Next Article in Journal
Neuropilin-1 as a Potential Biomarker of Prognosis and Invasive-Related Parameters in Liver and Colorectal Cancer: A Systematic Review and Meta-Analysis of Human Studies
Previous Article in Journal
Fatty Acid Signaling Impacts Prostate Cancer Lineage Plasticity in an Autocrine and Paracrine Manner
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Is Cell-Free DNA Testing in Pancreatic Ductal Adenocarcinoma Ready for Prime Time?

1
Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH 432120, USA
2
School of Medicine, The Ohio State University, Columbus, OH 432120, USA
3
Department of Internal Medicine, Kamineni Academy of Medical Sciences and Research Center, Hyderabad 500012, India
4
Department of Internal Medicine, Division of Medical Oncology at the Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(14), 3453; https://doi.org/10.3390/cancers14143453
Submission received: 16 June 2022 / Revised: 3 July 2022 / Accepted: 13 July 2022 / Published: 15 July 2022
(This article belongs to the Section Cancer Biomarkers)

Abstract

:

Simple Summary

Pancreatic cancer is a deadly cancer with limited treatment options. It is often detected in most people at stages where cure is not possible. There is no good test to know if a person will respond to treatment or if there is any disease beyond what can be seen by available imaging tests. Genetic material from the tumor is expected to float in the blood. Studying the alterations in the genetic material could help detect the tumor early, give an idea about its aggressiveness and response to available treatments, and facilitate the discovery of newer therapies. The focus of the studies so far has been on only one kind of genetic aberration, mutations, which has not given us great results. There is a need to explore another type of change known as methylation that could hold answers for managing pancreatic cancers better.

Abstract

Cell-free DNA (cfDNA) testing currently does not have a significant role in PDA management: it is insufficient to diagnose PDA, and its use is primarily restricted to identifying targetable mutations (if tissue is insufficient or unavailable). cfDNA testing has the potential to address critical needs in PDA management, such as pre-operative risk stratification (POR), prognostication, and predicting (and monitoring) treatment response. Prior studies have focused primarily on somatic mutations, specifically KRAS variants, and have shown limited success in addressing prognosis and POR. Recent studies have demonstrated the importance of other less prevalent mutations (ERBB2 and TP53), but no studies have provided reliable mutation panels for clinical use. Methylation aberrations in cfDNA (epigenetic markers) in PDA have been relatively less explored. However, early evidence has suggested they offer diagnostic and, to some extent, prognostic value. The inclusion of epigenetic markers of cfDNA adds another dimension to genomic testing and may open new therapeutic avenues beyond addressing critical areas of need in PDA treatment. For cfDNA to substantially influence PDA management, concerted efforts are required to include less frequent mutations and epigenetic markers. Furthermore, relying on KRAS mutations for PDA management will always be inadequate.

Graphical Abstract

1. Introduction

Pancreatic ductal adenocarcinoma (PDA) is a fatal cancer with dismal survival. The 5-year relative survival is 11% for all-stage PDA: 42% in localized tumors and 3% in metastatic tumors [1]. In the United States, PDA is expected to be tenth in incidence (>62,000) and third in mortality (approximately 50,000) among other malignancies in 2022 [1]. A lack of reliable screening protocols, the high frequency of diagnosis at advanced stages, the need for debilitating surgery with a very high rate of recurrence, the limited systemic treatment options (adjuvant therapy (AT), neoadjuvant therapy (NAT), and palliative treatment (PT)), and the absence of reliable biomarkers contribute to poor outcomes in PDA [2]. The current status of PDA management and the areas requiring critical attention, beyond the development of novel therapeutic agents, are discussed in this section.

1.1. Current Management of PDA: Treatment and Disease Monitoring

Surgical resection is offered for early-stage tumors (both resectable (R-PDA) and borderline resectable (BR-PDA) if feasible) as a curative treatment [3]. Unfortunately, 80–85% of patients present with advanced disease (aPDA), including metastatic (mPDA), and locally advanced (LA-PDA) tumors [4]. The recurrence rates after successful resection are very high (80%), and the median long-term survival (>10 years) is only 4% [5]. Chemotherapy (FOLFIRINOX or gemcitabine/nab-paclitaxel (G-NP)) remains the main component of PDA systemic management (for NAT, AT, or PT) [6,7].
The current standard of care uses a combination of imaging and carbohydrate antigen 19-9 (CA 19-9) levels to monitor treatment response in patients receiving systemic therapy and to detect recurrence during surveillance [8]. This approach is limited by not only the poor sensitivity of CA19-9 but also the latency and evolving diagnostic uncertainty surrounding radiographic changes associated with PDA [9,10]. Moreover, CA19-9 is an unreliable biomarker because it is elevated in benign diseases (such as acute cholangitis, cirrhosis, and cholestatic diseases) and biliary obstruction from tumors, and shows an absence of elevation in 5–10% of PDA cases [11,12,13]. Therefore, an urgent need exists for new techniques or biomarkers to aid in diagnosis, staging, and therapeutic decision-making for PDA. The newest first-line therapy for aPDA was approved more than a decade ago. Newer targets and agents to treat PDA are beyond the scope of this review. Other ways to improve outcomes are discussed below.

1.2. Critical Needs in Pancreatic Adenocarcinoma Management

A comprehensive approach to PDA should start with developing more effective tools to aid in (a) diagnosis (and screening); (b) prognostication to anticipate the outcomes at diagnosis, irrespective of disease stage; (c) pre-operative risk stratification (POR); and (d) predicting (choosing an appropriate chemotherapy combination, G-NP vs. FOLFIRINOX) and monitoring treatment response. Traditional genomic testing and personalized approaches have not revolutionized PDA management as expected. The gold standard method of diagnosis (particularly for non-metastatic PDAs and mPDA with inaccessible metastatic disease) is endoscopic ultrasound-guided fine-needle aspiration (EUS-FNA) [8]. This approach provides a limited number of cells for cytological analysis and is scarcely sufficient for diagnosis. It often fails to provide adequate tumor tissue to perform genomic testing, including next-generation sequencing (NGS). Alternatives to tissue testing, such as cell-free DNA (cfDNA) testing in blood or liquid biopsies, have garnered much public attention as a non-invasive technique that may aid in diagnosis, staging, and therapeutic decision-making for malignancies [14].
cfDNA includes normal cell DNA, tumor cell DNA (ctDNA), circulating tumor cells (CTCs), and exosomal DNA, which can be detected in non-tissue sources such as blood, pancreatic juice, and bile [15]. Genetic aberrations in cfDNA from the blood of patients with cancer, specifically somatic mutations, are accessible to treating physicians, but their use is currently limited to the identification of targetable mutations cfDNA; testing in its current form cannot serve as a method for biomarker analysis. However, methylation changes or epigenetic markers in blood cfDNA are slowly gaining prominence. Most studies on cfDNA in PDA have involved detection and/or quantitative analysis of KRAS variants (mut-KRAS) in blood cfDNA. Among the cfDNA epigenetic markers, changes in methylation patterns have been more studied than histone modifications. In this review, cfDNA denotes cfDNA from the blood unless another source (bile or pancreatic juice) is specified. The current roles of somatic mutations (mostly mut-KRAS variants) and methylation changes detected in the cfDNA in PDA management are discussed in the review, with a focus on the predictive, prognostic, and POR value. The diagnostic value of epigenetic markers is discussed in depth, because of the limited relevant information in the literature.

2. Detection of cfDNA in PDA

Tumor cells in the cardiovascular system were first described in 1869, when Australian pathologist Thomas Ashworth observed cells in a post-mortem blood sample that were morphologically identical to those in the >30 subcutaneous tumors found in the same patient [16]. This seminal work suggested that the cells must pass through the circulatory system for tumors to metastasize. This was the first written report suggesting that cells from a tumor could be found within the circulatory system. Almost a century later, in 1955, the surgeon H.C. Engell corroborated Ashworth’s observations and observed morphologically identical cells in the bloodstream of a patient with colorectal cancer [17]. The prognostic and diagnostic value of these cells, termed CTCs, was unclear at the time. The first report of nucleic acids found in circulating plasma was by Mandel and Metais in 1948 [18]. Over the past decade, research interest has been renewed in this circulating material, because studies have demonstrated that these markers can aid in the diagnosis of a variety of cancers.
Another study in 1977 demonstrated a correlation between patients with malignancy and the absolute concentration of cfDNA in the serum [19]. In that study, Leon et al. demonstrated elevated concentrations of detectable cfDNA in serum from patients with lymphoma, lung, ovary, uterus, and cervical tumors, which decreased after disease-directed radiation therapy. It was not until 1994 that the first point mutation was detected in cfDNA [20]. With advances in sequencing technologies, the first somatic point mutation was identified in the NRAS gene, isolated from cfDNA in patients with myelodysplastic syndrome and acute myeloid leukemia. As sequencing technologies have advanced, both epigenetic modifications and RNA components of cfDNA can be identified [21]. The mechanism of cfDNA release by cells into the circulation remains not fully understood. A set of preliminary studies using samples from patients with colorectal cancer have postulated that cfDNA arises from products associated with necrosis, active secretion of DNA (exosomes and DNA–lipoprotein complexes), and apoptosis, as the sizes of sequenced cfDNA fragments correspond to those after DNA cleavage events associated with necrosis and apoptosis, and the sizes of fragments found in exosomes [22].
The mere presence and concentration of cfDNA are insufficient to guide clinical decisions, because many other conditions beyond cancer alter levels of cfDNA, including inflammatory conditions, cerebrovascular accidents, exercise, smoking, and trauma [23]. Because PDAs are notoriously difficult to diagnose via EUS-FNA, the detection of cfDNA provides an alternative method for PDA diagnosis. Numerous studies have demonstrated that cfDNA is detectable in the blood of patients with PDA across various stages of the disease, through methods including traditional polymerase chain reaction (PCR), pre-developed assay reagents (PDAR), PDAR-restriction fragment length polymorphism (PDAR-RFLP), digital droplet PCR (ddPCR), beads, emulsion, amplification, and magnetics PCR (BEAM-PCR), and NGS [14]. The first study to detect cfDNA in patients with PDA used PCR to amplify the KRAS oncogene in three patients and subsequently used traditional Sanger sequencing to identify mutant variations in the KRAS oncogenes (mut-KRAS) [24]. Studies have demonstrated that the detection of genetic aberrations in cfDNA is possible; more importantly, they have highlighted that these abnormalities are highly representative of those found in the primary tumor, thus suggesting that the somatic mutations identified in cfDNA are concordant with those found in tumor tissue [25,26].
Studies in the literature on cfDNA somatic mutations (Supplementary Figure S1) can be broadly divided into either total cfDNA (non-specific) mutations or specific mutations such as KRAS, TP53, and CDK2NA. The parameter of interest in these studies is the detection of mutation(s) or quantitative analysis of detected mutations or the mutant allelic fraction (MAF, a measure of the percentage of mutant alleles among all alleles in any given sample). Serial cfDNA testing studies to monitor treatment response, disease burden, or outcomes typically involved following one of these parameters. Similarly, methylation assays may be non-specific (such as total methylation levels) or specific to promoters (hypo- or hypermethylated), differentially methylated regions (DMRs), or differentially methylated CpG sites (DMPs). The concentration or degree of methylation in a promoter, measured as the methylation index (MI, methylated copy number/methylated copy number + unmethylated copy number), has been assessed in some studies. In this review, the available evidence is divided into sections according to current clinical needs to enable better understanding of the role of cfDNA testing.

2.1. cfDNA Testing in PDA, beyond KRAS

In an abstract presented at GI ASCO 2021 of a study including 1,009 patients from the database of Foundation One Medicine, 61% (613/1009) of the patients had at least one somatic alteration detected in cfDNA through comprehensive genomic profiling [26]. Among the detected alterations, TP53 was the most frequently altered (55%), followed by KRAS (40%) and CDK2NA (6.5%). The other mutations in the top ten were ATM (2.3%), PIK3CA (2.2%), PTEN, TERT, NF1, JAK2, and GNAS. The frequency of the last five mutations in this list was below 1.5%. The frequency of the altered mutations in the blood (cfDNA) correlated with the frequency of mutations detected in tissue in the available data from 81 patients [25]. KRAS, TP53, and CDK2NA were the top three mutated genes in tissue and blood. In a similar study (n = 23), the prevalence of detected mutations was higher, at 78% for both TP53 and KRAS [27]. This study used an NGS panel from a different company (Guardant Health, Redwood City, CA, USA) than the previous study. A study using ddPCR for a five-gene panel (n = 188) identified other mutations: BRCA2 (11.7%), KDR (13.8%), EGFR (13.3%), ERBB2 exon17 (13.3%), and ERBB2 exon27 (6.4%) [28]. The mut-KRAS rate was 72.3 % (G12V, G12D, and G12R rates were 34.5%, 52.1%, and 9%, respectively) in the same study.
A large sequencing-based study in 21,807 patients (25,578 blood samples) with treated late-stage cancers demonstrated the abundance of cfDNA in blood samples from patients with malignancy [29]. Alterations tested include single nucleotide variants, copy number variations, insertions and deletions, and fusions (from the Guardant Health panel). This comprehensive study examined >50 cancer types and had 1112 (4%) cfDNA samples from patients with PDA. Somatic alterations (total or non-specific) were detected in approximately 80% of the tested PDA samples; however, much larger studies might be required to understand their roles. In a similar but smaller study (n = 410 and 155 PDA), cfDNA was detected in >80% of aPDAs and 48% of localized PDAs (although further details on tumor stages were not reported) [30].
The concordance of the genetic alterations between tissue or EUS-FNA samples and cfDNA in PDA was studied in a meta-analysis (14 studies with 369 patients) published in 2019 [31]. Most studies included in this meta-analysis examined mut-KRAS (as expected), except for three studies that used multigene panels. Two studies analyzed CTCs, and the remainder performed cfDNA testing. Overall, the sensitivity and specificity of liquid biopsy to diagnose PDA compared with tissue specimens in this meta-analysis were 70% and 86%, respectively. Interestingly, when mut-KRAS alone was analyzed, the sensitivity decreased to 65%, but the specificity increased to 91%. The concordance rate in the study was 32%, while the mutations exclusively detected in the blood and tissue samples were just 30% and 38%, respectively. Because cfDNA contains genetic material from various sources (e.g., tumor cells, normal cells, and exosomes), its profiles are more likely to represent genetic variability and provide greater genomic insight into tumor heterogeneity than direct tissue sampling [32]. Although the low concordance in PDA may be disappointing, PDA is a cancer in which tissue samples are difficult to obtain. cfDNA testing can identify 62% of mutations (alterations), and 30% of them are different from those detected in tissues. Interestingly, a concordance rate of >75% has been found in studies of mut-KRAS alone [33,34]. Therefore, cfDNA testing can serve as an alternative in PDA patients.
The important takeaway from these PDA cfDNA studies is that mut-KRAS is detected in 40–50% of samples, but other less prevalent mutations make up the other 30–40%, and targetable mutations are rare in PDA. Therefore, to make cfDNA testing relevant in PDA, a concerted effort is needed to expand beyond mut-KRAS testing. Studying the clinical value of other mutations for prognosis or POR is difficult because large studies are required to indicate correlations. One strategy involves adding another layer to testing by using epigenetic markers.
Contrary to the common assumption that cfDNA can be detected only in advanced PDAs, many studies in the literature have reported its detection in early-stage tumors [35,36], thus not only improving POR (discussed below) but also aiding in diagnosis/screening (briefly discussed in Supplementary Table S1). In current clinical practice, diagnosis cannot be made based on cfDNA testing alone outside experimental settings, but it can provide support in rare cases in which the histological diagnosis is inconclusive and the suspicion for PDA is very high.

2.2. cfDNA Epigenetic Markers

Methylation in the promoter regions of genes is a major regulator of gene activation and inactivation [37]. Malignancy is often associated with hypermethylation of tumor suppressor genes, thus resulting in genetic inactivation, and hypomethylation of oncogenes, thus resulting in genetic activation [37]. Advances in sequencing technologies have also allowed for the identification of hypermethylation and hypomethylation patterns in cfDNA [14]. cfDNA epigenetic markers can be used for diagnostic purposes, as suggested by several retrospective studies summarized in Table 1 below. In some studies, cfDNA epigenetic markers from PDA patients have been compared with those from healthy controls (HC) or with those having benign diseases, such as cysts, chronic pancreatitis (CP), or both, to derive diagnostic markers.
In an early study, differentially expressed methylated cfDNA patterns were compared in patients with PDA (all stages) and normal patients [38]. The authors of the study curated a panel of five genes—CCND2, PLAU, SOCS1, THBS, and VHL—enabling differentiation between malignant and normal samples (HC) with a sensitivity and specificity of 76% and 59%, respectively. The methylation frequency of the genes in the panel was lower in PDA cfDNA than HC. The authors then curated a panel of cfDNA epigenetic markers that successfully discriminated between PDA and CP with a sensitivity and specificity of 91.2% and 90.8%, respectively [39]. Interestingly, methylation of NPTX2 was able to differentiate PDA from CP but not from HC [54,55]. In addition, greater p16 promoter methylation has been observed in PDA than HC. A genome-wide methylation study has demonstrated that hypermethylation of the SST gene can be detected in cfDNA across all stages of PDA development, with a sensitivity of 89% for PDA diagnosis [46]. Recently, multiple panels with reliable diagnostic accuracy have been reported for distinguishing PDA from HC or CP [42,44,45,56]. In one study using a 28-gene promoter panel, the number of methylated genes in the PDA group (8.41 vs. 4.74, p < 0.001) was significantly higher than that in the non-cancer group (HC + CP + acute pancreatitis).
Advances in methylome sequencing techniques have led to a dramatic increase in studies examining promoter hypermethylation in cfDNA. A recent study generated a panel of 51 features including both 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC) methylation patterns that can distinguish between patients with PDA and healthy individuals, with a sensitivity of 93.8%, specificity of 95.5%, and AUC of 0.997 [2]. The compound 5hmC, a derivative of 5mC, is a distinct epigenetic marker of transcriptional regulation of normal differentiation [57]. Its global loss and re-distribution are common features in various cancers, including PDA [58]. Individually, the sensitivity and specificity of 5mC alone are better than those of 5hmC. Interestingly, gene set enrichment analysis (GSEA) of 794 differentially hydroxymethylated genes (differential 5hmC) identified in cfDNA from patients with PDA demonstrated a similar genetic pattern to those commonly dysregulated after KRAS activation and TP53 inhibition in primary tumor tissue [43]. This finding suggests that these differentially methylated genes are likely to originate from the tumor itself. Most of the genes identified are associated with pancreatic development or function (GATA4, GATA6, PROX1, ONECUT1, and MEIS2), and malignant transformation (YAP1, TEAD1, PROX1, and IGF1). A predication model developed by using the top 65% of genes with high 5hmC variation has shown an impressive AUC of 0.92–0.94. Additionally, a model developed from 5hmC data on tissues has been reported to have an AUC of 0.88 when tested in cfDNA.
EpiPanGI Dx is another NGS-based methylome panel developed to aid in the diagnosis of PDA [50]. To develop this tool, researchers compared DMRs between tumor tissue and adjacent normal tissue, then cross-referenced these regions with methylated regions that were differentially identified in cfDNA from patients with malignancy and healthy patients. Overall, this assay has a predictive value of approximately 85% for the diagnosis of PDAs. Similar studies have provided other DMPs and DMRs for the diagnosis of PDA [59,60,61]. One group has demonstrated that methylation levels in terms of the MI of four genes, secreted protein acidic and rich in cysteine (SPARC), ubiquitin carboxy-terminal hydrolase L1 (UCHL1), neuronal pentraxin 2 (NPTX2), and proenkephalin (PENK), can distinguish between HC, CP, and PDA [47]. They have reported several interesting results (Table 1). The MI of all genes was higher in abnormal patients (CP and PDA) than HC, and that of SPARC distinguished early-stage PDA from CP, and thus may be useful for screening patients.
Shinjo et al. developed a panel of five markers (ADAMTS1, HOXA1, PCDH10, SEMA5A, and SPSB4) through tissue-based DNA methylation microarray analysis of PDA (n = 37), which has shown impressive diagnostic accuracy in The Cancer Genome Atlas (TCGA) dataset and an independent cohort (n = 146) [48]. For cfDNA, methylated DNA was identified with methyl-CpG binding (MBD) proteins, followed by ddPCR. The results of cfDNA testing were not encouraging (47 PDA and 14 HC). One of the five markers was positive in 49% (23/47) of patients with PDA, with a sensitivity and specificity of 49% and 86%, respectively. When mut-KRAS was added to the panel, the sensitivity improved to 69% but the specificity remained the same. Comparison of these markers in paired tissue and cfDNA samples (n = 29) indicated that all tissues had mut-KRAS and one of the five markers, whereas 19/29 (66%) cfDNA specimens had either the mut-KRAS or one of the markers; moreover, genome wide sequencing showed an 80% overlap of methylated regions and a significant correlation (R = 0.97) between cfDNA and tissue DNA methylation profiles. This study demonstrated the advantage of adding somatic mutation to epigenetic markers from all available sources in the diagnosis of PDA, and the reliable concordance of methylation profiles between cfDNA and tissues.
Overall, a cfDNA epigenetic marker can be a specific gene promoter or DMP/DMR or MI. The prevalence of some of these markers is as high as 90% in PDA and much lower in people without cancer (HC and CP), thus indicating their substantial advantages over somatic mutations [41,47]. Markers that can distinguish HC from PDA may be developed for screening, and those that can differentiate PDA from benign disease (cysts and CP) could be used in the diagnosis of suspected cases and surveillance.

3. Prognostic Value of cfDNA in PDA

Biomarkers to forecast outcomes are needed to decide on appropriate treatment approaches, particularly in patients who present with aPDA and poor performance status secondary to complications of metastatic disease, including liver failure and pain. These markers may also aid in the precise staging of early-stage PDA on imaging (discussed below). Most cfDNA studies in PDA have involved detection and quantification of mut-KRAS, because their prevalence is higher than those of other known mutations. In the past 4–5 years, the prognostic value of epigenetic markers has also been reported, but more evidence on them is needed.

3.1. cfDNA KRAS Mutations

KRAS is the most frequently mutated driver of PDA detected in tissue [62]. Multiple studies have detected KRAS mutations in cfDNA from both blood/serum and biliary fluid in patients with PDA. The poor prognostic implications of finding mut-KRAS in the cfDNA of patients with PDA were first reported in 1999 [63]. This early study examined KRAS codon 12 mutations in 44 patients with PDA. The 6-month (7% vs. 41%) and 12-month (0% vs. 24%) survival rates were significantly lower in patients with mut-KRAS than wild-type KRAS. Studies examining the prognostic value of specific KRAS mutations from cfDNA are summarized in Table 2.
Almost 22 years later, more than 3000 published studies have examined the prognostic roles of KRAS mutations in cfDNA. Mut-KRAS detection in cfDNA is typically associated with advanced stages and more distant metastasis than early stages [64,66,67,71,79]. One study identified these mutations in just 8% of patients with R-PDA but in 60% of patients with metastasis [79]. The detection of mut-KRAS in aPDAs indicates poorer outcomes, as demonstrated by multiple studies [64,67,73,74,76,80,82]. A recent meta-analysis of 48 studies examined the prognostic role of mut-KRAS detection in cfDNA to provide a more comprehensive summary [83]. The study examined 3524 patients across all stages of the disease, before and after treatment (both surgical and pharmacologic). Mut-KRAS detection was profoundly associated with the PDA outcomes, irrespective of the stage (hazard ratio (HR) = 2.42, 95% CI: 1.95–2.99 for OS and HR = 2.46, 95% CI: 2.01–3.00 for PFS) and in advanced stages (HR = 2.51, 95% CI: 1.90–3.31). The absence of detectable mutations (KRAS negativity) after either surgical or pharmacological treatment in patients with prior mut-KRAS detection has been found to indicate favorable prognosis (HR = 2.46, 95% CI: 2.01–3.00). Additionally, researchers have empirically demonstrated that, when combined with CA19-9, the detection and concentration of mut-KRAS in cfDNA have greater prognostic value than those of CA19-9 alone across all stages (HR = 2.08; 95% CI: 1.20–3.63) [33].
In addition to detection, quantification of mut-KRAS can also confer prognostic value. Among patients with mut-KRAS, higher concentrations and higher MAF often correlate with aPDA [79,84]. A multivariate analysis of factors influencing outcomes (n = 77, 35 R-PDA, 36 mPDA, and 6 LA-PDA) demonstrated that mut-KRAS concentrations >0.165 copies/L are associated with poorer OS [71]. Poorer PFS and OS were observed in these patients in the same study after univariate analysis (UVA). In contrast, KRAS fractional abundance (>0.415%) was associated with PFS in UVA but not in OS. Similarly, in another study, a higher mutational load (MAF with a cutoff of 0.351%) was associated with poor PFS (175 days vs. 85 days, HR = 2, p = 0.05) and OS (310 days vs. 142 days, HR = 2.2, p = 0.02) [80]. That study even demonstrated that an increasing MAF over time is a poor prognostic factor. Mohan et al. showed that, along with MAF, copy number gains are also associated with poor outcomes [73].
All mutations associated with KRAS genes do not uniformly influence outcomes. The specificity of the codon and particular point mutations are also influential. Variants from codon 12, specifically G12V, are associated with poorer prognosis than other aberrations [34,65,67]. In aPDA, G12D, G12V, and high T-regulatory cells (Tregs) are associated with poor outcomes independently, but the outcomes are much poorer in patients with G12V and high Tregs [68]. Furthermore, patients with G12R, compared with other variants of KRAS, have longer OS (20.4 vs. 14.5 months, HR = 0.67 (95% confidence interval (CI): 0.47–0.93)). However, in that study, only 4% of the patients had ctDNA NGS [70]. The source of the DNA (ctDNA vs. exosomal DNA) in cfDNA also affects patient outcomes. Bernard et al. noted that exosomal mutation (>5%) alone or in combination with ctDNA is an indicator of poor prognosis [66]. Overall, mut-KRAS detection, quantification, and specificity (of the point mutation) are important influences on prognostic value.

3.2. cfDNA KRAS with Other Mutations in PDA

Although most studies have focused on mut-KRAS, some studies have demonstrated that the detection of other mutations also has prognostic value [75,77,85,86]. Some cfDNA-NGS studies have distinguished the value of mut-KRAS and other mutations. In these studies, the influence of the concentration of all mutations (including mut-KRAS) detected in cfDNA on patient outcomes was assessed. At the end of a 28-week study period, the survival rate was 54% in mutation-positive patients compared with 90% in mutation-negative patients in one study [75]. Stirjker et al. also showed that detection and a higher MAF of multiple mutations independently predict poor outcomes [77].
In a study with 127 patients with PDA (53/127, mPDA), mut-KRAS detection was not found to be significantly associated with clinicopathological factors or survival [85]. However, higher concentrations (>62 ng/mL) of cfDNA are also associated with adverse features, such as distant metastatic disease (median of 92 ng/mL (15–239) vs. 58.7 ng/mL (15–240), p = 0.01), vascular invasion (median of 78 ng/mL (15–240) vs. 63 ng/mL (15–239), p = 0.03), and poorer OS (3 months in >62 ng/mL vs. 11 months in ≤62 ng ng/mL, p = 0.22, HR = 2.6). Higher cfDNA levels were also found to be an independent risk factor in multivariate analysis (HR = 2.8 (95% CI: 1.8–4.6), p = 0.01). This study first quantified cfDNA by spectrophotometry and used PCR to identify mut-KRAS (codon 12). Similar findings were reported in a recent meta-analysis of 38 studies (n = 3318), demonstrating that the detection of mut-KRAS both alone and in combination with other somatic mutations (mut-KRAS+) is associated with poor survival irrespective of disease stage [86]. In contrast, in aPDA, mut-KRAS positivity has been found to have no prognostic value, but mut-KRAS+ detection has been found to indicate poorer outcomes. Other studies have demonstrated that the detection of TP53 and HER2 exon 17, with or without mut-KRAS variants, is significantly associated with patient outcomes [27,28].

3.3. cfDNA Epigenetic Markers

cfDNA epigenetic markers have not been studied as extensively as somatic mutations in PDA (summarized in Table 3). Henriksen et al. tested a 28-gene panel (based on a literature search and their own pre-clinical studies) by methylation-specific PCR for diagnosis and subsequent staging and prognostication (in terms of survival) in PDA [42,87]. In a study of 95 patients with PDA published in 2017, the authors showed that patients with mPDA tend to have a higher number of hypermethylated genes (10.24, of their 28-gene panel) than patients with other stage tumors (stage I, 7/09; stage II, 7; and stage III, 6.77) [88]. Through multivariable regression analysis, they identified a set of genes indicating metastasis (mPDA vs. non-metastatic PDA) and advanced disease (mPDA and stage III vs. stage I/II) with reasonable sensitivity and specificity. The same group proposed risk stratification for staging and survival (summarized in Supplementary Table S2) [89]. To do so, they used the same 28-gene panel to identify potential predictors of patient outcomes by UVA and developed a prognostic prediction model to divide the patients into four risk groups by adding an American Society of Anesthesiologists score (ASA) of 3 and performance status (Eastern Cooperative Oncology Group or ECOG) >0. This stratification was applied to the whole group (all stages), stage I/II patients, and stage IV patients. Patients with more than ten hypermethylated genes also had poor prognosis.
In a post-hoc analysis of two clinical trials of patients with mPDA (Prodige 35 and Prodige 37), ctDNA positivity (detection of methylated HOXD8 and POU4F1 by PCR) was found to be a poor prognostic factor for survival [91]. Multivariate analysis has identified ctDNA positivity as an independent risk factor for PFS and OS. Other factors included for OS were CA 19-9 > 1366 U/mL and ECOG of 1–2 (compared with 0). Epigenetic changes at selected CpG sites in a group of promoter genes (p16, RARbeta, TNFRSF10C, APC, ACIN1, DAPK1, 3OST2, BCL2, and CD) were examined in whole blood samples of 30 patients with PDA (22/30 advanced stage tumors) [90]. Higher methylation levels at TNFRSF10C and ACIN1 have been found to correlate with poor survival. The former is also associated with peri-neural invasion.
Cao et al. have reported the use of epigenetic changes, 5mC, and 5hmC in the diagnosis of PDA [2]. This strategy contrasts with the examination of specific promoters in prior studies. The authors started with genome-wide profiling for 5mC and 5hmC of cfDNA in the first set of healthy individuals and patients with PDA to identify differentially methylated peaks (DMPs) for 5mC and differentially hydroxymethylated peaks (DhMPs) for 5hmC between them. By testing these DMPs and DhMPs in training and validation sets, they identified 24 and 27 significantly different markers for 5mC and 5hmC, respectively. The 5hmC markers alone were able to distinguish stage I tumors from stage II, III, or IV tumors, and the 5mC and 5hmC markers together predicted tumor size (<3 cm vs. >3 cm) and perineural invasion. Notably, these markers did not predict resectability, distant metastasis, vascular invasion, or nodal involvement of the tumor. The influence of methylation status in terms of MI on PDA outcomes has been reported by Singh et al. [47]. Higher MI of SPARC (>0.2) and NPTX2 (>0.34) has been found to be typical of metastatic cancer and poor survival. Advanced cancers (stage III/IV) had higher MI of UCLH1 (>0.42) than early stages (stage I/II). Overall, epigenetic markers can aid in precise staging and prediction of outcome, but larger studies are needed before these markers can be validated for use in clinical practice.

4. cfDNA for POR

The current standard of care for POR in PDA is to ascertain resectability through imaging (computerized tomography or CT scans) [92]. Unfortunately, imaging provides limited information and can lead to misdiagnosis of 20% of unresectable PDAs and 36% of locally advanced or metastatic tumors as R-PDAs [93,94]. In resected tumors, the recurrence rate is very high (69–90%), and long-term survival (>10 years) is merely 4% (outcomes of key AT studies are summarized in Supplementary Table S3) [5,95,96,97,98,99,100]. Addition of NAT (chemotherapy alone with/without radiation) has not been found to be significantly associated with differences in the resection rate and survival in R-PDA, BR-PDA, and LA-PDA [101]. Insufficient tumor tissue often prevents molecular testing in early-stage PDA pre-operatively. An alternative strategy using the biomarker CA 19-9 is not always dependable (as discussed above) [11,12,13]. Inadequate POR with standard imaging (and CA 19-9) would subject patients with unresectable, LA-PDA, and mPDA (with micro-metastasis) disease to unnecessary surgical attempts/interventions and prevent them from receiving life-extending systemic therapy for 2–3 months. One possible avenue involves the use of genomic testing to identify prognostic biomarkers. However, this approach has been limited by inadequate pre-operative tissue access. Very few studies have examined POR with cfDNA testing. Some have tested post-operative blood or compared pre-operative cfDNA results with post-operative samples (Table 4).
Pre-operative detection of KRAS mutation often correlates with poor outcomes, irrespective of resectability (Table 4) [71,81,84,102,103,104,105,106,107,108,109,110,111]. Alternatively, two studies have not found that pre-operative mut-KRAS detection is an independent risk factor for poor prognosis, but in both studies, the emergence of post-operative mut-KRAS was predictive of poorer outcomes [81,109]. An observational study performing cfDNA testing at the time of resection demonstrated that perioperative variations in the levels of mut-KRAS detected in cfDNA can be a valuable tool for prognostication after curative resection of PDA [107]. In that study, patients with pre-operative and/or intraoperative detection but no post-operative detection of circulating mutant KRAS had longer OS than those who were entirely KRAS-negative (pre/intra−/post – group) and patients with mut-KRAS detected in cfDNA after surgery (pre/intra+/−/post + group). Furthermore, another study reported that monitoring of post-operative levels of mut-KRAS in cfDNA with CA19-9 successfully predicted PFS in a panel of 25 patients who underwent curative resection [108]. The addition of other somatic mutations (TP53, SMAD4, NRAS, PIK3CA, and STK11) to KRAS has not been found to improve POR [112]. A recent meta-analysis of 38 studies evaluated the prognostic role of cfDNA in patients with PDA [86]. A subgroup analysis of five studies was performed to examine the prognostic value of pre- and post-operative total cfDNA levels after surgical resection. As expected, detectable postoperative levels of total cfDNA were associated with poor prognosis and greater risk of recurrence. However, the authors interestingly found that elevated levels of cfDNA pre-operatively were also associated with recurrence after surgical intervention.
To our knowledge, no studies have investigated epigenetic changes in cfDNA after surgical resection in PDA. A study in patients with gastric cancer demonstrated that genes that are initially methylated pre-operatively become demethylated after surgical resection, and that this demethylation correlates with favorable prognosis [113]. Furthermore, low levels of hypermethylated cfDNA post-operatively are correlated with improved OS and PFS in gastric cancers [114]. Similarly, methylation of IKZF1 and BCAT1 detected in cfDNA from patients with colorectal cancer who underwent colectomy was found to indicate a greater risk of residual disease and recurrence [115]. Together, these studies inspire hope for similar results in PDA.
Overall, pre-operative detection of mut-KRAS variants indicates poor outcomes in patients considered for resection. Prospective, randomized trials are needed to determine whether these variants could be used as surrogate markers to decide on (i) the use of NAT until the mut-KRAS is undetectable, (ii) continuing AT until mut-KRAS is negative, or (iii) closer surveillance in mut-KRAS positive patients after resection or completion of AT. Additionally, epigenetic markers should also be explored for POR.

5. Predictive Value of cfDNA Markers

Limited systemic options and poor prognosis hinder choosing the right chemotherapy combination for first-line treatment in mPDA and LA-PDA. Younger patients (<75 years) with good performance status (ECOG of 0–1) are typically treated with FOLFIRINOX based on the study population (PRODIGE 4/ACCORD 11 trial) unless patients have other contraindications to individual drugs (e.g., neuropathy for oxaliplatin), whereas other patients receive G-NP [6,7]. No reliable predictive biomarkers markers are available to guide the use of these combinations beyond these criteria.
The prevalence of available markers, such as BRCA 1/2 for platinum-based therapy and MSI-H for immune-checkpoint inhibitors, is very low (1–2%) [116,117]. To improve PDA outcomes, biomarkers are needed that can predict treatment response, detect treatment resistance early, and identify the benefit of rechallenging previous regimens (as in using EGFR antibodies in CRC) [118]. cfDNA testing has the potential to help in such situations. A seminal study by Leon et al. demonstrated that treatment of the underlying malignancy decreases the detectable cfDNA in patients with malignancy, thus suggesting that cfDNA has predictive value [19]. Other groups have demonstrated that cfDNA aberrations can outperform CEA in predicting the response to therapy and recurrence in patients with colorectal cancer [119].

cfDNA Somatic Mutations

The detection of somatic mutations in cfDNA at baseline and their disappearance after initiation of therapy has been consistent with stable to partial response in prior studies [72,78,81,120]. In the same studies, continued detection and/or re-emergence of mutations have been found to be reliable biomarkers for disease progression. In one study, PFS was found to be 5-fold greater in mut-KRAS negative patients than in those who continued to test positive after 4–8 weeks (PFS of 248.5 in positive vs. 50 days negative, p < 0.001) [78]. Watanabe et al. showed that its (mut-KRAS) detection after 6 months significantly correlates with unfavorable therapeutic responses to first line chemotherapy [81]. In a different study, TP53 and/or KRAS clearance, compared with a lack of clearance, have been associated with better patient response and outcomes [27].
In a study published in 2017, blood samples were collected from 27 patients before the first dose of chemotherapy (FOLFIRINOX or G-NP), on day 15 of the first cycle, and at subsequent clinic visits [69]. The mut-KRAS dynamics (changes in concentrations) in the samples collected 15 days after the first dose were indicative of treatment response and survival. Increased mut-KRAS levels correlated well with poor survival and ineffective therapy (increase vs. no increase or reduction, median PFS of 2.5 vs. 7.5 months, p = 0.03; median OS of 6.5 vs. 11.5 months, p = 0.009). Another study tracked mut-KRAS in cfDNA from 54 patients with aPDA, most of whom (37/54) received first-line gemcitabine (Gem)-based therapy (Gem-only, or Gem with erlotinib, axitinib, or everolimus) [120]. An early decrease in mut-KRAS concentrations 14 days after chemotherapy was associated with a radiographic response at 6 months. A key finding was that cfDNA can be used to predict response to chemotherapy after one cycle and can therefore be used evaluate the effectiveness (risk of adverse events vs. benefit) of first-line treatment. Cheng et al. demonstrated the predictive value of KRAS 12V in patients treated with G-NP [28].
A prospective observational study in 38 patients with PDA receiving first-line FOLFIRINOX demonstrated similar results [32]. In that study, cfDNA profiling was performed on blood samples drawn before treatment initiation and at the time of response assessment. Changes in cfDNA MAF directly correlated with treatment response and disease burden. In the described cases, apart from KRAS, the MAFs of other mutations that correlated with patient response in serial testing were for TP53, GNAS, SF3B1, CDK2NA, KEAP-1, BCL11A, PBRM1, RB1, ARID1A, and FGFR1. In an NGS-based study (n = 17 for detectable for detectable cfDNA), patients with low MAF and copy number variants were found to have either a complete or partial response to chemotherapy [121]. Systemic therapies received by patients in this group included FOLFIRINOX, G-NP, Gem-only, Gem-erlotinib, and capecitabine/oxaliplatin.
Monitoring levels of mut-KRAS cfDNA during neoadjuvant chemotherapy have also been demonstrated to predict which patients may eventually be candidates for surgical resection [66]. In a study of 34 patients with PDA (18/34 and 16/34 received G-NP and FOLFIRINOX, respectively; 26 had chemo-radiation, 22 with capecitabine, and 4 with Gem), 71% of patients who underwent resection showed a statically significant decrease in levels of mut-KRAS cfDNA compared with pre-treatment. In contrast, among the patients who did not undergo resection, 94% had no reduction in, or increased, mut-KRAS levels.
PDAs with homologous recombination deficiency (HRD) or DNA damage repair (DDR) deficiency are known to have a good outcome as they respond to platinum agents better (than PDAs without them) [122,123]. cfDNA studies that specifically looked at the prevalence of such rare mutations (somatic and germline) are limited, but commercially available panels cover most of them [124]. Detecting pathogenic mutations in DDR genes such as BRCA 1 or 2 early in diagnosis can help the treating physician consider platinum therapy (FOLFIRINOX or gemcitabine/cisplatin) in the first line and poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi) for maintenance [125,126,127,128]. A retrospective study (n = 11, 9 PDA) reported a reasonable response (complete response, partial response, and stable disease was noted in two, one, and two PDA patients, respectively) of chemotherapy-refractive HRD-positive PDA with ipilimumab/nivolumab [129]. Therefore, even though HRD-positive and DDR-positive PDAs are rare, efforts to identify them are encouraged, given their impact on treatment selection and, ultimately, the outcome.
To our knowledge, no studies have directly investigated aberrations in cfDNA methylation in response to chemotherapy in PDA. Studies have demonstrated that concentrations of methylated cfDNA at RASSF1A correlate with the response to neoadjuvant therapy in patients with breast cancer: patients with a decline in methylated RASSF1A cfDNA compared with pre-treatment often responded to neoadjuvant therapy [130]. A similar study in non-small cell lung cancer demonstrated that a decrease in methylated cfDNA in a ten-locus gene set predicts response to therapy and OS [131]. Furthermore, a study in patients with colorectal cancer demonstrated that treatment with regorafenib increases the total cfDNA; however, the concentration of total methylated cfDNA decreases [132]. This decrease in methylated cfDNA correlates with therapeutic response to treatment. Interestingly, the authors have also demonstrated that high methylated cfDNA levels during treatment correlate with minimal regorafenib response. Future studies are needed to assess the dynamics of methylation of cfDNA in response to therapy in patients with PDA to determine signatures and expression profiles that can predict response to therapy.

6. Conclusions

Efforts to improve outcomes in PDA should start with early detection, better prognostication methods to overcome the drawbacks of standard imaging and CA 19-9, dependable predictive biomarkers, and superior POR systems. Prognostic and predictive biomarkers can be exploited to develop newer targets or agents to treat PDA. The latter would help clinicians choose the optimal first-line agent for PT or NAT. The evidence provided above provides hope that cfDNA might provide an ideal source of biomarkers. The current evidence of cfDNA in PDA can be summarized as indicated in Figure 1.
Prior studies have demonstrated the feasibility of detecting genetic alterations (cfDNA) in bodily fluids (such as blood, pancreatic juice, and bile) with available technology (ddPCR or NGS) in PDA. cfDNA from blood, compared with other sources, provides an ideal non-invasive alternative to study such alterations, particularly for monitoring disease. Mutations, methylation alterations, and other genetic events such as microsatellite aberrations and loss of heterozygosity identified in cfDNA may serve as markers of malignant transformation and as biomarkers for the early stages of PDAs [2,32]. Substantial evidence supporting the use any markers for the screening and diagnosis of PDA is lacking. Nonetheless, these markers may help confirm the diagnosis if EUS-FNA is not conclusive, or if a patient has carcinoma of unknown origin with non-specific pathology.
NGS-based cfDNA mutation testing could be offered for patients with PDA to identify targetable mutations if tumor tissue is unavailable or insufficient. The reasonable concordance between tumor and cfDNA testing reported in earlier studies suggests that this method provides a reliable alternative tissue mutation profiling. Notably, the top ten mutations identified in the cfDNA of patients with PDA are currently not targetable. Epigenetic marker identification is not accessible outside experimental settings and currently has no demonstrated therapeutic value in PDA. Non-specific demethylating agents such as decitabine and azacitidine are under investigation in phase I or I/II clinical trials, which have not required specific epigenetic markers (in tissue or cfDNA) for patient enrollment [133].
cfDNA has a no significant utility as a biomarker in current clinical practice. Most studies focused on mut-KRAS variants (detected by PCR) before the era of NGS-based studies. Although some studies have demonstrated that mut-KRAS detection, concentration, and MAF can provide insight into outcomes and treatment responses in patients with PDA, other larger studies and meta-analyses have indicated the importance of other mutations [27,28,75,77,85,86]. Efforts should be made to identify those in cfDNAs and develop panels of specific mutations instead of relying on only mut-KRAS variants in future studies. cfDNA epigenetic studies to date have not addressed prognosis or treatment response sufficiently to support their use. High prevalence of epigenetic markers among the patients with PDA compared to HC gives them a distinct advantage over somatic mutations for diagnosis and screening. As additional methylation signatures for diagnosis are identified, their prognostic and predictive value must be assessed in future studies. All the facets of epigenetic markers, such as detection (non-specific markers such as 5mC and 5hmC and specific promoters), concentrations, and MI, should be considered in future investigations. Prior cfDNA testing studies have not tackled POR adequately, including the detection of micrometastasis, risk of recurrence, and need/duration of NAT, particularly in BR-PDA. Finally, cfDNA testing cannot aid in choosing the ideal first-line therapy option between FOLFIRINOX and G-NP or in identifying early resistance to therapy.
In conclusion, cfDNA testing appears promising in PDA management, but its utility is mainly restricted to identifying targetable mutations in current clinical practice. It is not ready for diagnosis, screening, prognostication, prediction treatment response, monitoring of disease burden, and POR. The low prevalence of mutations in cfDNA hinders research and the drawing of reliable conclusions. Studies have suggested that epigenetic biomarkers may aid in all the critical areas discussed above. Future studies should further pursue epigenetic biomarkers to identify factors that may be exploited for therapeutic purposes.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cancers14143453/s1, Figure S1: Summary of cfDNA studies in PDA, Table S1: Summary of studies examining somatic mutations for diagnosis of PDA with concordance rate compared to tissue, Table S2: Risk stratification based on 28-gene panel and performance status in PDA, Table S3: Outcomes in prominent adjuvant therapy trials in pancreatic ductal adenocarcinoma [35,36,134,135,136,137,138].

Author Contributions

Conceptualization, A.M.; writing—original draft preparation, A.S. and A.M.; writing—review and editing, S.A. and S.P.N. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Surveillance, Epidemiology, and End Results (SEER) Program. SEER*Stat Database: Incidence—SEER Research Data, 9 Registries, Nov 2019 Sub (1975–2017). Available online: www.seer.cancer.gov (accessed on 15 January 2022).
  2. Cao, F.; Wei, A.; Hu, X.; He, Y.; Zhang, J.; Xia, L.; Tu, K.; Yuan, J.; Guo, Z.; Liu, H.; et al. Integrated epigenetic biomarkers in circulating cell-free DNA as a robust classifier for pancreatic cancer. Clin. Epigenet. 2020, 12, 112. [Google Scholar] [CrossRef]
  3. Callery, M.P.; Chang, K.J.; Fishman, E.K.; Talamonti, M.S.; William Traverso, L.; Linehan, D.C. Pretreatment assessment of resectable and borderline resectable pancreatic cancer: Expert consensus statement. Ann. Surg. Oncol. 2009, 16, 1727–1733. [Google Scholar] [CrossRef] [PubMed]
  4. Mizrahi, J.D.; Surana, R.; Valle, J.W.; Shroff, R.T. Pancreatic cancer. Lancet 2020, 395, 2008–2020. [Google Scholar] [CrossRef]
  5. Jones, R.P.; Psarelli, E.-E.; Jackson, R.; Ghaneh, P.; Halloran, C.M.; Palmer, D.H.; Campbell, F.; Valle, J.W.; Faluyi, O.; O’Reilly, D.A.; et al. Patterns of Recurrence After Resection of Pancreatic Ductal Adenocarcinoma. JAMA Surg. 2019, 154, 1038. [Google Scholar] [CrossRef] [PubMed]
  6. Conroy, T.; Desseigne, F.; Ychou, M.; Bouché, O.; Guimbaud, R.; Bécouarn, Y.; Adenis, A.; Raoul, J.-L.; Gourgou-Bourgade, S.; De La Fouchardière, C.; et al. FOLFIRINOX versus Gemcitabine for Metastatic Pancreatic Cancer. N. Engl. J. Med. 2011, 364, 1817–1825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Von Hoff, D.D.; Ervin, T.; Arena, F.P.; Chiorean, E.G.; Infante, J.; Moore, M.; Seay, T.; Tjulandin, S.A.; Ma, W.W.; Saleh, M.N.; et al. Increased Survival in Pancreatic Cancer with nab-Paclitaxel plus Gemcitabine. N. Engl. J. Med. 2013, 369, 1691–1703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Tempero, M.A.; Malafa, M.P.; Al-Hawary, M.; Behrman, S.W.; Benson, A.B.; Cardin, D.B.; Chiorean, E.G.; Chung, V.; Czito, B.; Del Chiaro, M.; et al. Pancreatic Adenocarcinoma, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. 2021, 19, 439–457. [Google Scholar] [CrossRef] [PubMed]
  9. Poruk, K.E.; Gay, D.Z.; Brown, K.; Mulvihill, J.D.; Boucher, K.M.; Scaife, C.L.; Firpo, M.A.; Mulvihill, S.J. The clinical utility of CA 19-9 in pancreatic adenocarcinoma: Diagnostic and prognostic updates. Curr. Mol. Med. 2013, 13, 340–351. [Google Scholar] [CrossRef]
  10. Elbanna, K.Y.; Jang, H.J.; Kim, T.K. Imaging diagnosis and staging of pancreatic ductal adenocarcinoma: A comprehensive review. Insights Imaging 2020, 11, 58. [Google Scholar] [CrossRef] [Green Version]
  11. Tsen, A.; Barbara, M.; Rosenkranz, L. Dilemma of elevated CA 19-9 in biliary pathology. Pancreatology 2018, 18, 862–867. [Google Scholar] [CrossRef]
  12. Tempero, M.A.; Uchida, E.; Takasaki, H.; Burnett, D.A.; Steplewski, Z.; Pour, P.M. Relationship of carbohydrate antigen 19-9 and Lewis antigens in pancreatic cancer. Cancer Res. 1987, 47, 5501–5503. [Google Scholar]
  13. Liu, C.; Deng, S.; Jin, K.; Gong, Y.; Cheng, H.; Fan, Z.; Qian, Y.; Huang, Q.; Ni, Q.; Luo, G.; et al. Lewis antigen-negative pancreatic cancer: An aggressive subgroup. Int. J. Oncol. 2020, 56, 900–908. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Corcoran, R.B.; Chabner, B.A. Application of Cell-free DNA Analysis to Cancer Treatment. N. Engl. J. Med. 2018, 379, 1754–1765. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Chen, Q.; Zhang, Z.-H.; Wang, S.; Lang, J.-H. Circulating Cell-Free DNA or Circulating Tumor DNA in the Management of Ovarian and Endometrial Cancer. OncoTargets Ther. 2019, 12, 11517–11530. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Ashworth, T.R. A Case of Cancer in Which Cells Similar to Those in the Tumours Were Seen in the Blood after Death. Med. J. Aust. 1869, 14, 146–147. [Google Scholar]
  17. Engell, H.C. Cancer cells in the circulating blood; a clinical study on the occurrence of cancer cells in the peripheral blood and in venous blood draining the tumour area at operation. Acta Chir. Scand. Suppl. 1955, 201, 1–70. [Google Scholar] [PubMed]
  18. Mandel, P.; Metaise, P. Nuclear Acids In Human Blood Plasma. C. R. Seances Soc. Biol. Fil. 1948, 142, 241–243. [Google Scholar]
  19. Leon, S.A.; Shapiro, B.; Sklaroff, D.M.; Yaros, M.J. Free DNA in the serum of cancer patients and the effect of therapy. Cancer Res. 1977, 37, 646–650. [Google Scholar]
  20. Vasioukhin, V.; Anker, P.; Maurice, P.; Lyautey, J.; Lederrey, C.; Stroun, M. Point mutations of the N-ras gene in the blood plasma DNA of patients with myelodysplastic syndrome or acute myelogenous leukaemia. Br. J. Haematol. 1994, 86, 774–779. [Google Scholar] [CrossRef]
  21. Crowley, E.; Di Nicolantonio, F.; Loupakis, F.; Bardelli, A. Liquid biopsy: Monitoring cancer-genetics in the blood. Nat. Rev. Clin. Oncol. 2013, 10, 472–484. [Google Scholar] [CrossRef]
  22. Mouliere, F.; Thierry, A.R. The importance of examining the proportion of circulating DNA originating from tumor, microenvironment and normal cells in colorectal cancer patients. Expert Opin. Biol. Ther. 2012, 12 (Suppl. 1), S209–S215. [Google Scholar] [CrossRef] [PubMed]
  23. Gall, T.M.H.; Belete, S.; Khanderia, E.; Frampton, A.E.; Jiao, L.R. Circulating Tumor Cells and Cell-Free DNA in Pancreatic Ductal Adenocarcinoma. Am. J. Pathol. 2019, 189, 71–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Sorenson, G.D.; Pribish, D.M.; Valone, F.H.; Memoli, V.A.; Bzik, D.J.; Yao, S.L. Soluble normal and mutated DNA sequences from single-copy genes in human blood. Cancer Epidemiol. Biomark. Prev. 1994, 3, 67–71. [Google Scholar]
  25. George, B.; Haberberger, J.; Ferguson, N.L.; Gjoerup, O.; McGregor, K.; Hendifar, A.E.; Laheru, D.A.; Weekes, C.D.; Ross, J.S.; Hemmerich, A. Correlation between comprehensive genomic profiling (CGP) utilizing tissue-based testing (T-CGP) and cell-free DNA (cfDNA) in patients (pts) with pancreatic ductal adenocarcinoma (PDAC). J. Clin. Oncol. 2021, 39, 422. [Google Scholar] [CrossRef]
  26. George, B.; Haberberger, J.; Ferguson, N.L.; Gjoerup, O.; McGregor, K.; Hendifar, A.E.; Laheru, D.A.; Weekes, C.D.; Ross, J.S.; Hemmerich, A. Comprehensive genomic profiling (CGP) utilizing cell-free DNA (cfDNA) in patients (pts) with pancreatic ductal adenocarcinoma (PDAC). J. Clin. Oncol. 2021, 39, 421. [Google Scholar] [CrossRef]
  27. Botrus, G.; Fu, Y.; Sonbol, M.B.; Drusbosky, L.; Ahn, D.H.; Borad, M.J.; Starr, J.S.; Jones, J.C.; Raman, P.; Mody, K.; et al. Serial cell-free DNA (cfDNA) sampling in advanced pancreatic ductal adenocarcinoma (PDAC) patients may predict therapeutic outcome. J. Clin. Oncol. 2021, 39, 423. [Google Scholar] [CrossRef]
  28. Cheng, H.; Liu, C.; Jiang, J.; Luo, G.; Lu, Y.; Jin, K.; Guo, M.; Zhang, Z.; Xu, J.; Liu, L.; et al. Analysis of ctDNA to predict prognosis and monitor treatment responses in metastatic pancreatic cancer patients. Int. J. Cancer 2017, 140, 2344–2350. [Google Scholar] [CrossRef]
  29. Zill, O.A.; Banks, K.C.; Fairclough, S.R.; Mortimer, S.A.; Vowles, J.V.; Mokhtari, R.; Gandara, D.R.; Mack, P.C.; Odegaard, J.I.; Nagy, R.J.; et al. The Landscape of Actionable Genomic Alterations in Cell-Free Circulating Tumor DNA from 21,807 Advanced Cancer Patients. Clin. Cancer Res. 2018, 24, 3528–3538. [Google Scholar] [CrossRef] [Green Version]
  30. Bettegowda, C.; Sausen, M.; Leary, R.J.; Kinde, I.; Wang, Y.; Agrawal, N.; Bartlett, B.R.; Wang, H.; Luber, B.; Alani, R.M.; et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci. Transl. Med. 2014, 6, 224ra224. [Google Scholar] [CrossRef] [Green Version]
  31. Luchini, C.; Veronese, N.; Nottegar, A.; Cappelletti, V.; Daidone, M.G.; Smith, L.; Parris, C.; Brosens, L.A.A.; Caruso, M.G.; Cheng, L.; et al. Liquid Biopsy as Surrogate for Tissue for Molecular Profiling in Pancreatic Cancer: A Meta-Analysis Towards Precision Medicine. Cancers 2019, 11, 1152. [Google Scholar] [CrossRef] [Green Version]
  32. Wei, T.; Zhang, Q.; Li, X.; Su, W.; Li, G.; Ma, T.; Gao, S.; Lou, J.; Que, R.; Zheng, L.; et al. Monitoring Tumor Burden in Response to FOLFIRINOX Chemotherapy Via Profiling Circulating Cell-Free DNA in Pancreatic Cancer. Mol. Cancer Ther. 2019, 18, 196–203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Kim, M.K.; Woo, S.M.; Park, B.; Yoon, K.-A.; Kim, Y.H.; Joo, J.; Park, S.J.; Kong, S.-Y. Prognostic impact of KRAS mutation in cell-free DNA in patients with pancreatic cancer. Ann. Oncol. 2017, 28, V9. [Google Scholar] [CrossRef] [Green Version]
  34. Kinugasa, H.; Nouso, K.; Miyahara, K.; Morimoto, Y.; Dohi, C.; Tsutsumi, K.; Kato, H.; Matsubara, T.; Okada, H.; Yamamoto, K. Detection of K-rasgene mutation by liquid biopsy in patients with pancreatic cancer. Cancer 2015, 121, 2271–2280. [Google Scholar] [CrossRef] [PubMed]
  35. Brychta, N.; Krahn, T.; Von Ahsen, O. Detection of KRAS Mutations in Circulating Tumor DNA by Digital PCR in Early Stages of Pancreatic Cancer. Clin. Chem. 2016, 62, 1482–1491. [Google Scholar] [CrossRef] [Green Version]
  36. Cohen, J.D.; Li, L.; Wang, Y.; Thoburn, C.; Afsari, B.; Danilova, L.; Douville, C.; Javed, A.A.; Wong, F.; Mattox, A.; et al. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science 2018, 359, 926–930. [Google Scholar] [CrossRef] [Green Version]
  37. Ehrlich, M. DNA methylation in cancer: Too much, but also too little. Oncogene 2002, 21, 5400–5413. [Google Scholar] [CrossRef] [Green Version]
  38. Melnikov, A.A.; Scholtens, D.; Talamonti, M.S.; Bentrem, D.J.; Levenson, V.V. Methylation profile of circulating plasma DNA in patients with pancreatic cancer. J. Surg. Oncol. 2009, 99, 119–122. [Google Scholar] [CrossRef]
  39. Liggett, T.; Melnikov, A.; Yi, Q.L.; Replogle, C.; Brand, R.; Kaul, K.; Talamonti, M.; Abrams, R.A.; Levenson, V. Differential methylation of cell-free circulating DNA among patients with pancreatic cancer versus chronic pancreatitis. Cancer 2010, 116, 1674–1680. [Google Scholar] [CrossRef]
  40. Yi, J.M.; Guzzetta, A.A.; Bailey, V.J.; Downing, S.R.; Van Neste, L.; Chiappinelli, K.B.; Keeley, B.P.; Stark, A.; Herrera, A.; Wolfgang, C.; et al. Novel methylation biomarker panel for the early detection of pancreatic cancer. Clin. Cancer Res. 2013, 19, 6544–6555. [Google Scholar] [CrossRef] [Green Version]
  41. Eissa, M.A.L.; Lerner, L.; Abdelfatah, E.; Shankar, N.; Canner, J.K.; Hasan, N.M.; Yaghoobi, V.; Huang, B.; Kerner, Z.; Takaesu, F.; et al. Promoter methylation of ADAMTS1 and BNC1 as potential biomarkers for early detection of pancreatic cancer in blood. Clin. Epigenet. 2019, 11, 59. [Google Scholar] [CrossRef]
  42. Henriksen, S.D.; Madsen, P.H.; Larsen, A.C.; Johansen, M.B.; Drewes, A.M.; Pedersen, I.S.; Krarup, H.; Thorlacius-Ussing, O. Cell-free DNA promoter hypermethylation in plasma as a diagnostic marker for pancreatic adenocarcinoma. Clin. Epigenet. 2016, 8, 117. [Google Scholar] [CrossRef] [Green Version]
  43. Guler, G.D.; Ning, Y.; Ku, C.J.; Phillips, T.; McCarthy, E.; Ellison, C.K.; Bergamaschi, A.; Collin, F.; Lloyd, P.; Scott, A.; et al. Detection of early stage pancreatic cancer using 5-hydroxymethylcytosine signatures in circulating cell free DNA. Nat. Commun. 2020, 11, 5270. [Google Scholar] [CrossRef] [PubMed]
  44. Li, S.; Wang, L.; Zhao, Q.; Wang, Z.; Lu, S.; Kang, Y.; Jin, G.; Tian, J. Genome-Wide Analysis of Cell-Free DNA Methylation Profiling for the Early Diagnosis of Pancreatic Cancer. Front. Genet. 2020, 11, 596078. [Google Scholar] [CrossRef] [PubMed]
  45. Ying, L.; Sharma, A.; Chhoda, A.; Ruzgar, N.; Hasan, N.; Kwak, R.; Wolfgang, C.L.; Wang, T.H.; Kunstman, J.W.; Salem, R.R.; et al. Methylation-based Cell-free DNA Signature for Early Detection of Pancreatic Cancer. Pancreas 2021, 50, 1267–1273. [Google Scholar] [CrossRef] [PubMed]
  46. Manoochehri, M.; Wu, Y.; Giese, N.A.; Strobel, O.; Kutschmann, S.; Haller, F.; Hoheisel, J.D.; Moskalev, E.A.; Hackert, T.; Bauer, A.S. SST gene hypermethylation acts as a pan-cancer marker for pancreatic ductal adenocarcinoma and multiple other tumors: Toward its use for blood-based diagnosis. Mol. Oncol. 2020, 14, 1252–1267. [Google Scholar] [CrossRef] [Green Version]
  47. Singh, N.; Rashid, S.; Dash, N.R.; Gupta, S.; Saraya, A. Clinical significance of promoter methylation status of tumor suppressor genes in circulating DNA of pancreatic cancer patients. J. Cancer Res. Clin. Oncol. 2020, 146, 897–907. [Google Scholar] [CrossRef]
  48. Shinjo, K.; Hara, K.; Nagae, G.; Umeda, T.; Katsushima, K.; Suzuki, M.; Murofushi, Y.; Umezu, Y.; Takeuchi, I.; Takahashi, S.; et al. A novel sensitive detection method for DNA methylation in circulating free DNA of pancreatic cancer. PLoS ONE 2020, 15, e0233782. [Google Scholar] [CrossRef]
  49. Fujimoto, Y.; Suehiro, Y.; Kaino, S.; Suenaga, S.; Tsuyama, T.; Matsui, H.; Higaki, S.; Fujii, I.; Suzuki, C.; Hoshida, T.; et al. Combination of CA19-9 and Blood Free-Circulating Methylated RUNX3 May Be Useful to Diagnose Stage I Pancreatic Cancer. Oncology 2021, 99, 234–239. [Google Scholar] [CrossRef]
  50. Kandimalla, R.; Xu, J.; Link, A.; Matsuyama, T.; Yamamura, K.; Parker, M.I.; Uetake, H.; Balaguer, F.; Borazanci, E.; Tsai, S.; et al. EpiPanGI Dx: A Cell-free DNA Methylation Fingerprint for the Early Detection of Gastrointestinal Cancers. Clin. Cancer Res. 2021, 27, 6135–6144. [Google Scholar] [CrossRef]
  51. Vrba, L.; Futscher, B.W.; Oshiro, M.; Watts, G.S.; Menashi, E.; Hu, C.; Hammad, H.; Pennington, D.R.; Golconda, U.; Gavini, H.; et al. Liquid biopsy, using a novel DNA methylation signature, distinguishes pancreatic adenocarcinoma from benign pancreatic disease. Clin. Epigenet. 2022, 14, 28. [Google Scholar] [CrossRef]
  52. Li, X.B.; Ma, J.; Liu, Z.W.; He, W.F.; Li, Z.Z.; Cui, M.; Hao, H.; Zhou, G.P.; You, L.; Wang, J.; et al. Non-invasive detection of pancreatic cancer by measuring DNA methylation of Basonuclin 1 and Septin 9 in plasma. Chin. Med. J. 2019, 132, 1504–1506. [Google Scholar] [CrossRef]
  53. Melson, J.; Li, Y.; Cassinotti, E.; Melnikov, A.; Boni, L.; Ai, J.; Greenspan, M.; Mobarhan, S.; Levenson, V.; Deng, Y. Commonality and differences of methylation signatures in the plasma of patients with pancreatic cancer and colorectal cancer. Int. J. Cancer 2014, 134, 2656–2662. [Google Scholar] [CrossRef]
  54. Park, J.W.; Baek, I.H.; Kim, Y.T. Preliminary Study Analyzing the Methylated Genes in the Plasma of Patients with Pancreatic Cancer. Scand. J. Surg. 2012, 101, 38–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Park, J.K.; Ryu, J.K.; Yoon, W.J.; Lee, S.H.; Lee, G.Y.; Jeong, K.S.; Kim, Y.T.; Yoon, Y.B. The role of quantitative NPTX2 hypermethylation as a novel serum diagnostic marker in pancreatic cancer. Pancreas 2012, 41, 95–101. [Google Scholar] [CrossRef] [PubMed]
  56. Henriksen, S.D.; Stubbe, B.E.; Madsen, P.H.; Johansen, J.S.; Jensen, B.V.; Hansen, C.P.; Johansen, M.N.; Pedersen, I.S.; Krarup, H.; Thorlacius-Ussing, O. Cell-free DNA promoter hypermethylation as a diagnostic marker for pancreatic ductal adenocarcinoma—An external validation study. Pancreatology 2021, 21, 1081–1091. [Google Scholar] [CrossRef] [PubMed]
  57. Bhattacharyya, S.; Yu, Y.; Suzuki, M.; Campbell, N.; Mazdo, J.; Vasanthakumar, A.; Bhagat, T.D.; Nischal, S.; Christopeit, M.; Parekh, S.; et al. Genome-wide hydroxymethylation tested using the HELP-GT assay shows redistribution in cancer. Nucleic Acids Res. 2013, 41, e157. [Google Scholar] [CrossRef] [Green Version]
  58. Ficz, G.; Branco, M.R.; Seisenberger, S.; Santos, F.; Krueger, F.; Hore, T.A.; Marques, C.J.; Andrews, S.; Reik, W. Dynamic regulation of 5-hydroxymethylcytosine in mouse ES cells and during differentiation. Nature 2011, 473, 398–402. [Google Scholar] [CrossRef]
  59. Wang, Z.; Lu, Y.; Fornage, M.; Jiao, L.; Shen, J.; Li, D.; Wei, P. Identification of novel susceptibility methylation loci for pancreatic cancer in a two-phase epigenome-wide association study. Epigenetics 2022, 1–16. [Google Scholar] [CrossRef]
  60. Pedersen, K.S.; Bamlet, W.R.; Oberg, A.L.; De Andrade, M.; Matsumoto, M.E.; Tang, H.; Thibodeau, S.N.; Petersen, G.M.; Wang, L. Leukocyte DNA Methylation Signature Differentiates Pancreatic Cancer Patients from Healthy Controls. PLoS ONE 2011, 6, e18223. [Google Scholar] [CrossRef] [Green Version]
  61. Zhu, J.; Yang, Y.; Kisiel, J.B.; Mahoney, D.W.; Michaud, D.S.; Guo, X.; Taylor, W.R.; Shu, X.-O.; Shu, X.; Liu, D.; et al. Integrating Genome and Methylome Data to Identify Candidate DNA Methylation Biomarkers for Pancreatic Cancer Risk. Cancer Epidemiol. Biomark. Prev. 2021, 30, 2079–2087. [Google Scholar] [CrossRef]
  62. Bryant, K.L.; Mancias, J.D.; Kimmelman, A.C.; Der, C.J. KRAS: Feeding pancreatic cancer proliferation. Trends Biochem. Sci. 2014, 39, 91–100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Castells, A.; Puig, P.; Móra, J.; Boadas, J.; Boix, L.; Urgell, E.; Solé, M.; Capellà, G.; Lluís, F.; Fernández-Cruz, L.; et al. K-ras mutations in DNA extracted from the plasma of patients with pancreatic carcinoma: Diagnostic utility and prognostic significance. J. Clin. Oncol. 1999, 17, 578–584. [Google Scholar] [CrossRef] [PubMed]
  64. Adamo, P.; Cowley, C.M.; Neal, C.P.; Mistry, V.; Page, K.; Dennison, A.R.; Isherwood, J.; Hastings, R.; Luo, J.; Moore, D.A.; et al. Profiling tumour heterogeneity through circulating tumour DNA in patients with pancreatic cancer. Oncotarget 2017, 8, 87221–87233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Ako, S.; Nouso, K.; Kinugasa, H.; Dohi, C.; Matushita, H.; Mizukawa, S.; Muro, S.; Akimoto, Y.; Uchida, D.; Tomoda, T.; et al. Utility of serum DNA as a marker for KRAS mutations in pancreatic cancer tissue. Pancreatology 2017, 17, 285–290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Bernard, V.; Kim, D.U.; San Lucas, F.A.; Castillo, J.; Allenson, K.; Mulu, F.C.; Stephens, B.M.; Huang, J.; Semaan, A.; Guerrero, P.A.; et al. Circulating Nucleic Acids Are Associated With Outcomes of Patients With Pancreatic Cancer. Gastroenterology 2019, 156, 108–118.e4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Chen, H.; Tu, H.; Meng, Z.Q.; Chen, Z.; Wang, P.; Liu, L.M. K-ras mutational status predicts poor prognosis in unresectable pancreatic cancer. Eur. J. Surg. Oncol. 2010, 36, 657–662. [Google Scholar] [CrossRef] [PubMed]
  68. Cheng, H.; Luo, G.; Jin, K.; Fan, Z.; Huang, Q.; Gong, Y.; Xu, J.; Yu, X.; Liu, C. Kras mutation correlating with circulating regulatory T cells predicts the prognosis of advanced pancreatic cancer patients. Cancer Med. 2020, 9, 2153–2159. [Google Scholar] [CrossRef]
  69. Del Re, M.; Vivaldi, C.; Rofi, E.; Vasile, E.; Miccoli, M.; Caparello, C.; d’Arienzo, P.D.; Fornaro, L.; Falcone, A.; Danesi, R. Early changes in plasma DNA levels of mutant KRAS as a sensitive marker of response to chemotherapy in pancreatic cancer. Sci. Rep. 2017, 7, 7931. [Google Scholar] [CrossRef]
  70. Diehl, A.; Hannan, L.M.; Chiorean, E.G. Prognostic value of KRAS and PI3K pathway mutations for advanced pancreatic ductal adenocarcinoma (PDAC) patients (pts). J. Clin. Oncol. 2021, 39, 424. [Google Scholar] [CrossRef]
  71. Kim, M.K.; Woo, S.M.; Park, B.; Yoon, K.A.; Kim, Y.H.; Joo, J.; Lee, W.J.; Han, S.S.; Park, S.J.; Kong, S.Y. Prognostic Implications of Multiplex Detection of KRAS Mutations in Cell-Free DNA from Patients with Pancreatic Ductal Adenocarcinoma. Clin. Chem. 2018, 64, 726–734. [Google Scholar] [CrossRef] [Green Version]
  72. Van Laethem, J.L.; Riess, H.; Jassem, J.; Haas, M.; Martens, U.M.; Weekes, C.; Peeters, M.; Ross, P.; Bridgewater, J.; Melichar, B.; et al. Phase I/II Study of Refametinib (BAY 86-9766) in Combination with Gemcitabine in Advanced Pancreatic cancer. Target. Oncol. 2017, 12, 97–109. [Google Scholar] [CrossRef] [PubMed]
  73. Mohan, S.; Ayub, M.; Rothwell, D.G.; Gulati, S.; Kilerci, B.; Hollebecque, A.; Sun Leong, H.; Smith, N.K.; Sahoo, S.; Descamps, T.; et al. Analysis of circulating cell-free DNA identifies KRAS copy number gain and mutation as a novel prognostic marker in Pancreatic cancer. Sci. Rep. 2019, 9, 11610. [Google Scholar] [CrossRef] [PubMed]
  74. Perets, R.; Greenberg, O.; Shentzer, T.; Semenisty, V.; Epelbaum, R.; Bick, T.; Sarji, S.; Ben-Izhak, O.; Sabo, E.; Hershkovitz, D. Mutant KRAS Circulating Tumor DNA Is an Accurate Tool for Pancreatic Cancer Monitoring. Oncologist 2018, 23, 566–572. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Pishvaian, M.J.; Joseph Bender, R.; Matrisian, L.M.; Rahib, L.; Hendifar, A.; Hoos, W.A.; Mikhail, S.; Chung, V.; Picozzi, V.; Heartwell, C.; et al. A pilot study evaluating concordance between blood-based and patient-matched tumor molecular testing within pancreatic cancer patients participating in the Know Your Tumor (KYT) initiative. Oncotarget 2017, 8, 83446–83456. [Google Scholar] [CrossRef] [Green Version]
  76. Semrad, T.; Barzi, A.; Lenz, H.J.; Hutchins, I.M.; Kim, E.J.; Gong, I.Y.; Tanaka, M.; Beckett, L.; Holland, W.; Burich, R.A.; et al. Pharmacodynamic separation of gemcitabine and erlotinib in locally advanced or metastatic pancreatic cancer: Therapeutic and biomarker results. Int. J. Clin. Oncol. 2015, 20, 518–524. [Google Scholar] [CrossRef] [Green Version]
  77. Strijker, M.; Soer, E.C.; de Pastena, M.; Creemers, A.; Balduzzi, A.; Beagan, J.J.; Busch, O.R.; van Delden, O.M.; Halfwerk, H.; van Hooft, J.E.; et al. Circulating tumor DNA quantity is related to tumor volume and both predict survival in metastatic pancreatic ductal adenocarcinoma. Int. J. Cancer 2020, 146, 1445–1456. [Google Scholar] [CrossRef]
  78. Sugimori, M.; Sugimori, K.; Tsuchiya, H.; Suzuki, Y.; Tsuyuki, S.; Kaneta, Y.; Hirotani, A.; Sanga, K.; Tozuka, Y.; Komiyama, S.; et al. Quantitative monitoring of circulating tumor DNA in patients with advanced pancreatic cancer undergoing chemotherapy. Cancer Sci. 2020, 111, 266–278. [Google Scholar] [CrossRef]
  79. Takai, E.; Totoki, Y.; Nakamura, H.; Morizane, C.; Nara, S.; Hama, N.; Suzuki, M.; Furukawa, E.; Kato, M.; Hayashi, H.; et al. Clinical utility of circulating tumor DNA for molecular assessment in pancreatic cancer. Sci. Rep. 2015, 5, 18425. [Google Scholar] [CrossRef] [Green Version]
  80. Toledano-Fonseca, M.; Cano, M.T.; Inga, E.; Rodríguez-Alonso, R.; Gómez-España, M.A.; Guil-Luna, S.; Mena-Osuna, R.; de la Haba-Rodríguez, J.R.; Rodríguez-Ariza, A.; Aranda, E. Circulating Cell-Free DNA-Based Liquid Biopsy Markers for the Non-Invasive Prognosis and Monitoring of Metastatic Pancreatic Cancer. Cancers 2020, 12, 1754. [Google Scholar] [CrossRef]
  81. Watanabe, F.; Suzuki, K.; Tamaki, S.; Abe, I.; Endo, Y.; Takayama, Y.; Ishikawa, H.; Kakizawa, N.; Saito, M.; Futsuhara, K.; et al. Longitudinal monitoring of KRAS-mutated circulating tumor DNA enables the prediction of prognosis and therapeutic responses in patients with pancreatic cancer. PLoS ONE 2019, 14, e0227366. [Google Scholar] [CrossRef]
  82. Tjensvoll, K.; Lapin, M.; Buhl, T.; Oltedal, S.; Steen-Ottosen Berry, K.; Gilje, B.; Søreide, J.A.; Javle, M.; Nordgård, O.; Smaaland, R. Clinical relevance of circulating KRAS mutated DNA in plasma from patients with advanced pancreatic cancer. Mol. Oncol. 2016, 10, 635–643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Milin-Lazovic, J.; Madzarevic, P.; Rajovic, N.; Djordjevic, V.; Milic, N.; Pavlovic, S.; Veljkovic, N.; Milic, N.M.; Radenkovic, D. Meta-Analysis of Circulating Cell-Free DNA’s Role in the Prognosis of Pancreatic Cancer. Cancers 2021, 13, 3378. [Google Scholar] [CrossRef] [PubMed]
  84. Wang, Z.Y.; Ding, X.Q.; Zhu, H.; Wang, R.X.; Pan, X.R.; Tong, J.H. KRAS Mutant Allele Fraction in Circulating Cell-Free DNA Correlates With Clinical Stage in Pancreatic Cancer Patients. Front. Oncol. 2019, 9, 1295. [Google Scholar] [CrossRef] [PubMed]
  85. Singh, N.; Gupta, S.; Pandey, R.M.; Chauhan, S.S.; Saraya, A. High levels of cell-free circulating nucleic acids in pancreatic cancer are associated with vascular encasement, metastasis and poor survival. Cancer Investig. 2015, 33, 78–85. [Google Scholar] [CrossRef] [PubMed]
  86. Bunduc, S.; Gede, N.; Váncsa, S.; Lillik, V.; Kiss, S.; Dembrovszky, F.; Eróss, B.; Szakács, Z.; Gheorghe, C.; Mikó, A.; et al. Prognostic role of cell-free DNA biomarkers in pancreatic adenocarcinoma: A systematic review and meta-analysis. Crit. Rev. Oncol. Hematol. 2022, 169, 103548. [Google Scholar] [CrossRef]
  87. Henriksen, S.D.; Madsen, P.H.; Krarup, H.; Thorlacius-Ussing, O. DNA Hypermethylation as a Blood-Based Marker for Pancreatic Cancer: A Literature Review. Pancreas 2015, 44, 1036–1045. [Google Scholar] [CrossRef]
  88. Henriksen, S.D.; Madsen, P.H.; Larsen, A.C.; Johansen, M.B.; Pedersen, I.S.; Krarup, H.; Thorlacius-Ussing, O. Promoter hypermethylation in plasma-derived cell-free DNA as a prognostic marker for pancreatic adenocarcinoma staging. Int. J. Cancer 2017, 141, 2489–2497. [Google Scholar] [CrossRef] [Green Version]
  89. Henriksen, S.D.; Madsen, P.H.; Larsen, A.C.; Johansen, M.B.; Pedersen, I.S.; Krarup, H.; Thorlacius-Ussing, O. Cell-free DNA promoter hypermethylation in plasma as a predictive marker for survival of patients with pancreatic adenocarcinoma. Oncotarget 2017, 8, 93942–93956. [Google Scholar] [CrossRef] [Green Version]
  90. Dauksa, A.; Gulbinas, A.; Barauskas, G.; Pundzius, J.; Oldenburg, J.; El-Maarri, O. Whole Blood DNA Aberrant Methylation in Pancreatic Adenocarcinoma Shows Association with the Course of the Disease: A Pilot Study. PLoS ONE 2012, 7, e37509. [Google Scholar] [CrossRef] [Green Version]
  91. Pietrasz, D.; Wang-Renault, S.; Taieb, J.; Dahan, L.; Postel, M.; Durand-Labrunie, J.; Le Malicot, K.; Mulot, C.; Rinaldi, Y.; Phelip, J.-M.; et al. Prognostic value of circulating tumour DNA in metastatic pancreatic cancer patients: Post-hoc analyses of two clinical trials. Br. J. Cancer 2022, 126, 440–448. [Google Scholar] [CrossRef]
  92. Ryan, D.P.; Hong, T.S.; Bardeesy, N. Pancreatic Adenocarcinoma. N. Engl. J. Med. 2014, 371, 1039–1049. [Google Scholar] [CrossRef] [PubMed]
  93. Ta, R.; O’Connor, D.B.; Sulistijo, A.; Chung, B.; Conlon, K.C. The Role of Staging Laparoscopy in Resectable and Borderline Resectable Pancreatic Cancer: A Systematic Review and Meta-Analysis. Dig. Surg. 2019, 36, 251–260. [Google Scholar] [CrossRef]
  94. Park, S.; Jang, J.K.; Byun, J.H.; Kim, J.H.; Lee, S.S.; Kim, H.J.; Hong, S.B.; Park, S.H. CT in the prediction of margin-negative resection in pancreatic cancer following neoadjuvant treatment: A systematic review and meta-analysis. Eur. Radiol. 2021, 31, 3383–3393. [Google Scholar] [CrossRef]
  95. Groot, V.P.; Daamen, L.A.; He, J.; Wolfgang, C.L.; Molenaar, I.Q. Patterns of Recurrence after Surgery for Pancreatic Cancer. In Textbook of Pancreatic Cancer; Springer International Publishing: Berlin/Heidelberg, Germany, 2021; pp. 1153–1168. [Google Scholar]
  96. Paniccia, A.; Hosokawa, P.; Henderson, W.; Schulick, R.D.; Edil, B.H.; Mccarter, M.D.; Gajdos, C. Characteristics of 10-Year Survivors of Pancreatic Ductal Adenocarcinoma. JAMA Surg. 2015, 150, 701. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Oettle, H.; Post, S.; Neuhaus, P.; Gellert, K.; Langrehr, J.; Ridwelski, K.; Schramm, H.; Fahlke, J.; Zuelke, C.; Burkart, C.; et al. Adjuvant Chemotherapy With Gemcitabine vs Observation in Patients Undergoing Curative-Intent Resection of Pancreatic Cancer. JAMA 2007, 297, 267. [Google Scholar] [CrossRef] [PubMed]
  98. Neoptolemos, J.P.; Stocken, D.D.; Bassi, C.; Ghaneh, P.; Cunningham, D.; Goldstein, D.; Padbury, R.; Moore, M.J.; Gallinger, S.; Mariette, C.; et al. Adjuvant Chemotherapy With Fluorouracil Plus Folinic Acid vs Gemcitabine Following Pancreatic Cancer Resection. JAMA 2010, 304, 1073. [Google Scholar] [CrossRef]
  99. Sinn, M.; Bahra, M.; Liersch, T.; Gellert, K.; Messmann, H.; Bechstein, W.; Waldschmidt, D.; Jacobasch, L.; Wilhelm, M.; Rau, B.M.; et al. CONKO-005: Adjuvant Chemotherapy With Gemcitabine Plus Erlotinib Versus Gemcitabine Alone in Patients After R0 Resection of Pancreatic Cancer: A Multicenter Randomized Phase III Trial. J. Clin. Oncol. 2017, 35, 3330–3337. [Google Scholar] [CrossRef]
  100. Conroy, T.; Hammel, P.; Hebbar, M.; Ben Abdelghani, M.; Wei, A.C.; Raoul, J.-L.; Choné, L.; Francois, E.; Artru, P.; Biagi, J.J.; et al. FOLFIRINOX or Gemcitabine as Adjuvant Therapy for Pancreatic Cancer. N. Engl. J. Med. 2018, 379, 2395–2406. [Google Scholar] [CrossRef]
  101. Oba, A.; Ho, F.; Bao, Q.R.; Al-Musawi, M.H.; Schulick, R.D.; Del Chiaro, M. Neoadjuvant Treatment in Pancreatic Cancer. Front. Oncol. 2020, 10, 245. [Google Scholar] [CrossRef]
  102. Groot, V.P.; Mosier, S.; Javed, A.A.; Teinor, J.A.; Gemenetzis, G.; Ding, D.; Haley, L.M.; Yu, J.; Burkhart, R.A.; Hasanain, A.; et al. Circulating Tumor DNA as a Clinical Test in Resected Pancreatic Cancer. Clin. Cancer Res. 2019, 25, 4973–4984. [Google Scholar] [CrossRef]
  103. Lee, J.-S.; Rhee, T.-M.; Pietrasz, D.; Bachet, J.-B.; Laurent-Puig, P.; Kong, S.-Y.; Takai, E.; Yachida, S.; Shibata, T.; Lee, J.W.; et al. Circulating tumor DNA as a prognostic indicator in resectable pancreatic ductal adenocarcinoma: A systematic review and meta-analysis. Sci. Rep. 2019, 9, 16971. [Google Scholar] [CrossRef] [PubMed]
  104. Yamaguchi, T.; Uemura, K.; Murakami, Y.; Kondo, N.; Nakagawa, N.; Okada, K.; Seo, S.; Hiyama, E.; Takahashi, S.; Sueda, T. Clinical Implications of Pre- and Postoperative Circulating Tumor DNA in Patients with Resected Pancreatic Ductal Adenocarcinoma. Ann. Surg. Oncol. 2021, 28, 3135–3144. [Google Scholar] [CrossRef] [PubMed]
  105. Guo, S.; Shi, X.; Shen, J.; Gao, S.; Wang, H.; Shen, S.; Pan, Y.; Li, B.; Xu, X.; Shao, Z.; et al. Preoperative detection of KRAS G12D mutation in ctDNA is a powerful predictor for early recurrence of resectable PDAC patients. Br. J. Cancer 2020, 122, 857–867. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Hadano, N.; Murakami, Y.; Uemura, K.; Hashimoto, Y.; Kondo, N.; Nakagawa, N.; Sueda, T.; Hiyama, E. Prognostic value of circulating tumour DNA in patients undergoing curative resection for pancreatic cancer. Br. J. Cancer 2016, 115, 59–65. [Google Scholar] [CrossRef] [PubMed]
  107. Hipp, J.; Hussung, S.; Timme-Bronsert, S.; Boerries, M.; Biesel, E.; Fichtner-Feigl, S.; Fritsch, R.; Wittel, U.A. Perioperative cell-free mutant KRAS dynamics in patients with pancreatic cancer. Br. J. Surg. 2021, 108, 239–243. [Google Scholar] [CrossRef]
  108. Hussung, S.; Akhoundova, D.; Hipp, J.; Follo, M.; Klar, R.F.U.; Philipp, U.; Scherer, F.; von Bubnoff, N.; Duyster, J.; Boerries, M.; et al. Longitudinal analysis of cell-free mutated KRAS and CA 19-9 predicts survival following curative resection of pancreatic cancer. BMC Cancer 2021, 21, 49. [Google Scholar] [CrossRef]
  109. Nakano, Y.; Kitago, M.; Matsuda, S.; Nakamura, Y.; Fujita, Y.; Imai, S.; Shinoda, M.; Yagi, H.; Abe, Y.; Hibi, T.; et al. KRAS mutations in cell-free DNA from preoperative and postoperative sera as a pancreatic cancer marker: A retrospective study. Br. J. Cancer 2018, 118, 662–669. [Google Scholar] [CrossRef] [Green Version]
  110. Sausen, M.; Phallen, J.; Adleff, V.; Jones, S.; Leary, R.J.; Barrett, M.T.; Anagnostou, V.; Parpart-Li, S.; Murphy, D.; Kay Li, Q.; et al. Clinical implications of genomic alterations in the tumour and circulation of pancreatic cancer patients. Nat. Commun. 2015, 6, 7686. [Google Scholar] [CrossRef]
  111. Lee, B.; Lipton, L.; Cohen, J.; Tie, J.; Javed, A.A.; Li, L.; Goldstein, D.; Burge, M.; Cooray, P.; Nagrial, A.; et al. Circulating tumor DNA as a potential marker of adjuvant chemotherapy benefit following surgery for localized pancreatic cancer. Ann. Oncol. 2019, 30, 1472–1478. [Google Scholar] [CrossRef]
  112. Pietrasz, D.; Pécuchet, N.; Garlan, F.; Didelot, A.; Dubreuil, O.; Doat, S.; Imbert-Bismut, F.; Karoui, M.; Vaillant, J.C.; Taly, V.; et al. Plasma Circulating Tumor DNA in Pancreatic Cancer Patients Is a Prognostic Marker. Clin. Cancer Res. 2017, 23, 116–123. [Google Scholar] [CrossRef] [Green Version]
  113. Shin, D.G.; Kim, K.C.; Choi, S.-W.; Joo, H.J.; Baik, S.H.; Park, M.K. A Methylation profile of Circulating Cell Free DNA for the Early Detection of Gastric Cancer and the Effects after Surgical Resection. J. Clin. Exp. Oncol. 2016, 5, 1000151. [Google Scholar] [CrossRef]
  114. Ko, K.; Kananazawa, Y.; Yamada, T.; Kakinuma, D.; Matsuno, K.; Ando, F.; Kuriyama, S.; Matsuda, A.; Yoshida, H. Methylation status and long-fragment cell-free DNA are prognostic biomarkers for gastric cancer. Cancer Med. 2021, 10, 2003–2012. [Google Scholar] [CrossRef] [PubMed]
  115. Murray, D.H.; Symonds, E.L.; Young, G.P.; Byrne, S.; Rabbitt, P.; Roy, A.; Cornthwaite, K.; Karapetis, C.S.; Pedersen, S.K. Relationship between post-surgery detection of methylated circulating tumor DNA with risk of residual disease and recurrence-free survival. J. Cancer Res. Clin. Oncol. 2018, 144, 1741–1750. [Google Scholar] [CrossRef] [PubMed]
  116. Pilarski, R. The Role of BRCA Testing in Hereditary Pancreatic and Prostate Cancer Families. Am. Soc. Clin. Oncol. Educ. Book 2019, 39, 79–86. [Google Scholar] [CrossRef]
  117. Luchini, C.; Brosens, L.A.A.; Wood, L.D.; Chatterjee, D.; Shin, J.I.; Sciammarella, C.; Fiadone, G.; Malleo, G.; Salvia, R.; Kryklyva, V.; et al. Comprehensive characterisation of pancreatic ductal adenocarcinoma with microsatellite instability: Histology, molecular pathology and clinical implications. Gut 2021, 70, 148–156. [Google Scholar] [CrossRef]
  118. Mariani, S.; Puzzoni, M.; Liscia, N.; Impera, V.; Pretta, A.; Tolu, S.; Pireddu, A.G.; Persano, M.; Musio, F.; Donisi, C.; et al. Liquid biopsy-driven anti-EGFR rechallenge in patients with metastatic colorectal cancer. J. Clin. Oncol. 2021, 39, 3577. [Google Scholar] [CrossRef]
  119. Diehl, F.; Schmidt, K.; Choti, M.A.; Romans, K.; Goodman, S.; Li, M.; Thornton, K.; Agrawal, N.; Sokoll, L.; Szabo, S.A.; et al. Circulating mutant DNA to assess tumor dynamics. Nat. Med. 2008, 14, 985–990. [Google Scholar] [CrossRef]
  120. Kruger, S.; Heinemann, V.; Ross, C.; Diehl, F.; Nagel, D.; Ormanns, S.; Liebmann, S.; Prinz-Bravin, I.; Westphalen, C.B.; Haas, M.; et al. Repeated mutKRAS ctDNA measurements represent a novel and promising tool for early response prediction and therapy monitoring in advanced pancreatic cancer. Ann. Oncol. 2018, 29, 2348–2355. [Google Scholar] [CrossRef]
  121. Park, G.; Park, J.K.; Son, D.-S.; Shin, S.-H.; Kim, Y.J.; Jeon, H.-J.; Lee, J.; Park, W.-Y.; Lee, K.H.; Park, D. Utility of targeted deep sequencing for detecting circulating tumor DNA in pancreatic cancer patients. Sci. Rep. 2018, 8, 11631. [Google Scholar] [CrossRef]
  122. Pishvaian, M.J.; Blais, E.M.; Brody, J.R.; Sohal, D.; Hendifar, A.E.; Chung, V.; Mikhail, S.; Rahib, L.; Lyons, E.; Tibbetts, L.; et al. Outcomes in pancreatic adenocarcinoma (PDA) patients (pts) with genetic alterations in DNA damage repair (DDR) pathways: Results from the Know Your Tumor (KYT) program. J. Clin. Oncol. 2019, 37, 191. [Google Scholar] [CrossRef]
  123. Perkhofer, L.; Gout, J.; Roger, E.; Kude De Almeida, F.; Baptista Simões, C.; Wiesmüller, L.; Seufferlein, T.; Kleger, A. DNA damage repair as a target in pancreatic cancer: State-of-the-art and future perspectives. Gut 2021, 70, 606–617. [Google Scholar] [CrossRef] [PubMed]
  124. Casolino, R.; Paiella, S.; Azzolina, D.; Beer, P.A.; Corbo, V.; Lorenzoni, G.; Gregori, D.; Golan, T.; Braconi, C.; Froeling, F.E.M.; et al. Homologous Recombination Deficiency in Pancreatic Cancer: A Systematic Review and Prevalence Meta-Analysis. J. Clin. Oncol. 2021, 39, 2617–2631. [Google Scholar] [CrossRef] [PubMed]
  125. Golan, T.; Kanji, Z.S.; Epelbaum, R.; Devaud, N.; Dagan, E.; Holter, S.; Aderka, D.; Paluch-Shimon, S.; Kaufman, B.; Gershoni-Baruch, R.; et al. Overall survival and clinical characteristics of pancreatic cancer in BRCA mutation carriers. Br. J. Cancer 2014, 111, 1132–1138. [Google Scholar] [CrossRef] [PubMed]
  126. Sehdev, A.; Gbolahan, O.; Hancock, B.A.; Stanley, M.; Shahda, S.; Wan, J.; Wu, H.H.; Radovich, M.; O’Neil, B.H. Germline and Somatic DNA Damage Repair Gene Mutations and Overall Survival in Metastatic Pancreatic Adenocarcinoma Patients Treated with FOLFIRINOX. Clin. Cancer Res. 2018, 24, 6204–6211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. O’Reilly, E.M.; Lee, J.W.; Zalupski, M.; Capanu, M.; Park, J.; Golan, T.; Tahover, E.; Lowery, M.A.; Chou, J.F.; Sahai, V.; et al. Randomized, Multicenter, Phase II Trial of Gemcitabine and Cisplatin With or Without Veliparib in Patients With Pancreas Adenocarcinoma and a Germline BRCA/PALB2 Mutation. J. Clin. Oncol. 2020, 38, 1378–1388. [Google Scholar] [CrossRef] [PubMed]
  128. Palacio, S.; Mcmurry, H.S.; Ali, R.; Donenberg, T.; Silva-Smith, R.; Wideroff, G.; Sussman, D.A.; Rocha Lima, C.M.S.; Hosein, P.J. DNA damage repair deficiency as a predictive biomarker for FOLFIRINOX efficacy in metastatic pancreatic cancer. J. Gastrointest. Oncol. 2019, 10, 1133–1139. [Google Scholar] [CrossRef] [PubMed]
  129. Terrero, G.; Datta, J.; Dennison, J.; Sussman, D.A.; Lohse, I.; Merchant, N.B.; Hosein, P.J. Ipilimumab/Nivolumab Therapy in Patients With Metastatic Pancreatic or Biliary Cancer With Homologous Recombination Deficiency Pathogenic Germline Variants. JAMA Oncol. 2022, 8, 1. [Google Scholar] [CrossRef] [PubMed]
  130. Takahashi, H.; Kagara, N.; Tanei, T.; Naoi, Y.; Shimoda, M.; Shimomura, A.; Shimazu, K.; Kim, S.J.; Noguchi, S. Correlation of Methylated Circulating Tumor DNA with Response to Neoadjuvant Chemotherapy in Breast Cancer Patients. Clin. Breast Cancer 2017, 17, 61–69.e3. [Google Scholar] [CrossRef]
  131. Guo, D.; Yang, L.; Yang, J.; Shi, K. Plasma cell-free DNA methylation combined with tumor mutation detection in prognostic prediction of patients with non-small cell lung cancer (NSCLC). Medicine 2020, 99, e20431. [Google Scholar] [CrossRef]
  132. Amatu, A.; Schirripa, M.; Tosi, F.; Lonardi, S.; Bencardino, K.; Bonazzina, E.; Palmeri, L.; Patanè, D.A.; Pizzutilo, E.G.; Mussolin, B.; et al. High Circulating Methylated DNA Is a Negative Predictive and Prognostic Marker in Metastatic Colorectal Cancer Patients Treated With Regorafenib. Front. Oncol. 2019, 9, 622. [Google Scholar] [CrossRef] [Green Version]
  133. Martinez-Useros, J.; Martin-Galan, M.; Florez-Cespedes, M.; Garcia-Foncillas, J. Epigenetics of Most Aggressive Solid Tumors: Pathways, Targets and Treatments. Cancers 2021, 13, 3209. [Google Scholar] [CrossRef] [PubMed]
  134. Berger, A.W.; Schwerdel, D.; Costa, I.G.; Hackert, T.; Strobel, O.; Lam, S.; Barth, T.F.; Schröppel, B.; Meining, A.; Büchler, M.W.; et al. Detection of Hot-Spot Mutations in Circulating Cell-Free DNA From Patients With Intraductal Papillary Mucinous Neoplasms of the Pancreas. Gastroenterology 2016, 151, 267–270. [Google Scholar] [CrossRef] [PubMed]
  135. Le Calvez-Kelm, F.; Foll, M.; Wozniak, M.B.; Delhomme, T.M.; Durand, G.; Chopard, P.; Pertesi, M.; Fabianova, E.; Adamcakova, Z.; Holcatova, I.; et al. KRAS mutations in blood circulating cell-free DNA: A pancreatic cancer case-control. Oncotarget 2016, 7, 78827–78840. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Cohen, J.D.; Javed, A.A.; Thoburn, C.; Wong, F.; Tie, J.; Gibbs, P.; Schmidt, C.M.; Yip-Schneider, M.T.; Allen, P.J.; Schattner, M.; et al. Combined circulating tumor DNA and protein biomarker-based liquid biopsy for the earlier detection of pancreatic cancers. Proc. Natl. Acad. Sci. USA 2017, 114, 10202–10207. [Google Scholar] [CrossRef] [Green Version]
  137. Berger, A.W.; Schwerdel, D.; Reinacher-Schick, A.; Uhl, W.; Algul, H.; Friess, H.; Janssen, K.P.; Konig, A.; Ghadimi, M.; Gallmeier, E.; et al. A Blood-Based Multi Marker Assay Supports the Differential Diagnosis of Early-Stage Pancreatic Cancer. Theranostics 2019, 9, 1280–1287. [Google Scholar] [CrossRef]
  138. Liu, X.; Liu, L.; Ji, Y.; Li, C.; Wei, T.; Yang, X.; Zhang, Y.; Cai, X.; Gao, Y.; Xu, W.; et al. Enrichment of short mutant cell-free DNA fragments enhanced detection of pancreatic cancer. EBioMedicine 2019, 41, 345–356. [Google Scholar] [CrossRef] [Green Version]
Figure 1. cfDNA somatic mutations and epigenetic markers in pancreatic ductal adenocarcinoma. Summary of specific somatic mutations and epigenetic markers that can be detected in cfDNA which have been shown to confer prognostic, predicative, and preoperative risk stratification value in patients with pancreatic ductal adenocarcinoma.
Figure 1. cfDNA somatic mutations and epigenetic markers in pancreatic ductal adenocarcinoma. Summary of specific somatic mutations and epigenetic markers that can be detected in cfDNA which have been shown to confer prognostic, predicative, and preoperative risk stratification value in patients with pancreatic ductal adenocarcinoma.
Cancers 14 03453 g001
Table 1. cfDNA methylation marker studies to diagnose pancreatic ductal adenocarcinoma.
Table 1. cfDNA methylation marker studies to diagnose pancreatic ductal adenocarcinoma.
StudyStudy PopulationTechnology UsedTargetSensitivity and Specificity, Respectively
Cao, Wei, Hu, He, Zhang, Xia, Tu, Yuan, Guo, Liu, Xie, and Li [2]HC vs. PDAMethylome sequencing24-feature 5mC and 27-feature 5hmC model5mC + 5hmC—93.8% and 95.5% (AUC of 0.99)
5mC alone—87% and 82% (AUC of 0.97)
5hmC alone—78–85% and 99–100% (AUC of 0.99–0.96)
Melnikov et al. [38]HC vs. PDAMicroarray* CCND2, PLAU, SOCS1, THBS, and VHL76% and 59%
Liggett et al. [39]HC vs. PDA vs. CPSame microarray as Melnikov et al., 200914 promoters for PDA vs. CP
8 for HC and CP
91.2% and 90.8% (PDA vs. CP)
82% and 78%
Yi et al. [40]PDA vs. PanIN or pancreatitisPCRBNC1 and ADAMTS1Both—81% and 85%
BNC1—79% and 89%
ADAMTS1—48% and 92%
Eissa et al. [41]PDA vs. noncancerPCRBNC1 and ADAMTS1Combined AUC of 0.95
ADAMTS1—87.2% and 95.8%
(AUC = 0.91; 95%)
BNC1—64.1% and 93.7% (AUC = 0.79)
Both—97.3% and 91.6% (AUC = 0.95)
Henriksen et al. [42]PDA vs. HC + CP + acute pancreatitisPCRBMP3, RASSF1A, BNC1, MESTv2, TFPI2, APDA, SFRP1, and SFRP2
With age >65 years
76% and 83% (0.86)
Guler et al. [43]PDA vs. no PDACHIP seq** Gene set of
top 65%
differentially hydroxymethylated genes
Training set AUC = 0.92
Validation set 1 AUC = 0.921
Validation set 2 AUC = 0.943
Li et al. [44]HC vs. PDAMeDIP-seqTRIM73, FAM150A, EPB41L3, SIX3, MIR663, MAPT, LOC100128977, and LOC10013014893.2% and 95%
Ying et al. [45]HC vs. PDAPCRADAMTS1, BNC1, LRFN5, and PXDN100% and 90%
Manoochehri et al. [46]HC vs. PDAddPCRSST100% and 89%
Singh et al. [47]HC vs. CP vs. HCPCRSPRC, UCHL1, NPTX2, and PENKHC vs. CP + PDA, MI of all 4 are increased
HC vs. CP, MI of UCLH1, PENK, NPTX2 increased
CP vs. PDA, MI of SPARC, and NPTX2 increased
CP vs. early-stage PDA, MI of SPARC increased
Shinjo et al. [48]PDA vs. HCMBD-ddPCRADAMTS1, HOXA1, PCDH10, SEMA5A, and SPSB4Panel of 5 genes with 49% and 86%
1 of 5 in 49% of PDA
Panel of 5 genes + KRAS mutation in cfDNA with 68% and 86%
Fujimoto et al. [49]PDA vs. benign disease and HCPCRRUNX3RUNX3 alone: 50.9% and 93.5%
RUNX3 combined with CA19-9: 85.5% and 93.5% for all stages and 78% for stage I
Kandimalla et al. [50]PDA vs. HCGenome-wide DNA methylation sequencingEpiPanGiDxPredictive value of 85%
Vrba et al. [51]PDA vs. benign cystPCR10-promoter panel in mPDA100% and 95% (AUC of 0.999)
Li et al. [52]PDA vs. PanIN benign tumors and pancreatitisPCRBNC1
SEPT9 in
Stage I and II
Combined—65% and 87%
BCN1—50.9% and 88.7%
SEPT9—36.8% and 96.2%
Combined + CA 19-9 vs. CA19-9
alone—86% vs. 61.4% and 81.1% vs.
90.6%
Individually they have low CT compared to HC and benign disease
Melson et al. [53]PDA vs. HCPCR #VHL, MYF3, TMS, GPC3, and SRBC80% and 66% (AUC = 0.848)
Park et al. [54]PDA vs. CP PCRNPTX280% and 76%
Park et al. [55]PDA vs. HCPCRP16 Higher methylation in PDA than HC (86.7 ± 29.8 vs. 33.3 ± 0.00, p = 0.016)
PDA—pancreatic ductal adenocarcinoma; mPDA—metastatic PDA; AUC—area under the receiver operating characteristic curve; PCR—polymerase chain reaction, ddPCR—digital droplet PCR; ChIP-Seq—chromatin immunoprecipitation sequencing; MeDIP-Seq—methylated DNA immunoprecipitation sequencing; MBD-ddPCR—enrichment of methyl-CpGbinding (MBD) protein count-followed by ddPCR; CT—cycle thresholds; * panel of reduced methylation; ** combination of hypo and hyper methylated genes; # methylation sensitive restriction enzyme and multiplex PCR.
Table 2. Summary of studies determining prognostic value KRAS mutations in cfDNA.
Table 2. Summary of studies determining prognostic value KRAS mutations in cfDNA.
KRAS VariantsTechniqueFindings
G12A, G12C, G12D, G12R, G12S, G12V, and G13D [64]ddPCRDetection of mutant KRAS is associated with poor OS (197 days vs. 60 days, HR = 2.8, p = 0.018).
G12D, G12V, and G12R [65]ddPCRKRAS mutation at G12V conferred poorer OS compared to WT (p < 0.01).
No significance effect of KRAS G12D mutation.
G12A, G12C, G12D, G12R, G12S, G12V, and G13D [66]ddPCRMAF ≥ 5% of any variant was a poor predictor of PFS (HR = 2.28; 95% CI: 1.18–4.40; p = 0.014) and OS (HR = 3.46; 95% CI: 1.40–8.50; p = 0.007).
MAF peak above 1% was significantly associated with radiologic progression (p = 0.0003).
KRAS Codon 12 mutations [67]PCRDetection of KRAS mutations conferred shorter OS compared to WT (3.9 months vs. 10.2 months, p < 0.001).
Mutational burden could significantly correlate with TNM tumor staging (p = 0.033) and liver metastasis (p = 0.014).
KRAS mutations were a negative prognostic factor for survival (HR = 7.39; 95% CI: 3.69–14.89).
G12V and G12D [68]ddPCRG12V conferred poor OS (p = 0.001).
G12D conferred poor OS (p = 0.044).
G12D, G12V, G12R, and G13D [69]ddPCRIncreased mutational burden conferred poor PFS (2.5 vs. 7.5 months, p = 0.03) and OS (6.5 vs. 11.5 months, p = 0.009).
G12D, G12V, G12R, and G12C [70]NGSG12R mutation conferred favorable OS compared to WT (20.4 vs. 14.5 m, HR = 0.67 (95% CI: 0.47–0.93), p = 0.0215) and PFS on first-line therapy (12.2 vs. 6.8 m, HR 0.60 (95% CI 0.40–0.85), p = 0.004).
G12A, G12C, G12D, G12R, G12S, G12V, and G13D [71]ddPCRKRAS mutations concentration >0.165 copies/L had worse OS
median fractional abundance (>0.415%.)
G12V, G12D, and G12R in codon 12 KRAS [34]ddPCR mOS was significantly shorter in patients with KRAS mutant (276 days) compared with patients with WT KRAS (413 days) from cfDNA samples (p  =  0.02).
mOS was significantly shorter only in G12V variants compared to other KRAS mutants (219 days vs. 410 days, p = 0.006).
G12D, G12R, G12V, Q61H, Q61R, and A59G [72]BEAM-PCROverall response rate, disease control rate, mPFS, and mOS were higher in patients without detectable KRAS mutations (48% vs. 28%, 81% vs. 69%, 8.8 vs. 5.3 months, and 18.2 vs. 6.6 months, respectively).
KRAS codon 12 [73]ddPCRPatients with WT KRAS had better OS than mutant KRAS patients (10.6 months vs. 5.6 months, p < 0.05).
Patients with KRAS mutation and copy number gain had the worst prognosis with a mOS of 2.5 months (p ≤ 0.0001).
KRAS exon 12 [74]PCRUndetectable mutant KRAS conferred favorable OS (8 vs. 37.5 months from diagnosis, p < 0.004).
KRAS mutations: not specified [75]DNA-based Ion-Torrent NGS assays (ClearID)Presence of KRAS mutations in cfDNA was associated with reduced mOS (54% in mutation-positive versus 90% in mutation-negative, p < 0.05).
KRAS codon 12 and 13 [76]PCRPatients with KRAS mutations detected in cfDNA had significantly lower mPFS (1.8 vs. 4.6 months, p = 0.014) and mOS (3.0 vs. 10.5 months, p = 0.003) than those without detected plasma KRAS mutations.
G12A, G12C, G12D, G12V, G12R, G12S, and G13D [77]ddPCR and NGS amplicon panelDetectable mutant KRAS cfDNA was associated with poor OS (3.2 vs. 8.4 months, p = 0.005).
KRAS G12/13 mutations and KRAS Q61K [78]ddPCRDetectable mutant KRAS cfDNA was associated with poor PFS (308.5 vs. 168 days, p = 0.07).
G12D, G12R, G12V, and G13D [79]ddPCRHigher concentration with advanced stages (p = 0.0129).
RAS mutation (KRAS/NRAS codons 12, 13, 59, 61, 117, and 146) [80]BEAM-PCRHigher RAS MAF was associated with poor OS (142 vs. 310 days, p = 0.0261) and PFS (85 versus 175 days; p = 0.0556).
G12V, G12D, G12R, and Q61H [81]ddPCRThe mOS of patients with detectable mutant KRAS cfDNA was shorter (15.8 months vs. 33.7 months; p < 0.05)
G12D, G12V, G12R, G13D [32]Digital PCR and NGSPatients with multiple liver metastasis and poor mOS had higher mutant KRAS cfDNA MAF compared to those with fewer lesions (p < 0.05).
PCR—polymerase chain reaction; ddPCR—digital droplet PCR; BEAM-PCR—beads, emulsion, amplification, and magnetics PCR; NGS—next generation sequencing; WT—wild type; MAF—mutant allelic fraction; HR—hazards ratio; CI—confidence interval; mOS—median overall survival; mPFS—median progression-free survival.
Table 3. Summary of studies examining epigenetic markers in cfDNA relating to prognosis of patients with PDA.
Table 3. Summary of studies examining epigenetic markers in cfDNA relating to prognosis of patients with PDA.
Genes StudiedComparisonFindings
5mC and 5hmC pan-sequencing [2]Identify DMPs for 5mC and DhMPs for 5hmC5mC: No difference between resectable vs. unresectable PDA.
5hmC: Significant different between stage I vs. II/III/IV; no difference between resectable vs. unresectable.
5mC + 5hmC: Higher in tumor size <3 cm (vs. >3 cm) and PNI; no difference between resectable vs. unresectable; no significant differences in vascular invasion or positive lymph node metastasis were found in resectable PDA patients.
SPARC
UCLH1
PENK
NPTX2 [47]
Low vs. high methylation indexSPARC: Higher in stage IV and poor survival (3 vs. 6 m); Lower in resectable (p = 0.02).
NPTX2: Higher in stage IV, met dz and poor survival (3 vs. 9 m) (p = 0.04).
UCLH1: Higher in stage III/IV vs. I/II (p = 0.034).
28-gene panel for staging [88]Number of methylated genesStage I: 7.09 (95% CI: 5.51–8.66).
Stage II: 7.00 (95% CI: 5.93–8.07).
Stage III: 6.77 (95% CI: 5.08–8.46).
Stage IV: 10.24 (95% CI: 8.88–11.60).
The number of methylated genes at stage IV was significantly higher compared to stage I/II/III PDA (p = 0.0002).
Specific promotersThe prediction model (SEPT9v2, SST, ALX4, CDKN2B, HIC1, MLH1, NEUROG1, and BNC1) enabled the differentiation of stage IV from stage I-III disease (AUC of 0.87 (cut point: 0.55); sensitivity of 74%, specificity of 87%)). Model (MLH1, SEPT9v2, BNC1, ALX4, CDKN2B, NEUROG1, WNT5A, and TFPI2) enabled the differentiation of stage I-II from stage III-IV disease (AUC of 0.82 (cut point: 0.66); sensitivity of 73%, specificity of 80%)).
Same panel as above
[89]



Number of methylated genes





Risk stratification based on ASA= 3 and methylation of GSTP1, SFRP2, BNC1, SFRP1, TFPI2, and WNT5A
Patients with more than 10 hypermethylated genes had an HR of 2.03 (95% CI: 1.15–3.57).

Total group (all stages of tumor) HR compared to group 1:
Risk group 2: HR 2.65 (95% CI: 1.24–5.66);
Risk group 3: HR of 4.34 (95% CI: 1.98–9.51);
Risk group 4: HR of 21.19 (95% CI: 8.61–52.15).

Stage I–II (ASA = 3, SFRP2, and MESTv2):
Risk group 2: HR of 4.83 (95% CI: 2.01–11.57);
Risk group 3: HR of 9.12 (95% CI: 2.18–38.25);
Risk group 4: HR of 70.90 (95% CI: 12.63–397.96).

Stage IV (BMP3, NPTX2, SFRP1, and MGMT):
Risk group 2: HR of 5.23 (95% CI: 2.13–12.82).
ADAMTS1, HOXA1, PCDH10, SEMA5A, and SPSB4 ± KRAS mutations [48]Positive vs. negativeLarge tumor size and higher frequency of liver metastatic disease in cfDNA positive patients
p16, RARbeta, TNFRSF10C, APC, ACIN1, DAPK1, 3OST2, BCL2, and CD44 [90]Methylation levels in CpG promoter regionsThe highest tertile of methylation of ACIN1 was associated with shorter survival compared to the middle and the lowest tertile group (13 months vs. 17 months).
Highest tertile of TNFRSF10C was associated with shorter survival compared to the middle and the lowest tertile group (OS, 13 months vs. 22 months).
TNFRSF10C SN1 methylation was significantly associated with PNI (OR = 0.088).
HOXD8 and POU4F1 [91]DetectionMedian PFS and OS were 5.3 and 8.2 months in ctDNA-positive and 6.2 and 12.6 months in ctDNA-negative patients, respectively.
ctDNA positivity was more often associated with young age, high CA19-9 level, and neutrophils lymphocytes ratio.
ctDNA was confirmed as an independent prognostic marker for PFS (HR = 1.5, CI 95%: [1.03–2.18], p = 0.034) and OS (HR = 1.62, CI 95%: [1.05–2.5], p = 0.029).
5mC—methylated methylcytosine; 5hmC—5-hydroxymethyl cytosine; DMPs—differentially methylated peaks; DhMPs—differentially hydroxymethylated peaks; PNI—perineural invasion; ASA—American Society of Anesthesiologists score; PFS—progression-free survival; OS—overall survival; HR—hazards ratio; CI—confidence interval; OR—odds ratio.
Table 4. Preoperative risk stratification of pancreatic ductal adenocarcinoma.
Table 4. Preoperative risk stratification of pancreatic ductal adenocarcinoma.
StudyKRAS Variants TestedImpact of Preop DetectionImpact of Postop Detection
Groot et al., 2019 [102]G12V/12D/12R/Q61HSignificantPersistent—significant
Lee et al., 2019 [103]Codons 12/13/61Significant
Yamaguchi et al., 2021 [104]G12/12V/12RNot significant
Guo et al., 2020 [105]G12DNot studied
Hadano et al., 2016 [106]G12V/12D/12R
Kim et al., 2018 [71]G12A/12C/12D/12R/12S/12V/13D
Hipp et al., 2021 [107]G12D/12V/12R/12CNot SignificantSignificant
Hussung et al., 2021 [108]Codons 12/13/61Significant
Nakano et al., 2018 [109]Codons 12/13Conversion from wild type to mutation—significant
Wantanabe et al., 2019 [81]G12V/12D/12R/Q61HEmergence—significant
Sausen et al., 2015 [110]G12V/12D/12R/12V/12C/13DNot studiedSignificant
Preop—preoperative; Postop—post operative.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Sheel, A.; Addison, S.; Nuguru, S.P.; Manne, A. Is Cell-Free DNA Testing in Pancreatic Ductal Adenocarcinoma Ready for Prime Time? Cancers 2022, 14, 3453. https://doi.org/10.3390/cancers14143453

AMA Style

Sheel A, Addison S, Nuguru SP, Manne A. Is Cell-Free DNA Testing in Pancreatic Ductal Adenocarcinoma Ready for Prime Time? Cancers. 2022; 14(14):3453. https://doi.org/10.3390/cancers14143453

Chicago/Turabian Style

Sheel, Ankur, Sarah Addison, Surya Pratik Nuguru, and Ashish Manne. 2022. "Is Cell-Free DNA Testing in Pancreatic Ductal Adenocarcinoma Ready for Prime Time?" Cancers 14, no. 14: 3453. https://doi.org/10.3390/cancers14143453

APA Style

Sheel, A., Addison, S., Nuguru, S. P., & Manne, A. (2022). Is Cell-Free DNA Testing in Pancreatic Ductal Adenocarcinoma Ready for Prime Time? Cancers, 14(14), 3453. https://doi.org/10.3390/cancers14143453

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop