Preprint
Review

This version is not peer-reviewed.

Circulating Tumor DNA in Genitourinary Malignancies: Current Applications and Future Perspectives

Submitted:

10 June 2025

Posted:

12 June 2025

You are already at the latest version

Abstract
Circulating tumor DNA (ctDNA) analysis, a cornerstone of liquid biopsy, is rapidly emerging as a transformative tool in the management of genitourinary (GU) malignancies. This review synthesizes the current evidence-based clinical utility, critical challenges, and future perspectives of ctDNA across the spectrum of major GU cancers—including prostate, urothelial, renal cell, and testicular germ cell tumors. We explore the foundational science and evolving technologies underpinning ctDNA detection and analysis, and detail its diverse clinical applications. These include its role in early detection and diagnosis, risk stratification, prognostication, real-time monitoring of therapeutic response, identification of minimal residual disease (MRD), and the elucidation of resistance mechanisms, comparing its performance with existing standard-of-care biomarkers and imaging. Advanced applications—such as the assessment of tumor mutational burden (TMB), microsatellite instability (MSI), ctDNA methylation patterns, characterization of tumor heterogeneity, and the integration of ctDNA with other liquid biopsy analytes and imaging modalities—are also discussed for their potential to provide deeper biological insights. Despite its promise, the widespread clinical implementation of ctDNA testing faces significant scientific, technical, logistical, and economic hurdles. These include the need for enhanced assay sensitivity, rigorous standardization, comprehensive clinical validation, and strategies for interpreting complex genomic data such as variants of uncertain significance (VUS) and clonal hematopoiesis of indeterminate potential (CHIP)). Furthermore, paramount ethical, legal, and social implications (ELSI) must be proactively addressed. Looking ahead, anticipated technological breakthroughs, the integration of artificial intelligence/machine learning, the development of malignancy-specific ctDNA assays, and innovative clinical trial designs are poised to overcome current limitations. These advancements are expected to unlock the full transformative potential of ctDNA, potentially revolutionizing GU cancer care by enabling more precise, personalized, and effective patient management across the disease continuum. This review provides clinicians and practitioners with a comprehensive overview to navigate the evolving landscape of ctDNA in GU oncology.
Keywords: 
;  ;  ;  ;  ;  ;  

Introduction

Circulating tumor DNA (ctDNA) consists of deoxyribonucleic acid fragments actively or passively released into the bloodstream from tumor cells, primarily through mechanisms such as apoptosis, necrosis, and active secretion [1,2,3,4]. These tumor-derived fragments constitute a minor fraction of the total cell-free DNA (cfDNA) found in circulation, the majority of which originates from normal, non-neoplastic cells [5,6]. Although the presence of tumor DNA in blood has been recognized for decades, recent technological advancements have been pivotal in enabling the comprehensive characterization and clinical utilization of this analyte [7]. The analysis of ctDNA has propelled the development of “liquid biopsy,” a minimally invasive diagnostic paradigm that offers substantial advantages over traditional tissue biopsies. These advantages include ease of performance, the capacity for frequent serial sampling to monitor dynamic tumor changes, and a potentially more representative assessment of intra-tumor heterogeneity by capturing genetic material from both primary and metastatic sites [7,8]. The significance of ctDNA as a biomarker lies in its ability to carry tumor-specific genetic and epigenetic alterations, thereby providing a real-time reflection of the tumor's genomic landscape [8,9].
The biological characteristics of ctDNA are crucial for its detection and clinical interpretation. Notably, ctDNA fragments are often shorter (e.g., around 145-150 base pairs) compared to cfDNA derived from non-malignant cells (e.g., 160-180 base pairs), a feature that can be exploited to enhance detection sensitivity [10,11,12]. The quantity and detectability of ctDNA in circulation are influenced by various tumor-related factors, including tumor size, stage, proliferative activity, and metastatic burden, with higher levels generally observed in patients with advanced-stage disease [13,14,15,16]. Furthermore, ctDNA possesses a relatively short half-life, estimated to range from approximately 16 minutes to 2.5 hours [12,15]. This rapid turnover allows ctDNA levels to dynamically reflect changes in tumor burden, making it a valuable tool for real-time disease monitoring and assessment of treatment response [7,17]. Molecularly, ctDNA encapsulates a wealth of tumor-specific information, including point mutations, copy number variations (CNVs), chromosomal rearrangements, and epigenetic modifications such as DNA methylation patterns [1,4,18,19]. This ability to reflect the genomic and epigenomic status of both primary and metastatic lesions underscores its potential to provide a more comprehensive molecular profile than single-site tissue biopsies [20,21].
The reliable detection, quantification, and comprehensive profiling of ctDNA from patient plasma, particularly in the context of genitourinary malignancies, are critically dependent on meticulous pre-analytical procedures and sophisticated analytical technologies. Pre-analytical variables significantly impact the quality and quantity of isolated ctDNA [22]. Key considerations include the choice of blood collection tubes, with specialized cfDNA BCTs (Blood Collection Tubes) demonstrating superiority over standard EDTA tubes in preserving sample integrity and reducing genomic DNA contamination, especially when immediate processing is not feasible [23]. Plasma is the preferred sample type over serum to minimize contamination from white blood cell lysis [24]. Standardized protocols for plasma separation, typically involving double centrifugation steps, are recommended to further reduce genomic DNA background [23]. Storage conditions and DNA extraction methodologies also profoundly influence ctDNA yield and quality, with various commercial kits offering different efficiencies [24,25].
Following optimal sample preparation, a range of analytical technologies are employed for ctDNA analysis. Polymerase chain reaction (PCR)-based methods, including quantitative PCR (qPCR), digital PCR (dPCR), droplet digital PCR (ddPCR), and BEAMing (Beads, Emulsion, Amplification, and Magnetics), offer high sensitivity for detecting known, pre-specified mutations, even at very low allele frequencies [2,24,26,27]. These targeted approaches are cost-effective and provide rapid results, making them suitable for monitoring specific alterations [28,29]. However, their primary limitation is the inability to discover novel or unexpected genomic alterations [2]. In contrast, next-generation sequencing (NGS) platforms, encompassing targeted panels, whole-exome sequencing (WES), and whole-genome sequencing (WGS), enable broader, unbiased interrogation of the ctDNA landscape, facilitating the identification of a wide array of genetic and epigenetic changes, including novel mutations and complex structural variants [6,8]. NGS technologies, often coupled with advanced bioinformatics algorithms for error correction and data interpretation, are becoming increasingly integral for comprehensive ctDNA profiling [5,12,30].
Despite these technological advancements, several inherent technical challenges persist. The typically low abundance of ctDNA, particularly in early-stage disease or minimal residual disease settings, necessitates assays with exceptionally high sensitivity and specificity to distinguish true tumor-derived signals from background noise and technical artifacts [11,31,32]. The presence of clonal hematopoiesis of indeterminate potential (CHIP) can also confound the interpretation of variants detected in cfDNA. Ensuring the analytical validity, clinical validity, and clinical utility of ctDNA assays requires ongoing efforts in standardization, rigorous validation across diverse clinical cohorts, and the development of robust bioinformatics pipelines [20,33]. Addressing these challenges is paramount for the successful integration of ctDNA analysis into routine clinical practice for guiding the management of genitourinary malignancies.

Clinical Applications of ctDNA in Genitourinary Malignancies

Circulating tumor DNA (ctDNA) analysis is rapidly establishing its role as a transformative biomarker in the clinical management of genitourinary (GU) malignancies, offering a minimally invasive avenue to interrogate tumor biology and dynamics [15,34]. The relative ease of obtaining “liquid biopsies,” primarily from blood and, in some GU contexts, urine, underscores its practical advantages for cancers such as prostate, bladder, and renal cell carcinoma [15]. The clinical utility of ctDNA in GU oncology is multifaceted, encompassing early detection, prognostication, identification of therapeutic targets, real-time monitoring of treatment efficacy, detection of minimal residual disease (MRD), and the early identification of emergent drug resistance mechanisms [15,35,36,37]. These applications are underpinned by ctDNA's ability to reflect tumor-related molecular features, including specific mutations and methylation changes [38].

Prostate Cancer

In prostate cancer, ctDNA analysis is proving particularly insightful for patients with advanced disease, especially castration-resistant prostate cancer (CRPC). Clinicians can utilize ctDNA to identify specific genomic alterations, such as androgen receptor (AR) gene amplifications or mutations (e.g., L702H, T878A), which are associated with response or resistance to AR-targeted therapies like abiraterone and enzalutamide [15,39,40]. For instance, AR gene gain detected in plasma ctDNA has been linked to poorer survival outcomes and reduced PSA decline in CRPC patients [39]. The PREMIERE trial further underscored this, showing significantly worse outcomes for patients with AR gain treated with enzalutamide [39].
Beyond AR alterations, ctDNA can reveal defects in DNA repair genes (e.g., BRCA2, ATM) and TP53 mutations, which are also associated with rapid resistance and poor outcomes with AR-targeted therapies [40,41]. This molecular information can guide therapy selection and stratification [37,42]. Furthermore, ctDNA analysis is being explored to understand and monitor neuroendocrine transformation, a resistant phenotype in prostate cancer [40]. While plasma DNA concentrations are generally higher in metastatic versus localized prostate cancer [6], factors like bone metastases and elevated LDH/PSA correlate with higher ctDNA levels [43], informing the interpretation of ctDNA results.

Urothelial (Bladder) Cancer

For urothelial cancer, particularly muscle-invasive bladder cancer (MIBC), ctDNA analysis is a powerful tool for early identification of disease recurrence post-cystectomy or trimodal therapy, often preceding imaging evidence by a median of 96–101 days [44,45]. Studies report high sensitivity (up to 100%) and specificity (98–100%) for ctDNA in predicting metastatic recurrence [44,45,46]. This early warning system allows for potential preemptive interventions. In patients receiving neoadjuvant chemotherapy, ctDNA detection accurately reflected disease persistence and predicted recurrence [45].
In non-muscle invasive bladder cancer (NMIBC), ctDNA levels can track treatment response, with high levels predicting disease progression [45]. For metastatic disease, ctDNA provides a comprehensive genomic snapshot, identifying actionable alterations like FGFR3, ERCC2, ERBB2 mutations, and tumor mutational burden (TMB) that can guide therapy [45,47]. Notably, ctDNA status may help identify patients most likely to benefit from adjuvant immunotherapy, as suggested by a study involving atezolizumab [40]. The ease of obtaining ctDNA from urine further enhances its utility in bladder cancer surveillance and monitoring [15,48].

Renal Cell Carcinoma (RCC)

The application of ctDNA in RCC is promising but faces challenges, primarily concerning assay sensitivity and detection limits [49]. Despite this, ctDNA is being investigated for earlier RCC detection, prognostication, real-time treatment response assessment, and MRD monitoring [15]. High-sensitivity MRD assays hold the potential to refine neoadjuvant and adjuvant treatment strategies [46]. While sensitivity in the adjuvant setting can vary (41–81% reported in some tumor-informed assays), specificity remains high (around 98–100%) [46]. Clinical trials are actively working to validate the utility of ctDNA analysis across different RCC stages, utilizing both plasma and urine samples [49].

Testicular Germ Cell Tumors (TGCT)

Research into ctDNA applications for TGCT is more nascent but growing (Patel et al., 2024). High-sensitivity MRD assays are being applied, offering the potential to improve disease monitoring and complement traditional serum tumor markers like AFP, hCG, and LDH [46]. Tumor-informed MRD testing in TGCT appears to maintain high specificity, crucial for reliable disease detection [46]. As with other GU cancers, ctDNA analysis could revolutionize neoadjuvant and adjuvant treatment approaches by providing a more sensitive measure of treatment efficacy and early relapse detection than current standards [46]. Prospective validation is ongoing.

Overarching Clinical Applications and Comparison to Standard of Care

Across GU malignancies, ctDNA offers distinct advantages:
  • Early Detection and Diagnosis: ctDNA can detect tumor-specific genetic or epigenetic (e.g., methylation) alterations, potentially before clinical manifestation or with greater sensitivity than some protein biomarkers [37,38]. Methylation patterns may even help identify the tissue of origin [37].
  • Risk Stratification and Prognostication: Post-treatment ctDNA positivity is a strong independent predictor of recurrence, often with months of lead time over imaging [40,50,51,52]. This allows for better identification of high-risk patients who may benefit from intensified or novel adjuvant strategies [53].
  • Real-Time Monitoring of Therapeutic Response: The short half-life of ctDNA allows for dynamic tracking of tumor burden and molecular changes during therapy [54,55]. Declines in ctDNA levels frequently correlate with treatment efficacy and improved outcomes, sometimes preceding radiological response [52,56,57,58,59].
  • Detection of Minimal Residual Disease (MRD): ctDNA is highly sensitive for MRD detection, guiding decisions about adjuvant therapy and surveillance intensity [40,46,60].
  • Identification of Resistance Mechanisms: Serial ctDNA analysis can detect the emergence of resistance mutations (e.g., to targeted therapies or chemotherapy) earlier than clinical progression, allowing for timely treatment adjustments [37,55,61,62]. It also helps characterize tumor heterogeneity [37].
Compared to traditional protein biomarkers, ctDNA generally offers greater cancer specificity and sensitivity for early recurrence detection [50]. While ctDNA analysis is currently pricier and less standardized than some conventional tests [50], its ability to provide comprehensive genomic information and real-time insights is unparalleled. It is envisioned that ctDNA will complement, and in some cases, potentially replace aspects of imaging for treatment monitoring and MRD detection [63]. However, establishing optimal sampling times, assay standardization, and clear VAF thresholds for clinical decisions remain active areas of research and require further validation in prospective trials[1,54].

Advanced and Integrative Applications

Beyond the detection of single-gene mutations, ctDNA analysis is evolving to provide a more comprehensive and dynamic understanding of tumor biology in GU malignancies. These advanced applications, including the assessment of broader genomic features and the integration of ctDNA data with other biomarkers and imaging modalities, are paving the way for more nuanced patient stratification. They also provide refined treatment monitoring and a more profound insight into tumor evolution. For clinicians, these developments promise to enhance personalized treatment strategies and improve patient management.

Unveiling Complex Genomic Landscapes

The utility of ctDNA extends to assessing complex genomic markers such as tumor mutational burden (TMB) and microsatellite instability (MSI). These features, detectable in blood samples, have emerged as important predictive biomarkers for response to immune checkpoint inhibitors across various cancers, including potentially GU malignancies [64]. The ability to non-invasively determine TMB or MSI status can aid clinicians in identifying patients most likely to benefit from immunotherapy, without the need for invasive tissue biopsies.
Epigenetic alterations, particularly DNA methylation patterns, represent another layer of information accessible through ctDNA analysis. Tumor-specific genomic methylation patterns can be identified in ctDNA and offer distinct advantages; for instance, methylation markers often exhibit remarkable stability throughout tumor development, making them reliable for tracking disease even amidst tumor heterogeneity or clonal evolution [65,66]. In the context of pediatric renal tumors, such as Wilms tumor and clear cell sarcoma of the kidney, ctDNA methylation patterns have shown utility in distinguishing between different tumor types [66,67]. Furthermore, hypermethylation of specific tumor suppressor genes (e.g., RASSF1A) detected in ctDNA via sensitive methods like droplet digital PCR (ddPCR) is being explored as a potential pan-tumor marker, which could have relevance for GU cancers [68].

Capturing Tumor Heterogeneity and Tracking Clonal Evolution

A critical advantage of ctDNA analysis is its potential to capture the spatial and temporal heterogeneity of a patient's cancer, which is often missed by single-site tissue biopsies [40,69]. By sampling DNA fragments released from various tumor sites, ctDNA can provide a more global representation of the tumor's genetic landscape, including subclonal alterations [66,70]. This is crucial for understanding clonal evolution, particularly under therapeutic pressure [48].
Longitudinal ctDNA monitoring allows clinicians to track these evolutionary dynamics in real-time [71,72]. For example, in non-small cell lung cancer, serial ctDNA analysis has revealed the emergence of multiple resistance mechanisms following EGFR inhibitor therapy, highlighting significant intra-patient heterogeneity [73]. Similar applications in GU oncology could enable the early detection of resistance to targeted therapies or chemotherapy, often months before radiographic progression, thereby informing timely treatment adjustments [74].

Integrating ctDNA with Other Liquid Biopsy Analytes for a Holistic View

To achieve an even more comprehensive understanding of tumor biology, ctDNA analysis is increasingly being integrated with other liquid biopsy analytes, such as circulating tumor cells (CTCs) and exosomes [75,76]. This multi-analyte approach allows each biomarker to compensate for the limitations of others, providing complementary data that can enhance overall diagnostic accuracy and monitoring capabilities [6].
CTCs, as intact tumor cells, offer unique phenotypic and cellular information (e.g., protein expression) that complements the genetic data from ctDNA [75]. The combination of CTC and ctDNA analysis has shown improved diagnostic performance in cancers like bladder cancer [77,78]. Similarly, exosomes carry functional protein and RNA cargo, offering insights beyond genomic alterations [75]. Tumor-educated platelets are another emerging analyte providing information about the tumor microenvironment [75].
Advanced technologies like next-generation sequencing (NGS) facilitate the simultaneous profiling of ctDNA and CTC-derived material, while machine learning algorithms help integrate these multiomic datasets [6,79,80]. While challenges such as ensuring concordance between different analytes exist [81], the synergistic information gained from integrated liquid biopsies holds significant promise for refining treatment stratification and monitoring in GU cancers [6].

Complementing Imaging with Molecular Insights

The integration of ctDNA analysis with conventional imaging modalities (e.g., PET, MRI, CT scans) offers a powerful synergy for cancer assessment. Molecular information derived from ctDNA complements the anatomical and functional data provided by imaging, leading to a more precise understanding of tumor burden, heterogeneity, and therapeutic response [82,83]. This combined approach is particularly valuable for personalized medicine in GU oncology, where timely and accurate monitoring can guide critical treatment decisions [84].
However, clinicians should be aware that the correlation between ctDNA levels and imaging findings is not always direct. For example, studies in melanoma have indicated that ctDNA detection rates at disease progression can sometimes be limited compared to PET imaging [83,85]. Despite such instances, the distinct yet complementary nature of information from ctDNA and imaging generally enhances the overall clinical picture, facilitating more informed patient management.

Challenges and Ethical Considerations

While circulating ctDNA testing holds considerable promise for revolutionizing the management of GU malignancies, its widespread and equitable integration into routine clinical practice is currently impeded by a confluence of significant scientific, technical, logistical, economic, and ethical challenges. For clinicians to confidently utilize ctDNA for applications such as early detection, treatment monitoring, or prognostication in prostate, bladder, renal, and testicular cancers, these multifaceted barriers must be systematically addressed. This section will delineate these critical challenges and the paramount ethical, legal, and social implications that require proactive consideration to ensure responsible innovation and safeguard patient well-being.

Scientific and Technical Hurdles

A primary scientific challenge lies in the inherent biology of ctDNA, particularly its often extremely low concentration in systemic circulation, especially in early-stage disease or settings of MRD [86]. This scarcity, sometimes amounting to only a few mutant molecules per blood draw, pushes the limits of current detection technologies. It can lead to tumors needing to reach a considerable size (1–3 cm) before ctDNA is reliably detectable, a point at which they may already be visible via conventional imaging [15,87].
Pre-analytical variability significantly impacts the reliability of ctDNA results. Factors such as the type of blood collection tube, sample processing times (e.g., plasma separation from EDTA tubes within six hours to prevent genomic DNA contamination from leukocytes), transport, storage conditions, and DNA isolation methods can all introduce inconsistencies that affect the quality and quantity of ctDNA recovered [88,89,90,91,92]. These logistical demands pose practical challenges for clinical laboratories.
Analytically, distinguishing true, low-frequency tumor-derived mutations from technical artifacts (e.g., PCR or sequencing errors) and biological noise is a major hurdle [86]. Cross-platform evaluations have revealed substantial variability in the performance of different ctDNA assays, particularly for variants below a 0.5% allele frequency, with false negatives (missed mutations) being more common than false positives [93]. A significant biological confounder is clonal hematopoiesis of indeterminate potential (CHIP), where somatic mutations in blood cells can mimic tumor-derived variants, potentially leading to false-positive results if not appropriately accounted for through matched leukocyte DNA sequencing [86,94,95]. Furthermore, while detection of point mutations is improving, the reliable identification of more complex but clinically actionable genomic alterations, such as copy number variations, structural rearrangements, and epigenetic modifications, in ctDNA remains a developing area [96,97,98].
The overarching lack of standardization across the entire ctDNA workflow—from pre-analytical procedures to analytical methodologies (including sequencing platforms and bioinformatic pipelines) and post-analytical interpretation and reporting—is a critical impediment [75,89,90,99]. External quality assessments have highlighted concerning error rates, emphasizing the urgent need for globally adopted standard operating procedures, rigorous quality control, and proficiency testing programs to ensure the analytical validity and reproducibility essential for clinical trust [100].

Clinical Validation and Regulatory Oversight

Beyond technical proficiency, the path to routine clinical use is contingent upon robust evidence of clinical validity (the test's ability to accurately identify a clinical condition) and clinical utility (the likelihood that the test will lead to improved patient outcomes) [101]. Currently, there is an insufficient evidence-based framework from large-scale, prospective clinical trials to definitively establish the clinical utility of ctDNA testing for many proposed applications in GU oncology. These applications could include routine early cancer detection or guiding adjuvant therapy based on MRD status [15,98,102,103]. This evidence gap makes it challenging to convince regulatory agencies to approve tests, payers to provide reimbursement, and clinicians to adopt them widely [101].
The interpretation of complex genomic data derived from ctDNA also poses a significant challenge for clinicians. Distinguishing driver mutations from passenger mutations or variants of uncertain significance (VUS), understanding the implications of CHIP, and interpreting clonal evolution patterns require specialized expertise [3,63,86,104,105,106]. Inconsistent reporting formats and a lack of standardized interpretation guidelines further complicate this [75,107].

Economic and Health System Implementation Barriers

The economic implications of ctDNA testing are substantial. High costs associated with sequencing, specialized equipment, data analysis, and expert interpretation can create significant financial burdens for healthcare systems and patients, potentially limiting access, especially in resource-constrained environments [108,109,110,111]. Insufficient insurance coverage, often stemming from the aforementioned lack of definitive clinical utility and cost-effectiveness data, remains a critical bottleneck [75,103,112,113]. Robust health economic models demonstrating improved outcomes and value are needed to support broader payer coverage [75].
Practical implementation within healthcare systems also faces hurdles, including the need for appropriate laboratory infrastructure, trained personnel, and seamless integration of genomic data into electronic health records (EHRs) with clinical decision support tools [114,115,116]. Limited provider knowledge regarding genomic medicine further complicates the adoption and appropriate use of ctDNA testing [111,116,117,118].

Ethical, Legal, and Social Implications (ELSI)

The increasing use of ctDNA testing brings to the forefront critical ELSI considerations. Patient privacy and the security of sensitive genomic data are paramount, necessitating robust data protection measures and compliance with regulations like HIPAA and GDPR [119]. The potential for genetic discrimination in employment or insurance based on ctDNA findings is a significant concern that requires strong legal safeguards [109,116,117].
The psychosocial impact on patients receiving complex or uncertain ctDNA results (e.g., VUS, incidental findings of CHIP or germline mutations) can be substantial, leading to anxiety and confusion [107,116,120]. Comprehensive informed consent processes that articulate the potential findings, limitations, and implications of testing are essential [121,122], alongside access to genetic counseling and psychological support [104].
Furthermore, as artificial intelligence and machine learning are increasingly used in ctDNA data analysis, issues of algorithmic fairness and bias must be addressed to prevent exacerbation of health disparities [119,123]. Finally, underdeveloped regulatory frameworks for these rapidly evolving technologies create uncertainty and necessitate proactive policy development to ensure responsible innovation and patient protection [3,4,109,110,124].
Addressing these multifaceted challenges requires a concerted, multidisciplinary effort involving researchers, clinicians, regulatory bodies, diagnostic developers, payers, and patient advocates. Establishing standardized protocols, conducting rigorous prospective validation studies, developing comprehensive educational programs, ensuring equitable access, and creating robust ethical and regulatory frameworks are crucial steps towards realizing the full clinical potential of ctDNA testing [125,126,127,128]. This will transform care for patients with genitourinary malignancies.

Future Perspectives

The journey of ctDNA from a promising research concept to a clinically impactful tool in GU oncology is rapidly advancing. While current applications are already demonstrating value, the full transformative potential of ctDNA is anticipated to be unlocked through a confluence of technological breakthroughs, sophisticated analytical approaches including artificial intelligence (AI) and machine learning (ML). This includes the development of more optimized and potentially malignancy-specific assays, and innovative clinical trial designs. These advancements are poised to overcome existing limitations in sensitivity, specificity, and clinical utility, thereby revolutionizing early cancer detection, personalizing treatment selection, enabling more dynamic response monitoring, and ultimately improving outcomes for patients with prostate, bladder, renal, and testicular cancers.

Anticipated Technological Breakthroughs and Assay Optimization

Future advancements in ctDNA analysis will likely focus on enhancing assay sensitivity and specificity, allowing for the detection of even more minute quantities of tumor-derived DNA. This is particularly true in early-stage disease and minimal residual disease (MRD) settings, where ctDNA levels are often exceedingly low [13]. Technologies are continuously improving to enable comprehensive genomic profiling from ctDNA, moving beyond single-nucleotide variants (SNVs) to reliably detect a wider array of alterations including copy-number aberrations (CNAs), complex structural variants, and tumor-specific genomic methylation patterns [66,129]. The analysis of ctDNA fragmentation patterns (fragmentomics) is also emerging as a powerful, non-mutation-based approach for cancer detection and characterization [80,130].
The development of novel assays targeting specific molecular hallmarks of GU cancers, such as unique methylation signatures or gene fusions, will further refine diagnostic and monitoring capabilities [66,68,131]. Moreover, the push towards lower-cost, decentralized, and potentially point-of-care ctDNA testing platforms could significantly improve accessibility and turnaround times, facilitating more widespread clinical integration [75,132].

The Role of Artificial Intelligence and Machine Learning

The sheer volume and complexity of data generated from ctDNA sequencing, especially when integrated with other biomarkers, necessitate advanced analytical tools. Artificial intelligence (AI) and machine learning (ML) algorithms are becoming indispensable for interpreting these large datasets [6]. AI/ML can enhance the detection of rare variants, improve the accuracy of ctDNA quantification, distinguish tumor-derived signals from background noise (including CHIP), and identify complex patterns in ctDNA fragmentation or methylation profiles that correlate with clinical outcomes [6,80,130]. For instance, ML models can integrate multiomic data from ctDNA, circulating tumor cells (CTCs), and exosomes to provide a more holistic and predictive assessment of tumor status [6].

Towards Optimized and Potentially Malignancy-Specific Assays

Given the biological diversity of GU malignancies and variations in ctDNA shedding characteristics [13], the future will likely see the development of ctDNA assays that are further optimized, and potentially tailored, for specific GU cancer types. Such assays would focus on genomic regions and alteration types most relevant to the biology and clinical behavior of prostate, bladder, renal, or testicular cancers, thereby maximizing sensitivity and clinical relevance. This could involve panels enriched for genes commonly mutated or epigenetically altered in these specific malignancies, leading to more precise diagnostic, prognostic, and predictive information.

Innovative Clinical Trial Designs and Validation Pathways

To firmly establish the clinical utility of ctDNA-guided interventions, innovative and robust clinical trial designs are essential. Future trials will increasingly incorporate ctDNA analysis as a primary or secondary endpoint for early assessment of treatment response, detection of MRD, and identification of resistance mechanisms, potentially accelerating drug development and approval pathways [72,133]. Adaptive trial designs, where treatment strategies are modified in real-time based on ctDNA dynamics, represent a promising avenue. Prospective, large-scale validation studies are crucial to demonstrate how ctDNA testing can lead to tangible improvements in patient outcomes, such as overall survival or quality of life, and to generate the evidence needed for guideline endorsement and payer coverage.

Transformative Impact Across the GU Cancer Care Continuum

These anticipated advancements are set to significantly enhance the role of ctDNA across the entire continuum of GU cancer care:
  • Earlier Detection: Improved assay sensitivity, coupled with the analysis of methylation patterns and fragmentomics, holds the promise of detecting GU cancers at earlier, even asymptomatic stages, potentially before they are identifiable by conventional imaging [40,130,134].
  • Personalized Treatment Selection: More comprehensive genomic profiling via ctDNA will enable more precise genotype-guided therapy selection, matching patients with the most effective targeted therapies or immunotherapies based on their tumor's unique molecular signature [40,64,135,136,137].
  • Dynamic and Real-Time Monitoring: Longitudinal ctDNA analysis will offer clinicians a non-invasive means to dynamically track tumor burden, assess treatment response more rapidly and sensitively than imaging alone, and monitor clonal evolution in real-time [69,71,72,138,139].
  • Refined MRD Assessment: Ultrasensitive ctDNA assays will improve the detection of MRD after curative-intent therapy, allowing for more accurate risk stratification and guidance of adjuvant treatment decisions [73,140,141,142,143].
  • Proactive Resistance Management: The ability to detect emerging resistance mutations in ctDNA, often months before clinical progression, will allow for proactive adjustments in treatment strategies, potentially circumventing or delaying overt resistance [73,140,141,142,143].
Yes, Dr. Chaux, I believe I have a comprehensive understanding of the preceding sections and the overall themes we've developed. I can now draft the "Conclusions" section for your review article. This will synthesize the key points discussed, reiterate the significance of ctDNA in genitourinary oncology, acknowledge the challenges, and offer a forward-looking perspective, all tailored for clinicians and practitioners and based on the information and sources we've utilized.

Conclusions

ctDNA analysis has unequivocally emerged as a groundbreaking innovation in oncology, with profound implications for the diagnosis, management, and monitoring of GU malignancies. This review has underscored the expanding clinical utility of ctDNA across prostate, urothelial, renal, and testicular cancers, demonstrating its potential to provide real-time, personalized molecular insights that can complement and, in some instances, enhance traditional diagnostic and monitoring paradigms. From facilitating earlier detection and more precise risk stratification to enabling dynamic treatment response assessment, identification of MRD, and the timely recognition of resistance mechanisms, ctDNA-based liquid biopsies are progressively informing critical clinical decision points [15,35,36,37].
The capacity of ctDNA to reflect not only single-gene mutations but also broader genomic landscapes—including TMB, MSI, and epigenetic alterations like methylation patterns—further amplifies its clinical value [64,66]. Moreover, the integration of ctDNA data with other liquid biopsy analytes (e.g., circulating tumor cells, exosomes) and conventional imaging modalities promises a more holistic and nuanced understanding of tumor biology and patient status [6,75,82,83].
Despite this considerable progress, the path to widespread, equitable, and routine clinical implementation of ctDNA testing in GU oncology is paved with significant challenges. Technical hurdles, including the detection of ultra-low ctDNA concentrations, pre-analytical variability, and the need for robust bioinformatics, remain [49,86,90]. The critical lack of standardization across assay methodologies and reporting practices hinders inter-laboratory comparability and broader clinical adoption [75,100]. Furthermore, rigorous prospective clinical validation is essential to definitively establish the clinical utility of ctDNA-guided interventions in improving patient outcomes[98,101]. Economic considerations, including test costs and reimbursement frameworks, also pose substantial barriers [75,108,112]. Critically, the ELSI—encompassing patient privacy, data security, potential for discrimination, informed consent, and the psychosocial impact of complex genomic results—must be proactively addressed to ensure responsible innovation and patient protection [101,107,116,119].
Looking ahead, the continued evolution of ctDNA technology, including anticipated breakthroughs in assay sensitivity and the development of malignancy-specific panels, holds immense promise. The integration of AI and ML will be pivotal for analyzing complex ctDNA datasets and extracting clinically actionable insights [6,80]. Innovative clinical trial designs are crucial for validating ctDNA as a surrogate endpoint and for demonstrating its impact on patient care pathways.
The transformative potential of ctDNA in GU cancer care is undeniable. As these technologies mature and current limitations are systematically overcome through collaborative research, standardization efforts, and thoughtful policy development, ctDNA analysis is poised to become an indispensable component of precision oncology. Its integration into routine clinical practice will likely lead to earlier diagnoses, more tailored therapeutic strategies, enhanced monitoring capabilities, and ultimately, improved survival and quality of life for patients with genitourinary malignancies. The continued commitment of the scientific and clinical community will be paramount in realizing this future.

References

  1. Palve S, Bhardwaj Y, Dusane J, Wankhade P. Blood-borne biomarkers: CT-DNA ushers in a new era of cancer detection. Int J Pharm Sci Drug Res.
  2. Russano M, Napolitano A, Ribelli G, Iuliani M, Simonetti S, Citarella F, et al. Liquid biopsy and tumor heterogeneity in metastatic solid tumors: the potentiality of blood samples. J Exp Clin Cancer Res. 2020, 39, 95. [Google Scholar] [CrossRef] [PubMed]
  3. Turabi K, Klute K, Radhakrishnan P. Decoding the dynamics of circulating tumor DNA in liquid biopsies. Cancers (Basel). 2024, 16, 2432. [Google Scholar] [CrossRef] [PubMed]
  4. Zhao Y, Zhang Z, Qiu JH, Li RY, Sun ZG. Catching cancer signals in the blood: Innovative pathways for early esophageal cancer diagnosis. World J Gastroenterol. 2025, 31, 101838. [Google Scholar]
  5. Li M, Xie S, Lu C, Zhu L, Zhu L. Application of data science in circulating tumor DNA detection: A promising avenue towards liquid biopsy. Front Oncol. 2021, 11, 692322. [Google Scholar] [CrossRef]
  6. Moon GY, Dalkiran B, Park HS, Shin D, Son C, Choi JH, et al. Dual biomarker strategies for liquid biopsy: Integrating circulating tumor cells and circulating tumor DNA for enhanced tumor monitoring. Biosensors (Basel). 2025, 15, 74. [Google Scholar]
  7. Yi X, Ma J, Guan Y, Chen R, Yang L, Xia X. The feasibility of using mutation detection in ctDNA to assess tumor dynamics. Int J Cancer. 2017, 140, 2642–2647. [Google Scholar] [CrossRef]
  8. Lin C, Liu X, Zheng B, Ke R, Tzeng CM. Liquid biopsy, ctDNA diagnosis through NGS. Life (Basel). 2021, 11, 890. [Google Scholar]
  9. Ji H, Long X, Gu J, Jin J, Mao X, Wang Z, et al. Longitudinal monitoring of plasma circulating tumour DNA enables the prediction of early relapse in patients with non-Hodgkin lymphoma: A case series. Diagnostics (Basel). 2021, 11, 2055. [Google Scholar] [CrossRef] [PubMed]
  10. Mair R, Mouliere F, Smith CG, Chandrananda D, Gale D, Marass F, et al. Measurement of plasma cell-free mitochondrial tumor DNA improves detection of glioblastoma in patient-derived orthotopic xenograft models. Cancer Res. 2019, 79, 220–230. [Google Scholar] [CrossRef]
  11. Underhill, HR. Leveraging the fragment length of circulating tumour DNA to improve molecular profiling of solid tumour malignancies with next-generation sequencing: A pathway to advanced non-invasive diagnostics in precision oncology? Mol Diagn Ther. 2021, 25, 389–408. [Google Scholar] [CrossRef]
  12. Wang Y, Wang Z, Gang X, Wang G. Liquid biopsy in prostate cancer: current status and future challenges of clinical application. Aging Male. 2021, 24, 58–71. [Google Scholar] [CrossRef] [PubMed]
  13. Bettegowda C, Sausen M, Leary R, Kinde I, Agrawal N, Bartlett B, et al. Abstract 5606, Detection of circulating tumor DNA in early and late stage human malignancies. Cancer Res. 2014, 74 (19_Supplement), 5606–5606. [CrossRef]
  14. Diaz LA Jr, Bardelli A. Liquid biopsies: genotyping circulating tumor DNA. J Clin Oncol. 2014, 32, 579–586. [Google Scholar] [CrossRef]
  15. Gerke MB, Jansen CS, Bilen MA. Circulating tumor DNA in genitourinary cancers: Detection, prognostics, and therapeutic implications. Cancers (Basel). 2024, 16, 2280. [Google Scholar] [CrossRef]
  16. Wang H, Zhang Y, Zhang H, Cao H, Mao J, Chen X, et al. Liquid biopsy for human cancer: cancer screening, monitoring, and treatment. MedComm. 2024, 5, e564. [Google Scholar] [CrossRef]
  17. Dawson SJ, Tsui DWY, Murtaza M, Biggs H, Rueda OM, Chin SF, et al. Analysis of circulating tumor DNA to monitor metastatic breast cancer. N Engl J Med. 2013, 368, 1199–1209. [Google Scholar] [CrossRef]
  18. Stejskal P, Goodarzi H, Srovnal J, Hajdúch M, van ’t Veer LJ, Magbanua MJM. Circulating tumor nucleic acids: biology, release mechanisms, and clinical relevance. Mol Cancer. 2023, 22, 15. [Google Scholar] [CrossRef] [PubMed]
  19. Warton K, Mahon KL, Samimi G. Methylated circulating tumor DNA in blood: power in cancer prognosis and response. Endocr Relat Cancer. 2016, 23, R157–71. [Google Scholar] [CrossRef]
  20. Cheng F, Su L, Qian C. Circulating tumor DNA: a promising biomarker in the liquid biopsy of cancer. Oncotarget. 2016, 7, 48832–48841. [Google Scholar] [CrossRef]
  21. Meriranta L, Alkodsi A, Pasanen A, Lepistö M, Mapar P, Blaker YN, et al. Molecular features encoded in the ctDNA reveal heterogeneity and predict outcome in high-risk aggressive B-cell lymphoma. Blood. 2022, 139, 1863–1877. [Google Scholar] [CrossRef]
  22. Danesi R, Lo YMD, Oellerich M, Beck J, Galbiati S, Re MD, et al. What do we need to obtain high quality circulating tumor DNA (ctDNA) for routine diagnostic test in oncology? - Considerations on pre-analytical aspects by the IFCC workgroup cfDNA. Clin Chim Acta. 2021, 520, 168–171. [Google Scholar] [CrossRef] [PubMed]
  23. Sherwood JL, Corcoran C, Brown H, Sharpe AD, Musilova M, Kohlmann A. Optimised pre-analytical methods improve KRAS mutation detection in circulating tumour DNA (ctDNA) from patients with non-small cell lung cancer (NSCLC). PLoS One. 2016, 11, e0150197. [Google Scholar]
  24. Hollanda CN, Gualberto ACM, Motoyama AB, Pittella-Silva F. Advancing leukemia management through liquid biopsy: Insights into biomarkers and clinical utility. Cancers (Basel) [Internet]. 2025, 17(9). Available from: https://www.mdpi.com/2072-6694/17/9/1438.
  25. Meddeb R, Dache ZAA, Thezenas S, Otandault A, Tanos R, Pastor B, et al. Quantifying circulating cell-free DNA in humans. Sci Rep. 2019, 9, 5220. [Google Scholar] [CrossRef] [PubMed]
  26. Asante DB, Tierno D, Grassi G, Scaggiante B. Circulating tumour DNA for ovarian cancer diagnosis and treatment monitoring: What perspectives for clinical use? Int J Mol Sci [Internet]. 2025 Feb 22 [cited 2025 May 22];26(5). Available from: https://www.mdpi.com/1422-0067/26/5/1889.
  27. van Ginkel JH, Huibers MMH, van Es RJJ, de Bree R, Willems SM. Droplet digital PCR for detection and quantification of circulating tumor DNA in plasma of head and neck cancer patients. BMC Cancer. 2017, 17, 428. [Google Scholar]
  28. Aiyer S, Kim TH, Collier K, Pollock R, Verschraegen C, Stover DG, et al. Unlocking the potential of ctDNA in sarcomas: A review of recent advances. Cancers (Basel). 2025, 17, 1040. [Google Scholar] [CrossRef]
  29. Yin H, Zhang M, Zhang Y, Zhang X, Zhang X, Zhang B. Liquid biopsies in cancer. Liquid biopsies in cancer. Mol Biomed. 2025.
  30. Bohers E, Viailly PJ, Jardin F. CfDNA sequencing: Technological approaches and bioinformatic issues. Pharmaceuticals (Basel). 2021, 14, 596. [Google Scholar] [CrossRef]
  31. Deveson IW, Gong B, Lai K, LoCoco JS, Richmond TA, Schageman J, et al. Evaluating the analytical validity of circulating tumor DNA sequencing assays for precision oncology. Nat Biotechnol. 2021, 39, 1115–1128. [Google Scholar] [CrossRef]
  32. Gao Y, Zhao H, An K, Liu Z, Hai L, Li R, et al. Whole-genome bisulfite sequencing analysis of circulating tumour DNA for the detection and molecular classification of cancer. Clin Transl Med. 2022, 12, e1014. [Google Scholar] [CrossRef]
  33. Merker JD, Oxnard GR, Compton C, Diehn M, Hurley P, Lazar AJ, et al. Circulating tumor DNA analysis in patients with cancer: American society of clinical oncology and College of American Pathologists joint review. J Clin Oncol. 2018, 36, 1631–1641. [Google Scholar] [CrossRef] [PubMed]
  34. Nakamura Y, Shitara K. Development of circulating tumour DNA analysis for gastrointestinal cancers. ESMO Open. 2020, 5 (Suppl 1), e000600. [CrossRef]
  35. Eibl RH, Schneemann M. Cell-free DNA as a biomarker in cancer. Extracell Vesicles Circ Nucl Acids. 2022, 3, 195–215. [Google Scholar]
  36. Lee JS, Cho EH, Kim B, Hong J, Kim YG, Kim Y, et al. Clinical practice guideline for blood-based circulating tumor DNA assays. Ann Lab Med. 2024, 44, 195–209. [Google Scholar] [CrossRef]
  37. Sorbini M, Carradori T, Togliatto GM, Vaisitti T, Deaglio S. Technical advances in circulating cell-free DNA detection and analysis for personalized medicine in patients’ care. Biomolecules [Internet]. 2024, 14(4). Available from: https://www.mdpi.com/2218-273X/14/4/498.
  38. Wang Z, Wu Q. Advancements in non-invasive diagnosis of gastric cancer. World J Gastroenterol. 2025.
  39. Conteduca V, Wetterskog D, Sharabiani M, Grande E, Fernández-Pérez M, Jayaram A, et al. Androgen receptor gene status in plasma DNA associates with worse outcome on enzalutamide or abiraterone for castration-resistant prostate cancer: a multi-institution correlative biomarker study. Ann Oncol. 2017.
  40. Hasenleithner SO, Speicher M. A clinician’s handbook for using ctDNA throughout the patient journey. Mol Cancer [Internet]. 2022, 21. Available from: https://molecular-cancer.biomedcentral.com/articles/10.1186/s12943-022-01551-7.
  41. Annala M, Vandekerkhove G, Khalaf D, Taavitsainen S, Beja K, Warner E, et al. Circulating tumor DNA genomics correlate with resistance to abiraterone and enzalutamide in prostate cancer. Cancer Discov. 2018, 8, 444–457. [CrossRef]
  42. Jänne P, Oxnard G, Thress K, Alden R, Lawrance R. Association between plasma genotyping and outcomes of treatment with osimertinib (AZD9291) in advanced non-small-cell lung cancer. J Clin Oncol. 2016, 34, 3375–3382.
  43. Belic J, Graf R, Bauernhofer T, Cherkas Y, Ulz P, Waldispuehl-Geigl J, et al. Genomic alterations in plasma DNA from patients with metastasized prostate cancer receiving abiraterone or enzalutamide. Int J Cancer. 2018, 143, 1236–1248. [Google Scholar] [CrossRef]
  44. Christensen E, Birkenkamp-Demtröder K, Sethi H, Shchegrova S, Salari R, Nordentoft I, et al. Early detection of metastatic relapse and monitoring of therapeutic efficacy by ultra-deep sequencing of plasma cell-free DNA in patients with urothelial bladder carcinoma. J Clin Oncol. 2019, 37, 1547–1557. [Google Scholar] [CrossRef]
  45. Li S, Xin K, Pan S, Wang Y, Zheng J, Li Z, et al. Blood-based liquid biopsy: insights into early detection, prediction, and treatment monitoring of bladder cancer. Cell Mol Biol Lett. 2023, 28, 28. [Google Scholar]
  46. Patel KR, Rais-Bahrami S, Basu A. High sensitivity ctDNA assays in genitourinary malignancies: current evidence and future directions. Oncologist. 2024, 29, 731–737. [Google Scholar] [CrossRef] [PubMed]
  47. Vandekerkhove G, Todenhöfer T, Annala M, Struss W, Wong A, Beja K, et al. Circulating tumor DNA reveals clinically actionable somatic genome of metastatic bladder cancer. Clin Cancer Res. 2017, 23, 6487–6497. [Google Scholar] [CrossRef] [PubMed]
  48. Sabit H, Attia MG, Mohamed N, Taha PS, Ahmed N, Osama S, et al. Beyond traditional biopsies: the emerging role of ctDNA and MRD on breast cancer diagnosis and treatment. Discov Oncol. 2025, 16, 271. [Google Scholar] [CrossRef]
  49. kimi AA. Circulating tumor DNA (ctDNA) in kidney cancer: A narrative review. Société Int d’urologie J [Internet]. 2023, Available from: https://www.mdpi.com/2563-6499/4/4/287.
  50. Duffy MJ, Crown J. Circulating tumor DNA as a biomarker for monitoring patients with solid cancers: Comparison with standard protein biomarkers. Clin Chem. 2022, 68, 1381–1390. [Google Scholar] [CrossRef]
  51. Moding EJ, Nabet BY, Alizadeh AA, Diehn M. Detecting liquid remnants of solid tumors: Circulating tumor DNA minimal residual disease. Cancer Discov. 2021, 11, 2968–2986. [Google Scholar] [CrossRef]
  52. Zollinger DR, Rivers E, Fine A, Huang Y, Son J, Kalyan A, et al. Analytical validation of a novel comprehensive genomic profiling informed circulating tumor DNA monitoring assay for solid tumors. PLoS One. 2024, 19, e0302129. [Google Scholar]
  53. Tie J, Cohen JD, Wang Y, Christie M, Simons K, Lee M, et al. Circulating tumor DNA analyses as markers of recurrence risk and benefit of adjuvant therapy for stage III colon cancer. JAMA Oncol. 2019, 5, 1710–1717. [Google Scholar] [CrossRef]
  54. Cheng ML, Pectasides E, Hanna GJ, Parsons HA, Choudhury AD, Oxnard GR. Circulating tumor DNA in advanced solid tumors: Clinical relevance and future directions. CA Cancer J Clin. 2021, 71, 176–190. [Google Scholar] [CrossRef]
  55. Fernandez SV, Tan YF, Rao S, Fittipaldi P, Sheriff F, Borghaei H, et al. Validation of a molecular diagnostic test for circulating tumor DNA by next-gen sequencing. Int J Mol Sci [Internet]. 2023, 24. Available from: https://www.mdpi.com/1422-0067/24/21/15779.
  56. Magbanua MJM, Brown-Swigart L, Wu HT, Hirst GL, Yau C, Wolf DM, et al. Circulating tumor DNA in neoadjuvant treated breast cancer reflects response and survival [Internet]. bioRxiv. medRxiv; 2020. Available from: http://medrxiv.org/lookup/doi/10.1101/2020.02.03.20019760.
  57. Kansara M, Bhardwaj N, Thavaneswaran S, Xu C, Lee JK, Chang LB, et al. Early circulating tumor DNA dynamics as a pan-tumor biomarker for long-term clinical outcome in patients treated with durvalumab and tremelimumab. Mol Oncol. 2022, 17, 298–311. [Google Scholar]
  58. Vega DM, Nishimura KK, Zariffa N, Thompson JC, Hoering A, Cilento V, et al. Changes in circulating tumor DNA reflect clinical benefit across multiple studies of patients with non–small-cell lung cancer treated with immune checkpoint inhibitors. JCO Precis Oncol [Internet]. 2022, 6. Available from: https://ascopubs.org/doi/10.1200/PO.21.00372.
  59. Zhang Q, Luo J, Wu S, Si H, Gao C, Xu W, et al. Prognostic and predictive impact of circulating tumor DNA in patients with advanced cancers treated with immune checkpoint blockade. Cancer Discov. 2020, 10, 1842–1853. [Google Scholar] [CrossRef]
  60. Chaudhuri AA, Chabon JJ, Lovejoy AF, Newman AM, Stehr H, Azad TD, et al. Early detection of molecular residual disease in localized lung cancer by circulating tumor DNA profiling. Cancer Discov. 2017, 7, 1394–1403. [Google Scholar] [CrossRef]
  61. Ma F, Zhu W, Guan Y, Yang L, Xia X, Chen S, et al. ctDNA dynamics: a novel indicator to track resistance in metastatic breast cancer treated with anti-HER2 therapy. Oncotarget. 2016, 7, 66020–66031. [Google Scholar] [CrossRef]
  62. Tie J, Kinde I, Wang Y, Wong H, Roebert J, Christie M, et al. Circulating tumor DNA as an early marker of therapeutic response in patients with metastatic colorectal cancer. Circulating tumor DNA as an early marker of therapeutic response in patients with metastatic colorectal cancer. Ann Oncol. 2015.
  63. Siravegna G, Mussolin B, Venesio T, Marsoni S, Seoane J, Dive C, et al. How liquid biopsies can change clinical practice in oncology. Ann Oncol. 2019, 30, 1580–1590. [Google Scholar] [CrossRef]
  64. Sivapalan L, Murray J, Canzoniero JV, Landon BV, Jackson J, Scott SL, et al. Liquid biopsy approaches to capture tumor evolution and clinical outcomes during cancer immunotherapy. J Immunother Cancer [Internet]. 2023, 11. Available from: https://jitc.bmj.com/lookup/doi/10.1136/jitc-2022-005924.
  65. Li L, Sun Y. Circulating tumor DNA methylation detection as biomarker and its application in tumor liquid biopsy: advances and challenges. MedComm. 2024, 5, e766. [Google Scholar] [CrossRef] [PubMed]
  66. Walz AL, Maschietto M, Crompton B, Evageliou N, Dix D, Tytgat G, et al. Tumor biology, biomarkers, and liquid biopsy in pediatric renal tumors. Pediatr Blood Cancer [Internet]. 2023, 70. Available from: https://onlinelibrary.wiley.com/doi/10.1002/pbc.30130.
  67. Charlton J, Williams RD, Weeks M, Sebire NJ, Popov S, Vujanic G, et al. Methylome analysis identifies a Wilms tumor epigenetic biomarker detectable in blood. Genome Biol. 2014, 15, 434. [Google Scholar]
  68. van Zogchel LMJ, Lak N, Verhagen O, Tissoudali A, Nuru MG, Gelineau NU, et al. Novel circulating hypermethylated RASSF1A ddPCR for liquid biopsies in patients with pediatric solid tumors. JCO Precis Oncol [Internet]. 2021, 5. Available from: https://ascopubs.org/doi/10.1200/PO.21.00130.
  69. Papalexis P, Georgakopoulou V, Drossos P, Thymara E, Nonni A, Lazaris AC, et al. Precision medicine in breast cancer (Review). Mol Clin Oncol [Internet]. 2024, 21. Available from: http://www.spandidos-publications.com/10.3892/mco.2024.2776.
  70. Madanat-Harjuoja L, Renfro L, Klega K, Tornwall B, Thorner A, Nag A, et al. Circulating tumor DNA as a biomarker in patients with stage III and IV Wilms tumor: Analysis from a Children’s Oncology Group trial, AREN0533. J Clin Oncol. 2022, 40, 3047–3056.
  71. Mattos-Arruda LD, Mattos-Arruda LD, Mattos-Arruda LD, Weigelt B, Cortés J, Won H, et al. Capturing intra-tumor genetic heterogeneity by de novo mutation profiling of circulating cell-free tumor DNA: a proof-of-principle. Ann Oncol. 2014, 25, 1729–1735. [Google Scholar] [CrossRef] [PubMed]
  72. Yeh P, Hunter T, Sinha D, Ftouni S, Wallach E, Jiang DHS, et al. Circulating tumour DNA reflects treatment response and clonal evolution in chronic lymphocytic leukaemia. Nat Commun [Internet]. 2017, 8. Available from: https://www.nature.com/articles/ncomms14756.
  73. Chabon J, Simmons A, Lovejoy A, Esfahani MS, Newman AM, Haringsma HJ, et al. Circulating tumour DNA profiling reveals heterogeneity of EGFR inhibitor resistance mechanisms in lung cancer patients. Nat Commun. 2016, 7, 11815. [Google Scholar] [CrossRef]
  74. Corcoran R, Chabner B. Application of Cell-free DNA Analysis to Cancer Treatment. N Engl J Med. 2018.
  75. Bamodu OA, Chung CC, Pisanic TR 2nd. Harnessing liquid biopsies: Exosomes and ctDNA as minimally invasive biomarkers for precision cancer medicine. J Liq Biopsy. 2023, 2, 100126. [Google Scholar]
  76. Hench IB, Hench J, Tolnay M. Liquid biopsy in clinical management of breast, lung, and colorectal cancer. Front Med (Lausanne) [Internet]. 2018, 5. Available from: http://journal.frontiersin.org/article/10.3389/fmed.2018.00009/full.
  77. Carrasco R, Ingelmo-Torres M, Trullas R, Roldán F, Rodriguez-Carunchio L, Juez L, et al. Tumor-agnostic circulating tumor DNA testing for monitoring muscle-invasive bladder cancer. Int J Mol Sci. 2023, 24, 16578. [Google Scholar] [CrossRef]
  78. Zhou Y, Wang R, Zeng M, Liu S. Circulating tumor DNA: a revolutionary approach for early detection and personalized treatment of bladder cancer. Front Pharmacol [Internet]. 2025, 16. Available from: https://www.frontiersin.org/articles/10.3389/fphar.2025.1551219/full.
  79. Onidani K, Shoji H, Kakizaki T, Yoshimoto S, Okaya S, Miura N, et al. Monitoring of cancer patients via next-generation sequencing of patient-derived circulating tumor cells and tumor DNA. Cancer Sci. 2019, 110, 2590–2599. [Google Scholar] [CrossRef] [PubMed]
  80. Peneder P, Stütz A, Surdez D, Krumbholz M, Semper S, Chicard M, et al. Multimodal analysis of cell-free DNA whole-genome sequencing for pediatric cancers with low mutational burden. Nat Commun [Internet]. 2021, 12. Available from: https://www.nature.com/articles/s41467-021-23445-w.
  81. Xie J, Hu B, Gong Y, He S, Lin J, Huang Q, et al. A comparative study on ctDNA and tumor DNA mutations in lung cancer and benign cases with a high number of CTCs and CTECs. J Transl Med [Internet]. 2023, 21. Available from: https://translational-medicine.biomedcentral.com/articles/10.1186/s12967-023-04746-8.
  82. Khanyile R, Chipiti T, Hull R, Dlamini Z. Radiogenomic landscape of metastatic endocrine-positive breast cancer resistant to aromatase inhibitors. Cancers (Basel) [Internet]. 2025, 17. Available from: https://www.mdpi.com/2072-6694/17/5/808.
  83. Mishra M, Ahmed R, Das DK, Pramanik DD, Dash SK, Pramanik A. Recent advancements in the application of circulating tumor DNA as biomarkers for early detection of cancers. ACS Biomater Sci Eng. 2024, 10, 4740–4756. [Google Scholar] [CrossRef] [PubMed]
  84. García-Pardo M, Makarem M, Li JJ, Kelly D, Leighl N. Integrating circulating-free DNA (cfDNA) analysis into clinical practice: opportunities and challenges. Br J Cancer. 2022, 127, 592–602. [Google Scholar] [CrossRef]
  85. Marsavela G, McEvoy A, Pereira M, Reid A, Al-Ogaili Z, Warburton L, et al. Detection of clinical progression through plasma ctDNA in metastatic melanoma patients: a comparison to radiological progression. Br J Cancer. 2021, 126, 401–408. [Google Scholar]
  86. Godsey JH, Silvestro A, Barrett J, Bramlett K, Chudova DI, Deras I, et al. Generic protocols for the analytical validation of Next-Generation Sequencing-based ctDNA assays: A joint consensus recommendation of the BloodPAC’s Analytical Variables Working Group. Clin Chem. 2020, 66, 1156–1166. [Google Scholar] [CrossRef]
  87. Fiala C, Diamandis E. Utility of circulating tumor DNA in cancer diagnostics with emphasis on early detection. BMC Med [Internet]. 2018, 16. Available from: https://bmcmedicine.biomedcentral.com/articles/10.1186/s12916-018-1157-9.
  88. Sotoudeh Anvari M, Gharib A, Abolhasani M, Azari-yam A, Gharalari FH, Safavi M, et al. Pre-analytical practices in the molecular diagnostic tests, A concise review. Iran J Pathol. 2020, 16, 1–19. [Google Scholar]
  89. Haselmann V, Hedtke M, Neumaier M. Liquid profiling for cancer patient stratification in precision medicine—current status and challenges for successful implementation in standard care. Diagnostics (Basel) [Internet]. 2022, 12. Available from: https://www.mdpi.com/2075-4418/12/3/748.
  90. Horgan D, Čufer T, Gatto F, Lugowska I, Verbanac D, Carvalho Â, et al. Accelerating the development and validation of liquid biopsy for early cancer screening and treatment tailoring. Healthcare [Internet]. 2022, 10. Available from: https://www.mdpi.com/2227-9032/10/9/1714.
  91. Hum M, Lee ASG. DNA methylation in breast cancer: early detection and biomarker discovery through current and emerging approaches. J Transl Med. 2025, 23, 465. [Google Scholar] [CrossRef] [PubMed]
  92. Meddeb R, Pisareva E, Thierry A. Guidelines for the preanalytical conditions for analyzing circulating cell-free DNA. Clin Chem. 2019, 65, 623–633. [Google Scholar] [CrossRef]
  93. Stetson DB, Ahmed A, Xu X, Nuttall B, Lubinski TJ, Johnson JH, et al. Orthogonal comparison of four plasma NGS tests with tumor suggests technical factors are a major source of assay discordance. JCO Precis Oncol. 2019, 3, 1–9. [Google Scholar]
  94. Huang Z, Fu Y, Yang H, Zhou Y, Shi M, Li Q, et al. Liquid biopsy in T-cell lymphoma: biomarker detection techniques and clinical application. Mol Cancer [Internet]. 2024, 23. Available from: https://molecular-cancer.biomedcentral.com/articles/10.1186/s12943-024-01947-7.
  95. Razavi P, Li BT, Brown DN, Jung B, Hubbell E, Shen R, et al. High-intensity sequencing reveals the sources of plasma circulating cell-free DNA variants. News@nat,Com. 2019, 25, 1928–1937.
  96. Diaz L, Sausen M, Fisher G, Velculescu V. Insights into therapeutic resistance from whole-genome analyses of circulating tumor DNA. Oncotarget. 2013, 4, 1856–1857. [Google Scholar] [CrossRef] [PubMed]
  97. Leary R, Sausen M, Diaz L, Velculescu V. Cancer detection using whole-genome sequencing of cell free DNA. Oncotarget. 2013, 4, 1119–1120. [Google Scholar] [CrossRef] [PubMed]
  98. Sausen M, Parpart S, Diaz L. Circulating tumor DNA moves further into the spotlight. Genome Med. 2014, 6, 35–35. [Google Scholar] [CrossRef]
  99. Ho HY, Chung KSK, Kan CM, Wong SCC. Liquid biopsy in the clinical management of cancers. Int J Mol Sci. 2024, 25, 8594. [Google Scholar] [CrossRef] [PubMed]
  100. Keppens C, Dequeker E, Patton S, Normanno N, Fenizia F, Butler R, et al. International pilot external quality assessment scheme for analysis and reporting of circulating tumour DNA. BMC Cancer [Internet]. 2018, 18. Available from: https://bmccancer.biomedcentral.com/articles/10.1186/s12885-018-4694-x.
  101. Dong L, Wang W, Li A, Kansal R, Chen Y, Chen H, et al. Clinical next generation sequencing for precision medicine in cancer. Curr Genomics. 2015, 16, 253–263. [Google Scholar] [CrossRef]
  102. Liu M, Mu T, Gu J, Xu M, Chen S. The method of minimal residual disease detection with circulating tumor DNA and its clinical applications in colorectal cancer. Cancer Rep. 2025, 8, e70167. [Google Scholar] [CrossRef]
  103. Zhang H, Wang L, Wu H. Liquid biopsy in ovarian cancer in China and the world: current status and future perspectives. Front Oncol [Internet]. 2023, 13. Available from: https://www.frontiersin.org/articles/10.3389/fonc.2023.1276085/full.
  104. Desai A, Vázquez TA, Arce KM, Corassa M, Mack PC, Gray JE, et al. CtDNA for the evaluation and management of EGFR-mutant non-small cell lung cancer. Cancers (Basel) [Internet]. 2024, 16. Available from: https://www.mdpi.com/2072-6694/16/5/940.
  105. Hasenleithner SO, Heitzer E. Liquid profiling for patients with advanced cancer is ready for clinical integration. Liquid profiling for patients with advanced cancer is ready for clinical integration. Memo. 2024.
  106. Hu Y, Ulrich B, Supplee JG, Kuang Y, Lizotte PH, Feeney N, et al. False-positive plasma genotyping due to clonal hematopoiesis. Clin Cancer Res. 2018, 24, 4437–4443. [Google Scholar] [CrossRef]
  107. Sheriff S, Saba M, Patel R, Fisher G, Schroeder T, Arnolda G, et al. A scoping review of factors influencing the implementation of liquid biopsy for cancer care. J Exp Clin Cancer Res [Internet]. 2025, 44. Available from: https://jeccr.biomedcentral.com/articles/10.1186/s13046-025-03322-w.
  108. Adhit KK, Wanjari A, Menon S, K S. Liquid Biopsy: An Evolving Paradigm for Non-invasive Disease Diagnosis and Monitoring in Medicine. Cureus. 2023.
  109. Alqahtani S, Alqahtani T, Venkatesan K, Sivadasan D, Ahmed R, Elfadil H, et al. Unveiling Pharmacogenomics Insights into Circular RNAs: Toward Precision Medicine in Cancer Therapy. Biomolecules. 2025.
  110. Ashif M, Akram R. Revolutionizing cancer care: Liquid biopsy redefining surgical biopsy paradigms. Int j multidiscip res [Internet]. 2024, Available from: https://www.ijfmr.com/research-paper.php?id=13089.
  111. Wang Y, Shao W, Li H, Zhao P, Tian L, Zhang L, et al. Clinical application of minimal residual disease detection by ctDNA testing in non-small cell lung cancer: a narrative review. Transl Lung Cancer Res. 2025, 14, 1007–1020. [Google Scholar] [CrossRef] [PubMed]
  112. Trosman J, Weldon C, Kurian A, Pasquinelli M, Kircher S, Martin N, et al. Perspectives of private payers on multicancer early-detection tests: informing research, implementation, and policy. Health Affairs Scholar [Internet]. 2023, 1. Available from: https://academic.oup.com/healthaffairsscholar/article/doi/10.1093/haschl/qxad005/7203674.
  113. Wu S, Thawani R. Tumor-agnostic therapies in practice: Challenges, innovations, and future perspectives. Cancers (Basel) [Internet]. 2025, 17. Available from: https://www.mdpi.com/2072-6694/17/5/801.
  114. Conran CA, Brendler C, Xu J. Personalized prostate cancer care: from screening to treatment. Asian J Androl. 2016, 18, 505–508. [Google Scholar] [CrossRef]
  115. Houwink E, Sollie A, Numans M, Cornel M. Proposed roadmap to stepwise integration of genetics in family medicine and clinical research. Clin Transl Med. 2013, 2, 5–5. [Google Scholar] [CrossRef]
  116. Tommel J, Kenis D, Lambrechts N, Brohet R, Swysen J, Mollen L, et al. Personal genomes in practice: Exploring citizen and healthcare professionals’ perspectives on personalized genomic medicine and personal health data spaces using a mixed-methods design. Genes (Basel) [Internet]. 2023, 14. Available from: https://www.mdpi.com/2073-4425/14/4/786.
  117. Houwink E, van Luijk SV, Henneman L, van der Vleuten CVD, Dinant GJ, Cornel M. Genetic educational needs and the role of genetics in primary care: a focus group study with multiple perspectives. BMC Fam Pract. 2011, 12, 5–5. [Google Scholar] [CrossRef]
  118. Pasic M, Samaan S, Yousef G. Genomic medicine: new frontiers and new challenges. Clin Chem. 2013, 59, 158–167. [CrossRef] [PubMed]
  119. An L, Liu Y, Liu Y. Advancements in Circulating Tumor Cell Detection for Early Cancer Diagnosis: An Integration of Machine Learning Algorithms with Microfluidic Technologies. Biosensors. 2025.
  120. Wang Y, Feng W. Cancer-related psychosocial challenges. Gen Psychiatr [Internet]. 2022, 35. Available from: https://gpsych.bmj.com/lookup/doi/10.1136/gpsych-2022-100871.
  121. Callier SL, Abudu R, Mehlman M, Singer M, Neuhauser D, Anan CC, et al. Ethical, legal, and social implications of personalized genomic medicine research: Current literature and suggestions for the future. Z-Medicine eJournal [Internet]. 2016, Available from: https://onlinelibrary.wiley.com/doi/10.1111/bioe.12285.
  122. Lemke A, Wolf WA, Hebert-Beirne J, Smith ME. Public and biobank participant attitudes toward genetic research participation and data sharing. Public Health Genomics. 2010, 13, 368–377. [Google Scholar] [CrossRef]
  123. Chua IS, Gaziel-Yablowitz M, Korach Z, Kehl K, Levitan N, Arriaga Y, et al. Artificial intelligence in oncology: Path to implementation. Cancer Med. 2021, 10, 4138–4149. [Google Scholar] [CrossRef] [PubMed]
  124. Pando A, Trivedi R, Pauwels J, Nowakowska J, Cavina B, Falkman L, et al. Unlocking the promise of liquid biopsies in precision oncology. The Journal of Liquid Biopsy [Internet]. 2024, 3. Available from: https://linkinghub.elsevier.com/retrieve/pii/S295019542400016X.
  125. Anderson MW, Schrijver I. Next generation DNA sequencing and the future of genomic medicine. Genes (Basel). 2010, 1, 38–69. [Google Scholar] [CrossRef] [PubMed]
  126. Manolio T, Chisholm R, Ozenberger B, Roden D, Williams MS, Wilson R, et al. Implementing genomic medicine in the clinic: the future is here. Genet Med. 2013, 15, 258–267. [Google Scholar] [CrossRef]
  127. Milko L, Khoury M. Editorial: DNA-based population screening for precision public health. Front Genet [Internet]. 2022, 13. Available from: https://www.frontiersin.org/articles/10.3389/fgene.2022.1061329/full.
  128. Wijetunga N, Yahalom J, Imber B, FitzGerald T, Hilal L, Hong JH, et al. The art of war: using genetic insights to understand and harness radiation sensitivity in hematologic malignancies. Front Oncol [Internet]. 2025, 14. Available from: https://www.frontiersin.org/articles/10.3389/fonc.2024.1478078/full.
  129. Adalsteinsson VA, Ha G, Freeman SS, Choudhury AD, Stover DG, Parsons HA, et al. Scalable whole-exome sequencing of cell-free DNA reveals high concordance with metastatic tumors. Nat Commun. 2017, 8, 1324. [Google Scholar] [CrossRef]
  130. Mathios D, Johansen JS, Cristiano S, Medina J, Phallen J, Larsen K, et al. Detection and characterization of lung cancer using cell-free DNA fragmentomes. Nat Commun [Internet]. 2021, 12. Available from: https://www.nature.com/articles/s41467-021-24994-w.
  131. Ueno-Yokohata H, Okita H, Nakasato K, Hishiki T, Shirai R, Tsujimoto S, et al. Preoperative diagnosis of clear cell sarcoma of the kidney by detection of BCOR internal tandem duplication in circulating tumor DNA. Genes Chromosomes Cancer. 2018, 57, 525–529. [Google Scholar] [CrossRef] [PubMed]
  132. Spreafico A, Hansen A, Abdul Razak AA, Bedard P, Siu L. The future of clinical trial design in oncology. Cancer Discov. 2021, 11, 822–837. [Google Scholar] [CrossRef] [PubMed]
  133. Betz M, Massard V, Gilson P, Witz A, Dardare J, Harlé A, et al. ESR1 gene mutations and liquid biopsy in ER-positive breast cancers: A small step forward, a giant leap for personalization of endocrine therapy? Cancers (Basel). 2023, 15, 5169. [Google Scholar] [CrossRef]
  134. Chen X, Gole J, Gore A, He Q, Lu M, Min J, et al. Non-invasive early detection of cancer four years before conventional diagnosis using a blood test. Nat Commun [Internet]. 2020, 11. Available from: https://www.nature.com/articles/s41467-020-17316-z.
  135. Aggarwal C, Thompson JC, Black TA, Katz SI, Fan R, Yee SS, et al. Clinical implications of plasma-based genotyping with the delivery of personalized therapy in metastatic non-small cell lung cancer. JAMA Oncol. 2019, 5, 173–180. [Google Scholar] [CrossRef] [PubMed]
  136. Brannon AR, Jayakumaran G, Diosdado M, Patel J, Razumova A, Hu Y, et al. Enhanced specificity of clinical high-sensitivity tumor mutation profiling in cell-free DNA via paired normal sequencing using MSK-ACCESS. Nat Commun. 2021.
  137. Mack P, Banks K, Espenschied C, Burich RA, Zill OA, Lee CE, et al. Spectrum of driver mutations and clinical impact of circulating tumor DNA analysis in non–small cell lung cancer: Analysis of over 8000 cases. Cancer. 2020, 126, 3219–3228. [Google Scholar] [CrossRef]
  138. Moser T, Waldispuehl-Geigl J, Belic J, Weber S, Zhou Q, Hasenleithner SO, et al. On-treatment measurements of circulating tumor DNA during FOLFOX therapy in patients with colorectal cancer. NPJ Precis Oncol [Internet]. 2020, 4. Available from: https://www.nature.com/articles/s41698-020-00134-3.
  139. Weber S, van der Leest P, Donker HC, Schlange T, Timens W, Tamminga M, et al. Dynamic changes of circulating tumor DNA predict clinical outcome in patients with advanced non–small-cell lung cancer treated with Immune checkpoint inhibitors. JCO Precis Oncol. 2021, 5, 1540–1553. [Google Scholar]
  140. Azad TD, Chaudhuri AA, Fang P, Qiao Y, Esfahani MS, Chabon JJ, et al. Circulating tumor DNA analysis for detection of minimal residual disease after chemoradiotherapy for localized esophageal cancer. Gastroenterology. 2020, 158, 494–505.e6. [Google Scholar] [CrossRef]
  141. Dagogo-Jack I, Brannon A, Ferris LA, Campbell CD, Lin JJ, Schultz KR, et al. Tracking the Evolution of Resistance to ALK Tyrosine Kinase Inhibitors through Longitudinal Analysis of Circulating Tumor DNA. Tracking the Evolution of Resistance to ALK Tyrosine Kinase Inhibitors through Longitudinal Analysis of Circulating Tumor DNA. JCO Precis Oncol. 2018.
  142. Recondo G, Bahcall M, Spurr LF, Che J, Ricciuti B, Leonardi G, et al. Molecular mechanisms of acquired resistance to MET tyrosine kinase inhibitors in patients with MET Exon 14–mutant NSCLC. Clin Cancer Res. 2020, 26, 2615–2625. [Google Scholar] [CrossRef] [PubMed]
  143. Zhou Q, Perakis SO, Ulz P, Mohan S, Riedl J, Talakic E, et al. Cell-free DNA analysis reveals POLR1D-mediated resistance to bevacizumab in colorectal cancer. Genome Med [Internet]. 2020, 12. Available from: https://genomemedicine.biomedcentral.com/articles/10.1186/s13073-020-0719-6.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

Disclaimer

Terms of Use

Privacy Policy

Privacy Settings

© 2025 MDPI (Basel, Switzerland) unless otherwise stated