ABSTRACT
Background
Head and neck cancers (HNCs) are too often diagnosed at advanced stages when outcomes are poor. Additionally, robust tools for the early detection of recurrence remain elusive. These gaps drive interest in so‐called liquid biopsy approaches for HNC detection, prognostication, and surveillance. Molecular heterogeneity presents challenges to liquid biopsy testing, but emerging approaches provide promising avenues toward clinical utility.
Methods
We review the latest developments in HNC liquid biopsies, provide perspectives on viral‐associated and nonviral‐associated cancers, and assess various biofluids, analytes, and molecular profiling approaches.
Results
Liquid biopsy assays targeting viral DNA from peripheral blood plasma have established clinical performance, and utility studies are ongoing, serving as a blueprint for other emerging assays.
Conclusion
The use of multiple biofluid sources and analytes may improve detection sensitivity and clinical applicability. Standardization and harmonization of analysis methods will be critical for enhancing biomarker discovery and enabling reliable clinical implementation.
Keywords: head and neck cancer, liquid biopsy, squamous cell carcinoma
Abbreviations
- CAPP‐seq
Cancer Personalized Profiling by deep sequencing
- CD
cluster of differentiation
- cfDNA
cell‐free DNA
- CTC
circulating tumor cell
- ctDNA
circulating tumor DNA
- dPCR
digital polymerase chain reaction
- EBV
Epstein–Barr virus
- EDTA
ethylenediaminetetraacetic acid
- EV
extracellular vesicles
- HNC
head and neck cancer
- HNSCC
head and neck squamous cell carcinoma
- HPV
human papillomavirus
- MBD‐seq
methyl‐CpG binding proteins capture
- MeDIP‐Seq
Methylated DNA Immunoprecipitation Sequencing
- MRD
molecular residual disease
- MSP
methylation‐specific PCR
- NGS
next‐generation sequencing
- NPC
nasopharyngeal carcinoma
- OPSCC
oropharyngeal squamous cell carcinoma
- OSCC
oral squamous cell carcinoma
1. Introduction
Head and neck cancer (HNC), including the most common histological subtype, head and neck squamous cell carcinoma (HNSCC), is a disease with profound functional implications for patients due to its anatomical location and side effects from treatment. Screening and surveillance are limited to clinical examinations and radiographic studies, which can miss signs of low‐volume cancers. Diagnosis is made through tissue biopsy, which is invasive and restricted to a limited number of sites due to patient tolerance. For these reasons, detection is often delayed, leading to a worsened prognosis.
Molecular analysis of blood and other bodily fluids (i.e., “liquid biopsy”) could lead to improvements in HNC diagnosis and optimized patient outcomes. Errors from tissue sampling (i.e., geographic misses) and tumor accessibility can be mitigated. Serial testing during and after treatment provides an opportunity for monitoring treatment response, detecting molecular residual disease (MRD), and identifying mechanisms of emerging treatment resistance. These applications of liquid biopsy could become relevant for making timely treatment decisions as well as supporting the development of new interventional strategies.
In this review, we discuss the pathogenesis of HNCs as it pertains to molecular features that can be leveraged for liquid biopsies, such as oncogenic viruses and specific genetic and epigenetic processes. We next describe various analytes (e.g., circulating tumor cells and cell‐free DNA) and biofluids (e.g., blood and saliva) that serve as sources for putative liquid biopsy biomarkers. We summarize evidence from studies on both viral‐associated and nonviral‐associated HNCs for clinical applications such as detection, monitoring, and prognostication. Finally, we discuss the current challenges and the future direction for unlocking the potential of liquid biopsies in HNC, including the potential of multi‐analyte and multi‐fluid assessments.
2. HNC Subtypes and Molecular Pathogenesis
Worldwide, HNC affects 900 000 people annually, with around 450 000 deaths every year [1]. Major anatomical subtypes of HNC include cancers of the oral cavity, lip, oropharynx, nasopharynx, larynx, and salivary glands (Figure 1). Geographic differences in HNC subtypes include a higher incidence of cancers of the nasopharynx in Southeast Asia and of the oral cavity in the Indian subcontinent [2]. Major causative agents for the development of HNCs include tobacco smoking, alcohol consumption, smokeless tobacco use, and betel nut chewing, along with infectious, environmental, and occupational exposures [3, 4, 5, 6, 7]. Genetic factors [8, 9, 10] and immunological health [11, 12] play important roles in HNC oncogenesis. Common oncogenic viruses include Epstein–Barr virus (EBV) in nasopharyngeal carcinoma (NPC) [13] and human papillomavirus (HPV) in oropharyngeal squamous cell carcinoma (OPSCC) [14]. Emerging liquid biopsy approaches have had to account for this heterogeneity of HNC pathogenesis.
FIGURE 1.

Head and neck cancer liquid biopsies and their composition. Graphical representation of common cancers seen in the head and neck with etiologic origins. Tumors at these sites can shed biomarkers into blood, saliva, and draining lymphatic fluid, with the components of each depicted above.
3. Liquid Biopsy Analytes and Biofluids
Sources of liquid biopsies can be categorized according to the type of analyte and the biofluid under study. This section will review each as it pertains to HNCs.
3.1. Liquid Biopsy Analytes
Major classes of liquid biopsy analytes include intact cells (i.e., circulating tumor cells [CTCs]) and various cell‐free tumor‐derived biomolecules such as cell‐free DNA (cfDNA), extracellular vesicles (EVs), cell‐free RNAs, and proteomic biomarkers. The focus of this section will be on CTCs, cfDNA, and EVs, whereas studies on cell‐free RNA and proteomic biomarkers have recently been reviewed elsewhere [15, 16, 17].
3.1.1. CTCs
The potential for cancer cells to be detectable in the circulation of patients with solid tumors has been recognized for over 150 years [18]. In many disease settings, the relative scarcity of CTCs—often representing less than one cell for every 10 million white blood cells in patients with known metastatic disease [19]—has presented technical challenges to their study. But recent advances in microfluidics, imaging, and high throughput molecular profiling have revealed that CTCs can reflect biological features of primary and metastatic tumors, and that their abundance can be associated with disease burden and risk of metastatic spread. CTCs can be passively shed from primary tumors and metastatic deposits, and they can also represent active invasion as part of the metastatic cascade [20]. As described in more detail below, the presence and abundance of CTCs in HNC patients are correlated with aggressive biology and poor prognosis [20, 21].
CTCs can be identified through selective isolation from the bloodstream or by screening cells for molecular features without selection. CTC isolation techniques rely on physical (e.g., size) and biological (e.g., distinct surface antigens) features that differ between cancer cells and leukocytes [22]. Isolation techniques for CTCs have been reviewed elsewhere [23]. In HNC, most studies on CTCs have employed selective isolation based on epithelial markers and examined associations with prognosis and recurrence [24, 25]. A common drawback to this approach is the inability to detect cancer cells that have lost epithelial markers through epithelial‐mesenchymal transition. For this reason, some studies are expanding the panel of antibodies for CTC selection [26]. To date, CTCs remain most readily detectable in advanced stages of HNC, with few if any cells detectable in early stages.
3.1.2. Circulating Tumor DNA (ctDNA)
DNA can be released by cells into biological fluids, including the peripheral circulation, in both pathological and physiological conditions [27]. This cfDNA—and ctDNA, the tumor‐derived portion of cfDNA—is a convenient source of potential biomarkers for diagnostic, prognostic, and predictive applications in oncology. cfDNA has a short half‐life of around 6 h and is highly fragmented [28, 29]. ctDNA often only represents a small fraction of the cfDNA that is shed into the circulation, and this fraction is dependent upon the tumor burden and stage, rate of cellular turnover, as well as response to therapy (Figure 2). The molecular makeup of ctDNA reflects the genetic and epigenetic composition of tumor cells, including somatic mutations and aberrant DNA methylation and chromatin structure [30].
FIGURE 2.

Tumor burden detectability utilizing liquid biopsies. Schematic view of ctDNA prior to diagnosis and after treatment. The fraction of plasma cfDNA that consists of ctDNA generally increases as disease progresses, where screening may allow for subclinical disease detection. Definitive treatment is generally associated with a decrease in ctDNA levels, and posttreatment surveillance may allow for the detection of molecular residual disease (MRD). Relapse of disease is generally associated with a rebound in ctDNA levels and may serve to guide therapeutic decision making.
With such a low abundance of ctDNA, sensitive detection methods are required. PCR‐based methods such as digital PCR (dPCR) can have high analytical sensitivity for specific genetic markers (e.g., mutations, methylated cytosines, or viral sequences) but are relatively limited in their ability to simultaneously interrogate multiple targets [31]. In contrast, next‐generation sequencing (NGS) technologies are capable of multiplexing across wide genomic regions [32] and are increasingly capable of interrogating a variety of molecular features within cfDNA.
Current guidelines from the European Society for Medical Oncology recommend that validated ctDNA testing can be used routinely in practice for the genotyping of advanced diseases for lung, breast, gastrointestinal cancer, and others [33]. For HNCs, however, mutation‐based precision medicine and targeted therapies are currently lacking. Nonetheless, as discussed in detail below, ctDNA testing continues to be investigated for other applications in HNCs, including early detection, prognostication, and monitoring.
3.1.3. EVs
EVs are lipid bilayered particles of various sizes that are released by cells into the extracellular environment and act as mediators of intercellular communication [34]. Exosomes represent a subset of EVs that range from 30 to 150 nm in diameter [35, 36, 37] and can transport various biomolecules (e.g., proteins, metabolites miRNA, mRNA, as well as genomic and mitochondrial DNA) out of and between cells [38, 39]. Exosomes have been detected in most biological fluids, including blood and saliva, and hold potential for being utilized as liquid biomarkers in HNSCC [40]. Though they exist in both healthy and diseased states, their presence in the tumor microenvironment has been implicated in tumor invasion, metastasis, and immunologic response to SCC [41, 42, 43]. Additionally, exosomes are able to transfer bioactive molecules between tumor cells, immune cells, and stromal cells, allowing for cancer cells to escape immune surveillance and induce immune tolerance [44].
3.2. Biofluid Sources for Liquid Biopsies
While many different biofluids are being studied as a source for liquid biopsies, blood and saliva are the most relevant and widely examined for HNC (Figure 1) [32, 45]. Recently, surgical drain lymph fluid has also been suggested to have potential utility for MRD detection and will be briefly reviewed.
3.2.1. Blood
Blood is in direct contact with all anatomical sites that give rise to HNC as well as draining nodal basins and distant organs at risk of harboring metastatic deposits. Whole blood (or buffy coat following centrifugation) is used for CTC isolation, whereas serum or plasma is used for cfDNA/ctDNA and EV/exosome analyses. Plasma is generally preferred over serum for cfDNA/ctDNA studies because there is less contamination from leukocytes [46, 47]. Plasma collection tubes also have cation chelators such as EDTA, which block nuclease activity and preserve cfDNA. In recent years, cell stabilizing agents have been added to blood collection tubes to reduce the contamination from leukocytes further and extend the acceptable time from blood draw to processing and storage [48, 49, 50].
3.2.2. Saliva
Saliva is a biological fluid derived from the salivary glands, gingival crevicular fluid, epithelial and inflammatory cell debris, bacteria, nasal and bronchial secretions, and blood [51, 52, 53]. Compared to blood, saliva could provide a more sensitive source for the detection of primary tumors due to the direct shedding of cells and cellular material. However, normal epithelial cells are also shed into saliva and could interfere with the specific detection of tumor cells. The source of cfDNA and EVs in saliva is more complex, as these smaller biomolecules can be secreted into saliva from blood components in addition to direct shedding [54, 55, 56, 57]. There may be less concern for leukocyte contamination in saliva compared to blood [58, 59, 60]. Moreover, whether distinct sizes of cfDNA and EVs in saliva may be relatively enriched for tumor‐derived content is an area of active study [61, 62, 63]. However, the enhanced sensitivity of more proximally collected biomarkers has already been suggested [64].
3.2.3. Lymphatic Fluid
Lymphatic fluid, or lymph, has recently been identified as a source for liquid biopsies in HNCs [65]. Draining lymph has been reported to contain tumor‐derived cfDNA. Surgical drains allow for the collection of lymph‐enriched fluid and are routinely placed after neck dissection in patients with HNCs, allowing for ease of collection and postsurgical MRD detection.
4. Viral‐Associated HNC
4.1. EBV‐Driven NPC
One of the best‐studied liquid biopsy biomarkers in HNC is plasma EBV DNA in patients with NPC. EBV is an oncogenic DNA virus that is found in a variety of cancer types, including NPC as well as certain gastric cancers and lymphomas. EBV‐associated NPC is endemic to Southeast Asia but is also found at lower rates around the world [13].
4.1.1. Detection
In a landmark study, EBV DNA was found to be detectable in the plasma of patients with NPC at the time of diagnosis, with higher levels associated with more advanced stages of disease [66]. Plasma samples were analyzed for the presence of EBV DNA through quantitative PCR (qPCR), targeting the EBNA‐1 and BamHI‐W regions. EBV DNA was detectable in the plasma of 97% of NPC patients prior to oncologic intervention, and copy numbers were 10 times higher in those patients with more advanced disease than those with early‐stage disease. This work suggested that quantitative analysis of plasma EBV DNA may be useful for detecting early‐stage NPC.
Subsequently, plasma EBV DNA detection was investigated as a screening modality for asymptomatic individuals [67]. Among 20 000 participants, persistently positive plasma EBV levels had sensitivity and specificity of 97.1% and 98.6%, respectively. Additionally, there was a notable shift toward earlier stage NPC diagnoses compared to historical controls. With a positive predictive value (PPV) of just 11%, the group later proposed sequencing‐based metrics to boost this metric. They found that integrating fragment size profiles of plasma EBV DNA molecules produced a PPV of 19.6% [68]. Moreover, plasma EBV DNA methylation profiling enabled differentiation of NPC, EBV‐associated lymphoma, and infectious mononucleosis, and integrating all of these metrics improved the PPV to 35.1% [69]. These works together show the great potential of plasma EBV DNA analysis as a screening test for endemic EBV‐driven NPC, with more work required, including comparisons with other promising approaches such as those that incorporate nasopharyngeal brushings [70].
Apart from plasma, urine as a liquid biopsy for EBV DNA detection in NPC has been investigated; however, low transrenal excretion only allows for detection in those patients exhibiting high levels within plasma [71]. While EBV DNA methylation studies of saliva in patients with NPC have been performed, sensitivity was not as high as that in plasma [72]. Therefore, plasma remains the dominant biofluid source for EBV DNA detection in patients with NPC.
4.1.2. Monitoring and Surveillance
In addition to its use in NPC screening, plasma EBV DNA has been assessed for its role in monitoring and surveillance. Since the first reports by Lo et al. over 25 years ago [73, 74], numerous studies have evaluated the association between plasma EBV DNA levels at baseline and throughout the treatment course with clinical events. A meta‐analysis of 14 studies involving more than 7836 cases found that patients with plasma EBV DNA levels that were high prior to initiation of treatment, patients with detectable levels mid‐ and posttreatment, and patients with slow clearance after treatment all experienced poor clinical outcomes with overall survival hazard ratios of 2.81, 3.29, 4.26, and 3.58, respectively [75]. In general, the prognostic value increased during and after treatment as compared to pretreatment. Moreover, the associations with distant metastasis were stronger than local or regional failure [75]. These results confirmed that plasma EBV DNA levels, especially posttreatment levels, have clinical validity as a prognostic biomarker and could help guide clinical decision making.
Based on these reproducible findings, recent investigations have focused on utilizing post‐chemoradiotherapy plasma EBV levels to guide adjuvant systemic treatment decisions. As the value of adjuvant chemotherapy remains controversial, a prospective randomized controlled trial sought to evaluate the benefit of adjuvant cisplatin and gemcitabine vs. observation in patients with residual detectable plasma EBV DNA 6–8 weeks after concurrent chemoradiotherapy [76]. With a median of 6.6 years follow‐up, there was no significant difference in 5‐year relapse‐free survival between the arms. This unexpected result could indicate the difficulty in sterilizing residual treatment‐refractory cancer cells after chemoradiotherapy. As cisplatin was included in both the concurrent and adjuvant phases of the trial, cross‐resistance may have developed that hindered the efficacy of the adjuvant regimen. Other studies such as NRG‐HN001 are evaluating regimens with less potential for cross‐resistance in this setting.
In contrast to the adjuvant setting, there has been greater progress in demonstrating potential clinical utility for plasma EBV DNA in the neoadjuvant setting. In a previously completed multi‐center randomized trial that established neoadjuvant cisplatin and gemcitabine as a standard treatment for locally advanced NPC [77], ad hoc analysis suggested that the benefit of neoadjuvant treatment was restricted to patients with pretreatment plasma EBV DNA ≥ 4000 copies/mL (5‐year overall survival 85.9% vs. 70.8%). Strikingly, there was no apparent benefit among patients with pretreatment EBV DNA < 4000 copies/mL (5‐year overall survival 90.6% vs. 91.4%). Future trials will need to replicate these findings in a prospective manner to confirm the specific thresholds and strength of the biomarker interaction.
Compared to the standard qPCR assay used in the vast majority of studies, more sensitive detection of plasma EBV DNA could provide greater prognostic value in the posttreatment setting. Emerging sequencing‐based methods could drive down technical detection limits by over 100‐fold. Indeed, sequencing‐based analysis of post‐chemoradiotherapy plasma EBV DNA in NPC patients from the aforementioned randomized adjuvant study [76] demonstrates higher sensitivity for both local (85.5%) and distant recurrences (97.1%) [78]. The added prognostic value of targeted sequencing over qPCR could lead to more informed clinical treatment decisions, but practical trade‐offs such as cost and turnaround time, which are both generally higher with sequencing‐based assays, also need to be taken into consideration when implementing these tests into clinical trials and standard practice.
4.1.3. EBV DNA Test Standardization
As EBV DNA testing becomes more broadly utilized, there has been a need for standardization of assay methods [79]. Le et al. 2013 assessed tests across different laboratories to harmonize international protocols, reagents, as well as cut‐off criteria for ctDNA detection. Further, in 2015, the National Cancer Institute convened a workshop on the harmonization of EBV testing for NPC to create standards for its appropriate clinical validation [80]. Although there continues to be a need to improve standardization efforts, the current progress with EBV DNA may serve as a blueprint for other assay types.
4.2. HPV‐Driven Oropharyngeal Carcinoma
The discovery that HPV infection can be a driver of oropharyngeal carcinogenesis, among other cancers, has provided the opportunity for major advancement in both detection and treatment. Compared to HPV‐negative OPSCC, HPV‐associated OPSCC is characterized by significantly better treatment responses and clinical outcomes [81]. While there are well over 150 different strains of HPV, 90% of HPV‐associated OPSCCs are attributed to the high‐risk HPV genotype 16 [14]. HPV16 and other high‐risk subtypes drive carcinogenesis in part by integrating their double‐stranded DNA into the host genome [14]. This integration event can lead to dysregulated expression of viral E6 and E7 oncogenes, which promote cellular proliferation, genetic instability, and remove cell cycle checkpoints, thereby promoting malignant transformation [82, 83].
4.2.1. Detection
HPV can be detected in tumor tissues by PCR, in situ hybridization, and immunohistochemistry targeting the surrogate marker p16 [84]. Unfortunately, most cases of OPSCC are initially detected through the presentation of enlarged lymph nodes in the neck, indicating lymphatic spread has already occurred, and diagnosed through tissue biopsy of the neck node and/or primary lesion. Currently, liquid biopsy approaches are being investigated for less invasive detection and diagnosis.
Plasma HPV DNA is being extensively studied for a range of liquid biopsy applications in OPSCC. An early study using qPCR detected HPV16 DNA in peripheral blood serum from only 9% of patients with HPV‐associated OPSCC [85]. Subsequent studies utilizing plasma instead of serum have shown higher sensitivities in a manner dependent on disease burden as determined by clinical stage [86, 87, 88, 89, 90, 91, 92, 93]. Incorporating HPV types other than HPV16, including HPV 18, 31, 33, and 35, can also boost sensitivity. One multi‐institutional prospective study found sensitivity of 89% and specificity of 97% among patients treated with chemoradiotherapy using dPCR [86]. Other notable cross‐sectional studies have found sensitivities of 89%–96% with similarly high specificity [87, 88]. Importantly, patients with primary‐only disease display lower detection rates compared to those who have nodal metastases, and N category is a strong driver of plasma HPV DNA levels.
The lower sensitivity for early‐stage disease has motivated the development of newer techniques for plasma HPV DNA detection. NGS provides a means for simultaneous detection of HPV DNA fragments derived from the entire ~8 kb genome, potentially boosting sensitivity by up to ~50‐fold compared to PCR‐based techniques. Indeed, studies using cell lines and patient plasma samples have confirmed higher analytical and clinical sensitivity when targeting the full HPV genome using hybrid capture NGS [94, 95]. In mostly early‐stage cases of HPV+ OPSCC, HPV genome NGS produced sensitivity of 99% and specificity of 98%, which compared favorably to dPCR [95]. Recognizing the additional cost and complexity of NGS assays compared to PCR, future studies are needed to define the clinical scenarios that require this additional boost in sensitivity to provide meaningful benefits in clinical utility.
Despite the reported association with clinical stage, plasma HPV DNA levels vary considerably among patients even after accounting for the known burden of disease. This variability may be explained in part by different rates of shedding and clearance from the circulation [96, 97] as well as the number of viral genome copies within malignant cells [98]. Moreover, viral DNA integration into the host genome is negatively associated with plasma HPV DNA levels and has also been linked with worse outcomes in OPSCC [99, 100]. Thus, the relationship between pretreatment plasma HPV DNA levels, disease burden, and prognosis is complex and multifactorial.
Beyond plasma cfDNA, EVs have also been assessed for their capacity to serve as a diagnostic biomarker of HPV‐associated OPSCC. In one study, DNA from EVs and total plasma cfDNA were both assessed using dPCR [101]. Sensitivity for the detection of locally advanced HPV‐associated OPSCC favored total plasma cfDNA (91%) over EVs (42%), indicating that EVs may not be as effective for assessing HPV DNA. EVs also contain RNA and other molecules that could be cancer‐specific, so additional work is needed to systematically compare these different analytes.
Saliva has also been evaluated for its utility in the detection of HPV‐associated OPSCC. A number of groups have employed PCR to assess for salivary HPV DNA in patients with HPV‐positive OPSCC. A cross‐sectional investigation assessed copy numbers of E6 and E7 together in salivary rinses with matched tumor samples and found a high specificity of 98.3% but only 30.4% sensitivity [102]. In a separate study with slightly different methods, sensitivity was higher at 81.4% with 96.4% specificity, indicating that the methodology of saliva collection is an important determinant of test performance [103].
The utility of salivary HPV16 DNA as a screening modality was recently evaluated in a prospective study of otherwise healthy individuals [104]. Participants with persistently positive results after 30 months were assessed by an otolaryngologist. Of the 650 participants recruited, 12 had persistent HPV16 DNA, and 1 of these was found to have a small primary lesion that was positive on pathology for HPV‐associated OPSCC. Longer follow‐up would be required to determine how many of these individuals with persistently detectable salivary HPV16 DNA eventually go on to develop invasive cancer.
Taken together, these studies indicate the promise of HPV DNA detection for liquid biopsy applications. Future studies are needed to define the clinical utility as a diagnostic or screening biomarker.
4.2.2. Monitoring and Surveillance
As the optimal strategy for posttreatment surveillance remains controversial, plasma HPV DNA detection has been proposed due to its noninvasive and quantitative properties. One of the first studies to examine dPCR for this purpose enrolled 22 patients with advanced HPV‐associated OPSCC [105]. Plasma HPV DNA levels correlated positively with the total tumor burden prior to and during treatment, where all patients who experienced clinical evidence of treatment response exhibited drops in plasma HPV copy numbers. The decrease in plasma HPV cfDNA was also shown to precede clinical response by 15 days. Conversely, patients with disease progression all showed either persistent or increasing levels of HPV DNA.
In the setting of nonmetastatic HPV‐associated OPSCC, plasma HPV DNA could be a useful tool for surveillance following chemoradiotherapy. An early demonstration of this strategy was shown in a cohort of 115 patients, 28 of whom developed a positive plasma HPV DNA test [106]. The median lead time between a positive plasma test and biopsy‐proven recurrence was 3.9 months, and the PPV of consecutive positive results was 94%. Subsequently, a multi‐center study of 1076 patients with HPV‐associated OPSCC demonstrated that only 80 (7.43%) had plasma HPV DNA‐positivity during surveillance after definitive therapy, with the majority (74%) of these positive results occurring in patients with no clinical evidence of disease [107]. Estimates of patient‐level positive and negative predictive values in this surveillance setting were 95% and 98%, respectively, albeit with limited follow‐up duration [107, 108].
With the promising results seen with plasma HPV DNA in the surveillance setting, there has been interest in using on‐treatment kinetics to adapt treatment intensity during chemoradiotherapy. Cao et al. observed a rapid decline in plasma HPV DNA levels in 14 patients during the course of chemoradiotherapy [93]. Chera et al. examined plasma HPV DNA kinetics weekly among 103 patients treated with chemoradiotherapy and found that rapid clearance 4 weeks into treatment correlated with favorable prognosis [86]. These intriguing results remain to be replicated, and the clinical utility for guiding treatment intensity will need to be evaluated in prospective studies.
Beyond plasma, HPV DNA in saliva and oral rinses has also been evaluated for monitoring of disease recurrence. In a two‐institution prospective study, qPCR was performed for the detection of high‐risk HPV subtypes in oral rinses after treatment for HPV‐associated HNSCC. Two‐year overall survival was only 65% in those patients with persistent high‐risk HPV DNA in oral rinses vs. 95% in those patients without [109]. Another study focusing on only HPV‐associated OPSCC patients found that persistent HPV16 DNA in oral rinses was strongly associated with recurrence, with specificity of 100% but modest sensitivity of 26% [110].
In an effort to boost sensitivity for posttreatment surveillance, a number of studies have evaluated dual plasma‐saliva liquid biopsies [56, 111, 112]. One study found that pretreatment sensitivity of HPV qPCR rose to 76% when analyzing both biofluids from 52.8% and 67.3% when analyzing just saliva or plasma, respectively [56]. Moreover, overall survival was reduced in patients who exhibited posttreatment HPV DNA in saliva, whereas survival was not affected if HPV DNA was detected in plasma alone. In terms of recurrence, combined plasma and saliva posttreatment samples were 90.7% specific and 69.5% sensitive in predicting recurrent disease in 3 years. These results were replicated in a subsequent prospective clinical trial with 87% specificity and 65% sensitivity for the combined HPV DNA test [111]. The mean lead time for a positive test when compared to clinically detected recurrence was 122 days, indicating that clinical implementation of dual liquid biopsies for disease detection holds promise.
To further address the limited sensitivity of HPV DNA detection in the posttreatment setting, another liquid biopsy source, lymph, has been introduced [65]. Plasma and paired lymphatic drainage fluid were assessed postsurgical resection, and cell‐free HPV DNA was found to be enriched in lymph compared to plasma and correlated with advanced nodal stage. While further work is needed to validate these findings, this proof‐of‐concept study demonstrates that lymph analysis could enable more sensitive HPV DNA detection for MRD applications. In particular, lymph may provide a window into regional recurrence risk while saliva and plasma reflect local and distant recurrence risk, respectively.
5. Nonviral‐Associated HNC
Nonviral HNCs present different challenges with respect to biomarker elucidation. While viral DNA offers convenient molecular targets for detection that are often present in multiple copies per cell, the heterogeneous nature of molecular aberrations driving nonviral HNCs necessitates more complex assays for liquid biopsy applications.
5.1. Mutational Profiling as a Means of Cancer Detection and Monitoring
Carcinogenesis is characterized by the accumulation of mutations that promote oncogene expression and/or inhibit the function of tumor suppressors. Mutations may be in the form of single‐nucleotide variants, insertions, deletions, and large chromosomal structural variants, and may be both inherited and acquired. Landmark advancements in genomic sequencing technologies have revealed hundreds of oncologic drivers [113, 114, 115]. Genes important for cell survival and proliferation (TP53, HRAS, EGFR, PTEN, and PIK3CA), cell‐cycle control (CDKN2A and CCND1), cellular differentiation (NOTCH1), reactive oxygen species scavenging (NFE2L2 and KEAP1), and adhesions and invasion signaling (FAT1) have all been implicated in nonviral‐associated HNSCC. Table 1 summarizes liquid biopsy studies investigating mutational targets in plasma, saliva, and oral rinses.
TABLE 1.
Mutational DNA liquid biopsy targets for HNSCCs.
| Study | Country | Patients | Tstage (early, late) | HPV status | Detection method | Tumor‐informed | Genes | Personalized panel | Sample type | Detection | Clinical context |
|---|---|---|---|---|---|---|---|---|---|---|---|
|
Wang et al., 2015 [116] |
USA | 93 HNSCC | 20, 73 | 40% HPV+ | Multiplex PCR | Yes | TP53, PIK3CA, CDKN2A, FBXW7, HRAS, NRAS | No | Plasma | 87% | Diagnosis, recurrence |
| No | Saliva | 76% | |||||||||
|
Perdomo et al., 2017 [117] |
France | 36 HNSCC | 14, 22 | HPV− | Sequencing | Yes | TP53, NOTCH1, CDKN2A, CASP8, PTEN | No | Plasma | 42% | Detection, diagnosis |
| 37 HNSCC | 0, 37 | TP53 | No | Oral rinse | 70% | ||||||
|
Schwaederle et al., 2017 [118] |
USA | 25 HNSCC | — | — | NGS | No | 70‐gene panel | No | Plasma | 88% | Detection, diagnosis |
|
Egyud et al., 2019 [119] |
USA | 7 HNSCC | 1, 6 | — | NGS | Yes | TP53, ARID1B, ATM, CDK8, FANCA, RASA1, SMARCA4, XRCC2, BCL10 | No | Plasma | 43% | Recurrence |
|
Mes et al., 2020 [120] |
Netherlands | 40 HNSCC | 6, 34 | 25% | NGS | Yes | TP53, NOTCH1, HRAS, CDKN2A, PTEN, PIK3CA, AJUBA, CASP8, FAT1, FBXW7, KMT2D, NSD1 | No | Plasma | 67% | Detection, diagnosis |
|
Bai et al., 2020 [121] |
China | 7 HNSCC | 0, 7 | — | NGS | Yes | TP53, PIK3CA | No | Plasma | 71% | Monitoring |
|
Galot et al., 2020 [122] |
Belgium | 39 HNSCC | 0, 39 | 12.8% | NGS | Yes | 604‐gene panel | No | Plasma | 51% | Recurrence, metastasis |
|
Hilke et al., 2020 [123] |
Germany | 20 HNSCC | 0, 20 | 15% | NGS | Yes | 327‐gene panel | No | Plasma | 85% | Monitoring |
|
Khandelwal et al., 2020 [124] |
USA | 22 OPSCC | 0, 22 | 50% | Sequencing | Yes | 56‐gene panel | No | Plasma | — | Prognosis, progression |
|
Porter et al. 2020 [125] |
USA | 48 HNSCC | 8, 29 | 25% | NGS | Yes | 73‐gene panel | No | Plasma | 83% | Recurrence, metastasis |
|
Wilson et al., 2021 [126] |
USA | 75 HNSCC | 28, 47 | 26.7% | NGS | Yes | 73‐gene panel | No | Plasma | 65.3% | Prognosis |
|
Wu et al., 2021 [127] |
China | 27 HNSCC | 9, 18 | HPV— | NGS | Yes | 1021‐gene panel | No | Plasma | 70.4% | Recurrence |
| No | Saliva | 63% | |||||||||
|
Cui et al., 2021 [128] |
Korea | 11 OSCC | 4, 7 | — | NGS | Yes | 493‐gene panel | No | Plasma | 40% | Recurrence |
| No | Saliva | 90.9% | |||||||||
|
Lee et al., 2021 [129] |
Korea | 7 HNSCC | 3, 4 | — | Multiplex PCR | Yes | 275‐gene panel | No | Saliva | 85.7% | Diagnosis, recurrence |
|
D'Cruz et al., 2021 [130] |
India | 15 OSCC | — | — | PCR | Yes | TP53 | No | Salivary rinse | 66.7% | Diagnosis |
|
Shanmugam et al., 2021 [131] |
USA | 121 OSCC | 53, 68 | — | NGS | Yes | TP53, PIK3CA, FAT1, CDKN2A, NOTCH1, CASP8, HRAS | No | Salivary rinse | 95.9% | Diagnosis, recurrence |
|
Burgener et al., 2021 [132] |
Canada | 30 HNSCC | 11, 19 | HPV— | CAPP‐seq | No | 389‐gene panel | No | Plasma | 66.7% | Recurrence, prognosis |
|
Rapado‐González et al., 2023 [133] |
Spain | 3 HNSCC | 0, 3 | HPV— | NGS | Yes | 170‐gene panel | No | Plasma | 100% | Detection |
| dPCR | EP300, NOTCH1, TP53 | Saliva | 33.3% | ||||||||
|
Flach et al., 2022 [134] |
Germany | 17 HNSCC | 0, 17 | HPV— | WES and Multiplex PCR | Yes | 48‐gene panel | Yes | Plasma | 100% | Detection, recurrence |
|
Honoré et al., 2023 [135] |
Belgium | 53 HNSCC | 12, 41 | 19% | NGS | No | 26‐gene panel + E6, E7 | No | Plasma | 77.4% | Detection, recurrence, prognosis |
|
Ahmed et al., 2024 [136] |
England | 11 OSCC | 1, 10 | — | PanelSeq | Yes | TP53, CDKN2A, KDM6B, NSD1, DNAH7, PIK3CA | No | Saliva | 82% | Detection |
|
Sanz‐Garcia, et al., 2024 [137] |
Canada | 32 HNSCC | 32 | 53% | NGS | Yes |
RaDaR CAPP‐seq HPV‐seq |
Yes | Plasma |
RaDaR 88% CAPP‐seq 51.7% |
Detection, recurrence |
| Hanna et al., 2024 [138] | USA | 116 HNSCC | 34, 82 | HPV— | WES | Yes | 16‐gene panel | Yes | Plasma | 91% | Recurrence, prognosis |
|
Sim et al., 2025 [139] |
USA | 24 HNSCC | 4, 20 | 4% (42% n/a) | WGS | Yes | MAESTRO | No | Plasma | 92.9% | Recurrence, prognosis |
Abbreviations: CAPP‐seq = Cancer Personalized Profiling by deep sequencing; MAESTRO = minor allele‐enriched sequencing through recognition oligonucleotides; NGS = next‐generation sequencing; WES = whole‐exome sequencing.
A notable study by Wang et al. assessed multi‐source liquid biopsies in the detection of mutations in HNSCC patients [116]. This observational study utilized both cfDNA from plasma and total salivary DNA for the detection of HPV DNA and somatic mutations using multiplex PCR targeting potential oncogenes. Plasma alone had a sensitivity of 87%, while saliva alone had a sensitivity of 76%; when combined, sensitivity improved to 96%. Interestingly, salivary DNA provided 100% sensitivity in oral squamous cell carcinoma (OSCC) patients, highlighting the value of interrogating both sources of liquid biopsy material.
These findings have been corroborated by another small study of 11 OSCC patients in which both plasma and saliva were interrogated using an NGS‐based technique [128]. Sensitivity was 27% in plasma and 91% in saliva when analyzed in isolation, but 100% when combined. Recurrent disease was detected by the combined liquid biopsy with 100% sensitivity. It is noted that in all cases of recurrent disease, detection by liquid biopsy was made prior to the emergence of clinical and radiographic findings, with up to 6 months lead time. While larger studies are needed, these findings indicate that mutational profiling within multiple biofluids holds promise for detection and monitoring of nonviral HNC.
Apart from multiple liquid biopsy sources, tumor tissue analysis has been investigated as a means for designing personalized, bespoke ctDNA assays. Flach et al. interrogated plasma cfDNA of patients with HPV‐negative HNSCC following tumor whole‐exome sequencing for personalized variant selection; multiplex amplicon NGS of tumor‐specific variants achieved high sensitivity [134]. In all patients that recurred, ctDNA was detected prior to clinically observed progression, with lead times of 108–253 days. Similarly, Sanz‐Garcia et al. found higher sensitivity with a tumor‐informed bespoke ctDNA assay as compared to a tissue‐agnostic panel‐based NGS such as CAPP‐seq [137], and Honore et al. observed lower sensitivity using a tumor‐agnostic 26‐gene NGS panel.
Taken together, this work highlights that tumor‐informed mutation‐based ctDNA assays—while more complex and costly than tissue‐agnostic assays—more accurately detect disease recurrence in nonviral HNSCC. In the absence of tissue, analysis of multiple biofluid sources could provide a means for driving greater sensitivity.
5.2. Methylation Profiling as a Means of Cancer Detection and Monitoring
In addition to mutations, DNA methylation has emerged as a promising class of liquid biopsy biomarkers for HNC. Cancer cells harbor hypermethylation of CpG‐dense regions (i.e., CpG islands) and hypomethylation of CpG‐sparse regions across the genome. Of these, hypermethylated CpG islands located in the vicinity of gene promoters have been the most commonly investigated regions for ctDNA detection due to their potential for cancer specificity with a high signal‐to‐noise ratio in cfDNA [140, 141]. Many cancers show more instances of conserved CpG island hypermethylation than mutated sequences, which could drive greater sensitivity for ctDNA detection in HNC [142]. Table 2 summarizes liquid biopsy studies investigating methylation changes in DNA in plasma, saliva, and oral rinses. Until recently, methylation‐specific PCR (MSP) has been the most commonly utilized technique. In a large observational study utilizing multi‐source liquid biopsies, Carvalho et al. paired serum and oral rinse samples from patients with HNSCC, and healthy participants were analyzed using quantitative MSP. Detection of MINT31, MGMT, CCNA1, and CDKN2A in serum and oral rinse resulted in 84.5% and 85.0% sensitivity, respectively, and 50% and 30% specificity [151]. This group made note that differential patterns of methylation in normal subjects between the serum and salivary liquid biopsies existed, which influenced the utility of single markers.
TABLE 2.
Methylated DNA liquid biopsy targets for HNSCCs.
| Study | Country | Patients | T category (T1–2, T3–4, unknown) | Detection method | Tumor‐informed | Methylated targets | Personalized panel | Sample detection | Clinical context |
|---|---|---|---|---|---|---|---|---|---|
|
Sanchez‐Cespedes et al., 2000 [143] |
USA | 23 HNSCC | 4, 19 | MSP | Yes | P16, MGMT, GSTp1DAPK | No |
Tumor: 42% Serum: p16 (Sens 31%), MGMT (Sens 48%), DAPK (Sens 18%) |
Detection |
|
Rosas et al., 2001 [144] |
USA | 30 HNSCC | 7, 23 | MSP | Yes | p16, MGMT, DAPK | No |
Tumor: 56% Oral rinse: Sens 36.7% |
Detection |
|
Wong et al., 2003 [145] |
China | 20 HNSCC | — | MSP | Yes | p15, p16 | No | Plasma: p15 (Sens 60%, Spec 50%), p16 (Sens 65%, Spec 80%) | Detection |
|
Nakahara et al., 2005 [146] |
Japan | 17 OSCC | 9, 8 | MSP | Yes | p16 | No |
Tumor: 64% Serum: Sens 54.5% |
Detection |
|
Righini et al., 2007 [147] |
France | 90 HNSCC | 29, 61 | MSP | Yes | p16, MGMT, DAPK, RASSF1A, TIMP3, ECAD | No |
Tumor: Sens 77% Oral Rinse (n = 60): Sens 82% |
Detection, recurrence |
|
Franzmann et al., 2007 [148] |
USA | 102 HNSCC | 40, 62 | MSP | No | CD44 | No | Oral rinse: Sens 62%–70% | Detection, recurrence |
|
Viet et al., 2007 [149] |
USA | 14 OSCC/dysplasia | — | Methylight | Yes | p16, MGMT, p15, APC, ECAD | No | Saliva: Sens 7%–35% | Detection |
|
Viet et al., 2008 [150] |
USA | 13 OSCC | 7, 6 | GoldenGate Methylation Array | Yes | GARB3, IL11, INSR, NOTCH3, NTRK3, PXN | No | Saliva: Sens 77% Spec 87% | Detection, monitoring |
|
Carvalho et al., 2008 [151] |
USA | 176 HNSCC | — | qMSP | Yes | MINT31, MGMT, CCNA1, p16 | No | Serum: Sens 84.5%, Spec 50% | Detection |
| No | Oral rinse: Sens 85%, Spec 30% | ||||||||
|
Pattani et al., 2010 [152] |
USA | 161 OSCC/dysplasia | — | qMSP | No | EDNRB | No | Oral rinse: Sens 71%, Spec 58% | Detection |
|
Demokan et al., 2010 [153] |
USA | 71 HNSCC | 23, 48 | qMSP | Yes | EDNRB, KIF1A | No | Oral rinse: Sens 77%, Spec 93% | Detection, prognosis |
|
Guerrero‐Preston et al., 2011 [154] |
USA and Spain | 16 OSCC | — | Human Methylation27 BeadChip | Yes | HOXA9, NID2 | No | Oral rinse: Sens 94%, Spec 97% | Detection |
|
Nagata et al., 2012 [155] |
Japan | 34 OSCC | 24, 10 | MSP | No | ECAD, TMEFF2, RARb, MGMT | No | Oral rinse: Sens 100%, Spec 87.5% | Detection |
|
Liu et al., 2012 [156] |
China | 32 OSCC | 18, 14 | qMSP | Yes | DAPK | No | Blood: Sen 52.2% Spec 86.6% | Detection |
| No | Oral rinse: Sens 3.4% | ||||||||
|
Kusumoto et al., 2012 [157] |
Japan | 10 OSCC | — | MSP | No | CDKN2A | No | Oral rinse: Sens 40%, Spec 100% | Detection |
|
Ovchinnikov et al., 2012 [158] |
Australia | 143 HNSCC | — | Nested MSP | No | RASSF1A, DAPK1, p16 | No | Saliva: Sens 80%, Spec 87% | Detection |
|
Rettori et al., 2013 [159] |
Brazil | 146 HNSCC | 58, 88 | qMSP | Yes | CCNA1, TIMP3 | No | Oral rinse: Sens 14%, Spec 93% | Detection |
|
Puttipanyalears et al., 2013 [160] |
Thailand | 43 OSCC | 14, 29 | Combined‐bisulfite restriction analysis (hypomethylation) | No | Alu | No | Oral rinse: Sens 87%, Spec 57% | Detection |
|
Schussel et al., 2013 [161] |
USA | 191 HNSCC | — | qMSP | No | EDNRB, DCC | No | Oral rinse: Sens 75%, Spec 48% | Detection |
|
Ovchinnikov et al., 2014 [162] |
Australia | 46 HNSCC | — | MSP | No | MED15, PCQAP5′ | No | Saliva: Sens 70%, Spec 63% | Detection |
|
Gaykalova et al., 2015 [163] |
USA | 59 HNSCC | 15, 41 | HumanMethylation27 BeadChips | Yes | ZNF160 | No | Oral rinse: Sens 17%, Spec 100% | Detection |
|
Langevin et al., 2015 [164] |
USA | OSCC/OPSCC | 36, 65 | HumanMethylation450 BeadArray | No | 22 CpG island classifier | No | Oral rinse: Sens 64.7%, Spec 96.0% | Detection |
|
Lim et al., 2016 [165] |
Australia | 88 HNSCC | 32, 36, 20 | MSP | No | RASSF1a, p16 INK4a , TIMP3, PCQAP5′, PCQAP3′ | No | Saliva: Sens 71%, Spec 80% | Detection |
|
Mydlarz et al., 2016 [166] |
USA | 100 HNSCC | 20, 80 | qMSP | No | EDNRB | No | Serum: Sens 10%, Spec 100% | Detection |
|
Ferlazzo et al., 2017 [167] |
Italy | 58 OSCC | — | MSP | No | p16, MGMT | No | Saliva: Sens 21% | Detection |
|
De Vos et al., 2017 [168] |
Germany | 278 HNSCC | 140, 131, 19 | dMSP | No | SEPT9, SHOX2 | No | Plasma: Sens 64%–65%, Spec 90%–91% | Detection |
|
Schröck et al., 2017 [169] |
Germany | 141 HNSCC | — | dMSP | No | SEPT9, SHOX2 | No | Plasma: Sens 52%, Spec 95% | Detection, prognosis, recurrence |
|
Cheng et al., 2018 [170] |
Taiwan | 94 OSCC | — | qMSP | No | ZNF582 | No | Oral rinse: Sens 65%, Spec 75% | Detection |
|
Puttipanyalears et al., 2018 [171] |
Thailand | 42 OSCC/24 OPSCC | 17, 25 (OSCC)/7, 17 (OPSCC) | qMSP | No | TRH | No |
Oral rinse: OSCC Sens 88%, Spec 93% OPSCC Sens 83%, Spec 93% |
Detection |
|
Liyanage et al., 2019 [172] |
Sri Lanka | 54 OSCC/34 OPSCC | 8, 23, 23 (OSCC)/2, 13, 19 (OPSCC) | MSP | No | p16, RASSF1a, TIMP3, PCQAP/MED15 | No |
Saliva: OSCC Sens 92%, Spec 92% OPSCC Sens 100%, Spec 92% |
Detection |
|
De Jesus et al., 2020 [173] |
Brazil | 15 OPSCC | — | dPCR | Yes | CCNA1, DAPK, CDH8, TIMP3 | No |
Tumor: 71% Plasma: Sens 73.3%, Spec 100% |
Detection |
|
Srisuttee et al., 2020 [174] |
Thailand | 43 OSCC | — | qMSP | No | NID2 | No | Oral rinse: Sens 79%, Spec 100% | Detection |
|
Shen et al., 2020 [175] |
USA | 21 OPSCC | 2, 19 | qMSP | No | PAX5, EDNRB | No | Oral rinse: Sens 95%, Spec 90% | Detection, recurrence |
|
Gonzalez‐Perez et al., 2020 [176] |
Columbia | 43 OSCC | 24, 19 | MSP | No | p16, RASSF1a | No | Saliva: Sens 54%, Spec 88% | Detection, prognosis |
|
Burgener et al., 2021 [132] |
Canada | 30 HNSCC | 11, 19 | MeDIPSeq | No | DMR: 941 hypermethylated regions, 56 hypomethylated regions | No | Plasma | Recurrence, prognosis |
|
Patel et al., 2023 [177] |
USA | 8 OSCC | 3, 5 | MBD‐seq | No |
Detection: PENK, NXPH1, ZIK1, TBXT, CDO1 Prognosis: ZIK1, IRF4, PCDH17, PENK |
No | Plasma | Detection, prognosis |
|
Rapado‐González et al., 2024 [178] |
Spain | 4 OSCC | 4, 0 | MethylationEPIC BeadChip | No | A2BP1, ANK1, ALDH1A2, GFRA1, TTYH1, PDE4B | No | Saliva: Sens 75%–100%, Spec 85%–100% | Detection |
|
Liu et al., 2025 [179] |
Canada | 325 HNSCC | 81, 68 | MeDIPSeq | No | DMR panel | No | Plasma: Sens 91%, Spec 88% | Recurrence |
Abbreviations: dPCR = digital polymerase chain reaction; DMR = differentially methylated regions; MBD‐seq = methyl‐CpG binding proteins capture; MeDIP‐Seq = Methylated DNA Immunoprecipitation sequencing; MSP = methylation‐specific PCR; sens = sensitivity; spec = specificity.
As MSP is limited in the number of targets that can be simultaneously interrogated, recent studies have applied NGS for broader profiling of HNC‐specific DNA methylation biomarkers. Burgener et al. compared cell‐free Methylated DNA ImmunoPrecipitation and high throughput sequencing (cfMeDIP‐seq) with mutation‐based NGS from plasma cfDNA [132]. In a cohort of HPV‐negative HNSCC, pretreatment ctDNA levels determined by cfMeDIP‐seq and mutation‐based NGS were highly correlated, and posttreatment persistence of methylated ctDNA was strongly correlated with recurrence. Interestingly, DNA methylation biomarkers associated with clinical outcomes when measured in tumor tissue were also strongly prognostic in plasma cfDNA in a manner that was independent of ctDNA levels. These results suggest that the value of epigenetic profiles of cfDNA can extend beyond detection and quantification to more phenotypic characterization of ctDNA, which could contribute to clinical utility by boosting prognostic performance. Importantly, many of the results from this study were replicated in a larger cohort of patients consisting of both HPV‐associated and HPV‐negative HNSCC [179], indicating that this approach could be generalizable across subtypes of HNC.
5.3. CTCs in Nonviral HNSCC
CTCs are relatively scarce in HNSCC, with only late‐stage disease demonstrating consistent detection [180]. In one study of patients with Stages II–IV HNSCC, E48 RNA, an epithelial marker, was detected by RT‐PCR in 35% of bone marrow samples and 10% of blood samples, with only late‐stage patients demonstrating detectability. In another study, peripheral pretreatment blood from HNSCC patients was subjected to CTC enrichment using EpCAM antibodies [181]. Of the 18 HNSCC patients studied, only 8 (44%) had detectable CTCs, 7 of whom had late‐stage disease. The CellSearch technique also targets EpCAM‐positive cells, along with cytokeratins 8, 18, and 19, and is coupled with flow cytometry [182]. In a multi‐center prospective study of patients with advanced‐stage HNSCC, 40% of pretreatment blood samples had detectable CTCs.
While CTCs have low sensitivity in early‐stage disease, multiple studies have shown potential prognostic value. CTC detectability was prognostic among patients with recurrent or metastatic HNSCC [183] as well as locally advanced OSCC [184, 185]. Other studies have evaluated dynamic changes in CTC levels in patients with HNSCC [186]. A reduction in CTC levels 2–4 weeks after treatment initiation correlated with better progression‐free survival and overall survival. Taken together, detectable CTCs appear to reflect a group of patients with highly aggressive disease behavior with poor prognosis.
5.4. EVs in Nonviral HNSCC
Apart from cfDNA and CTCs, EVs have also been assessed in liquid biopsies of HNC patients. Large EVs that are 100–1000 nm in size, termed microvesicles, have been quantified within saliva from newly diagnosed OSCC patients [187]. Purified salivary microvesicles were quantified by flow cytometry with higher counts in patients with OSCC than healthy controls or patients with benign oral disease. Salivary microvesicle counts were also correlated with disease burden as measured by lymph node status and clinical stage. While these findings are of interest, further work is needed on salivary EVs as a liquid biopsy.
EVs can also be quantified from peripheral blood plasma. In one study, plasma exosomes were measured pre‐, mid‐, and posttreatment in HNSCC patients receiving cetuximab and ipilimumab with radiotherapy [188]. Tumor‐derived exosomes were isolated by size‐exclusion chromatography, differentiating them from T‐cell‐derived exosomes via immunocapture Cluster of Differentiation (CD)3‐positivity. Levels of tumor‐derived exosomes increased from pretreatment levels among those patients with disease recurrence. In another study, exosomal levels of the immunosuppressive molecule, programmed death‐ligand 1 (PD‐L1), were correlated with clinical stage [189]. These data highlight the potential of evaluating specific targetable molecules within exosomes that may inform prognosis and/or therapeutic targeting.
6. Future Opportunities for Clinical Impact
With a growing catalog of methodologies available for HNC liquid biopsy applications (Table 3), major gaps remain in establishing clinical utility and optimal implementation of specific tests. Here, we propose a number of topics for future investigation.
TABLE 3.
Feature table of common detection platforms for HNC liquid biopsies.
| Detection methods and liquid biopsy source | Sensitivity | Advantages | Disadvantages | References | |
|---|---|---|---|---|---|
| qPCR | Plasma cfDNA | 9%–97% |
|
|
[73, 85, 91, 102, 103, 130, 190] |
| Saliva cfDNA | 30%–81% | ||||
| Digital PCR | Plasma cfDNA | 12%–100% |
|
|
[88, 89, 90, 91, 92, 133, 168, 169, 173, 191, 192] |
| Saliva cfDNA | 33%–73% | ||||
| Antibody enrichment | Blood CTCs | 40%–44% |
|
|
[181, 182] |
| Methylation‐specific PCR and qPCR | Plasma cfDNA | 18%–71% |
|
|
[143, 144, 145, 147, 148, 151, 152, 153, 155, 156, 159, 161, 162, 165, 167, 170, 171, 172, 174, 175, 176, 193, 194] |
| Saliva cfDNA | 3%–100% | ||||
| Next‐generation sequencing | Plasma cfDNA | 40%–100% |
|
|
[65, 95, 118, 119, 120, 121, 122, 123, 125, 126, 127, 128, 129, 130, 133, 134, 135, 136, 137, 138] |
| Saliva cfDNA | 63%–96% | ||||
6.1. Multimodal Liquid Biopsy Profiling
In this review, we have summarized the diversity of molecular analytes and biofluid sources in HNCs. Evaluating multiple analytes and/or biofluids may boost test performance and expand clinical utility. A number of studies have already begun to conduct multimodal liquid biopsy assessments. For instance, Burgener et al. [132] and Sanz‐Garcia et al. [137] both profiled multiple molecular targets (i.e., mutations, viral sequences, and/or aberrant DNA methylation) within ctDNA, while Wang et al. [116] and Earland et al. [65] both evaluated multiple biofluids. These are just a few examples of multimodal profiling approaches that could allow for a high‐resolution snapshot of disease burden, biology, and behavior. One tempting application where such multimodal profiling could have a major clinical impact is in the setting of post‐operative MRD detection, where complementary tests of saliva, lymph, and peripheral blood could be used to address the risk of local, regional, and distant relapse, respectively.
6.2. Protocol Standardization for Saliva
Techniques for peripheral blood handling for the purposes of liquid biopsy are now well established [195]. A similar degree of standardization would be of great benefit for saliva and lymph fluids. For saliva, different groups have used various combinations of whole saliva, saliva rinses, and oral swabs [196, 197, 198]. Typically, saliva is collected in a receptacle that can be subjected to centrifugation to isolate or remove the cellular components. DNA should be isolated within hours of collection to avoid degradation, or else preserved with fixative solutions to allow for batch processing. However, most fixative solutions contain lysis buffers that lead to a release of genomic DNA, which can dilute cfDNA. Unfortunately, the lack of standardization of saliva collection and analyte isolation procedures makes cross‐study comparisons difficult.
The ideal saliva receptacle should allow for the isolation of different liquid biopsy analytes. Cell lysing agents should be avoided so that cellular components and cfDNA can be analyzed separately. A DNA stabilizing agent would allow for delayed processing and improve clinical workflows. Finally, an antimicrobial agent would decrease bacterial outgrowth. Saliva has a unique microbiome that is altered in patients with HNC [199, 200, 201], and an accurate representation of salivary bacteria could lead to additional promising research directions [202].
6.3. Clinical Use of HNC Liquid Biopsies
Moving liquid biopsies into clinical use for HNC requires a number of considerations. The distribution of disease in local, regional, and distant sites will impact the potential utility of different tests. Due to its proximity to primary tumors, saliva may be best utilized for screening and local disease assessment. Many HNC patients experience treatment‐related xerostomia, which can impede salivary collection. Peripheral blood may be better suited for posttreatment surveillance in these patients and has demonstrated higher sensitivity for detecting metastatic spread. While sampling of peripheral blood can be easily repeated, lymph fluid has thus far only been studied in the immediate post‐operative period while surgical drains are in situ.
6.4. Implementation of HNC Liquid Biopsies Into Clinical Practice
To drive broad adoption of new tests, clinical utility studies are needed to demonstrate improvements in disease‐related outcomes, toxicity, and/or health system costs. To date, strong evidence of clinical utility for liquid biopsy testing in HNC has remained elusive. This gap is reflected in the absence of such tests in NCCN guidelines (v.4.2005). Prospective clinical trials are currently testing the potential for plasma EBV DNA and HPV DNA to lead to improved outcomes in NPC and OPSCC, respectively, but caution should be exercised until definitive results are available. The failure of adjuvant chemotherapy to improve relapse‐free survival in NPC patients with detectable plasma EBV DNA after chemoradiotherapy [76] serves as an important reminder that rigorously conducted interventional studies are needed.
Early investigations have pointed to the potential for cost savings from implementing liquid biopsy tests in certain clinical settings. For instance, detecting recurrence of HPV‐driven OPSCC in the surveillance setting could result in reduced costs by decreasing the need for other diagnostic tests [203]. Future investigations could follow the blueprint of ctDNA in Stage II colon cancer and Oncotype DX in breast cancer, which provide prognostic information that leads to cost savings related to reductions in unnecessary therapy in favorable risk patients [204, 205]. As the cost of sequencing modalities decreases, accessibility and cost‐effectiveness of emerging tests may continue to improve.
For patients to have access to liquid biopsy tests, clinical labs must be capable of providing specialized services. The success of implementation depends on reimbursement, assay cost, complexity, turnaround time requirements, testing volume, and regulatory scrutiny. Centralized lab testing often addresses several of these considerations through increased scale and lower variability, but can lead to limited access. Test standardization and harmonization between multiple clinical labs could increase accessibility; recent efforts to harmonize plasma EBV DNA testing illustrate the potential for this approach.
The future is bright for liquid biopsy research in HNC. Existing technologies are already making their way into clinical practice, with much work remaining to establish their optimal clinical settings and utility. Emerging technologies that target new analytes and biofluids have the potential to revolutionize how HNC patients are diagnosed and managed.
Conflicts of Interest
Scott V. Bratman declares ownership and leadership of Adela and patents/licensing with Roche and Adela. The other author declares no conflicts of interest.
Brooks P. J. and Bratman S. V., “Liquid Biopsies in Head and Neck Cancers: Recent Developments Across Biofluids, Analytes, and Molecular Features,” Head & Neck 47, no. 11 (2025): 3150–3171, 10.1002/hed.70009.
Data Availability Statement
Data sharing is not applicable to this article as no new data were created or analyzed in this study.
References
- 1. Global Cancer Observatory , “International Agency for Research on Cancer [Internet],” World Health Organization, accessed February 26, 2022, https://gco.iarc.fr/.
- 2. Bray F., Ren J. S., Masuyer E., and Ferlay J., “Global Estimates of Cancer Prevalence for 27 Sites in the Adult Population in 2008,” International Journal of Cancer 132 (2013): 1133–1145. [DOI] [PubMed] [Google Scholar]
- 3. Wyss A., Hashibe M., Chuang S.‐C., et al., “Cigarette, Cigar, and Pipe Smoking and the Risk of Head and Neck Cancers: Pooled Analysis in the International Head and Neck Cancer Epidemiology Consortium,” American Journal of Epidemiology 178, no. 5 (2013): 679–690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. De Stefani E., Boffetta P., Oreggia F., Fierro L., and Hard M. M., “Hard Liquor Drinking Is Associated With Higher Risk of Cancer of the Oral Cavity and Pharynx Than Wine Drinking. A Case‐Control Study in Uruguay,” Oral Oncology 34 (1998): 99. [DOI] [PubMed] [Google Scholar]
- 5. Wyss A. B., Hashibe M., Lee Y.‐C. A., et al., “Smokeless Tobacco Use and the Risk of Head and Neck Cancer: Pooled Analysis of US Studies in the INHANCE Consortium,” American Journal of Epidemiology 184, no. 10 (2016): 703–716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Guha N., Warnakulasuriya S., Vlaanderen J., and Straif K., “Betel Quid Chewing and the Risk of Oral and Oropharyngeal Cancers: A Meta‐Analysis With Implications for Cancer Control,” International Journal of Cancer 135 (2014): 1433–1443. [DOI] [PubMed] [Google Scholar]
- 7. Sheikh M., Shakeri R., Poustchi H., et al., “Opium Use and Subsequent Incidence of Cancer: Results From the Golestan Cohort Study,” Lancet Global Health 8 (2020): 649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Lacko M., Braakhuis B. J., Sturgis E. M., et al., “Genetic Susceptibility to Head and Neck Squamous Cell Carcinoma,” International Journal of Radiation Oncology, Biology, Physics 89, no. 1 (2014): 38–48. [DOI] [PubMed] [Google Scholar]
- 9. Jenkins G., O'Byrne K. J., Panizza B., and Richard D. J., “Genome Stability Pathways in Head and Neck Cancers,” International Journal of Genomics and Proteomics 2013 (2013): 464720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Lesseur C., Diergaarde B., Olshan A. F., et al., “Genome‐Wide Association Analyses Identify New Susceptibility Loci for Oral Cavity and Pharyngeal Cancer,” Nature Genetics 48, no. 12 (2016): 1544–1550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Mj P., Jl R., Sh C., Sy N., Sy K., and Lee Y. S., “De Novo Head and Neck Cancer Arising in Solid Organ Transplantation Recipients: The Asan Medical Center Experience,” Auris, Nasus, Larynx 45 (2018): 838–845. [DOI] [PubMed] [Google Scholar]
- 12. Douglas C. M., Jethwa A. R., Hasan W., et al., “Long‐Term Survival of Head and Neck Squamous Cell Carcinoma After Bone Marrow Transplant,” Head and Neck 42, no. 11 (2020): 3389–3395, https://onlinelibrary.wiley.com/doi/abs/10.1002/hed.26402. [DOI] [PubMed] [Google Scholar]
- 13. Tsao S. W., Tsang C. M., and Lo K. W., “Epstein‐Barr Virus Infection and Nasopharyngeal Carcinoma,” Philosophical Transactions of the Royal Society of London. Series B, Biological Sciences 372, no. 1732 (2017): 20160270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Gillison W. L., Koch W. M., Capone R. B., et al., “Evidence for a Causal Association Between Human Papillomavirus and a Subset of Head and Neck Cancers,” Journal of the National Cancer Institute 92, no. 9 (2000): 709–720. [DOI] [PubMed] [Google Scholar]
- 15. Kabzinski J., Kucharska‐Lusina A., and Majsterek I., “RNA‐Based Liquid Biopsy in Head and Neck Cancer,” Cells 12, no. 14 (2023): 1916–1943, 10.3390/cells12141916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Amenábar J. M., Da Silva B. M., and Punyadeera C., “Salivary Protein Biomarkers for Head and Neck Cancer,” Expert Review of Molecular Diagnostics 20, no. 3 (2020): 305–313. [DOI] [PubMed] [Google Scholar]
- 17. Konings H., Stappers S., Geens M., et al., “A Literature Review of the Potential Diagnostic Biomarkers of Head and Neck Neoplasms,” Frontiers in Oncology 10 (2020): 1020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Ashworth T. R., “A Case of Cancer in Which Cells Similar to Those in the Tumors Were Seen in the Blood After Death,” Australasian Medical Journal 14 (1869): 146–149. [Google Scholar]
- 19. Krebs M. G., Metcalf R. L., Carter L., Brady G., Blackhall F. H., and Dive C., “Molecular Analysis of Circulating Tumour Cells—Biology and Biomarkers,” Nature Reviews. Clinical Oncology 11 (2014): 129–144. [DOI] [PubMed] [Google Scholar]
- 20. Singh M., Yelle N., Venugopal C., and Singh S. K., “EMT: Mechanisms and Therapeutic Implications,” Pharmacology & Therapeutics 182 (2018): 80–94. [DOI] [PubMed] [Google Scholar]
- 21. Garrel R., Mazel M., Perriard F., et al., “Circulating Tumor Cells as a Prognostic Factor in Recurrent or Metastatic Head and Neck Squamous Cell Carcinoma: The CIRCUTEC Prospective Study,” Clinical Chemistry 65, no. 10 (2019): 1267–1275. [DOI] [PubMed] [Google Scholar]
- 22. Lin D., Shen L., Luo M., et al., “Circulating Tumor Cells: Biology and Clinical Significance,” Signal Transduction and Targeted Therapy 6, no. 1 (2021): 404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Yu M., Stott S., Toner M., Maheswaran S., and Haber D. A., “Circulating Tumor Cells: Approaches to Isolation and Characterization,” Journal of Cell Biology 192 (2011): 373–382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Patel S., Shah K., Mirza S., Shah K., and Rawal R., “Circulating Tumor Stem Like Cells in Oral Squamous Cell Carcinoma: An Unresolved Paradox,” Oral Oncology 62 (2016): 139–146. [DOI] [PubMed] [Google Scholar]
- 25. Harouaka R., Kang Z., Zheng S. Y., and Cao L., “Circulating Tumor Cells: Advances in Isolation and Analysis, and Challenges for Clinical Applications,” Pharmacology & Therapeutics 141 (2014): 209–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Gorges T. M., Tinhofer I., Drosch M., et al., “Circulating Tumour Cells Escape From EpCAM‐Based Detection due to Epithelial‐to‐Mesenchymal Transition,” BMC Cancer 12 (2012): 178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Thierry A. R., El Messaoudi S., Gahan P. B., Anker P., and Stroun M., “Origins, Structures, and Functions of Circulating DNA in Oncology,” Cancer Metastasis Reviews 35 (2016): 347–376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Thierry A. R., Mouliere F., Gongora C., et al., “Origin and Quantification of Circulating DNA in Mice With Human Colorectal Cancer Xenografts,” Nucleic Acids Research 38, no. 18 (2010): 6159–6175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. van der Vaart M. and Pretorius P. J., “Circulating DNA. Its Origin and Fluctuation,” Annals of the New York Academy of Sciences 1137 (2008): 18–26. [DOI] [PubMed] [Google Scholar]
- 30. Penny L., Main S. C., De Michino S. D., and Bratman S. V., “Chromatin‐ and Nucleosome‐Associated Features in Liquid Biopsy: Implications for Cancer Biomarker Discovery,” Biochemistry and Cell Biology 102, no. 4 (2024): 291–298. [DOI] [PubMed] [Google Scholar]
- 31. Chang Y., Tolani B., Nie X., Zhi X., Hu M., and He B., “Review of the Clinical Applications and Technological Advances of Circulating Tumor DNA in Cancer Monitoring,” Therapeutics and Clinical Risk Management 13 (2017): 1363–1374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Siravegna G., Marsoni S., Siena S., and Bardelli A., “Integrating Liquid Biopsies Into the Management of Cancer,” Nature Reviews Clinical Oncology 14, no. 9 (2017): 531–548. [DOI] [PubMed] [Google Scholar]
- 33. Pascual J., Attard G., Bidard F.‐C., et al., “ESMO Recommendations on the Use of Circulating Tumour DNA Assays for Patients With Cancer: A Report From the ESMO Precision Medicine Working Group,” Annals of Oncology 33, no. 8 (2022): 750–768. [DOI] [PubMed] [Google Scholar]
- 34. Kalluri R. and McAndrews K. M., “The Role of Extracellular Vesicles in Cancer,” Cell 186, no. 8 (2023): 1610–1626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Kalluri R., “The Biology and Function of Exosomes in Cancer,” Journal of Clinical Investigation 126 (2016): 1208–1215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Thakur B. K., Zhang H., Becker A., et al., “Double‐Stranded DNA in Exosomes: A Novel Biomarker in Cancer Detection,” Cell Research 24 (2014): 766–769. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Milane L., Singh A., Mattheolabakis G., Suresh M., and Amiji M. M., “Exosome Mediated Communication Within the Tumor Microenvironment,” Journal of Controlled Release 219, no. 219 (2015): 278–294. [DOI] [PubMed] [Google Scholar]
- 38. Lee Y., El Andaloussi S., and Wood M. J., “Exosomes and Microvesicles: Extracellular Vesicles for Genetic Information Transfer and Gene Therapy,” Human Molecular Genetics 21, no. R1 (2012): R125–R134. [DOI] [PubMed] [Google Scholar]
- 39. Zhang Y., Liang F., Zhang D., Qi S., and Liu Y., “Metabolites as Extracellular Vesicle Cargo in Health, Cancer, Pleural Effusion, and Cardiovascular Diseases: An Emerging Field of Study to Diagnostic and Therapeutic Purposes,” Biomedicine & Pharmacotherapy 157 (2023): 114046. [DOI] [PubMed] [Google Scholar]
- 40. Harding C. V., Heuser J. E., and Stahl P. D., “Exosomes: Looking Back Three Decades and Into the Future,” Journal of Cell Biology 200 (2013): 367–371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. van der Pol E., Böing A. N., Harrison P., Sturk A., and Nieuwland R., “Classification, Functions, and Clinical Relevance of Extracellular Vesicles,” Pharmacological Reviews 64 (2012): 676–705. [DOI] [PubMed] [Google Scholar]
- 42. Wahlgren J., Karlson T. D., Brisslert M., et al., “Plasma Exosomes Can Deliver Exogenous Short Interfering RNA to Monocytes and Lymphocyte,” Nucleic Acids Research 40 (2012): E130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Rashed M. H., Bayraktar E., Helal G. K., et al., “Exosomes: From Garbage Bins to Promising Therapeutic Targets,” International Journal of Molecular Sciences 18 (2017): 538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Liu J., Ren L., Li S., et al., “The Biology, Function, and Applications of Exosomes in Cancer,” Acta Pharmaceutica Sinica B 11, no. 9 (2021): 2783–2797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Arantes L. M. R., De Carvalho A., Melendez M. E., and Lopes Carvalho A., “Serum, Plasma and Saliva Biomarkers for Head and Neck Cancer,” Expert Review of Molecular Diagnostics 18, no. 1 (2018): 85–112. [DOI] [PubMed] [Google Scholar]
- 46. Lee J.‐S., Kim M., Seong M.‐W., Kim H.‐S., Lee Y. K., and Kang H. J., “Plasma vs. Serum in Circulating Tumor DNA Measurement: Characterization by DNA Fragment Sizing and Digital Droplet Polymerase Chain Reaction,” Clinical Chemistry and Laboratory Medicine 58, no. 4 (2020): 527–532. [DOI] [PubMed] [Google Scholar]
- 47. Pittella‐Silva F., Chin Y. M., Chan H. T., et al., “Plasma or Serum: Which Is Preferable for Mutation Detection in Liquid Biopsy?,” Clinical Chemistry 66, no. 7 (2020): 946–957. [DOI] [PubMed] [Google Scholar]
- 48. Zhao Y., Li Y., Chen P., Li S., Luo J., and Xia H., “Performance Comparison of Blood Collection Tubes as Liquid Biopsy Storage System for Minimizing cfDNA Contamination From Genomic DNA,” Journal of Clinical Laboratory Analysis 33 (2019): e22670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Maass K. K., Schad P. S., Finster A. M., et al., “From Sampling to Sequencing: A Liquid Biopsy Pre‐Analytic Workflow to Maximize Multi‐Layer Genomic Information From a Single Tube,” Cancers 13, no. 12 (2021): 3002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Das K., Fernando M. R., Basiaga S., Wigginton S. M., and Williams T., “Effects of a Novel Cell Stabilizing Reagent on DNA Amplification by PCR as Compared to Traditional Stabilizing Reagents,” Acta Histochemica 116 (2014): 55–60. [DOI] [PubMed] [Google Scholar]
- 51. Kaufman E. and Lamster I. B., “The Diagnostic Applications of Saliva—A Review,” Critical Reviews in Oral Biology and Medicine 13, no. 2 (2002): 197–212. [DOI] [PubMed] [Google Scholar]
- 52. Lee Y. H. and Wong D. T., “Saliva: An Emerging Biofluid for Early Detection of Diseases,” American Journal of Dentistry 22, no. 4 (2009): 241–248. [PMC free article] [PubMed] [Google Scholar]
- 53. Liu J. and Duan Y., “A Potential Media for Disease Diagnostics and Monitoring,” Oral Oncology 48, no. 7 (2012): 569–577. [DOI] [PubMed] [Google Scholar]
- 54. Schafer C. A., Schafer J. J., Yakob M., De Lima P. O., Camargo P., and Wong D. T. W., “Saliva Diagnostics: Utilizing Oral Fluids to Determine Health Status,” Monographs in Oral Science 24 (2014): 88–98. [DOI] [PubMed] [Google Scholar]
- 55. Lim Y., Sun C. X., Tran P., and Punyadeera C., “Salivary Epigenetic Biomarkers in Head and Neck Squamous Cell Carcinomas,” Biomarkers in Medicine 10, no. 3 (2016): 301–313. [DOI] [PubMed] [Google Scholar]
- 56. Ahn S. M., Chan J. Y. K., Zhang Z., et al., “Saliva and Plasma Quantitative Polymerase Chain Reaction‐Based Detection and Surveillance of Human Papillomavirus‐Related Head and Neck Cancer,” JAMA Otolaryngology. Head & Neck Surgery 140, no. 9 (2014): 846–854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Bettegowda C., Sausen M., Leary R. J., et al., “Detection of Circulating Tumor DNA in Early‐ and Late‐Stage Human Malignancies,” Science Translational Medicine 6 (2014): 224ra24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Topkas E., Keith P., Dimeski G., Cooper‐White J., and Punyadeera C., “Evaluation of Saliva Collection Devices for the Analysis of Proteins,” Clinica Chimica Acta 413, no. 13–14 (2012): 1066–1070. [DOI] [PubMed] [Google Scholar]
- 59. Schulz B. L., Cooper‐White J., and Punyadeera C. K., “Saliva Proteome Research: Current Status and Future Outlook,” Critical Reviews in Biotechnology 33, no. 3 (2013): 246–259. [DOI] [PubMed] [Google Scholar]
- 60. Gemmell C. H., Sefton M. V., and Yeo E. L., “Platelet‐Derived Microparticle Formation Involves Glycoprotein IIb‐IIIa. Inhibition by RGDS and a Glanzmann's Thrombasthenia Defect,” Journal of Biological Chemistry 268, no. 20 (1993): 14586–14589. [PubMed] [Google Scholar]
- 61. Rapado‐González Ó., López‐Cedrún J. L., Lago‐Lestón R. M., et al., “Integrity and Quantity of Salivary Cell‐Free DNA as a Potential Molecular Biomarker in Oral Cancer: A Preliminary Study,” Journal of Oral Pathology & Medicine 51, no. 5 (2022): 429–435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Heitzer E., Auinger L., and Speicher M. R., “Cell‐Free DNA and Apoptosis: How Dead Cells Inform About the Living,” Trends in Molecular Medicine 26, no. 5 (2020): 519–528. [DOI] [PubMed] [Google Scholar]
- 63. Summerer I., Niyazi M., Unger K., et al., “Changes in Circulating microRNAs After Radiochemotherapy in Head and Neck Cancer Patients,” Radiation Oncology 8 (2013): 296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Hanna G. J., Lau C. J., Mahmood U., et al., “Salivary HPV DNA Informs Locoregional Disease Status in Advanced HPV‐Associated Oropharyngeal Cancer,” Oral Oncology 95 (2019): 120–126. [DOI] [PubMed] [Google Scholar]
- 65. Earland N., Semenkovich N. P., Ramirez R. J., et al., “Sensitive MRD Detection From Lymphatic Fluid After Surgery in HPV‐Associated Oropharyngeal Cancer,” Clinical Cancer Research 30, no. 7 (2024): 1409–1421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Lo Y. M., Chan L. Y., Lo K.‐W., et al., “Quantitative Analysis of Cell‐Free Epstein‐Barr Virus DNA in Plasma of Patients With Nasopharyngeal Carcinoma,” Cancer Research 59, no. 6 (1999): 1188–1191. [PubMed] [Google Scholar]
- 67. Chan K. C. A., Woo J. K. S., King A., et al., “Analysis of Plasma Epstein–Barr Virus DNA to Screen for Nasopharyngeal Cancer,” New England Journal of Medicine 377, no. 6 (2017): 513–522. [DOI] [PubMed] [Google Scholar]
- 68. Lam W. K. J., Jiang P., Chan K. C. A., et al., “Sequencing‐Based Counting and Size Profiling of Plasma Epstein‐Barr Virus DNA Enhance Population Screening of Nasopharyngeal Carcinoma,” Proceedings of the National Academy of Sciences of the United States of America 115, no. 22 (2018): E5115–E5124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Lam W. K. J., Jiang P., Chan K. C. A., et al., “Methylation Analysis of Plasma DNA Informs Etiologies of Epstein‐Barr Virus‐Associated Diseases,” Nature Communications 10, no. 1 (2019): 3256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Zheng X.‐H., Li X.‐Z., Tang C.‐L., et al., “Detection of Epstein–Barr Virus DNA Methylation as Tumor Markers of Nasopharyngeal Carcinoma Patients in Saliva, Oropharyngeal Swab, Oral Swab, and Mouthwash,” MedComm (London) 5, no. 9 (2024): e673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Chan K. C. A., Leung S. F., Yeung S. W., Chan A. T. C., and Lo Y. M. D., “Quantitative Analysis of the Transrenal Excretion of Circulating EBV DNA in Nasopharyngeal Carcinoma Patients,” Clinical Cancer Research 14, no. 15 (2008): 4809–4813. [DOI] [PubMed] [Google Scholar]
- 72. Zheng X.‐H., Deng C.‐M., Zhou T., et al., “Saliva Biopsy: Detecting the Difference of EBV DNA Methylation in the Diagnosis of Nasopharyngeal Carcinoma,” International Journal of Cancer 153, no. 4 (2023): 882–892. [DOI] [PubMed] [Google Scholar]
- 73. Lo Y. M. D., Chan L. Y. S., Chan A. T. C., et al., “Quantitative and Temporal Correlation Between Circulating Cell‐Free Epstein‐Barr Virus DNA and Tumor Recurrence in Nasopharyngeal Carcinoma,” Cancer Research 59, no. 21 (1999): 5452–5455. [PubMed] [Google Scholar]
- 74. Lo Y. M., Chan A. T., Chan L. Y., et al., “Molecular Prognostication of Nasopharyngeal Carcinoma by Quantitative Analysis of Circulating Epstein‐Barr Virus DNA,” Cancer Research 60, no. 24 (2000): 6878–6881. [PubMed] [Google Scholar]
- 75. Zhang W., Chen Y., Chen L., et al., “The Clinical Utility of Plasma Epstein‐Barr Virus DNA Assays in Nasopharyngeal Carcinoma: The Dawn of a New Era?: A Systematic Review and Meta‐Analysis of 7836 Cases,” Medicine 94, no. 20 (2015): e845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Chan A. T. C., Hui E. P., Ngan R. K.‐C., et al., “Analysis of Plasma Epstein‐Barr Virus DNA in Nasopharyngeal Cancer After Chemoradiation to Identify High‐Risk Patients for Adjuvant Chemotherapy: A Randomized Controlled Trial,” Journal of Clinical Oncology 36, no. 31 (2018): 3091–3100. [DOI] [PubMed] [Google Scholar]
- 77. Zhang Y., Chen L., Hu G.‐Q., et al., “Final Overall Survival Analysis of Gemcitabine and Cisplatin Induction Chemotherapy in Nasopharyngeal Carcinoma: A Multicenter, Randomized Phase III Trial,” Journal of Clinical Oncology 40, no. 22 (2022): 2420–2425. [DOI] [PubMed] [Google Scholar]
- 78. Chan D. C., Lam W. K., Hui E. P., et al., “Improved Risk Stratification of Nasopharyngeal Cancer by Targeted Sequencing of Epstein–Barr Virus DNA in Post‐Treatment Plasma,” Annals of Oncology 33, no. 8 (2022): 794–803. [DOI] [PubMed] [Google Scholar]
- 79. Le Q.‐T., Zhang Q., Cao H., et al., “An International Collaboration to Harmonize the Quantitative Plasma Epstein‐Barr Virus DNA Assay for Future Biomarker‐Guided Trials in Nasopharyngeal Carcinoma,” Clinical Cancer Research 19, no. 8 (2013): 2208–2215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Kim K. Y., Le Q.‐T., Yom S. S., et al., “Current State of PCR‐Based Epstein‐Barr Virus DNA Testing for Nasopharyngeal Cancer,” Journal of the National Cancer Institute 109, no. 4 (2017): djx007, 10.1093/jnci/djx007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Ang K. K., Harris J., Wheeler R., et al., “Human Papillomavirus and Survival of Patients With Oropharyngeal Cancer,” New England Journal of Medicine 363, no. 1 (2010): 24–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Shukla S., Mahata S., Shishodia G., et al., “Physical State & Copy Number of High Risk Human Papillomavirus Type 16 DNA in Progression of Cervical Cancer,” Indian Journal of Medical Research 139, no. 4 (2014): 531–543. [PMC free article] [PubMed] [Google Scholar]
- 83. Jeon S., Allen‐Hoffmann B. L., and Lambert P. F., “Integration of Human Papillomavirus Type 16 Into the Human Genome Correlates With a Selective Growth Advantage of Cells,” Journal of Virology 69, no. 5 (1995): 2989–2997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Augustin J. G., Lepine C., Morini A., et al., “HPV Detection in Head and Neck Squamous Cell Carcinomas: What Is the Issue?,” Frontiers in Oncology 10 (2020): 1751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Capone R. B., Pai S. I., Koch W. M., et al., “Detection and Quantitation of Human Papillomavirus (HPV) DNA in the Sera of Patients With HPV‐Associated Head and Neck Squamous Cell Carcinoma,” Clinical Cancer Research 6 (2000): 4171–4175. [PubMed] [Google Scholar]
- 86. Chera B. S., Kumar S., Beaty B. T., et al., “Rapid Clearance Profile of Plasma Circulating Tumor HPV Type 16 DNA During Chemoradiotherapy Correlates With Disease Control in HPV‐Associated Oropharyngeal Cancer,” Clinical Cancer Research 25, no. 15 (2019): 4682–4690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Damerla R. R., Lee N. Y., You D., Soni R., Shah R., and Reyngold M., “Detection of Early Human Papillomavirus‐Associated Cancers by Liquid Biopsy,” JCO Precision Oncology 3 (2019): 1–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Rettig E. M., Wang A. A., Tran N.‐A., et al., “Association of Pretreatment Circulating Tumor Tissue‐Modified Viral HPV DNA With Clinicopathologic Factors in HPV‐Positive Oropharyngeal Cancer,” JAMA Otolaryngology. Head & Neck Surgery 148, no. 12 (2022): 1120–1130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Veyer D., Wack M., Mandavit M., et al., “HPV Circulating Tumoral DNA Quantification by Droplet‐Based Digital PCR: A Promising Predictive and Prognostic Biomarker for HPV‐Associated Oropharyngeal Cancers,” International Journal of Cancer 147, no. 4 (2020): 1222–1227. [DOI] [PubMed] [Google Scholar]
- 90. Akashi K., Sakai T., Fukuoka O., et al., “Usefulness of Circulating Tumor DNA by Targeting Human Papilloma Virus‐Derived Sequences as a Biomarker in p16‐Positive Oropharyngeal Cancer,” Scientific Reports 12, no. 1 (2022): 1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Rutkowski T. W., Mazurek A. M., Śnietura M., et al., “Circulating HPV16 DNA May Complement Imaging Assessment of Early Treatment Efficacy in Patients With HPV‐Positive Oropharyngeal Cancer,” Journal of Translational Medicine 18, no. 1 (2020): 167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Tanaka H., Takemoto N., Horie M., et al., “Circulating Tumor HPV DNA Complements PET‐CT in Guiding Management After Radiotherapy in HPV‐Related Squamous Cell Carcinoma of the Head and Neck,” International Journal of Cancer 148, no. 4 (2021): 995–1005. [DOI] [PubMed] [Google Scholar]
- 93. Cao H., Banh A., Kwok S., et al., “Quantitation of Human Papillomavirus DNA in Plasma of Oropharyngeal Carcinoma Patients,” International Journal of Radiation Oncology, Biology, Physics 82, no. 3 (2012): e351–e358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Leung E., Han K., Zou J., et al., “HPV Sequencing Facilitates Ultrasensitive Detection of HPV Circulating Tumor DNA,” Clinical Cancer Research 27, no. 21 (2021): 5857–5868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Bryan M. E., Aye L., Das D., et al., “Direct Comparison of Alternative Blood‐Based Approaches for Early Detection and Diagnosis of HPV‐Associated Head and Neck Cancers,” Clinical Cancer Research (2025): OF1–OF11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Moser T., Kühberger S., Lazzeri I., Vlachos G., and Heitzer E., “Bridging Biological cfDNA Features and Machine Learning Approaches,” Trends in Genetics 39, no. 4 (2023): 285–307. [DOI] [PubMed] [Google Scholar]
- 97. Rostami A., Lambie M., Yu C. W., Stambolic V., Waldron J. N., and Bratman S. V., “Senescence, Necrosis, and Apoptosis Govern Circulating Cell‐Free DNA Release Kinetics,” Cell Reports 31, no. 13 (2020): 107830. [DOI] [PubMed] [Google Scholar]
- 98. Hu Z., Zhu D., Wang W., et al., “Genome‐Wide Profiling of HPV Integration in Cervical Cancer Identifies Clustered Genomic Hot Spots and a Potential Microhomology‐Mediated Integration Mechanism,” Nature Genetics 47, no. 2 (2015): 158–163. [DOI] [PubMed] [Google Scholar]
- 99. Bratman S. V., Waldron J. N., and Liu F.‐F., “Human Papillomavirus Genotypes Conferring Poor Prognosis in Head and Neck Squamous Cell Carcinoma—Reply,” JAMA Oncology 3, no. 1 (2017): 125–126. [DOI] [PubMed] [Google Scholar]
- 100. Koneva L. A., Zhang Y., Virani S., et al., “HPV Integration in HNSCC Correlates With Survival Outcomes, Immune Response Signatures, and Candidate Drivers,” Molecular Cancer Research 16, no. 1 (2018): 90–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Nguyen B., Meehan K., Pereira M. R., et al., “A Comparative Study of Extracellular Vesicle‐Associated and Cell‐Free DNA and RNA for HPV Detection in Oropharyngeal Squamous Cell Carcinoma,” Scientific Reports 10, no. 1 (2020): 1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Zhao M., Rosenbaum E., Carvalho A. L., et al., “Feasibility of Quantitative PCR‐Based Saliva Rinse Screening of HPV for Head and Neck Cancer,” International Journal of Cancer 117, no. 4 (2005): 605–610. [DOI] [PubMed] [Google Scholar]
- 103. Ekanayake Weeramange C., Liu Z., Hartel G., et al., “Salivary High‐Risk Human Papillomavirus (HPV) DNA as a Biomarker for HPV‐Driven Head and Neck Cancers,” Journal of Molecular Diagnostics 23, no. 10 (2021): 1334–1342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Tang K. D., Vasani S., Menezes L., et al., “Oral HPV16 DNA as a Screening Tool to Detect Early Oropharyngeal Squamous Cell Carcinoma,” Cancer Science 111, no. 10 (2020): 3854–3861. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Hanna G. J., Supplee J. G., Kuang Y., et al., “Plasma HPV Cell‐Free DNA Monitoring in Advanced HPV‐Associated Oropharyngeal Cancer,” Annals of Oncology 29, no. 9 (2018): 1980–1986. [DOI] [PubMed] [Google Scholar]
- 106. Chera B. S., Kumar S., Shen C., et al., “Plasma Circulating Tumor HPV DNA for the Surveillance of Cancer Recurrence in HPV‐Associated Oropharyngeal Cancer,” Journal of Clinical Oncology 38, no. 10 (2020): 1050–1058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Berger B. M., Hanna G. J., Posner M. R., et al., “Detection of Occult Recurrence Using Circulating Tumor Tissue Modified Viral HPV DNA Among Patients Treated for HPV‐Driven Oropharyngeal Carcinoma,” Clinical Cancer Research 28, no. 19 (2022): 4292–4301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Hanna G. J., Roof S. A., Jabalee J., et al., “Negative Predictive Value of Circulating Tumor Tissue Modified Viral (TTMV)‐HPV DNA for HPV‐Driven Oropharyngeal Cancer Surveillance,” Clinical Cancer Research 29, no. 20 (2023): 4306–4313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Fakhry C., Blackford A. L., Neuner G., et al., “Association of Oral Human Papillomavirus DNA Persistence With Cancer Progression After Primary Treatment for Oral Cavity and Oropharyngeal Squamous Cell Carcinoma,” JAMA Oncology 5, no. 7 (2019): 985–992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Rettig E. M., Wentz A., Posner M. R., et al., “Prognostic Implication of Persistent Human Papillomavirus Type 16 DNA Detection in Oral Rinses for Human Papillomavirus‐Related Oropharyngeal Carcinoma,” JAMA Oncology 1, no. 7 (2015): 907–915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Califano J., Yousef A., Mostafa H., et al., “Lead Time to Recurrence After Posttreatment Plasma and Saliva HPV DNA Testing in Patients With Low‐Risk HPV Oropharynx Cancer,” JAMA Otolaryngology. Head & Neck Surgery 149, no. 9 (2023): 812–819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Ferrier S. T., Tsering T., Sadeghi N., Zeitouni A., and Burnier J. V., “Blood and Saliva‐Derived ctDNA Is a Marker of Residual Disease After Treatment and Correlates With Recurrence in Human Papillomavirus‐Associated Head and Neck Cancer,” Cancer Medicine 12, no. 15 (2023): 15777–15787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. The Cancer Genome Atlas Network , “Comprehensive Genomic Characterization of Head and Neck Squamous Cell Carcinomas,” Nature 517, no. 7536 (2015): 576–582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Stransky N., Egloff A. M., Tward A. D., et al., “The Mutational Landscape of Head and Neck Squamous Cell Carcinoma,” Science 333, no. 6046 (2011): 1157–1160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Agrawal N., Frederick M. J., Pickering C. R., et al., “Exome Sequencing of Head and Neck Squamous Cell Carcinoma Reveals Inactivating Mutations in NOTCH1,” Science 333, no. 6046 (2011): 1154–1157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Wang Y., Springer S., Mulvey C. L., et al., “Detection of Somatic Mutations and HPV in the Saliva and Plasma of Patients With Head and Neck Squamous Cell Carcinomas,” Science Translational Medicine 7, no. 293 (2015): 293ra104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Perdomo S., Avogbe P. H., Foll M., et al., “Circulating Tumor DNA Detection in Head and Neck Cancer: Evaluation of Two Different Detection Approaches,” Oncotarget 8, no. 42 (2017): 72621–72632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Schwaederle M., Chattopadhyay R., Kato S., et al., “Genomic Alterations in Circulating Tumor DNA From Diverse Cancer Patients Identified by Next‐Generation Sequencing,” Cancer Research 77, no. 19 (2017): 5419–5427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Egyud M., Sridhar P., Devaiah A., et al., “Plasma Circulating Tumor DNA as a Potential Tool for Disease Monitoring in Head and Neck Cancer,” Head & Neck 41, no. 5 (2019): 1351–1358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Mes S. W., Brink A., Sistermans E. A., et al., “Comprehensive Multiparameter Genetic Analysis Improves Circulating Tumor DNA Detection in Head and Neck Cancer Patients,” Oral Oncology 109 (2020): 104852. [DOI] [PubMed] [Google Scholar]
- 121. Bai J., Zhang S., Li X., Zhang Y., and Liu X., “Next‐Generation Sequencing of Plasma Cell‐Free DNA for Treatment Monitoring in Advanced Head and Neck Cancer Patients,” Clinical Laboratory 66, no. 7 (2020), 10.7754/Clin.Lab.2019.191150. [DOI] [PubMed] [Google Scholar]
- 122. Galot R., van Marcke C., Helaers R., et al., “Liquid Biopsy for Mutational Profiling of Locoregional Recurrent and/or Metastatic Head and Neck Squamous Cell Carcinoma,” Oral Oncology 104 (2020): 104631. [DOI] [PubMed] [Google Scholar]
- 123. Hilke F. J., Muyas F., Admard J., et al., “Dynamics of Cell‐Free Tumour DNA Correlate With Treatment Response of Head and Neck Cancer Patients Receiving Radiochemotherapy,” Radiotherapy and Oncology 151 (2020): 182–189. [DOI] [PubMed] [Google Scholar]
- 124. Khandelwal A. R., Greer A. H., Hamiter M., et al., “Comparing Cell‐Free Circulating Tumor DNA Mutational Profiles of Disease‐Free and Nonresponders Patients With Oropharyngeal Squamous Cell Carcinoma,” Laryngoscope Investigative Otolaryngology 5, no. 5 (2020): 868–878. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Porter A., Natsuhara M., Daniels G. A., et al., “Next Generation Sequencing of Cell Free Circulating Tumor DNA in Blood Samples of Recurrent and Metastatic Head and Neck Cancer Patients,” Translational Cancer Research 9, no. 1 (2020): 203–209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Wilson H. L., R. B. D'Agostino, Jr. , Meegalla N., et al., “The Prognostic and Therapeutic Value of the Mutational Profile of Blood and Tumor Tissue in Head and Neck Squamous Cell Carcinoma,” Oncologist 26, no. 2 (2021): e279–e289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Wu P., Xie C., Yang L., et al., “The Genomic Architectures of Tumour‐Adjacent Tissues, Plasma and Saliva Reveal Evolutionary Underpinnings of Relapse in Head and Neck Squamous Cell Carcinoma,” British Journal of Cancer 125, no. 6 (2021): 854–864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Cui Y., Kim H.‐S., Cho E. S., et al., “Longitudinal Detection of Somatic Mutations in Saliva and Plasma for the Surveillance of Oral Squamous Cell Carcinomas,” PLoS One 16, no. 9 (2021): e0256979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Lee Y. C., Kim S. I., Kwon H., Bae J.‐S., and Lee S., “Circulating Tumor DNA in the Saliva of Patients With Head and Neck Cancer: A Pilot Report,” Oral Diseases 27, no. 6 (2021): 1421–1425. [DOI] [PubMed] [Google Scholar]
- 130. D'Cruz A., Dechamma P. N., Saldanha M., Maben S., Shetty P., and Chakraborty A., “Non‐Invasive Saliva‐Based Detection of Gene Mutations in Oral Cancer Patients by Oral Rub and Rinse Technique,” Asian Pacific Journal of Cancer Prevention 22, no. 10 (2021): 3287–3291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Shanmugam A., Hariharan A. K., Hasina R., et al., “Ultrasensitive Detection of Tumor‐Specific Mutations in Saliva of Patients With Oral Cavity Squamous Cell Carcinoma,” Cancer 127, no. 10 (2021): 1576–1589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Burgener J. M., Zou J., Zhao Z., et al., “Tumor‐Naive Multimodal Profiling of Circulating Tumor DNA in Localized Head and Neck Squamous Cell Carcinoma,” Clinical Cancer Research 27, no. 15 (2021): 4230–4244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Rapado‐González Ó., Brea‐Iglesias J., Rodríguez‐Casanova A., et al., “Somatic Mutations in Tumor and Plasma of Locoregional Recurrent and/or Metastatic Head and Neck Cancer Using a Next‐Generation Sequencing Panel: A Preliminary Study,” Cancer Medicine 12, no. 6 (2023): 6615–6622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Flach S., Howarth K., Hackinger S., et al., “Liquid BIOpsy for MiNimal RESidual DiSease Detection in Head and Neck Squamous Cell Carcinoma (LIONESS)—A Personalised Circulating Tumour DNA Analysis in Head and Neck Squamous Cell Carcinoma,” British Journal of Cancer 126, no. 8 (2022): 1186–1195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Honoré N., van Marcke C., Galot R., et al., “Tumor‐Agnostic Plasma Assay for Circulating Tumor DNA Detects Minimal Residual Disease and Predicts Outcome in Locally Advanced Squamous Cell Carcinoma of the Head and Neck,” Annals of Oncology 34, no. 12 (2023): 1175–1186. [DOI] [PubMed] [Google Scholar]
- 136. Ahmed A. A., Sborchia M., Bye H., et al., “Mutation Detection in Saliva From Oral Cancer Patients,” Oral Oncology 151 (2024): 106717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Sanz‐Garcia E., Zou J., Avery L., et al., “Multimodal Detection of Molecular Residual Disease in High‐Risk Locally Advanced Squamous Cell Carcinoma of the Head and Neck,” Cell Death and Differentiation 31, no. 4 (2024): 460–468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Hanna G. J., Dennis M. J., Scarfo N., et al., “Personalized ctDNA for Monitoring Disease Status in Head and Neck Squamous Cell Carcinoma,” Clinical Cancer Research 30, no. 15 (2024): 3329–3336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Sim E. S., Rhoades J., Xiong K., et al., “Early Postoperative Minimal Residual Disease Detection With MAESTRO Is Associated With Recurrence and Worse Survival in Patients With Head and Neck Cancer,” Clinical Cancer Research (2025): OF1–OF9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Elhamamsy A. R., “DNA Methylation Dynamics in Plants and Mammals: Overview of Regulation and Dysregulation,” Cell Biochemistry and Function 34, no. 5 (2016): 289–298. [DOI] [PubMed] [Google Scholar]
- 141. Jamshidi A., Liu M. C., Klein E. A., et al., “Evaluation of Cell‐Free DNA Approaches for Multi‐Cancer Early Detection,” Cancer Cell 40, no. 12 (2022): 1537–1549.e12. [DOI] [PubMed] [Google Scholar]
- 142. Jones P. A., “Functions of DNA Methylation: Islands, Start Sites, Gene Bodies and Beyond,” Nature Reviews. Genetics 13, no. 7 (2012): 484–492. [DOI] [PubMed] [Google Scholar]
- 143. Sanchez‐Cespedes M., Esteller M., Wu L., et al., “Gene Promoter Hypermethylation in Tumors and Serum of Head and Neck Cancer Patients,” Cancer Research 60, no. 4 (2000): 892–895. [PubMed] [Google Scholar]
- 144. Rosas S. L., Koch W., da Costa Carvalho M. G., et al., “Promoter Hypermethylation Patterns of p16, O6‐Methylguanine‐DNA‐Methyltransferase, and Death‐Associated Protein Kinase in Tumors and Saliva of Head and Neck Cancer Patients,” Cancer Research 61, no. 3 (2001): 939–942. [PubMed] [Google Scholar]
- 145. Wong T.‐S., Man M. W.‐L., Lam A. K.‐Y., Wei W. I., Kwong Y.‐L., and Yuen A. P.‐W., “The Study of p16 and p15 Gene Methylation in Head and Neck Squamous Cell Carcinoma and Their Quantitative Evaluation in Plasma by Real‐Time PCR,” European Journal of Cancer 39, no. 13 (2003): 1881–1887. [DOI] [PubMed] [Google Scholar]
- 146. Nakahara Y., Shintani S., Mihara M., Hino S., and Hamakawa H., “Detection of p16 Promoter Methylation in the Serum of Oral Cancer Patients,” International Journal of Oral and Maxillofacial Surgery 35, no. 4 (2006): 362–365. [DOI] [PubMed] [Google Scholar]
- 147. Righini C. A., de Fraipont F., Timsit J.‐F., et al., “Tumor‐Specific Methylation in Saliva: A Promising Biomarker for Early Detection of Head and Neck Cancer Recurrence,” Clinical Cancer Research 13, no. 4 (2007): 1179–1185. [DOI] [PubMed] [Google Scholar]
- 148. Franzmann E. J., Reategui E. P., Pedroso F., et al., “Soluble CD44 Is a Potential Marker for the Early Detection of Head and Neck Cancer,” Cancer Epidemiology, Biomarkers & Prevention 16, no. 7 (2007): 1348–1355. [DOI] [PubMed] [Google Scholar]
- 149. Viet C. T., Jordan R. C. K., and Schmidt B. L., “DNA Promoter Hypermethylation in Saliva for the Early Diagnosis of Oral Cancer,” Journal of the California Dental Association 35, no. 12 (2007): 844–849. [PubMed] [Google Scholar]
- 150. Viet C. T. and Schmidt B. L., “Methylation Array Analysis of Preoperative and Postoperative Saliva DNA in Oral Cancer Patients,” Cancer Epidemiology, Biomarkers & Prevention 17, no. 12 (2008): 3603–3611. [DOI] [PubMed] [Google Scholar]
- 151. Carvalho A. L., Jeronimo C., Kim M. M., et al., “Evaluation of Promoter Hypermethylation Detection in Body Fluids as a Screening/Diagnosis Tool for Head and Neck Squamous Cell Carcinoma,” Clinical Cancer Research 14 (2008): 97–107. [DOI] [PubMed] [Google Scholar]
- 152. Pattani K. M., Zhang Z., Demokan S., et al., “Endothelin Receptor Type B Gene Promoter Hypermethylation in Salivary Rinses Is Independently Associated With Risk of Oral Cavity Cancer and Premalignancy,” Cancer Prevention Research 3, no. 9 (2010): 1093–1103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Demokan S., Chang X., Chuang A., et al., “KIF1A and EDNRB Are Differentially Methylated in Primary HNSCC and Salivary Rinses,” International Journal of Cancer 127, no. 10 (2010): 2351–2359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Guerrero‐Preston R., Soudry E., Acero J., et al., “NID2 and HOXA9 Promoter Hypermethylation as Biomarkers for Prevention and Early Detection in Oral Cavity Squamous Cell Carcinoma Tissues and Saliva,” Cancer Prevention Research 4, no. 7 (2011): 1061–1072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Nagata S., Hamada T., Yamada N., et al., “Aberrant DNA Methylation of Tumor‐Related Genes in Oral Rinse: A Noninvasive Method for Detection of Oral Squamous Cell Carcinoma,” Cancer 118, no. 17 (2012): 4298–4308. [DOI] [PubMed] [Google Scholar]
- 156. Liu Y., Zhou Z.‐T., He Q.‐B., and Jiang W.‐W., “DAPK Promoter Hypermethylation in Tissues and Body Fluids of Oral Precancer Patients,” Medical Oncology 29, no. 2 (2012): 729–733. [DOI] [PubMed] [Google Scholar]
- 157. Kusumoto T., Hamada T., Yamada N., et al., “Comprehensive Epigenetic Analysis Using Oral Rinse Samples: A Pilot Study,” Journal of Oral and Maxillofacial Surgery 70, no. 6 (2012): 1486–1494. [DOI] [PubMed] [Google Scholar]
- 158. Ovchinnikov D. A., Cooper M. A., Pandit P., et al., “Tumor‐Suppressor Gene Promoter Hypermethylation in Saliva of Head and Neck Cancer Patients,” Translational Oncology 5, no. 5 (2012): 321–326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Rettori M. M., De carvalho A. C., Bomfim longo A. L., et al., “Prognostic Significance of TIMP3 Hypermethylation in Post‐Treatment Salivary Rinse From Head and Neck Squamous Cell Carcinoma Patients,” Carcinogenesis 34, no. 1 (2013): 20–27. [DOI] [PubMed] [Google Scholar]
- 160. Puttipanyalears C., Subbalekha K., Mutirangura A., and Kitkumthorn N., “Alu Hypomethylation in Smoke‐Exposed Epithelia and Oral Squamous Carcinoma,” Asian Pacific Journal of Cancer Prevention 14, no. 9 (2013): 5495–5501. [DOI] [PubMed] [Google Scholar]
- 161. Schussel J., Zhou X. C., Zhang Z., et al., “EDNRB and DCC Salivary Rinse Hypermethylation Has a Similar Performance as Expert Clinical Examination in Discrimination of Oral Cancer/Dysplasia Versus Benign Lesions,” Clinical Cancer Research 19, no. 12 (2013): 3268–3275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Ovchinnikov D. A., Wan Y., Coman W. B., et al., “DNA Methylation at the Novel CpG Sites in the Promoter of MED15/PCQAP Gene as a Biomarker for Head and Neck Cancers,” Biomarker Insights 9 (2014): 53–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Gaykalova D. A., Vatapalli R., Wei Y., et al., “Outlier Analysis Defines Zinc Finger Gene Family DNA Methylation in Tumors and Saliva of Head and Neck Cancer Patients,” PLoS One 10, no. 11 (2015): e0142148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164. Langevin S. M., Eliot M., Butler R. A., et al., “CpG Island Methylation Profile in Non‐Invasive Oral Rinse Samples Is Predictive of Oral and Pharyngeal Carcinoma,” Clinical Epigenetics 7 (2015): 125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Lim Y., Wan Y., Vagenas D., et al., “Salivary DNA Methylation Panel to Diagnose HPV‐Positive and HPV‐Negative Head and Neck Cancers,” BMC Cancer 16, no. 1 (2016): 749. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Mydlarz W. K., Hennessey P. T., Wang H., Carvalho A. L., and Califano J. A., “Serum Biomarkers for Detection of Head and Neck Squamous Cell Carcinoma,” Head & Neck 38, no. 1 (2016): 9–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Ferlazzo N., Currò M., Zinellu A., et al., “Influence of MTHFR Genetic Background on p16 and MGMT Methylation in Oral Squamous Cell Cancer,” International Journal of Molecular Sciences 18, no. 4 (2017): 724–734, https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5412310/. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168. de Vos L., Gevensleben H., Schröck A., et al., “Comparison of Quantification Algorithms for Circulating Cell‐Free DNA Methylation Biomarkers in Blood Plasma From Cancer Patients,” Clinical Epigenetics 9 (2017): 125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169. Schröck A., Leisse A., de Vos L., et al., “Free‐Circulating Methylated DNA in Blood for Diagnosis, Staging, Prognosis, and Monitoring of Head and Neck Squamous Cell Carcinoma Patients: An Observational Prospective Cohort Study,” Clinical Chemistry 63, no. 7 (2017): 1288–1296. [DOI] [PubMed] [Google Scholar]
- 170. Cheng S.‐J., Chang C.‐F., Ko H.‐H., et al., “Hypermethylated ZNF582 and PAX1 Genes in Mouth Rinse Samples as Biomarkers for Oral Dysplasia and Oral Cancer Detection,” Head & Neck 40, no. 2 (2018): 355–368. [DOI] [PubMed] [Google Scholar]
- 171. Puttipanyalears C., Arayataweegool A., Chalertpet K., et al., “TRH Site‐Specific Methylation in Oral and Oropharyngeal Squamous Cell Carcinoma,” BMC Cancer 18, no. 1 (2018): 786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172. Liyanage C., Wathupola A., Muraleetharan S., Perera K., Punyadeera C., and Udagama P., “Promoter Hypermethylation of Tumor‐Suppressor Genes p16INK4a, RASSF1A, TIMP3, and PCQAP/MED15 in Salivary DNA as a Quadruple Biomarker Panel for Early Detection of Oral and Oropharyngeal Cancers,” Biomolecules 9, no. 4 (2019): 148, 10.3390/biom9040148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173. de Jesus L. M., Dos Reis M. B., Carvalho R. S., et al., “Feasibility of Methylated ctDNA Detection in Plasma Samples of Oropharyngeal Squamous Cell Carcinoma Patients,” Head and Neck 42, no. 11 (2020): 3307–3315. [DOI] [PubMed] [Google Scholar]
- 174. Srisuttee R., Arayataweegool A., Mahattanasakul P., et al., “Evaluation of NID2 Promoter Methylation for Screening of Oral Squamous Cell Carcinoma,” BMC Cancer 20, no. 1 (2020): 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Shen S., Saito Y., Ren S., et al., “Targeting Viral DNA and Promoter Hypermethylation in Salivary Rinses for Recurrent HPV‐Positive Oropharyngeal Cancer,” Otolaryngology and Head and Neck Surgery 162, no. 4 (2020): 512–519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176. González‐Pérez L.‐V., Isaza‐Guzmán D.‐M., Arango‐Pérez E.‐A., and Tobón‐Arroyave S.‐I., “Analysis of Salivary Detection of P16INK4A and RASSF1A Promoter Gene Methylation and Its Association With Oral Squamous Cell Carcinoma in a Colombian Population,” Journal of Clinical and Experimental Dentistry 12, no. 5 (2020): e452–e460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Patel K. B., Padhya T. A., Huang J., et al., “Plasma Cell‐Free DNA Methylome Profiling in Pre‐ and Post‐Surgery Oral Cavity Squamous Cell Carcinoma,” Molecular Carcinogenesis (2023): 493–502, https://onlinelibrary.wiley.com/doi/10.1002/mc.23501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178. Rapado‐González Ó., Costa‐Fraga N., Bao‐Caamano A., et al., “Genome‐Wide DNA Methylation Profiling in Tongue Squamous Cell Carcinoma,” Oral Diseases 30 (2024): 259–271, 10.1111/odi.14444. [DOI] [PubMed] [Google Scholar]
- 179. Liu G., Huang S. H., Ailles L., et al., “Clinical Validation of a Tissue‐Agnostic Genome‐Wide Methylome Enrichment Molecular Residual Disease Assay for Head and Neck Malignancies,” Annals of Oncology 36, no. 1 (2025): 108–117. [DOI] [PubMed] [Google Scholar]
- 180. Brakenhoff R. H., Stroomer J. G., ten Brink C., et al., “Sensitive Detection of Squamous Cells in Bone Marrow and Blood of Head and Neck Cancer Patients by E48 Reverse Transcriptase‐Polymerase Chain Reaction,” Clinical Cancer Research 5, no. 4 (1999): 725–732. [PubMed] [Google Scholar]
- 181. Wirtschafter A., Benninger M. S., Moss T. J., Umiel T., Blazoff K., and Worsham M. J., “Micrometastatic Tumor Detection in Patients With Head and Neck Cancer: A Preliminary Report,” Archives of Otolaryngology – Head & Neck Surgery 128, no. 1 (2002): 40–43. [DOI] [PubMed] [Google Scholar]
- 182. Nichols A. C., Lowes L. E., Szeto C. C. T., et al., “Detection of Circulating Tumor Cells in Advanced Head and Neck Cancer Using the CellSearch System,” Head and Neck 34, no. 10 (2012): 1440–1444. [DOI] [PubMed] [Google Scholar]
- 183. Grisanti S., Almici C., Consoli F., et al., “Circulating Tumor Cells in Patients With Recurrent or Metastatic Head and Neck Carcinoma: Prognostic and Predictive Significance,” PLoS One 9, no. 8 (2014): e103918. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184. Inhestern J., Oertel K., Stemmann V., et al., “Prognostic Role of Circulating Tumor Cells During Induction Chemotherapy Followed by Curative Surgery Combined With Postoperative Radiotherapy in Patients With Locally Advanced Oral and Oropharyngeal Squamous Cell Cancer,” PLoS One 10, no. 7 (2015): e0132901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185. Zhang X., Weeramange C. E., Hughes B. G. M., et al., “Circulating Tumour Cells Predict Recurrences and Survival in Head and Neck Squamous Cell Carcinoma Patients,” Cellular and Molecular Life Sciences 81, no. 1 (2024): 233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186. Wang H.‐M., Wu M.‐H., Chang P.‐H., et al., “The Change in Circulating Tumor Cells Before and During Concurrent Chemoradiotherapy Is Associated With Survival in Patients With Locally Advanced Head and Neck Cancer,” Head & Neck 41, no. 8 (2019): 2676–2687. [DOI] [PubMed] [Google Scholar]
- 187. Zhong W.‐Q., Ren J.‐G., Xiong X.‐P., et al., “Increased Salivary Microvesicles Are Associated With the Prognosis of Patients With Oral Squamous Cell Carcinoma,” Journal of Cellular and Molecular Medicine 23, no. 6 (2019): 4054–4062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188. Theodoraki M.‐N., Yerneni S., Gooding W. E., et al., “Circulating Exosomes Measure Responses to Therapy in Head and Neck Cancer Patients Treated With Cetuximab, Ipilimumab, and IMRT,” Oncoimmunology 8, no. 7 (2019): 1593805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189. Theodoraki M.‐N., Yerneni S. S., Hoffmann T. K., Gooding W. E., and Whiteside T. L., “Clinical Significance of PD‐L1+ Exosomes in Plasma of Head and Neck Cancer Patients PD‐L1+ Exosomes in Plasma of HNC Patients,” Clinical Cancer Research 24, no. 4 (2018): 896–905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190. Mazurek A. M., Rutkowski T., Fiszer‐Kierzkowska A., Małusecka E., and Składowski K., “Assessment of the Total cfDNA and HPV16/18 Detection in Plasma Samples of Head and Neck Squamous Cell Carcinoma Patients,” Oral Oncology 54 (2016): 36–41. [DOI] [PubMed] [Google Scholar]
- 191. Lin L.‐H., Cheng H.‐W., and Liu C.‐J., “Droplet Digital Polymerase Chain Reaction for Detection and Quantification of Cell‐Free DNA TP53 Target Somatic Mutations in Oral Cancer,” Cancer Biomarkers 33, no. 1 (2022): 29–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192. van Ginkel J. H., Huibers Mmh M., van Es R. J. J., de Bree R., and Willems S. M., “Droplet Digital PCR for Detection and Quantification of Circulating Tumor DNA in Plasma of Head and Neck Cancer Patients,” BMC Cancer 17, no. 1 (2017): 428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193. Wong T.‐S., Kwong D. L.‐W., Sham J. S.‐T., Wei W. I., Kwong Y.‐L., and Yuen A. P.‐W., “Quantitative Plasma Hypermethylated DNA Markers of Undifferentiated Nasopharyngeal Carcinoma,” Clinical Cancer Research 10, no. 7 (2004): 2401–2406. [DOI] [PubMed] [Google Scholar]
- 194. Chang H. W., Chan A., Kwong D. L. W., Wei W. I., Sham J. S. T., and Yuen A. P. W., “Detection of Hypermethylated RIZ1 Gene in Primary Tumor, Mouth, and Throat Rinsing Fluid, Nasopharyngeal Swab, and Peripheral Blood of Nasopharyngeal Carcinoma Patient,” Clinical Cancer Research 9, no. 3 (2003): 1033–1038. [PubMed] [Google Scholar]
- 195. Meddeb R., Pisareva E., and Thierry A. R., “Guidelines for the Preanalytical Conditions for Analyzing Circulating Cell‐Free DNA,” Clinical Chemistry 65, no. 5 (2019): 623–633. [DOI] [PubMed] [Google Scholar]
- 196. Rogers N. L., Cole S. A., Lan H.‐C., Crossa A., and Demerath E. W., “New Saliva DNA Collection Method Compared to Buccal Cell Collection Techniques for Epidemiological Studies,” American Journal of Human Biology 19, no. 3 (2007): 319–326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197. Matthews A. M., Kaur H., Dodd M., et al., “Saliva Collection Methods for DNA Biomarker Analysis in Oral Cancer Patients,” British Journal of Oral & Maxillofacial Surgery 51, no. 5 (2013): 394–398. [DOI] [PubMed] [Google Scholar]
- 198. Brooks P. J., Malkin E., and Bratman S. V., “Determination of Best Practices for Salivary DNA Collection and Isolation for Use in Cancer Detection” In preparation.
- 199. Amerongen A. V. N. and Veerman E. C. I., “Saliva—The Defender of the Oral Cavity,” Oral Diseases 8, no. 1 (2002): 12–22. [DOI] [PubMed] [Google Scholar]
- 200. Nagy K. N., Sonkodi I., Szöke I., Nagy E., and Newman H. N., “The Microflora Associated With Human Oral Carcinomas,” Oral Oncology 34, no. 4 (1998): 304–308. [PubMed] [Google Scholar]
- 201. Bolz J., Dosá E., Schubert J., and Eckert A. W., “Bacterial Colonization of Microbial Biofilms in Oral Squamous Cell Carcinoma,” Clinical Oral Investigations 18, no. 2 (2014): 409–414. [DOI] [PubMed] [Google Scholar]
- 202. Sami A., Elimairi I., Stanton C., Ross R. P., and Ryan C. A., “The Role of the Microbiome in Oral Squamous Cell Carcinoma With Insight Into the Microbiome–Treatment Axis,” International Journal of Molecular Sciences 21, no. 21 (2020): 8061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203. Lin M. G., Zhu A., Read P. W., Garneau J., and McLaughlin C., “Novel HPV Associated Oropharyngeal Squamous Cell Carcinoma Surveillance DNA Assay Cost Analysis,” Laryngoscope 133, no. 11 (2023): 3006–3012. [DOI] [PubMed] [Google Scholar]
- 204. Berdunov V., Cuyun Carter G., Laws E., et al., “Cost‐Effectiveness Analysis of the Oncotype DX Breast Recurrence Score Test From a US Societal Perspective,” ClinicoEconomics and Outcomes Research 16 (2024): 471–482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205. Tie J., Wang Y., Lo S. N., et al., “Circulating Tumor DNA Analysis Guiding Adjuvant Therapy in Stage II Colon Cancer: 5‐Year Outcomes of the Randomized DYNAMIC Trial,” Nature Medicine 31 (2025): 1509–1518, 10.1038/s41591-025-03579-w. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Data sharing is not applicable to this article as no new data were created or analyzed in this study.
