction, and microfluidic platforms. These technologies enhance the sensitivity and specificity of cancer detection, allowing for the identification of minimal residual disease (MRD) and tracking of tumor evolution over time (Fu et al., 2024; Garzarelli et al., 2025) [4][32]. The ability to detect tumor markers from easily accessible biological fluids like blood and saliva not only improves patient compliance but also expands the scope of screening to a broader population, which is essential for preventive oncology (Bao et al., 2024) [2].
In colorectal cancer (CRC), for instance, liquid biopsies have demonstrated prognostic significance by revealing genetic mutations and epigenetic alterations that correlate with treatment responses and disease recurrence (Liatsou et al., 2025) [33]. The identification of ctDNA in post-surgical patients is particularly noteworthy, as it has been associated with a higher risk of recurrence, underscoring the potential of liquid biopsies in guiding personalized therapeutic strategies.
Despite these advancements, challenges remain, including the need for standardized methodologies and improved assay sensitivity to detect early-stage cancers effectively. Nonetheless, ongoing research and technological innovations continue to enhance the capabilities of liquid biopsy, paving the way for its integration into routine clinical practice (Connal et al., 2023) [34].
In summary, liquid biopsy significantly improves cancer diagnosis through its non-invasive nature, ability to provide real-time insights into tumor biology, and the integration of advanced detection technologies. Its application in early detection not only promises better patient outcomes but also heralds a new era in personalized medicine, where treatments can be tailored based on the molecular characteristics of individual tumors.
3.2 Monitoring Treatment Response
Liquid biopsy represents a transformative advancement in oncology, particularly in the context of monitoring treatment responses in cancer patients. This innovative approach enables the analysis of circulating tumor components, such as circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), and extracellular vesicles (EVs), which are present in bodily fluids like blood and urine. The noninvasive nature of liquid biopsy allows for repeated sampling, thereby facilitating real-time monitoring of tumor dynamics and treatment efficacy without the need for invasive tissue biopsies.
One of the significant clinical applications of liquid biopsy is its role in assessing treatment responses. For instance, liquid biopsy can provide early indications of how a patient is responding to therapy, potentially even before conventional imaging techniques can detect changes. This capability is crucial, as it allows clinicians to make timely decisions regarding treatment adjustments, thereby optimizing therapeutic strategies. Studies have demonstrated that liquid biopsy can detect minimal residual disease (MRD) and monitor for emerging resistance mechanisms during treatment, which is vital for ensuring that patients receive the most effective interventions [1][35].
Moreover, the integration of advanced technologies, such as next-generation sequencing (NGS) and digital polymerase chain reaction (dPCR), has significantly enhanced the sensitivity and specificity of liquid biopsy tests. These technologies enable the detection of somatic variants and actionable genomic alterations in tumors, allowing for a more precise understanding of the tumor's molecular profile. Such insights can guide clinicians in selecting targeted therapies and predicting treatment outcomes [4][36].
Despite the promising advantages of liquid biopsy in monitoring treatment responses, several challenges remain. These include ensuring high sensitivity and specificity in detecting analytes, particularly in early-stage cancers, and the complexities associated with tumor heterogeneity. The interpretation of liquid biopsy results must also consider the dynamic nature of tumors and the potential presence of subclonal populations, which can complicate the assessment of treatment efficacy [1][6].
In conclusion, liquid biopsy enhances cancer diagnosis and treatment monitoring by providing a noninvasive, dynamic, and comprehensive method for evaluating tumor responses to therapy. The ongoing advancements in liquid biopsy technologies and methodologies hold the promise of significantly improving patient outcomes in cancer care through personalized treatment strategies and timely interventions [2][7].
3.3 Detecting Minimal Residual Disease
Liquid biopsy represents a significant advancement in the field of oncology, particularly in the context of detecting minimal residual disease (MRD). This non-invasive method analyzes various biomarkers, such as circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), and extracellular vesicles (EVs), present in bodily fluids, primarily blood. The application of liquid biopsy in detecting MRD is critical for improving cancer diagnosis, monitoring treatment responses, and guiding personalized therapies.
One of the primary advantages of liquid biopsy is its ability to provide real-time insights into tumor dynamics. For instance, in colorectal cancer (CRC), the detection of ctDNA has been shown to correlate with the presence of MRD following surgical intervention. Studies indicate that the identification of ctDNA in patients post-surgery is associated with a heightened risk of recurrence and a poor prognosis, underscoring its potential utility in assessing treatment effectiveness and tailoring subsequent therapeutic strategies (Liatsou et al., 2025) [33]. Furthermore, the prognostic significance of CTCs in early-stage CRC has been established, indicating their role in predicting disease recurrence and survival outcomes (Liatsou et al., 2025) [33].
In the context of soft tissue sarcomas, liquid biopsy is being explored as a minimally invasive technique for monitoring MRD. The serial assessment of ctDNA and CTCs may help identify patients at the highest risk of relapse following curative-intent interventions (Kournoutas & Siontis, 2025) [37]. This is particularly relevant given the heterogeneous nature of sarcomas and their propensity to develop treatment resistance, making early detection of residual disease crucial for improving patient outcomes.
Moreover, the integration of liquid biopsy with advanced technologies, such as artificial intelligence (AI) and machine learning (ML), enhances its sensitivity and specificity. These technologies can assist in interpreting complex data amidst tumor heterogeneity, thereby improving the accuracy of MRD detection (Fu et al., 2024) [4]. The potential to combine liquid biopsy with other diagnostic modalities further strengthens its role in precision oncology, allowing for a more comprehensive understanding of tumor biology and treatment response.
Despite the promising applications of liquid biopsy in detecting MRD, several challenges remain. Issues such as assay standardization, cost-effectiveness, and the need for extensive research to validate clinical utility must be addressed to facilitate its widespread adoption in clinical practice (Fu et al., 2024) [4]. Nonetheless, the ongoing innovations in liquid biopsy technology and methodology hold the potential to significantly enhance early cancer detection and the personalization of treatment strategies, ultimately leading to improved patient outcomes across various malignancies.
4 Advantages Over Traditional Biopsies
4.1 Non-Invasiveness
Liquid biopsy represents a significant advancement in cancer diagnostics, particularly due to its non-invasive nature compared to traditional tissue biopsies. Traditional biopsies involve the extraction of tissue from the primary tumor, which can be invasive, painful, and carry risks of complications. In contrast, liquid biopsy allows for the analysis of biomarkers from bodily fluids such as blood, saliva, or urine, making it a more patient-friendly option.
One of the primary advantages of liquid biopsy is its ability to provide real-time information about the tumor's dynamics and genetic characteristics without the need for invasive procedures. This technique analyzes circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), and other components such as extracellular vesicles and tumor-educated platelets. These biomarkers reflect the tumor's heterogeneity and molecular profile, offering insights that may not be available from a single tissue sample [1][3].
The non-invasive nature of liquid biopsy facilitates repeated sampling over time, which is particularly valuable for monitoring treatment responses and disease progression. This capability allows clinicians to make timely adjustments to treatment plans based on the evolving characteristics of the tumor, thereby enhancing personalized medicine approaches [1][38].
Moreover, liquid biopsy has been shown to improve the sensitivity and specificity of cancer detection. The use of advanced technologies, such as next-generation sequencing, enhances the detection of genetic alterations in cfDNA, making it a reliable method for early diagnosis and prognosis [2][19]. In cases of non-small cell lung carcinoma (NSCLC), for instance, liquid biopsy has been recognized for its role in understanding tumor staging, heterogeneity, and therapeutic resistance [38].
Overall, the non-invasive nature of liquid biopsy not only reduces the physical burden on patients but also opens avenues for earlier detection and continuous monitoring of cancer, which are critical for improving patient outcomes and survival rates [39][40]. As research progresses, liquid biopsy is expected to further revolutionize cancer diagnostics and management by providing comprehensive, real-time insights into tumor biology.
4.2 Real-Time Monitoring
Liquid biopsy represents a transformative advancement in oncology, providing significant advantages over traditional tissue biopsies in cancer diagnosis and management. This innovative technique enables the non-invasive analysis of tumor-derived components such as circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), and extracellular vesicles (EVs) from bodily fluids like blood, urine, and saliva.
One of the primary benefits of liquid biopsy is its ability to facilitate early cancer detection and real-time monitoring of tumor dynamics. Unlike traditional biopsies, which require invasive procedures to obtain tissue samples from tumors, liquid biopsies can be performed through simple blood draws, significantly reducing patient discomfort and risk associated with surgical interventions. This non-invasive nature allows for repeated sampling over time, which is essential for monitoring disease progression, treatment responses, and detecting recurrences at an early stage [1][4].
The real-time monitoring capability of liquid biopsies enhances personalized medicine by providing continuous insights into the molecular profile of tumors. For instance, the analysis of ctDNA can reveal mutations that are critical for tailoring specific therapies, enabling clinicians to adjust treatment plans based on the evolving characteristics of the tumor [11]. Furthermore, the ability to track minimal residual disease and tumor evolution through liquid biopsies allows for timely interventions, which can significantly improve patient outcomes [1].
Liquid biopsy also offers a comprehensive view of tumor heterogeneity, which is often challenging to capture with traditional biopsies that may only sample a small portion of the tumor [2]. By analyzing multiple biomarkers simultaneously, liquid biopsy can provide a more accurate representation of the tumor's genetic landscape, aiding in the identification of actionable targets for therapy [1].
Moreover, recent advancements in biosensing technologies have further enhanced the sensitivity and specificity of liquid biopsies, enabling the detection of biomarkers at very low concentrations [11]. This is crucial for early-stage cancer detection, where the presence of tumor markers may be minimal yet indicative of disease [3].
In summary, liquid biopsy improves cancer diagnosis by offering a less invasive, real-time monitoring solution that enhances the precision of treatment strategies. Its ability to provide continuous insights into tumor biology while overcoming the limitations of traditional biopsies positions it as a pivotal tool in the evolution of personalized oncology [1][2][4].
4.3 Comprehensive Tumor Profiling
Liquid biopsy represents a transformative advancement in cancer diagnostics, offering several advantages over traditional tissue biopsies, particularly in terms of comprehensive tumor profiling. This technique involves the analysis of biofluids such as blood, urine, saliva, and other bodily fluids to extract and evaluate various tumor-related biomarkers, including circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), microRNAs, and extracellular vesicles.
One of the most significant advantages of liquid biopsy is its non-invasive nature, which allows for repeated sampling without the risks associated with surgical procedures. Traditional biopsies often require invasive tissue extraction from primary tumors, which can be painful, carry a risk of complications, and may not be feasible for tumors located in difficult-to-reach areas. In contrast, liquid biopsies can be performed more frequently, enabling continuous monitoring of tumor dynamics, treatment responses, and disease progression over time[2].
Moreover, liquid biopsies provide a more comprehensive assessment of tumor heterogeneity. Unlike traditional biopsies that may only capture a small portion of the tumor's genetic landscape, liquid biopsies analyze genetic material shed from all tumor sites into the bloodstream. This allows for a more representative profile of the tumor's molecular characteristics, potentially leading to better-informed treatment decisions and personalized therapeutic strategies[19].
The technological advancements in liquid biopsy methodologies, including next-generation sequencing and the integration of artificial intelligence, have significantly enhanced the sensitivity and specificity of these tests. This enables the detection of minimal residual disease and tracking of tumor evolution, which are crucial for timely interventions[1]. Additionally, the ability to combine multiple biomarkers, such as ctDNA, CTCs, and extracellular vesicles, into a multimodal approach offers a more holistic view of the tumor's behavior and treatment response[4].
In summary, liquid biopsy improves cancer diagnosis by providing a non-invasive, comprehensive, and real-time analysis of tumor characteristics, facilitating early detection and ongoing monitoring, which are essential for effective cancer management and personalized medicine. As the field continues to evolve, the integration of liquid biopsy into clinical practice holds the potential to significantly enhance patient outcomes and revolutionize cancer care[41].
5 Challenges and Limitations
5.1 Technical Challenges
Liquid biopsy represents a significant advancement in cancer diagnostics, providing a non-invasive method for the early detection and monitoring of various cancers. The technique primarily involves the analysis of circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), extracellular vesicles (EVs), and tumor-educated platelets (TEPs) present in bodily fluids such as blood, urine, and saliva. This innovative approach offers real-time insights into tumor dynamics, treatment responses, and disease progression, potentially enabling personalized treatment strategies and timely interventions [1][2][4].
However, the implementation of liquid biopsy in clinical practice is not without its challenges, particularly in the context of technical limitations. One of the foremost technical challenges is ensuring the sensitivity and specificity of liquid biopsy assays, especially for early-stage cancers. Detecting analytes in the presence of low tumor burden or subclonal populations can significantly impact the reliability of results. Tumor heterogeneity further complicates the analysis, as different tumor cells may shed varying amounts of biomarkers into circulation [1][4].
Moreover, the presence of background noise from non-tumor-derived components in bodily fluids can obscure the detection of relevant biomarkers. This necessitates the development of highly sensitive and selective detection methods, which are still under research and optimization [2][7]. The integration of artificial intelligence (AI) and machine learning (ML) techniques has shown promise in enhancing the diagnostic accuracy of liquid biopsies by enabling the analysis of complex data sets and improving the interpretation of results [4].
Another significant challenge is the standardization of methodologies across different laboratories and clinical settings. Variability in sample collection, processing, and analysis techniques can lead to inconsistent results, which hampers the clinical utility of liquid biopsy [2][9]. Additionally, the lack of established protocols for the evaluation of liquid biopsy results in clinical decision-making poses another barrier to its widespread adoption [7].
In summary, while liquid biopsy holds great potential to transform cancer diagnostics by providing a less invasive, real-time alternative to traditional tissue biopsies, its successful implementation is contingent upon overcoming significant technical challenges. These include improving sensitivity and specificity, addressing tumor heterogeneity, standardizing methodologies, and integrating advanced technologies for data analysis. Ongoing research and collaboration among experts in the field are essential to realize the full potential of liquid biopsy in precision oncology [1][2][4].
5.2 Standardization and Validation
Liquid biopsy has emerged as a significant advancement in cancer diagnostics, providing a noninvasive approach to detect and monitor cancer through the analysis of biomarkers in bodily fluids such as blood, urine, and saliva. This technique enables the profiling of circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), microRNAs, and extracellular vesicles, which are pivotal in understanding tumor dynamics and tailoring personalized treatment strategies. The advantages of liquid biopsy include early detection of cancer, real-time monitoring of treatment responses, and reduced patient risk compared to traditional tissue biopsies [2][42].
However, despite its promising potential, the clinical integration of liquid biopsy faces several challenges and limitations. One major issue is the lack of standardization in methodologies and protocols. Variability in sample collection, processing, and analysis can lead to inconsistencies in results, which complicates the establishment of reliable Standard Operating Procedures (SOPs) for clinical use [43]. Furthermore, the clinical utility of specific assays remains to be clearly defined, which hinders the adoption of liquid biopsy in routine practice [4][43].
The need for rigorous validation of liquid biopsy techniques is paramount. As the technology evolves, ensuring that tests are both sensitive and specific is crucial for accurate cancer detection and monitoring. This is particularly challenging due to tumor heterogeneity, which can affect biomarker expression and, consequently, the reliability of liquid biopsy results [4]. Moreover, the integration of liquid biopsy into clinical workflows necessitates collaboration among laboratories to harmonize practices and mitigate inter-laboratory variability [43].
Quality control is another critical aspect that must be addressed to enhance the credibility of liquid biopsy tests. Regular participation in external quality assessment schemes is essential for laboratories offering liquid biopsy testing, as it helps identify deficiencies and facilitates improvements in service quality [43]. Accreditation of laboratories based on established standards, such as ISO15189 in Europe or CLIA/CAP in the United States, is also vital for ensuring the reliability of liquid biopsy results [43].
In summary, while liquid biopsy presents a revolutionary approach to cancer diagnosis with numerous advantages, its successful implementation in clinical practice is contingent upon overcoming significant challenges related to standardization, validation, and quality control. Continued research and collaboration among stakeholders in the field will be essential to fully realize the potential of liquid biopsy in improving cancer diagnosis and patient outcomes [4][43].
5.3 Ethical Considerations
Liquid biopsy represents a transformative advancement in cancer diagnosis, primarily by offering a non-invasive alternative to traditional tissue biopsies. This innovative approach allows for the analysis of various biomarkers, including circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), microRNAs, and extracellular vesicles (EVs), extracted from biofluids such as blood, urine, and saliva. The advantages of liquid biopsy in cancer diagnosis are multifaceted.
Firstly, liquid biopsy facilitates early detection of cancers, which is crucial for improving patient outcomes. It enables real-time monitoring of tumor dynamics and provides insights into treatment responses, allowing for timely adjustments in therapy. This is particularly significant given that many cancers do not have established screening programs, and early-stage detection can significantly enhance survival rates and reduce the financial burden associated with advanced-stage treatments[34].
Moreover, liquid biopsy is characterized by its non-invasive nature, which minimizes risks associated with surgical procedures. This aspect not only improves patient comfort but also allows for more frequent testing, enabling continuous monitoring of disease progression and treatment efficacy[44]. The incorporation of advanced technologies, such as microfluidics and artificial intelligence, further enhances the sensitivity and specificity of liquid biopsies, making them more reliable for clinical applications[7].
However, despite these advantages, liquid biopsy faces several challenges and limitations. One significant issue is the sensitivity required to detect early-stage cancers, as current methodologies may not consistently achieve the necessary levels of detection for all tumor types[1]. Additionally, tumor heterogeneity presents a challenge in accurately assessing the molecular profile of a patient's cancer, as variations in tumor cell populations can affect the results of liquid biopsies[4].
Another limitation is the lack of standardized protocols for liquid biopsy testing, which can lead to variability in results across different laboratories and clinical settings[9]. The interpretation of liquid biopsy results can also be complex, particularly in the context of tumor evolution and the presence of subclonal populations, which may not be adequately captured by current techniques[1].
Ethical considerations surrounding liquid biopsy include issues related to patient consent, data privacy, and the potential for psychological impacts stemming from the results. As liquid biopsies provide information about a patient's genetic and molecular profile, ensuring the confidentiality and ethical use of this sensitive data is paramount. Additionally, the implications of incidental findings, which may arise during testing, necessitate careful ethical considerations regarding how such information is communicated to patients and how it may affect their treatment decisions[9].
In conclusion, while liquid biopsy holds great promise for improving cancer diagnosis through early detection and continuous monitoring, addressing the associated challenges and ethical considerations is essential for its successful integration into clinical practice. Continued advancements in technology and collaborative research will be crucial in overcoming these hurdles and maximizing the potential of liquid biopsy in oncology.
6 Future Directions
6.1 Advances in Technology
Liquid biopsy represents a transformative approach in cancer diagnosis, providing several advantages over traditional tissue biopsy methods. It allows for the non-invasive analysis of various biomarkers found in body fluids, such as blood, urine, saliva, and cerebrospinal fluid. This method facilitates the real-time monitoring of tumor dynamics, treatment responses, and disease progression, which is critical for timely interventions and personalized treatment strategies.
One of the primary benefits of liquid biopsy is its ability to detect circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), extracellular vesicles (EVs), and tumor-educated platelets (TEPs). These biomarkers offer insights into the tumor's genetic landscape and can reflect extensive malignant features that may not be present in primary tumors, thereby enhancing diagnostic accuracy. Recent advancements in artificial intelligence (AI) have further improved the diagnostic precision of liquid biopsy by integrating multimodal data from various biomarkers, providing a more comprehensive view of tumor characteristics [1].
Technological innovations, particularly in microfluidics, have significantly advanced the capabilities of liquid biopsy. Microfluidic platforms enable real-time observations and the precise, sensitive detection of biomarkers at microscale levels. The integration of microfluidics with advanced technologies, such as CRISPR gene editing and nanotechnology, has the potential to enhance the sensitivity and specificity of liquid biopsies, making them more effective for early cancer diagnosis [7].
Moreover, liquid biopsy offers the potential for serial assessments, allowing for continuous monitoring of treatment responses and the identification of emerging resistance mechanisms. This capability is particularly beneficial in oncology, where treatment regimens may need to be adjusted based on the evolving tumor profile [45]. The use of liquid biopsy can also reduce the need for invasive tissue sampling, making it a more patient-friendly option, especially in cases where traditional biopsies are challenging or impossible [36].
Despite these advancements, challenges remain in the widespread implementation of liquid biopsy. Issues such as the sensitivity of detecting analytes in early-stage cancers and the need for standardized methodologies must be addressed to fully realize the potential of liquid biopsy in clinical settings [2]. Future research and technological developments will likely focus on overcoming these challenges, ultimately aiming to establish liquid biopsy as a standard diagnostic tool in cancer care.
In summary, liquid biopsy enhances cancer diagnosis through its non-invasive nature, ability to provide real-time insights into tumor biology, and integration with cutting-edge technologies. As research progresses, the field is poised for significant advancements that will further refine and expand the applications of liquid biopsy in oncology.
6.2 Integration into Clinical Practice
Liquid biopsy represents a transformative advancement in oncology, significantly enhancing cancer diagnosis through various mechanisms. This non-invasive technique allows for the analysis of circulating tumor components, including circulating tumor DNA (ctDNA), circulating tumor RNA (ctRNA), circulating tumor cells (CTCs), and extracellular vesicles (EVs), thereby providing real-time insights into tumor biology and dynamics.
One of the primary benefits of liquid biopsy is its potential for early cancer detection. By identifying specific biomarkers associated with malignancies, liquid biopsy can facilitate the diagnosis of cancer at stages that are often undetectable through conventional imaging or tissue biopsies. This capability is particularly crucial for improving patient outcomes, as early intervention can lead to better prognosis and treatment efficacy [35].
In addition to early detection, liquid biopsy plays a vital role in cancer staging and prognosis evaluation. It enables the monitoring of tumor progression and response to treatment, thereby informing clinicians about the effectiveness of therapeutic strategies. This real-time monitoring capability allows for timely adjustments to treatment plans, which is essential in the context of personalized medicine [42].
The integration of liquid biopsy into clinical practice is further supported by advancements in genomic and molecular technologies, such as next-generation sequencing (NGS) and digital polymerase chain reaction (dPCR). These technologies enhance the sensitivity and specificity of liquid biopsy, allowing for the detection of somatic variants and actionable genomic alterations in tumors [35]. Furthermore, the incorporation of artificial intelligence (AI) and machine learning (ML) in analyzing liquid biopsy data has shown promise in improving diagnostic accuracy and enabling multimodal approaches that combine various biomarkers [1].
Despite its advantages, several challenges remain in the widespread adoption of liquid biopsy in clinical settings. These include the need for standardization of techniques, interpretation of results amidst tumor heterogeneity, and ensuring accessibility and affordability of tests [4]. Addressing these challenges is crucial for integrating liquid biopsy into routine clinical workflows effectively.
Looking forward, the future of liquid biopsy is poised for further innovations, with expectations for enhanced standardization and the application of multi-omics coanalysis. The ongoing research aims to solidify the role of liquid biopsy in precision medicine, thereby improving cancer diagnosis and treatment management [42]. As technological advancements continue to emerge, liquid biopsy is likely to become an indispensable tool in oncology, enabling a more personalized approach to cancer care that aligns with the principles of precision medicine.
6.3 Potential for Personalized Medicine
Liquid biopsy has emerged as a transformative approach in oncology, enhancing cancer diagnosis through its non-invasive nature and ability to provide real-time insights into tumor biology. By analyzing circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), extracellular vesicles (EVs), and other biomarkers in biofluids, liquid biopsy offers a comprehensive profile of the tumor's genetic and molecular characteristics, facilitating early detection and personalized treatment strategies.
The technique of liquid biopsy allows for the assessment of tumor dynamics, treatment response, and disease progression, making it a powerful tool for precision oncology. For instance, it can detect minimal residual disease and track tumor evolution, which are critical for tailoring therapies to individual patients based on their unique tumor profiles [4]. Moreover, advancements in artificial intelligence (AI) and multi-omics approaches have significantly improved the diagnostic accuracy of liquid biopsies, integrating various biomarkers to provide a more holistic view of tumor characteristics [1].
Despite its advantages, the application of liquid biopsy faces challenges, particularly in detecting analytes in early-stage cancers and evaluating the tumor molecular fraction. Factors such as tumor burden, molecular fraction, and the presence of subclones can affect the sensitivity and specificity of the analysis [1]. Therefore, ongoing research and technological innovations are essential to overcome these hurdles and fully realize the potential of liquid biopsy in cancer care.
Looking towards the future, liquid biopsy is poised to revolutionize cancer diagnostics and treatment by enabling more personalized medicine approaches. Its ability to provide rapid, non-invasive, and cost-effective diagnostics could lead to timely interventions tailored to the individual patient's needs [2]. The integration of liquid biopsy into clinical workflows is expected to enhance patient outcomes by facilitating early detection, continuous monitoring, and the customization of therapeutic strategies based on the molecular profiles of tumors [46].
In conclusion, liquid biopsy represents a significant advancement in cancer diagnosis, with the potential to enhance personalized medicine through its non-invasive methodology and real-time insights into tumor biology. As technological challenges are addressed and methodologies are standardized, liquid biopsy is likely to become an integral component of precision oncology, paving the way for improved patient management and outcomes.
7 Conclusion
Liquid biopsy represents a significant advancement in cancer diagnostics, transforming how we detect, monitor, and manage cancer. The primary findings highlight the non-invasive nature of liquid biopsy, which allows for real-time monitoring of tumor dynamics through the analysis of circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), and exosomes. This approach not only facilitates early detection of cancer but also supports personalized treatment strategies by providing insights into tumor heterogeneity and genetic mutations. The current state of research indicates a growing acceptance of liquid biopsy in clinical settings, particularly for cancers like non-small cell lung carcinoma and colorectal cancer, where early intervention can lead to improved outcomes. However, challenges such as standardization of methodologies, technical limitations, and ethical considerations remain. Future research should focus on overcoming these barriers, enhancing the sensitivity and specificity of liquid biopsy tests, and integrating these methodologies into routine clinical practice. The potential for liquid biopsy to revolutionize cancer care is immense, promising a future where diagnostics and treatment are tailored to the individual characteristics of each patient's tumor.
References
- [1] Claudio Antonio Coppola;Simona De Summa;Giuseppina Matera;Brunella Pilato;Debora Traversa;Stefania Tommasi. Liquid Biopsy: The Challenges of a Revolutionary Approach in Oncology.. International journal of molecular sciences(IF=4.9). 2025. PMID:40507825. DOI: 10.3390/ijms26115013.
- [2] Ying Bao;Dejing Zhang;Huihui Guo;Wenxue Ma. Beyond blood: Advancing the frontiers of liquid biopsy in oncology and personalized medicine.. Cancer science(IF=4.3). 2024. PMID:38308498. DOI: 10.1111/cas.16097.
- [3] Lu Chang;Jinming Li;Rui Zhang. Liquid biopsy for early diagnosis of non-small cell lung carcinoma: recent research and detection technologies.. Biochimica et biophysica acta. Reviews on cancer(IF=8.3). 2022. PMID:35436521. DOI: 10.1016/j.bbcan.2022.188729.
- [4] Sidney W Fu;Cong Tang;Xiaohui Tan;Sudhir Srivastava. Liquid biopsy for early cancer detection: technological revolutions and clinical dilemma.. Expert review of molecular diagnostics(IF=3.6). 2024. PMID:39360748. DOI: 10.1080/14737159.2024.2408744.
- [5] Raffaele Palmirotta;Domenica Lovero;Paola Cafforio;Claudia Felici;Francesco Mannavola;Eleonora Pellè;Davide Quaresmini;Marco Tucci;Franco Silvestris. Liquid biopsy of cancer: a multimodal diagnostic tool in clinical oncology.. Therapeutic advances in medical oncology(IF=4.2). 2018. PMID:30181785. DOI: 10.1177/1758835918794630.
- [6] Rudy N Zalzal;Najla Fakhruddin;Rami Mahfouz. Liquid Biopsy's Role in Head and Neck Tumors: Changing Paradigms in the Era of Precision Medicine.. Diagnostics (Basel, Switzerland)(IF=3.3). 2025. PMID:40941749. DOI: 10.3390/diagnostics15172262.
- [7] Osama H Abusara;Ahmed S A Ali Agha;Sanaa K Bardaweel. Advancements and innovations in liquid biopsy through microfluidic technology for cancer diagnosis.. The Analyst(IF=3.3). 2025. PMID:40181713. DOI: 10.1039/d5an00105f.
- [8] Narshone Soda;Bernd H A Rehm;Prashant Sonar;Nam-Trung Nguyen;Muhammad J A Shiddiky. Advanced liquid biopsy technologies for circulating biomarker detection.. Journal of materials chemistry. B(IF=5.7). 2019. PMID:31646316. DOI: 10.1039/c9tb01490j.
- [9] Anca-Narcisa Neagu;Pathea S Bruno;Claudiu-Laurentiu Josan;Natalie Waterman;Hailey Morrissiey;Victor T Njoku;Costel C Darie. In Search of Ideal Solutions for Cancer Diagnosis: From Conventional Methods to Protein Biomarkers in Liquid Biopsy.. Proteomes(IF=3.6). 2025. PMID:41133620. DOI: 10.3390/proteomes13040047.
- [10] Paola Ulivi;Stefano Indraccolo. Liquid Biopsies in Cancer Diagnosis, Monitoring and Prognosis.. Biomedicines(IF=3.9). 2022. PMID:36359268. DOI: 10.3390/biomedicines10112748.
- [11] Panagiota M Kalligosfyri;Eleni Lamprou;Despina P Kalogianni. Emerging Sensing Technologies for Liquid Biopsy Applications: Steps Closer to Personalized Medicine.. Sensors (Basel, Switzerland)(IF=3.5). 2024. PMID:39771639. DOI: 10.3390/s24247902.
- [12] A Markou;E Tzanikou;E Lianidou. The potential of liquid biopsy in the management of cancer patients.. Seminars in cancer biology(IF=15.7). 2022. PMID:35331850. DOI: 10.1016/j.semcancer.2022.03.013.
- [13] Areti Strati;Athina Markou;Evgenia Kyriakopoulou;Evi Lianidou. Detection and Molecular Characterization of Circulating Tumour Cells: Challenges for the Clinical Setting.. Cancers(IF=4.4). 2023. PMID:37046848. DOI: 10.3390/cancers15072185.
- [14] Shrikant B Mali. Molecular screening of head neck cancer.. Oral oncology(IF=3.9). 2023. PMID:37399708. DOI: 10.1016/j.oraloncology.2023.106481.
- [15] Tanvi H Visal;Petra den Hollander;Massimo Cristofanilli;Sendurai A Mani. Circulating tumour cells in the -omics era: how far are we from achieving the 'singularity'?. British journal of cancer(IF=6.8). 2022. PMID:35273384. DOI: 10.1038/s41416-022-01768-9.
- [16] Enrico Cortesi;Michela Palleschi;Valentina Magri;Giuseppe Naso. The promise of liquid biopsy in cancer: a clinical perspective.. Chinese journal of cancer research = Chung-kuo yen cheng yen chiu(IF=6.3). 2015. PMID:26543335. DOI: 10.3978/j.issn.1000-9604.2015.10.01.
- [17] Hang Yin;Manjie Zhang;Yu Zhang;Xuebing Zhang;Xia Zhang;Bin Zhang. Liquid biopsies in cancer.. Molecular biomedicine(IF=10.1). 2025. PMID:40108089. DOI: 10.1186/s43556-025-00257-8.
- [18] Michail Ignatiadis;Mark Lee;Stefanie S Jeffrey. Circulating Tumor Cells and Circulating Tumor DNA: Challenges and Opportunities on the Path to Clinical Utility.. Clinical cancer research : an official journal of the American Association for Cancer Research(IF=10.2). 2015. PMID:26527805. DOI: 10.1158/1078-0432.CCR-14-1190.
- [19] Shicai Liu;Jinke Wang. Current and Future Perspectives of Cell-Free DNA in Liquid Biopsy.. Current issues in molecular biology(IF=3.0). 2022. PMID:35735625. DOI: 10.3390/cimb44060184.
- [20] Arlou Kristina Angeles;Florian Janke;Simone Bauer;Petros Christopoulos;Anja Lisa Riediger;Holger Sültmann. Liquid Biopsies beyond Mutation Calling: Genomic and Epigenomic Features of Cell-Free DNA in Cancer.. Cancers(IF=4.4). 2021. PMID:34830770. DOI: 10.3390/cancers13225615.
- [21] Francesca Galardi;Francesca De Luca;Dario Romagnoli;Chiara Biagioni;Erica Moretti;Laura Biganzoli;Angelo Di Leo;Ilenia Migliaccio;Luca Malorni;Matteo Benelli. Cell-Free DNA-Methylation-Based Methods and Applications in Oncology.. Biomolecules(IF=4.8). 2020. PMID:33334040. DOI: 10.3390/biom10121677.
- [22] Sihao Du;Ke Cao;Yadong Yan;Yupeng Wang;Zhenshun Wang;Dongdong Lin. Developments and current status of cell-free DNA in the early detection and management of hepatocellular carcinoma.. Journal of gastroenterology and hepatology(IF=3.4). 2024. PMID:37990622. DOI: 10.1111/jgh.16416.
- [23] Yuyuan Zhang;Zaoqu Liu;Kun Ji;Xin Li;Caihong Wang;Zhigang Ren;Yang Liu;Xinju Chen;Xinwei Han;Lingfang Meng;Lifeng Li;Zhen Li. Clinical Application Value of Circulating Cell-free DNA in Hepatocellular Carcinoma.. Frontiers in molecular biosciences(IF=4.0). 2021. PMID:34660697. DOI: 10.3389/fmolb.2021.736330.
- [24] Rafael Colmenares;Noemí Álvarez;Santiago Barrio;Joaquín Martínez-López;Rosa Ayala. The Minimal Residual Disease Using Liquid Biopsies in Hematological Malignancies.. Cancers(IF=4.4). 2022. PMID:35267616. DOI: 10.3390/cancers14051310.
- [25] Joseph G Schroers-Martin;Ash A Alizadeh. Cell-Free DNA in Hematologic Malignancies.. JCO oncology practice(IF=4.6). 2024. PMID:39531844. DOI: 10.1200/OP-24-00648.
- [26] Adriana Alonso Novais;Guilherme Henrique Tamarindo;Luiz Gustavo de Almeida Chuffa;Debora Aparecida Pires de Campos Zuccari. Decoding Hidden Messengers: Proteomic Profiling of Exosomes in Mammary Cancer Research.. Biomedicines(IF=3.9). 2023. PMID:37893211. DOI: 10.3390/biomedicines11102839.
- [27] Dan Yu;Yixin Li;Maoye Wang;Jianmei Gu;Wenrong Xu;Hui Cai;Xinjian Fang;Xu Zhang. Exosomes as a new frontier of cancer liquid biopsy.. Molecular cancer(IF=33.9). 2022. PMID:35180868. DOI: 10.1186/s12943-022-01509-9.
- [28] Na Li;Cheng Cheng;Di Wu;Zixuan Song;Bei Wang;Guiying Li;Fang Yang. Immunofluorescent analysis of exosomes using a microchip filled with transparent antibody-conjugated beads for breast cancer liquid biopsy.. Analytica chimica acta(IF=6.0). 2025. PMID:40015783. DOI: 10.1016/j.aca.2025.343743.
- [29] Svetlana Tamkovich;Alexandra Borisova;Andrey Shevela;Alexander Chernyavskiy;Alyona Chernyshovа. Exosomal MicroRNA: Diagnostic Potential and Role in Breast Cancer Dissemination.. Molecules (Basel, Switzerland)(IF=4.6). 2025. PMID:41097280. DOI: 10.3390/molecules30193858.
- [30] K Auxzilia Preethi;Sushmaa Chandralekha Selvakumar;Kehinde Ross;Selvaraj Jayaraman;Deusdedit Tusubira;Durairaj Sekar. Liquid biopsy: Exosomal microRNAs as novel diagnostic and prognostic biomarkers in cancer.. Molecular cancer(IF=33.9). 2022. PMID:35172817. DOI: 10.1186/s12943-022-01525-9.
- [31] Haozhou Tang;Dan Yu;Jiahui Zhang;Maoye Wang;Min Fu;Yu Qian;Xiaoxin Zhang;Runbi Ji;Jianmei Gu;Xu Zhang. The new advance of exosome-based liquid biopsy for cancer diagnosis.. Journal of nanobiotechnology(IF=12.6). 2024. PMID:39380060. DOI: 10.1186/s12951-024-02863-0.
- [32] Valeria Garzarelli;Alessia Foscarini;Vanessa Indirli;Ilaria Menon;Diego Mangiullo;Tiziano Verri;Elisabetta Primiceri;Annamaria Nigro;Angelo Quattrini;Alessandro Romano;Maria Serena Chiriacò;Giuseppe Gigli;Francesco Ferrara. Lab on chips for liquid biopsy: a flexible and customized approach through microfabrication.. Biomedical microdevices(IF=3.3). 2025. PMID:40484881. DOI: 10.1007/s10544-025-00757-y.
- [33] Efstathia Liatsou;Ioannis Kollias;Maria Trapali;Diamantis I Tsilimigras;Maria Gavriatopoulou;Ioannis Ntanasis-Stathopoulos. Liquid Biopsies in the Early Diagnosis, Prognosis, and Tailored Treatment of Colorectal Cancer.. Cancers(IF=4.4). 2025. PMID:40149264. DOI: 10.3390/cancers17060927.
- [34] Siobhan Connal;James M Cameron;Alexandra Sala;Paul M Brennan;David S Palmer;Joshua D Palmer;Haley Perlow;Matthew J Baker. Liquid biopsies: the future of cancer early detection.. Journal of translational medicine(IF=7.5). 2023. PMID:36774504. DOI: 10.1186/s12967-023-03960-8.
- [35] Ho-Yin Ho;Kei-See Kasey Chung;Chau-Ming Kan;Sze-Chuen Cesar Wong. Liquid Biopsy in the Clinical Management of Cancers.. International journal of molecular sciences(IF=4.9). 2024. PMID:39201281. DOI: 10.3390/ijms25168594.
- [36] Marius E Mayerhoefer;Andreas Kienzle;Sungmin Woo;Hebert Alberto Vargas. Update on Liquid Biopsy.. Radiology(IF=15.2). 2025. PMID:40525978. DOI: 10.1148/radiol.241030.
- [37] Ioannis Kournoutas;Brittany L Siontis. Minimal Residual Disease in Metastatic Soft Tissue Sarcoma.. Current treatment options in oncology(IF=4.7). 2025. PMID:40072823. DOI: 10.1007/s11864-025-01303-x.
- [38] Maria Palmieri;Elisa Frullanti. Different Liquid Biopsies for the Management of Non-Small Cell Lung Cancer in the Mutational Oncology Era.. Medical sciences (Basel, Switzerland)(IF=4.4). 2023. PMID:36649045. DOI: 10.3390/medsci11010008.
- [39] Shlomi Madar;Reef Einoch Amor;Sharon Furman-Assaf;Eitan Friedman. Innovative Approaches to Early Detection of Cancer-Transforming Screening for Breast, Lung, and Hard-to-Screen Cancers.. Cancers(IF=4.4). 2025. PMID:40507348. DOI: 10.3390/cancers17111867.
- [40] Michael D Kessler;Nisha R Pawar;Stuart S Martin;Toni M Antalis;Timothy D O'Connor. Improving Cancer Detection and Treatment with Liquid Biopsies and ptDNA.. Trends in cancer(IF=17.5). 2018. PMID:30149882. DOI: 10.1016/j.trecan.2018.07.004.
- [41] Austin K Mattox;Chetan Bettegowda;Shibin Zhou;Nickolas Papadopoulos;Kenneth W Kinzler;Bert Vogelstein. Applications of liquid biopsies for cancer.. Science translational medicine(IF=14.6). 2019. PMID:31462507. DOI: 10.1126/scitranslmed.aay1984.
- [42] Jinghan Song;Xiong Ye;Hui Xiao. Liquid biopsy entering clinical practice: Past discoveries, current insights, and future innovations.. Critical reviews in oncology/hematology(IF=5.6). 2025. PMID:39756526. DOI: 10.1016/j.critrevonc.2025.104613.
- [43] Aliki Ntzifa;Evi Lianidou. Pre-analytical conditions and implementation of quality control steps in liquid biopsy analysis.. Critical reviews in clinical laboratory sciences(IF=5.5). 2023. PMID:37518938. DOI: 10.1080/10408363.2023.2230290.
- [44] Liwei Ma;Huiling Guo;Yunxiang Zhao;Zhibo Liu;Chenran Wang;Jiahao Bu;Ting Sun;Jianwei Wei. Liquid biopsy in cancer current: status, challenges and future prospects.. Signal transduction and targeted therapy(IF=52.7). 2024. PMID:39617822. DOI: 10.1038/s41392-024-02021-w.
- [45] Umberto Malapelle;Pasquale Pisapia;Francesco Pepe;Gianluca Russo;Mauro Buono;Alessandro Russo;Jorge Gomez;Ola Khorshid;Philip C Mack;Christian Rolfo;Giancarlo Troncone. The evolving role of liquid biopsy in lung cancer.. Lung cancer (Amsterdam, Netherlands)(IF=4.4). 2022. PMID:35998482. DOI: 10.1016/j.lungcan.2022.08.004.
- [46] Elena Ricciardi;Elena Giordani;Giovanna Ziccheddu;Italia Falcone;Patrizio Giacomini;Maurizio Fanciulli;Michelangelo Russillo;Marianna Cerro;Gennaro Ciliberto;Aldo Morrone;Antonino Guerrisi;Fabio Valenti. Metastatic Melanoma: Liquid Biopsy as a New Precision Medicine Approach.. International journal of molecular sciences(IF=4.9). 2023. PMID:36835424. DOI: 10.3390/ijms24044014.
MaltSci Intelligent Research Services
Search for more papers on MaltSci.com
liquid biopsy · cancer diagnosis · circulating tumor cells
© 2025 MaltSci
