Next Article in Journal
The Role of the Complement in Clear Cell Renal Carcinoma (ccRCC)—What Future Prospects Are There for Its Use in Clinical Practice?
Next Article in Special Issue
Management of Intrahepatic Cholangiocarcinoma: A Narrative Review
Previous Article in Journal
Using Flavonoid Substitution Status to Predict Anticancer Effects in Human Melanoma Cancers: An In Vitro Study
Previous Article in Special Issue
Deep Learning Applications in Pancreatic Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Anatomical and Biological Considerations to Determine Resectability in Pancreatic Cancer

by
Ingmar F. Rompen
1,2,
Joseph R. Habib
1,
Christopher L. Wolfgang
1 and
Ammar A. Javed
1,*
1
Department of Surgery, The NYU Grossman School of Medicine and NYU Langone Health, New York, NY 10016, USA
2
Department of General, Visceral, and Transplantation Surgery, Heidelberg University Hospital, 69120 Heidelberg, Germany
*
Author to whom correspondence should be addressed.
Cancers 2024, 16(3), 489; https://doi.org/10.3390/cancers16030489
Submission received: 14 November 2023 / Revised: 11 January 2024 / Accepted: 19 January 2024 / Published: 23 January 2024
(This article belongs to the Special Issue Surgical Management of Gastrointestinal Cancers)

Abstract

:

Simple Summary

Surgical candidacy in the treatment of pancreatic cancer is complex. There are multiple factors influencing if a patient is a good candidate for surgery with or without preceding treatment with chemotherapy. This review summarizes the importance of both anatomical (tumor size and its relation to important blood vessels to the liver and bowel) and biological (aggressiveness of the tumor) considerations.

Abstract

Pancreatic ductal adenocarcinoma (PDAC) remains associated with poor outcomes with a 5-year survival of 12% across all stages of the disease. These poor outcomes are driven by a delay in diagnosis and an early propensity for systemic dissemination of the disease. Recently, aggressive surgical approaches involving complex vascular resections and reconstructions have become more common, thus allowing more locally advanced tumors to be resected. Unfortunately, however, even after the completion of surgery and systemic therapy, approximately 40% of patients experience early recurrence of disease. To determine resectability, many institutions utilize anatomical staging systems based on the presence and extent of vascular involvement of major abdominal vessels around the pancreas. However, these classification systems are based on anatomical considerations only and do not factor in the burden of systemic disease. By integrating the biological criteria, we possibly could avoid futile resections often associated with significant morbidity. Especially patients with anatomically resectable disease who have a heavy burden of radiologically undetected systemic disease most likely do not derive a survival benefit from resection. On the contrary, we could offer complex resections to those who have locally advanced or oligometastatic disease but have favorable systemic biology and are most likely to benefit from resection. This review summarizes the current literature on defining anatomical and biological resectability in patients with pancreatic cancer.

1. Introduction

Pancreatic cancer is associated with a devastating five-year overall survival of 12% across all stages of the disease [1,2]. It is the third leading cause of cancer-related death with a rising incidence [2]. Due to the asymptomatic nature of the disease, only approximately 20% of patients are found to have resectable disease at the time of diagnosis [2,3]. Additionally, poor outcomes are driven by an early propensity for the systemic spread of the disease. This is evident by the fact that a significant proportion of patients undergoing resection will experience systemic recurrence of the disease [4,5,6]. Effective systemic control of the disease remains one of the strongest limitations to achieving cure and long-term survival.
Surgery remains the only curative therapy, with a modest survival benefit having been reported with the adjunct of multiagent system therapies. Historically, resectability has been based on anatomical considerations. The National Comprehensive Cancer Network (NCCN) guidelines define localized pancreatic cancer based on the anatomical relation between the tumor and surrounding vasculature into resectable, borderline resectable (BRPC), and locally advanced pancreatic cancer (LAPC) [7]. Advances in surgical technique and chemotherapy have increased surgical candidacy in patients with locally advanced disease, which accounts for approximately 30–40% of PDAC [8,9,10]. At specialized centers, up to 35–60% of LAPC patients can now undergo resection [11,12]. While surgical resection can provide cure and long-term survival in some patients, early recurrence with a progression of the disease is routinely seen in clinical practice [4]. In the latter cases, undetected microscopic residual disease and aggressive cancer biology may lead to early clinical recurrence and death, rendering a morbid local resection futile. Thus, considering the tumor biology to determine surgical candidacy is becoming increasingly important [13,14]. Institutions such as the international consensus meetings and the MD Anderson Cancer Center have realized the importance of tumor biology and come up with alternative criteria for resectability that factor in both anatomic and biological parameters [15,16].
Despite these considerations, significant variability exists in adherence to these guidelines across centers. Some centers advocate upfront resection to avoid progression of the disease and worsening of the patient’s condition during the neoadjuvant window while others prefer a neoadjuvant approach. The latter has the benefit of early treatment of undetected micrometastatic disease and the selection of surgical candidates by avoiding futile surgery in those who progress on systemic therapy and ensures that all patients receive systemic therapy. The variability in outcomes of patients regardless of their local stage suggests a role for tumor heterogeneity and tumor biology in driving these outcomes. Therefore, a precision approach is warranted where it is not only the anatomical staging of the disease but also a biological assessment that should determine resectability in these patients. In doing so, we would perform resections in patients who would derive survival benefit while avoiding potentially morbid operations in those with a heavy burden of systemic disease. This review aims to summarize and discuss the evidence on the anatomical and biological considerations of resectability for the management of pancreatic cancer.

2. Diagnostic Workup for Pancreatic Cancer

When assessing resectability, an adequate diagnostic workup is pivotal. The gold standard for pancreatic cancer diagnosis is a pancreas protocol computed tomography (PPCT) [7,17], which can help in assessing both the locoregional extent and the metastatic spread of disease to the liver, peritoneum, and lungs [18]. It is also highly sensitive and specific for vascular involvement with venous contact or infiltration presenting with obstruction or thrombosis of the vein, stenosis or irregularity of the vessel caliber, or a teardrop sign [19]. Similarly, arterial involvement of the superior mesenteric artery (SMA), celiac trunk (CA), or the common hepatic artery (CHA) can be appreciated on PPCT and is relevant to resectability and prognosis [19]. Furthermore, PPCT can help to assess the aberrant anatomy of hepatic arteries and stenosis of the celiac trunk, which are important for surgical planning. Even after neoadjuvant treatment, the CT scan can reliably distinguish between vascular infiltration and vascular contact through the “Halo” and “String” signs [20]. The “Halo” sign describes tumor infiltration of the lymphatic and neural tissue surrounding the SMA and, therefore, arterial encasement but without true infiltration [20]. This allows for the surgical planning of periadventitial divestment, thus skeletonizing the involved artery [21]. The “String” sign on the other hand suggests true arterial invasion, and therefore, surgical planning mandatorily should involve strategies for arterial resection and reconstruction [20]. The artery-first approach allows further exploration and identification of SMA involvement before the point of no return is reached [21,22,23]. Depending on the site of predicted involvement, different approaches can be chosen [24]. However, poor performance of the CT is reported for the detection of small metastases and nodal involvement with a diagnostic accuracy of 38–77% and a sensitivity of 14–24% [19]. Both CT and magnet resonance imaging (MRI) have similar sensitivity and specificity. Due to wider availability and the greater consistency of images within CT scans, MRI is typically used in patients with contraindications for CT [25]. Currently, FDG or FAPI PET/CT is not routinely used for pancreatic cancer workup [19]. An additional value is proposed for detecting lymph node involvement and metastatic disease [25]. Additionally, cinematic rendering has shown promise in the vascular mapping and identification of occult metastases in pancreatic cancer [26]. Following an image-based diagnosis of the disease, an endoscopic ultrasound (EUS) with a fine-needle aspiration (FNA) or brush cytology is used for histopathological confirmation of the disease, which is important for the initiation of neoadjuvant/induction therapy [19,27].
Recent studies have evaluated the role of laparoscopic staging before neoadjuvant treatment in LAPC for better assessment of occult metastases or before surgical exploration to assess resectability [28]. When performed in the pretreatment setting, occult metastases are detected in around 10–20% of patients with resectable or borderline resectable and up to 30% in locally advanced PDACs [29,30]. Additionally, intraoperative ultrasound can be used to assess the tumor location and vascular involvement during exploration [31]. The NCCN guidelines recommend a staging laparoscopy directly before a planned resection through laparotomy in all patients with a high suspicion of peritoneal or hepatic metastases or questionable resectability [7]. Thus, a staging laparoscopy should be considered in patients with CA19-9 > 150 U/mL, low-volume ascites, borderline resectable tumor, size > 3 cm, or suspicious lymphadenopathy.

3. Anatomical Assessment of Resectability and Implications for Treatment

Once the diagnosis of PDAC is established, tumor staging is performed using the American Joint Committee on Cancer (AJCC) TNM classification [32]. For localized PDACs, TNM staging has limited value for the assessment of resectability [32]. Therefore, the AHPBA [33], Alliance [34], NCCN [7], and the MD Anderson Cancer Center [35] have developed anatomical criteria that are primarily based on the extent of involvement of the surrounding major vessels (Table 1). Radiographically localized disease is divided into resectable, borderline resectable, and locally advanced disease. In brief, resectable tumors are those that can be resected without any additional resections and reconstructions of the vessels. Borderline resectable tumors often require additional resection and reconstruction of adjacent vessels or en bloc resection of infiltrated organs and are at a higher risk of margin-positive resections [13]. In locally advanced tumors, upfront resection is extremely challenging due to the extent of vascular involvement [36]. Major differences within the classifications are that the AHPBA classification defines tumors with contact to the SMV/portal vein as borderline resectable whereas, for all other relevant classifications, encasement contact <180° is considered the resectable stage. These anatomical criteria have a prognostic value and have implications on the treatment sequence [4,36].
For resectable tumors, upfront resection with adjuvant chemotherapy, is currently preferred [36]. However, it has been shown that approximately 40% of patients do not receive adjuvant therapy due to postoperative complications or early progression of the disease [37]. Due to a substantial number of patients being diagnosed with recurrence despite successful resection, there is an increasing belief that pancreatic cancer is a systemic disease even when diagnosed at an early stage [5]. Therefore, there may be a theoretical benefit for patients receiving neoadjuvant therapy in order to control disseminated disease early on [38]. On the contrary, others argue that there is a risk of progression of the disease during the neoadjuvant window that would render these patients unresectable or lead to a worsening of their physical condition during chemotherapy, preventing the patient from undergoing a possibly curative resection [36]. To date, there is no survival benefit shown within subgroup analyses for resectable PDAC in randomized trials comparing neoadjuvant treatment versus upfront surgery such as the PREOPANC1 trial (HR 0.79, 95%CI: 0.54–1.16, p = 0.23) [39]. The PRODIGE48/PANACHE-01 and NEONAX phase II trials showed the feasibility of perioperative cytotoxic treatment with FOLFIRINOX or gemcitabine plus nab-paclitaxel, respectively [40,41], but the preliminary results of the NORPANC trial showed opposite trends with worse survival rates despite an improvement in R0 resection rates after neoadjuvant treatment [42]. Further ongoing studies including the ALLIANCE (NCT 04340141) and PREOPANC-3 (NL75539.078.20) will add to the growing evidence for the treatment sequence in resectable PDACs.
For borderline resectable pancreatic cancers, there is a higher risk of a margin-positive resection when using a surgery-first approach [39]. It is reported that, of all patients started on neoadjuvant therapy, only approximately 65% will undergo resection, with a majority of the rest having progression of the disease [13]. Overall, the ESPAC-5 and PREOPANC trials showed a survival benefit with neoadjuvant treatment in borderline resectable patients [39,43]. In the PREOPANC-I study, a median survival benefit of 4.4 months (17.6 months versus 13.2 months, HR 0.62, 95%CI: 0.40–0.95) and an improved R0 resection rate (72% vs. 43%, p < 0.001) were observed in the intention-to-treat analysis comparing gemcitabine plus radiotherapy versus upfront surgery [39]. The survival benefit was present despite a lower resection rate, which was 61% in the neoadjuvant group and 72% in the upfront surgery group. Similar findings were seen within the ESPAC-5 trial, with chemotherapy outperforming chemoradiotherapy in most outcomes except R0 resection rates [43].
In locally advanced pancreatic cancers, induction therapy can potentially result in a downstaging of the disease and allowing resection in approximately 35–45% of the patients [44,45]. With 8–12 cycles of mFOLFIRINOX treatment at high-volume centers with experience in vascular resections, the resection rates can be up to 60% [11]. As shown by Hackert et al., resection leads to a significant improvement in survival (15.3 months after resection vs. 8.5 months after exploration alone, p < 0.001) [11]. Arguably, resection may have been performed more in patients with better response to therapy or smaller tumors; however, this indicates that, with appropriate patient selection, improved survival can be achieved. Favorable survival outcomes were also concluded in a multicenter study with propensity score matching (3-year OS of 31% in the resected group versus 11% in the non-resected group) [46] and in a study investigating patients with a favorable tumor biology (>40 months versus 21.4 months, p < 0.001) [47]. Further trials are awaited to determine the optimal length of induction treatment and to investigate the role of vascular surgery. The value of radiotherapy in pancreatic cancer is highly disputed; no evidence supports a survival benefit while it is anecdotally associated with more challenging resections due to postradiation tissue changes [36].
With an improvement in systemic control via the introduction of multiagent systemic therapies, a growing interest in resecting patients with oligometastatic disease (limited metastatic disease confined to a single distant organ) has been observed. Resection of oligometastatic cancer may be beneficial in a highly selected group of patients after induction therapy [48]. Multiple retrospective studies have reported a survival benefit after surgical resection versus no resection [49,50]. Nagai et al. from the Johns Hopkins University showed the feasibility of resection for oligometastatic PDAC to the liver with 38.1 months median survival after induction chemotherapy [49]. Furthermore, the series from the Heidelberg University Hospital showed a 5-year survival of 8.1% [50]. This suggests that a small cohort of patients with oligometastatic disease can achieve long-term survival and, hence, are optimal surgical candidates. As of now, the single-arm prospective HOLIPANC study is enrolling patients with oligometastatic liver PDAC to receive induction therapy followed by surgical exploration and resection when feasible [51]. Additionally, the randomized trial METAPANC (AIO-PAK-0219) is currently in the phase of patients acquisition for resection after induction therapy with a minimum of eight cycles of FOLFIRINOX versus continued FOLFIRINOX treatment.
Many of the aforementioned studies focus on the liver as the metastatic site; however, isolated lung oligometastatic pancreatic cancer is associated with a better prognosis than that with liver or peritoneal involvement [4,52,53]. The limited evidence that is available shows improved outcomes for primary surgery and for metastasectomy in lung-only synchronous or metachronous pancreatic cancer [52,53]. Stuart et al. reported a survival after recurrence of 30.8 months in the subgroup undergoing pulmonary metastasectomy versus 18.6 months in patients who were managed non-surgically [54]. Furthermore, an impressive median overall survival of 68.9 months after surgical treatment for lung-only recurrence was observed by Groot et al. [55]. Further studies are required to dissect the tumor biology in this cohort to identify biologically distinct subtypes. In doing so, in the future, we will be able to define cohorts that are most likely to benefit from surgical resection even in the setting of oligometastatic disease.

4. Biological Assessment of Resectability

The anatomical considerations described earlier are vital for surgical planning and predicting local resectability. However, the systemically aggressive nature of PDAC poses a biological challenge, and multiple considerations need to be made (Figure 1). Despite substantial improvements in surgical technique and perioperative care, patient selection, therefore, should consider the tumor biology and conditional factors [56,57]. Therefore, the IAP, the MD Anderson Cancer Center and JSHBPS have published several statements regarding biological considerations for resectability [15,16,58]. In the IAP international consensus statement, the ECOG score as a surrogate conditional factor for poor tolerance to therapy was integrated as well as evidence of lymph node metastases and a cutoff value for CA19-9 (500 µ/mL) as variables for upstaging anatomical resectable disease to borderline resectable pancreatic cancer [15]. The evidence for these considerations as well as future perspectives will be discussed subsequently.
Lymph node involvement does not alter local resectability in common guidelines but is represented in the biological considerations of the IAP international consensus statement [15,16,58]. In pathological evaluations, only approximately one out of three patients have no lymph node involvement [59]. A lymph node ratio > 0.2 is a strong predictor of systemic recurrence and, therefore, represents an unfavorable tumor biology [5,59]. Furthermore, there is a strong correlation to beneficial survival effects for adjuvant chemotherapy in node-positive PDAC patients [60]. Van Roessel et al. also showed that, after neoadjuvant therapy, only patients with pathologically node-positive disease benefit from further adjuvant treatment [61]. Therefore, lymph node positivity is a strong surrogate marker for systemic disease and unfavorable tumor biology. However, given that this information becomes available after resection, it has limited utility in determining resectability. As imaging modalities improve and the preoperative prediction of nodal disease becomes accurate, this could be integrated into the biological assessment of resectability.
Carbohydrate antigen 19-9 (CA19-9) is the most frequently used biomarker for the assessment of disease in pancreatic cancer. Its sensitivity is, however, limited by approximately 15–20% of the patients being non-producers due to having a Lewis antigen Alpha and Beta negative blood group [62]. Furthermore, often, coexistent biliary obstruction can elevate levels [28]. Nevertheless, elevated CA19-9 levels can be used as a surrogate marker for tumor burden and activity [62]. Obtaining a baseline CA19-9 can be used to predict long-term survival and correlates with R0 resections [62]. However, the most important function of CA19-9 is as a biomarker for monitoring treatment response and recurrence.
A baseline CA19-9 cutoff of 500 µ/mL was suggested by the IAP for upstaging resectable to borderline resectable PDAC [15,16]. A retrospective study of upfront-treated patients conducted by Kato et al. showed a worse prognosis for the borderline resectable stage due to biological criteria compared to resectable stages [63]. However, the prognosis was still significantly better than that for non-biological borderline tumors (NCCN borderline or resectable with ECOG > 2) [63]. The CA19-9 cutoff of 500 was not significant as a prognostic factor, but >1000 µ/mL was (OR 2.03, 95%CI: 1.45–2.84) [63]. In the PREOPANC study, there was no difference in treatment effect for patients below or above 500 µ/mL [39]. Again, validated in a combined study of two RCTs, patients with a CA19-9 > 500 µ/mL did not benefit, but patients with a CA19-9 below that threshold did actually have significant benefit from neoadjuvant treatment [64]. Since CA19-9 is a surrogate marker for tumor activity, high CA19-9 is expected in those with systemic disease [62]. However, a recent meta-analysis of recurrence patterns after neoadjuvant therapy suggests that the main benefit of neoadjuvant therapy is local control and, therefore, fewer local recurrences [65]. According to that analysis, there was no reduction in recurrences at distant sites, thus questioning the role of neoadjuvant therapy in systemic control [65]. As many studies are currently being conducted on neoadjuvant chemotherapy in resectable PDAC, further evidence on the value of CA19-9 in resectable stages is expected.
While the optimal cutoff value for upstaging resectable to borderline resectable and, thus, for having an expected benefit from neoadjuvant treatment, has still to be defined, pretreatment CA19-9 levels do not predict resectability in borderline resectable and locally advanced PDAC [66,67]. However, in a retrospective study conducted by Heger et al., a low ratio of pretreatment/posttreatment as well as lower posttreatment CA19-9 levels predicted resectability in borderline and locally advanced PDAC [67]. A posttreatment level of <91.8 U/mL predicted resectability with a 75% sensitivity and 77% specificity. A cutoff of 0.4 from posttreatment divided by pretreatment CA19-9 levels yielded a positive predictive value for resectability of 83% in patients treated with FOLFIRINOX [67]. R0 resections were achieved in 36.4% of the resected cohort, which also included oligo-metastasized staged patients. Interestingly, patients above the cutoffs did not benefit from resection versus exploration in terms of overall survival [67]. Hartlab et al. showed similar results in the NEOLAP trial; however, they stated an optimal cutoff at posttreatment level below 50 U/mL for predicting survival and <61 U/mL for R0 resection rate [66]. These results were further validated in a study from the Massachusetts General Hospital with the normalization of CA19-9 being associated with resectability [68]. Furthermore, a prospective study conducted by van Veldhuisen et al. showed a relevant benefit for adding >30% CA19-9 response to the RECIST criteria [69].
In summary, for a patient with a resectable PDAC, the value of pretreatment CA19-9 and its implications on treatment sequence have yet to be defined. In patients with borderline resectable, locally advanced, and oligometastatic cancer, the pretreatment level of CA19-9 does not predict resectability, but posttreatment level and changes between pre-and posttreatment values do. Therefore, the phase of neoadjuvant treatment provides a window of opportunity to assess tumor biology and allows the tumor to declare its aggressiveness. If a poor response is observed, the chemotherapy agents can be switched [70]. However, after exhausting all available therapies and if a poor response with a high propensity of systemic disease is observed, it could potentially indicate a biologically aggressive tumor, questioning the benefit from further cytotoxic or surgical treatments (Figure 2). Further studies are needed to define optimal cutoffs for CA19-9, such that they are standardized and broadly applicable in clinical practice.
Carcinoembryonic antigen (CEA) is another biomarker that is used as an adjunct to CA19-9 in the clinical setting. An elevated CEA shows associations with advanced tumor stages and poor prognosis [71]. However, for the assessment of resectability, CEA shows poor performance [72]. CEA is furthermore not elevated in approximately 50–60% of PDAC patients [73,74]. Therefore, its current value for the assessment of PDAC is limited, and the need for an alternative biomarker is urgent [73]. Other potential candidate biomarkers that could help assess the tumor biology and systemic burden of the disease have been identified and are being studied. The most reported biomarkers include TIMP-1, DUPAN-2, serum-MUC5AC, CA125, and CA242 [75,76]. While CA19-9 still outperforms the novel biomarkers, biomarker panels including or excluding CA19-9 have shown improvements in diagnostic accuracy [76]. Elevated levels of DUPAN-2 (>200 U/mL) have also shown to be of prognostic value in CA19-9 non-producers [76]. DUPAN-2 is a precursor of CA19-9 and is not further metabolized in Lewis negative blood groups and, therefore, serves as a potential substitute in CA19-9 non-producers [77]. Furthermore, CEA and CA125 are associated with tumor burden and therapeutic response in CA19-9 non-producers, making them potential monitoring biomarkers [78,79]. Inflammation, as a surrogate for tumor dynamics and immunologic antitumor response, has been studied. High neutrophil/lymphocyte ratio (NLR), platelet/lymphocyte ratio (PLR), and elevated c-reactive protein (CRP) have been found to be poor prognostic factors [80,81]. Interestingly, pre-operative NLR and PLR was inversely correlated with R0 resections in a retrospective study by Recio-Boiles et al. [82]. However, these predictive values and their correlations with resectability have to be interpreted with caution. First, there is no accepted cutoff, and there is a high variability within published studies [83]. Second, there is a high possibility of publication bias as many negative results are not published [83]. In summary, the literature in predicting resectability and prognosis with alternative serum protein biomarkers or cell ratios is sparse or not convincing to date.
Through tumor biopsies, additional information can be drawn about the primary tumor, and tumor grading and molecular assessment can be performed. For example, liver recurrence is associated with poor tumor differentiation and often occurs early after surgery [18]. However, even with the recently improved understanding of the mutational landscape, convincing targetable mutations have not yet been identified. PDACs usually arises from pancreatic intraepithelial neoplasia (PanIN) precursor lesions, while some arise from intraductal papillary mucinous neoplasms (IPMNs). In PanIN-derived pancreatic cancer, usually KRAS mutation (87%) is present with later mutations involving TP53 (62%), CDKN2A (16%), and SMAD4 (16%) [84,85]. SMAD4 mutations, loss of function of the CDKN2A tumor suppressor gene, and FGFR2 gene fusions are associated with a poor outcome after standard chemotherapeutic treatment in PDACs [86,87]. Bailey et al. described four different subtypes that differ in their evolution and aggressivity [88]. According to their mutational and expression profiles, PDACs can be subclassified into squamous, ADEX, pancreatic progenitor, and immunogenic subtypes [88]. One stand out for the prognosis is the epithelial subtype, which has considerable overlap with the quasi-mesenchymal subtype described by Collissons et al. [89] The squamous subtype is characterized by gene networks that are involved in inflammation, hypoxia response, metabolic reprogramming, TGF-B signaling, MYC pathway activation, autophagy, and upregulated expression of TP63 [88]. Many of those pathways are involved in epithelial–mesenchymal transition and tumor dormancy [90]. This leads to more adaptive tumor cells and results in more treatment failure and poorer outcomes compared to the classical subtypes [91]. Therefore, in future, subtyping may aid in treatment decisions concerning biological resectability.
Liquid biopsy has emerged as a promising technology that allows the non-invasive sampling of tumor fragments within the blood circulation including circulating tumor DNA (ctDNA), microRNA, tumor-derived exosomes, and circulating tumor cells (CTCs) [62]. The analysis of liquid biopsies can inform treatment decisions as it harbors specific information about the tumor biology such as specific mutations and epigenetic changes from the primary tumor or metastatic sites [62]. In the future, it can possibly overcome the drawback of tumor biopsies with limited identification of tumor heterogeneity as it represents the features of the systemic involvement of disease [92]. As a reliable marker for systemic disease, it most possibly fails to detect local-only disease, limiting its diagnostic value. Therefore, in a current meta-analysis, pooled sensitivity for ctDNA and CTC is low with 64% and 74% with a higher specificity of 92% and 83%, respectively [93].
Circulating tumor DNA is defined as the tumor-derived portion of cell free DNA within the circulation. Through apoptosis and necrosis within the tumor, ctDNA is released into the circulation [94]. The ctDNA-fragments are isolated, amplified, and analyzed with PCR or sequencing [92]. With an approximate mutation rate of 90%, KRAS (most polymorphisms G12D, G12V, and G12R) is the most important ctDNA marker, but panels of different mutations can also be used [92,94]. Low concentrations and mutations in premalignant lesions hamper its value as a diagnostic biomarker. However, ctDNA has shown valuable results as a diagnostic, monitoring, and prognostic biomarker in many cancer types [95,96,97]. When detected at baseline, it predicts worse progression-free and overall survival [98]. Longitudinal multigene ctDNA measurements furthermore predicted progression through increasing levels in 70% of patients with advanced PDAC in the study of Lapin et al. [98]. The lead time to radiologically determined progression was 19 days compared to 6 days in CA19-9 increase [98]. Therefore, it could possibly be used as a monitoring biomarker also in neoadjuvant treatment as ctDNA persistence was associated with treatment failure to cytotoxic agents [98].
In terms of predicting resectability, ctDNA, to date, has not been implemented in clinical decision making. Higher ctDNA levels are associated with vascular involvement and advanced local tumor size [99]. Furthermore, ctDNA was correlated with positive resection margins within the study of McDuff et al. [100]. In LAPC, ctDNA-positive patients had 44% R0 resections compared to 88% with ctDNA-negative findings (n = 29) [100]. In summary, since preoperative ctDNA positivity has shown to be a predictor of early recurrence and worse survival outcomes in resectable PDAC, its positivity could aid the selection of resectable PDAC patients who will benefit from neoadjuvant treatment compared to upfront resection [100,101].
Circulating tumor cells are tumor cells that have detached from the primary tumor or metastatic site and can be found within the blood stream. Epithelial–mesenchymal transition (EMT) can lead to cell migration, intravasation, and consequently, the presence of CTCs, which are key players in the early dissemination and metastatic seeding of pancreatic cancer cells [102,103]. The detection and isolation of CTCs can be performed by biomarker-mediated platforms such as microfluidic chips, magnetic beads, or size-sensitive microfiltration [104]. Analysis can then be performed, applying a wide range of methods including flow cytometry, immunofluorescence staining, and single-cell RNA sequencing (scRNAseq) [104]. As with ctDNA, the presence of CTCs with mesenchymal properties predicts recurrence after pancreatectomy [105,106]. This wide arrange of analytical method and the CTCs representing systemic disease of a heterogenous tumor may allow precision oncology in future. Anatomical resectable PDACs with positive transitional CTCs may benefit from neoadjuvant systemic treatment compared to upfront surgery.

5. Current Limitations of Assessment of Anatomical and Biological Resectability

To date, there are certain limitations to the assessment of biological resectability that urgently need to be addressed. First, the radiographic assessment of lymph node involvement is unreliable. The enlargement of lymph nodes beyond the cutoff of 1 cm is deemed as suspicious for lymph node involvement in cancer. Since the enlargement of lymph nodes can also be due to peritumoral inflammation and, on the other hand, metastatic lymph nodes can measure below the cutoff value of 10 mm, the poor performance of CT scans in terms of diagnostic accuracy (38–77%) and sensitivity (14–24%) is seen in clinical practice [19]. In the future, more adequate lymph node examinations, for example, through biopsies or PET-CT may fill the gap between the reliability of radiological to pathological assessment for the decision on neoadjuvant treatment in resectable PDACs [19].
Second, the assessment of treatment response is challenging. Radiological response assessment is performed through a CT scan using the Response Evaluation Criteria in Solid Tumors (RECIST) [107]. Progression according to the RECIST criteria encompasses a tumor growth of at least 20% in diameter or newly detected lesions; stable disease is defined by the absence of progression or regression; and regression is defined as a shrinkage of at least 30% in three directions [107]. The percentages of patients with progression, stable disease, partial, and complete response after induction treatment with mFOLFIRINOX were 16%, 59%, 16%, and <1%, respectively [19,28]. However, radiological assessment after neoadjuvant treatment underestimates resectability [108,109]. In the interpretation of radiological findings, the assessment of regression is challenging due to fibrotic and edematous posttreatment reactions that are hard to distinguish from viable tumors, especially at the tumor borders [19]. Therefore, the radiologic and pathologic evaluations of tumor response only show moderate agreement [110]. In a case series by Ferrone et al., 92% of patients with LAPC had an R0 resection even though imaging suggested non-resectability [109]. Furthermore, there are no reliable predictors of resectability after neoadjuvant or induction treatment for LAPC and borderline resectable PDAC [111]. Therefore, surgical exploration should be performed in all fit patients without progression or secondary metastatic disease after induction therapy at a high-volume center with experience in arterial divestment and resection [28,36].
Third, in current clinical practice, the biological treatment response is mainly determined by measurement of the CA19-9 levels [112]. Since alternative serum protein biomarkers have failed to compensate for the drawbacks of CA19-9, this represents an unsatisfactory state. Efforts are currently being undertaken, especially with liquid biopsies, to compensate for those shortcomings. For example, Meijer et al. have shown that the downregulation of microRNA-181a-5p can be used to monitor the response to FOLFIRINOX [113]. Further data on liquid biopsies, especially in the setting of neoadjuvant therapy, are much awaited. Molecular analyses have yet shown very few targetable lesions or implications about different treatments. Possibly, there is an advantage in immunotherapy for the immunogenic subtype that has more immune-cell infiltration compared to the other types being immunogenic cold tumors [88]. Other efforts toward personalized treatment, for example, better treatment response to platinum-based chemotherapy, are currently under investigation [114]. In the future, multianalyte panels encompassing a range of variables have the potential to mitigate the limitations associated with solitary biomarkers.

6. Conclusions

Assessment of local resectability based on presence and extent of vascular involvement of major abdominal vessels has prognostic value. However, due to advancements in surgical technique including vascular resections, biological considerations are becoming even more important for treatment decision making in patients with pancreatic cancer. Patients who present with anatomically resectable disease and unfavorable tumor biology may experience early recurrence and therefore potentially morbid resection might have limited values in these cases. Conversely, patients with anatomically advanced tumors exhibiting favorable tumor biology have the potential to attain cure and long-term survival through surgical intervention. Therefore, if treatment decisions are solely guided by anatomic criteria, a risk of surgically overtreating biologically aggressive diseases and undertreating those with biologically favorable profiles exists.
The assessment of tumor biology can be performed through surrogate markers indicative of advanced disease and systemic involvement such as CA19-9, ctDNA, and CTCs, but, to date, a majority of these biomarkers have performed insufficiently for predicting treatment benefit when analyzed alone. However, a combination of the aforementioned biomarkers could possibly define candidates who will benefit from resection compared to those who probably will develop early systemic recurrence and have dismal prognosis despite surgical treatment. Developing multianalyte composite tests based on these biomarkers is essential for defining optimal personalized treatment. Future guidelines combining anatomic and biologic features in the determination of resectability could optimize surgical candidacy such that patients undergoing resection derive maximum benefit from these operations. If a patient progresses anatomically or biological unresectability persists, this patient might not be an appropriate candidate for surgical resection. However, if a patient with LAPC with favorable tumor biology does not progress during induction chemotherapy, resection could be offered at specialized centers.

Author Contributions

Conceptualization, I.F.R., C.L.W. and A.A.J.; methodology, A.A.J.; software, I.F.R.; validation, J.R.H., A.A.J. and C.L.W.; formal analysis, NA; investigation, I.F.R.; resources, C.L.W.; data curation, I.F.R. and J.R.H.; writing—original draft preparation, I.F.R.; writing—review and editing, J.R.H., C.L.W. and A.A.J.; visualization, I.F.R. and A.A.J.; supervision, C.L.W.; project administration, A.A.J.; funding acquisition, NA. All authors have read and agreed to the published version of the manuscript.

Funding

Ingmar F. Rompen is supported by the Swiss National Science Foundation (SNSF, grant number 217684).

Conflicts of Interest

There are no conflicts of interest for any of the authors.

References

  1. Rahib, L.; Smith, B.D.; Aizenberg, R.; Rosenzweig, A.B.; Fleshman, J.M.; Matrisian, L.M. Projecting cancer incidence and deaths to 2030: The unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014, 74, 2913–2921. [Google Scholar] [CrossRef] [PubMed]
  2. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2020. CA Cancer J. Clin. 2020, 70, 7–30. [Google Scholar] [CrossRef] [PubMed]
  3. Bockhorn, M.; Uzunoglu, F.G.; Adham, M.; Imrie, C.; Milicevic, M.; Sandberg, A.A.; Asbun, H.J.; Bassi, C.; Buchler, M.; Charnley, R.M.; et al. Borderline resectable pancreatic cancer: A consensus statement by the International Study Group of Pancreatic Surgery (ISGPS). Surgery 2014, 155, 977–988. [Google Scholar] [CrossRef] [PubMed]
  4. Rompen, I.F.; Levine, J.; Habib, J.R.; Sereni, E.; Mughal, N.; Hewitt, D.B.; Sacks, G.D.; Welling, T.H.; Simeone, D.M.; Kaplan, B.; et al. Progression of Site-Specific Recurrence of Pancreatic Cancer and Implications for Treatment. Ann. Surg. 2023; online ahead of print. [Google Scholar] [CrossRef] [PubMed]
  5. Groot, V.P.; Rezaee, N.; Wu, W.; Cameron, J.L.; Fishman, E.K.; Hruban, R.H.; Weiss, M.J.; Zheng, L.; Wolfgang, C.L.; He, J. Patterns, Timing, and Predictors of Recurrence Following Pancreatectomy for Pancreatic Ductal Adenocarcinoma. Ann. Surg. 2018, 267, 936–945. [Google Scholar] [CrossRef] [PubMed]
  6. 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: A Secondary Analysis of the ESPAC-4 Randomized Adjuvant Chemotherapy Trial. JAMA Surg. 2019, 154, 1038–1048. [Google Scholar] [CrossRef] [PubMed]
  7. Chiorean, E.G.; Chiaro, M.D.; Tempero, M.A.; Malafa, M.P.; Benson, A.B.; Cardin, D.B.; Christensen, J.A.; Chung, V.; Czito, B.; Dillhoff, M.; et al. Ampullary Adenocarcinoma, Version 1.2023, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Canc. Netw. 2023, 21, 753–782. [Google Scholar] [CrossRef]
  8. Kinny-Köster, B.; van Oosten, F.; Habib, J.R.; Javed, A.A.; Cameron, J.L.; Lafaro, K.J.; Burkhart, R.A.; Burns, W.R.; He, J.; Fishman, E.K.; et al. Mesoportal bypass, interposition graft, and mesocaval shunt: Surgical strategies to overcome superior mesenteric vein involvement in pancreatic cancer. Surgery 2020, 168, 1048–1055. [Google Scholar] [CrossRef]
  9. Kinny-Köster, B.; Habib, J.R.; van Oosten, F.; Javed, A.A.; Cameron, J.L.; Burkhart, R.A.; Burns, W.R.; He, J.; Wolfgang, C.L. Conduits in Vascular Pancreatic Surgery: Analysis of Clinical Outcomes, Operative Techniques, and Graft Performance. Ann. Surg. 2023, 278, e94–e104. [Google Scholar] [CrossRef]
  10. Loos, M.; Kester, T.; Klaiber, U.; Mihaljevic, A.L.; Mehrabi, A.; Müller-Stich, B.M.; Diener, M.K.; Schneider, M.A.; Berchtold, C.; Hinz, U.; et al. Arterial Resection in Pancreatic Cancer Surgery: Effective After a Learning Curve. Ann. Surg. 2022, 275, 759–768. [Google Scholar] [CrossRef]
  11. Hackert, T.; Sachsenmaier, M.; Hinz, U.; Schneider, L.; Michalski, C.W.; Springfeld, C.; Strobel, O.; Jäger, D.; Ulrich, A.; Büchler, M.W. Locally Advanced Pancreatic Cancer: Neoadjuvant Therapy with Folfirinox Results in Resectability in 60% of the Patients. Ann. Surg. 2016, 264, 457–463. [Google Scholar] [CrossRef] [PubMed]
  12. Gemenetzis, G.; Groot, V.P.; Blair, A.B.; Laheru, D.A.; Zheng, L.; Narang, A.K.; Fishman, E.K.; Hruban, R.H.; Yu, J.; Burkhart, R.A.; et al. Survival in Locally Advanced Pancreatic Cancer after Neoadjuvant Therapy and Surgical Resection. Ann. Surg. 2019, 270, 340–347. [Google Scholar] [CrossRef] [PubMed]
  13. Javed, A.A.; Wright, M.J.; Siddique, A.; Blair, A.B.; Ding, D.; Burkhart, R.A.; Makary, M.; Cameron, J.L.; Narang, A.; Herman, J.; et al. Outcome of Patients with Borderline Resectable Pancreatic Cancer in the Contemporary Era of Neoadjuvant Chemotherapy. J. Gastrointest. Surg. 2019, 23, 112–121. [Google Scholar] [CrossRef] [PubMed]
  14. Oba, A.; Del Chiaro, M.; Fujii, T.; Okano, K.; Stoop, T.F.; Wu, Y.H.A.; Maekawa, A.; Yoshida, Y.; Hashimoto, D.; Sugawara, T.; et al. “Conversion surgery” for locally advanced pancreatic cancer: A position paper by the study group at the joint meeting of the International Association of Pancreatology (IAP) & Japan Pancreas Society (JPS) 2022. Pancreatology 2023, 23, 712–720. [Google Scholar] [CrossRef] [PubMed]
  15. Isaji, S.; Mizuno, S.; Windsor, J.A.; Bassi, C.; Fernández-del Castillo, C.; Hackert, T.; Hayasaki, A.; Katz, M.H.G.; Kim, S.-W.; Kishiwada, M.; et al. International consensus on definition and criteria of borderline resectable pancreatic ductal adenocarcinoma 2017. Pancreatology 2018, 18, 2–11. [Google Scholar] [CrossRef]
  16. Oba, A.; Del Chiaro, M.; Satoi, S.; Kim, S.W.; Takahashi, H.; Yu, J.; Hioki, M.; Tanaka, M.; Kato, Y.; Ariake, K.; et al. New criteria of resectability for pancreatic cancer: A position paper by the Japanese Society of Hepato-Biliary-Pancreatic Surgery (JSHBPS). J. Hepatobiliary Pancreat. Sci. 2022, 29, 725–731. [Google Scholar] [CrossRef]
  17. Lee, E.S.; Lee, J.M. Imaging diagnosis of pancreatic cancer: A state-of-the-art review. World J. Gastroenterol. 2014, 20, 7864–7877. [Google Scholar] [CrossRef]
  18. Tanaka, M.; Mihaljevic, A.L.; Probst, P.; Heckler, M.; Klaiber, U.; Heger, U.; Büchler, M.W.; Hackert, T. Meta-analysis of recurrence pattern after resection for pancreatic cancer. Br. J. Surg. 2019, 106, 1590–1601. [Google Scholar] [CrossRef]
  19. Søreide, K.; Rangelova, E.; Dopazo, C.; Mieog, S.; Stättner, S. Pancreatic cancer. Eur. J. Surg. Oncol. 2023, 49, 521–525. [Google Scholar] [CrossRef]
  20. Habib, J.R.; Kinny-Köster, B.; van Oosten, F.; Javed, A.A.; Cameron, J.L.; Lafaro, K.J.; Burkhart, R.A.; Burns, W.R.; He, J.; Thompson, E.D.; et al. Periadventitial dissection of the superior mesenteric artery for locally advanced pancreatic cancer: Surgical planning with the “halo sign” and “string sign”. Surgery 2021, 169, 1026–1031. [Google Scholar] [CrossRef]
  21. Hackert, T.; Strobel, O.; Michalski, C.W.; Mihaljevic, A.L.; Mehrabi, A.; Müller-Stich, B.; Berchtold, C.; Ulrich, A.; Büchler, M.W. The TRIANGLE operation—Radical surgery after neoadjuvant treatment for advanced pancreatic cancer: A single arm observational study. HPB 2017, 19, 1001–1007. [Google Scholar] [CrossRef] [PubMed]
  22. Hackert, T.; Werner, J.; Weitz, J.; Schmidt, J.; Büchler, M.W. Uncinate process first--a novel approach for pancreatic head resection. Langenbecks Arch. Surg. 2010, 395, 1161–1164. [Google Scholar] [CrossRef] [PubMed]
  23. Weitz, J.; Rahbari, N.; Koch, M.; Büchler, M.W. The “artery first” approach for resection of pancreatic head cancer. J. Am. Coll. Surg. 2010, 210, e1–e4. [Google Scholar] [CrossRef] [PubMed]
  24. Müller, P.C.; Müller, B.P.; Hackert, T. Contemporary artery-first approaches in pancreatoduodenectomy. Br. J. Surg. 2023, 110, 1570–1573. [Google Scholar] [CrossRef] [PubMed]
  25. Chu, L.C.; Fishman, E.K. Pancreatic ductal adenocarcinoma staging: A narrative review of radiologic techniques and advances. Int. J. Surg. 2023, 10–1097. [Google Scholar] [CrossRef]
  26. Javed, A.A.; Young, R.W.C.; Habib, J.R.; Kinny-Köster, B.; Cohen, S.M.; Fishman, E.K.; Wolfgang, C.L. Cinematic Rendering: Novel Tool for Improving Pancreatic Cancer Surgical Planning. Curr. Probl. Diagn. Radiol. 2022, 51, 878–883. [Google Scholar] [CrossRef]
  27. Asbun, H.J.; Conlon, K.; Fernandez-Cruz, L.; Friess, H.; Shrikhande, S.V.; Adham, M.; Bassi, C.; Bockhorn, M.; Büchler, M.; Charnley, R.M.; et al. When to perform a pancreatoduodenectomy in the absence of positive histology? A consensus statement by the International Study Group of Pancreatic Surgery. Surgery 2014, 155, 887–892. [Google Scholar] [CrossRef]
  28. van Veldhuisen, E.; van den Oord, C.; Brada, L.J.; Walma, M.S.; Vogel, J.A.; Wilmink, J.W.; del Chiaro, M.; van Lienden, K.P.; Meijerink, M.R.; van Tienhoven, G.; et al. Locally Advanced Pancreatic Cancer: Work-Up, Staging, and Local Intervention Strategies. Cancers 2019, 11, 976. [Google Scholar] [CrossRef]
  29. Allen, V.B.; Gurusamy, K.S.; Takwoingi, Y.; Kalia, A.; Davidson, B.R. Diagnostic accuracy of laparoscopy following computed tomography (CT) scanning for assessing the resectability with curative intent in pancreatic and periampullary cancer. Cochrane Database Syst. Rev. 2016, 7, Cd009323. [Google Scholar] [CrossRef]
  30. van Dongen, J.C.; Versteijne, E.; Bonsing, B.A.; Mieog, J.S.D.; de Hingh, I.; Festen, S.; Patijn, G.A.; van Dam, R.; van der Harst, E.; Wijsman, J.H.; et al. The yield of staging laparoscopy for resectable and borderline resectable pancreatic cancer in the PREOPANC randomized controlled trial. Eur. J. Surg. Oncol. 2023, 49, 811–817. [Google Scholar] [CrossRef]
  31. Michiels, N.; Doppenberg, D.; Groen, J.V.; van Veldhuisen, E.; Bonsing, B.A.; Busch, O.R.; Crobach, A.; van Delden, O.M.; van Dieren, S.; Farina, A.; et al. Intraoperative Ultrasound During Surgical Exploration in Patients with Pancreatic Cancer and Vascular Involvement (ULTRAPANC): A Prospective Multicenter Study. Ann. Surg. Oncol. 2023, 30, 3455–3463. [Google Scholar] [CrossRef] [PubMed]
  32. Amin, M.B.; Greene, F.L.; Edge, S.B.; Compton, C.C.; Gershenwald, J.E.; Brookland, R.K.; Meyer, L.; Gress, D.M.; Byrd, D.R.; Winchester, D.P. The Eighth Edition AJCC Cancer Staging Manual: Continuing to build a bridge from a population-based to a more “personalized” approach to cancer staging. CA Cancer J. Clin. 2017, 67, 93–99. [Google Scholar] [CrossRef] [PubMed]
  33. 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]
  34. Katz, M.H.; Shi, Q.; Ahmad, S.A.; Herman, J.M.; Marsh Rde, W.; Collisson, E.; Schwartz, L.; Frankel, W.; Martin, R.; Conway, W.; et al. Preoperative Modified FOLFIRINOX Treatment Followed by Capecitabine-Based Chemoradiation for Borderline Resectable Pancreatic Cancer: Alliance for Clinical Trials in Oncology Trial A021101. JAMA Surg. 2016, 151, e161137. [Google Scholar] [CrossRef] [PubMed]
  35. Varadhachary, G.R.; Tamm, E.P.; Abbruzzese, J.L.; Xiong, H.Q.; Crane, C.H.; Wang, H.; Lee, J.E.; Pisters, P.W.; Evans, D.B.; Wolff, R.A. Borderline resectable pancreatic cancer: Definitions, management, and role of preoperative therapy. Ann. Surg. Oncol. 2006, 13, 1035–1046. [Google Scholar] [CrossRef] [PubMed]
  36. Springfeld, C.; Ferrone, C.R.; Katz, M.H.G.; Philip, P.A.; Hong, T.S.; Hackert, T.; Büchler, M.W.; Neoptolemos, J. Neoadjuvant therapy for pancreatic cancer. Nat. Rev. Clin. Oncol. 2023, 20, 318–337. [Google Scholar] [CrossRef]
  37. Wu, W.; He, J.; Cameron, J.L.; Makary, M.; Soares, K.; Ahuja, N.; Rezaee, N.; Herman, J.; Zheng, L.; Laheru, D.; et al. The impact of postoperative complications on the administration of adjuvant therapy following pancreaticoduodenectomy for adenocarcinoma. Ann. Surg. Oncol. 2014, 21, 2873–2881. [Google Scholar] [CrossRef]
  38. Versteijne, E.; Vogel, J.A.; Besselink, M.G.; Busch, O.R.C.; Wilmink, J.W.; Daams, J.G.; van Eijck, C.H.J.; Groot Koerkamp, B.; Rasch, C.R.N.; van Tienhoven, G.; et al. Meta-analysis comparing upfront surgery with neoadjuvant treatment in patients with resectable or borderline resectable pancreatic cancer. Br. J. Surg. 2018, 105, 946–958. [Google Scholar] [CrossRef]
  39. Versteijne, E.; van Dam, J.L.; Suker, M.; Janssen, Q.P.; Groothuis, K.; Akkermans-Vogelaar, J.M.; Besselink, M.G.; Bonsing, B.A.; Buijsen, J.; Busch, O.R.; et al. Neoadjuvant Chemoradiotherapy Versus Upfront Surgery for Resectable and Borderline Resectable Pancreatic Cancer: Long-Term Results of the Dutch Randomized PREOPANC Trial. J. Clin. Oncol. 2022, 40, 1220–1230. [Google Scholar] [CrossRef]
  40. Seufferlein, T.; Uhl, W.; Kornmann, M.; Algül, H.; Friess, H.; König, A.; Ghadimi, M.; Gallmeier, E.; Bartsch, D.K.; Lutz, M.P.; et al. Perioperative or only adjuvant gemcitabine plus nab-paclitaxel for resectable pancreatic cancer (NEONAX)—A randomized phase II trial of the AIO pancreatic cancer group. Ann. Oncol. 2023, 34, 91–100. [Google Scholar] [CrossRef]
  41. Schwarz, L.; Bachet, J.-B.; Meurisse, A.; Bouché, O.; Assenat, E.; Piessen, G.; Terrebonne, E.; Hammel, P.; Regenet, N.; Turco, C.; et al. Resectable pancreatic adenocarcinoma neo-adjuvant FOLF(IRIN)OX-based chemotherapy: A multicenter, non-comparative, randomized, phase II trial (PANACHE01-PRODIGE48 study). J. Clin. Oncol. 2022, 40 (Suppl. S16), 4134. [Google Scholar] [CrossRef]
  42. Labori, K.J.; Bratlie, S.O.; Biörserud, C.; Björnsson, B.; Bringeland, E.; Elander, N.; Grønbech, J.E.; Haux, J.; Hemmingsson, O.; Nymo, L.; et al. Short-course neoadjuvant FOLFIRINOX versus upfront surgery for resectable pancreatic head cancer: A multicenter randomized phase-II trial (NORPACT-1). J. Clin. Oncol. 2023, 41 (Suppl. S17), LBA4005. [Google Scholar] [CrossRef]
  43. Ghaneh, P.; Palmer, D.; Cicconi, S.; Jackson, R.; Halloran, C.M.; Rawcliffe, C.; Sripadam, R.; Mukherjee, S.; Soonawalla, Z.; Wadsley, J.; et al. Immediate surgery compared with short-course neoadjuvant gemcitabine plus capecitabine, FOLFIRINOX, or chemoradiotherapy in patients with borderline resectable pancreatic cancer (ESPAC5): A four-arm, multicentre, randomised, phase 2 trial. Lancet Gastroenterol. Hepatol. 2023, 8, 157–168. [Google Scholar] [CrossRef] [PubMed]
  44. Kunzmann, V.; Siveke, J.T.; Algül, H.; Goekkurt, E.; Siegler, G.; Martens, U.; Waldschmidt, D.; Pelzer, U.; Fuchs, M.; Kullmann, F.; et al. Nab-paclitaxel plus gemcitabine versus nab-paclitaxel plus gemcitabine followed by FOLFIRINOX induction chemotherapy in locally advanced pancreatic cancer (NEOLAP-AIO-PAK-0113): A multicentre, randomised, phase 2 trial. Lancet Gastroenterol. Hepatol. 2021, 6, 128–138. [Google Scholar] [CrossRef]
  45. Fietkau, R.; Ghadimi, M.; Grützmann, R.; Wittel, U.A.; Jacobasch, L.; Uhl, W.; Croner, R.S.; Bechstein, W.O.; Neumann, U.P.; Waldschmidt, D.; et al. Randomized phase III trial of induction chemotherapy followed by chemoradiotherapy or chemotherapy alone for nonresectable locally advanced pancreatic cancer: First results of the CONKO-007 trial. J. Clin. Oncol. 2022, 40 (Suppl. S16), 4008. [Google Scholar] [CrossRef]
  46. Brada, L.J.H.; Daamen, L.A.; Magermans, L.G.; Walma, M.S.; Latifi, D.; van Dam, R.M.; de Hingh, I.H.; Liem, M.S.L.; de Meijer, V.E.; Patijn, G.A.; et al. Survival Benefit Associated with Resection of Locally Advanced Pancreatic Cancer after Upfront FOLFIRINOX Versus FOLFIRINOX Only: Multicenter Propensity Score-matched Analysis. Ann. Surg. 2021, 274, 729–735. [Google Scholar] [CrossRef]
  47. Ushida, Y.; Inoue, Y.; Oba, A.; Mie, T.; Ito, H.; Ono, Y.; Sato, T.; Ozaka, M.; Sasaki, T.; Saiura, A.; et al. Optimizing Indications for Conversion Surgery Based on Analysis of 454 Consecutive Japanese Cases with Unresectable Pancreatic Cancer Who Received Modified FOLFIRINOX or Gemcitabine Plus Nab-paclitaxel: A Single-Center Retrospective Study. Ann. Surg. Oncol. 2022, 29, 5038–5050. [Google Scholar] [CrossRef]
  48. Kaslow, S.R.; Sacks, G.D.; Berman, R.S.; Lee, A.Y.; Correa-Gallego, C. Natural History of Stage IV Pancreatic Cancer. Identifying Survival Benchmarks for Curative-intent Resection in Patients with Synchronous Liver-only Metastases. Ann. Surg. 2022, 278, e798–e804. [Google Scholar] [CrossRef]
  49. Nagai, M.; Wright, M.J.; Ding, D.; Thompson, E.D.; Javed, A.A.; Weiss, M.J.; Hruban, R.H.; Yu, J.; Burkhart, R.A.; He, J.; et al. Oncologic resection of pancreatic cancer with isolated liver metastasis: Favorable outcomes in select patients. J. Hepatobiliary Pancreat Sci. 2023, 30, 1025–1035. [Google Scholar] [CrossRef]
  50. Hackert, T.; Niesen, W.; Hinz, U.; Tjaden, C.; Strobel, O.; Ulrich, A.; Michalski, C.W.; Büchler, M.W. Radical surgery of oligometastatic pancreatic cancer. Eur. J. Surg. Oncol. 2017, 43, 358–363. [Google Scholar] [CrossRef]
  51. Gebauer, F.; Damanakis, A.I.; Popp, F.; Quaas, A.; Kütting, F.; Lutz, K.; Held, S.; Deuß, B.; Göser, T.; Waldschmidt, D.; et al. Study protocol of an open-label, single arm phase II trial investigating the efficacy, safety and quality of life of neoadjuvant chemotherapy with liposomal irinotecan combined with Oxaliplatin and 5-fluorouracil/Folinic acid followed by curative surgical resection in patients with hepatic Oligometastatic adenocarcinoma of the pancreas (HOLIPANC). BMC Cancer 2021, 21, 1239. [Google Scholar] [CrossRef]
  52. Ilmer, M.; Schiergens, T.S.; Renz, B.W.; Schneider, C.; Sargut, M.; Waligora, R.; Weniger, M.; Hartwig, W.; Ceyhan, G.O.; Friess, H.; et al. Oligometastatic pulmonary metastasis in pancreatic cancer patients: Safety and outcome of resection. Surg. Oncol. 2019, 31, 16–21. [Google Scholar] [CrossRef] [PubMed]
  53. Liu, Q.; Zhang, R.; Michalski, C.W.; Liu, B.; Liao, Q.; Kleeff, J. Surgery for synchronous and metachronous single-organ metastasis of pancreatic cancer: A SEER database analysis and systematic literature review. Sci. Rep. 2020, 10, 4444. [Google Scholar] [CrossRef] [PubMed]
  54. Stuart, C.M.; Kirsch, M.J.; Zhuang, Y.; Meguid, C.L.; Sugawara, T.; Colborn, K.L.; Messersmith, W.; Lieu, C.; Gleisner, A.L.; Del Chiaro, M.; et al. Pulmonary metastasectomy is associated with survival after lung-only recurrence in pancreatic cancer. Surgery 2023, 174, 654–659. [Google Scholar] [CrossRef] [PubMed]
  55. Groot, V.P.; Blair, A.B.; Gemenetzis, G.; Ding, D.; Burkhart, R.A.; van Oosten, A.F.; Molenaar, I.Q.; Cameron, J.L.; Weiss, M.J.; Yang, S.C.; et al. Isolated pulmonary recurrence after resection of pancreatic cancer: The effect of patient factors and treatment modalities on survival. HPB 2019, 21, 998–1008. [Google Scholar] [CrossRef] [PubMed]
  56. Rompen, I.F.; Merz, D.C.; Alhalabi, K.T.; Klotz, R.; Kalkum, E.; Pausch, T.M.; Strothmann, H.; Probst, P. Perioperative Drug Treatment in Pancreatic Surgery-A Systematic Review and Meta-Analysis. J. Clin. Med. 2023, 12, 1750. [Google Scholar] [CrossRef] [PubMed]
  57. Rompen, I.F.; Nienhuser, H.; Crnovrsanin, N.; Musa, J.; Haag, G.M.; Longerich, T.; Fiedler, T.; Muller-Stich, B.P.; Sisic, L.; Billeter, A.T. Clinical Characteristics and Oncological Outcomes of Surgically Treated Early-Onset Gastric Adenocarcinoma—A Retrospective Cohort Study. J. Cancer 2023, 14, 1470–1478. [Google Scholar] [CrossRef]
  58. Katz, M.H.; Pisters, P.W.; Evans, D.B.; Sun, C.C.; Lee, J.E.; Fleming, J.B.; Vauthey, J.N.; Abdalla, E.K.; Crane, C.H.; Wolff, R.A.; et al. Borderline resectable pancreatic cancer: The importance of this emerging stage of disease. J. Am. Coll. Surg. 2008, 206, 833–846; discussion 846–838. [Google Scholar] [CrossRef]
  59. Morales-Oyarvide, V.; Rubinson, D.A.; Dunne, R.F.; Kozak, M.M.; Bui, J.L.; Yuan, C.; Qian, Z.R.; Babic, A.; Da Silva, A.; Nowak, J.A.; et al. Lymph node metastases in resected pancreatic ductal adenocarcinoma: Predictors of disease recurrence and survival. Br. J. Cancer 2017, 117, 1874–1882. [Google Scholar] [CrossRef]
  60. Ivey, G.D.; Shoucair, S.; Delitto, D.J.; Habib, J.R.; Kinny-Köster, B.; Shubert, C.R.; Lafaro, K.J.; Cameron, J.L.; Burns, W.R.; Burkhart, R.A.; et al. Postoperative Chemotherapy is Associated with Improved Survival in Patients with Node-Positive Pancreatic Ductal Adenocarcinoma after Neoadjuvant Therapy. World J. Surg. 2022, 46, 2751–2759. [Google Scholar] [CrossRef]
  61. van Roessel, S.; van Veldhuisen, E.; Klompmaker, S.; Janssen, Q.P.; Abu Hilal, M.; Alseidi, A.; Balduzzi, A.; Balzano, G.; Bassi, C.; Berrevoet, F.; et al. Evaluation of Adjuvant Chemotherapy in Patients With Resected Pancreatic Cancer After Neoadjuvant FOLFIRINOX Treatment. JAMA Oncol. 2020, 6, 1733–1740. [Google Scholar] [CrossRef] [PubMed]
  62. Kinny-Köster, B.; Habib, J.R.; Wolfgang, C.L.; He, J.; Javed, A.A. Favorable tumor biology in locally advanced pancreatic cancer—Beyond CA19-9. J. Gastrointest. Oncol. 2021, 12, 2484–2494. [Google Scholar] [CrossRef] [PubMed]
  63. Kato, Y.; Yamada, S.; Tashiro, M.; Sonohara, F.; Takami, H.; Hayashi, M.; Kanda, M.; Kobayashi, D.; Tanaka, C.; Nakayama, G.; et al. Biological and conditional factors should be included when defining criteria for resectability for patients with pancreatic cancer. HPB 2019, 21, 1211–1218. [Google Scholar] [CrossRef] [PubMed]
  64. Doppenberg, D.; van Dam, J.L.; Han, Y.; Bonsing, B.A.; Busch, O.R.; Festen, S.; van der Harst, E.; de Hingh, I.H.; Homs, M.Y.V.; Kwon, W.; et al. Predictive value of baseline serum carbohydrate antigen 19-9 level on treatment effect of neoadjuvant chemoradiotherapy in patients with resectable and borderline resectable pancreatic cancer in two randomized trials. Br. J. Surg. 2023, 110, 1374–1380. [Google Scholar] [CrossRef]
  65. Schorn, S.; Demir, I.E.; Samm, N.; Scheufele, F.; Calavrezos, L.; Sargut, M.; Schirren, R.M.; Friess, H.; Ceyhan, G.O. Meta-analysis of the impact of neoadjuvant therapy on patterns of recurrence in pancreatic ductal adenocarcinoma. BJS Open 2018, 2, 52–61. [Google Scholar] [CrossRef] [PubMed]
  66. Hartlapp, I.; Valta-Seufzer, D.; Siveke, J.T.; Algül, H.; Goekkurt, E.; Siegler, G.; Martens, U.M.; Waldschmidt, D.; Pelzer, U.; Fuchs, M.; et al. Prognostic and predictive value of CA 19-9 in locally advanced pancreatic cancer treated with multiagent induction chemotherapy: Results from a prospective, multicenter phase II trial (NEOLAP-AIO-PAK-0113). ESMO Open 2022, 7, 100552. [Google Scholar] [CrossRef]
  67. Heger, U.; Sun, H.; Hinz, U.; Klaiber, U.; Tanaka, M.; Liu, B.; Sachsenmaier, M.; Springfeld, C.; Michalski, C.W.; Büchler, M.W.; et al. Induction chemotherapy in pancreatic cancer: CA 19-9 may predict resectability and survival. HPB 2020, 22, 224–232. [Google Scholar] [CrossRef]
  68. Servin-Rojas, M.; Fong, Z.V.; Fernandez-Del Castillo, C.; Ferrone, C.R.; Lee, H.; Lopez-Verdugo, F.; Qiao, G.; Rocha-Castellanos, D.M.; Lillemoe, K.D.; Qadan, M. Tumor Size Reduction and Serum Carbohydrate Antigen 19-9 Kinetics After Neoadjuvant FOLFIRINOX in Patients with Pancreatic Ductal Adenocarcinoma. Surgery 2024, 175, 471–476. [Google Scholar] [CrossRef]
  69. van Veldhuisen, E.; Vogel, J.A.; Klompmaker, S.; Busch, O.R.; van Laarhoven, H.W.M.; van Lienden, K.P.; Wilmink, J.W.; Marsman, H.A.; Besselink, M.G. Added value of CA19-9 response in predicting resectability of locally advanced pancreatic cancer following induction chemotherapy. HPB 2018, 20, 605–611. [Google Scholar] [CrossRef]
  70. Alva-Ruiz, R.; Yohanathan, L.; Yonkus, J.A.; Abdelrahman, A.M.; Gregory, L.A.; Halfdanarson, T.R.; Mahipal, A.; McWilliams, R.R.; Ma, W.W.; Hallemeier, C.L.; et al. Neoadjuvant Chemotherapy Switch in Borderline Resectable/Locally Advanced Pancreatic Cancer. Ann. Surg. Oncol. 2022, 29, 1579–1591. [Google Scholar] [CrossRef]
  71. van Manen, L.; Groen, J.V.; Putter, H.; Vahrmeijer, A.L.; Swijnenburg, R.-J.; Bonsing, B.A.; Mieog, J.S.D. Elevated CEA and CA19-9 serum levels independently predict advanced pancreatic cancer at diagnosis. Biomarkers 2020, 25, 186–193. [Google Scholar] [CrossRef] [PubMed]
  72. Schlieman, M.G.; Ho, H.S.; Bold, R.J. Utility of Tumor Markers in Determining Resectability of Pancreatic Cancer. Arch. Surg. 2003, 138, 951–956. [Google Scholar] [CrossRef] [PubMed]
  73. Distler, M.; Pilarsky, E.; Kersting, S.; Grützmann, R. Preoperative CEA and CA 19-9 are prognostic markers for survival after curative resection for ductal adenocarcinoma of the pancreas—A retrospective tumor marker prognostic study. Int. J. Surg. 2013, 11, 1067–1072. [Google Scholar] [CrossRef] [PubMed]
  74. Duraker, N.; Hot, S.; Polat, Y.; Höbek, A.; Gençler, N.; Urhan, N. CEA, CA 19-9, and CA 125 in the differential diagnosis of benign and malignant pancreatic diseases with or without jaundice. J. Surg. Oncol. 2007, 95, 142–147. [Google Scholar] [CrossRef] [PubMed]
  75. Kane, L.E.; Mellotte, G.S.; Mylod, E.; O’Brien, R.M.; O’Connell, F.; Buckley, C.E.; Arlow, J.; Nguyen, K.; Mockler, D.; Meade, A.D.; et al. Diagnostic Accuracy of Blood-based Biomarkers for Pancreatic Cancer: A Systematic Review and Meta-analysis. Cancer Res. Commun. 2022, 2, 1229–1243. [Google Scholar] [CrossRef] [PubMed]
  76. Omiya, K.; Oba, A.; Inoue, Y.; Kobayashi, K.; Wu, Y.H.A.; Ono, Y.; Sato, T.; Sasaki, T.; Ozaka, M.; Sasahira, N.; et al. Serum DUPAN-2 could be an Alternative Biological Marker for CA19-9 Non-secretors with Pancreatic Cancer. Ann. Surg. 2022, 277, e1278–e1283. [Google Scholar] [CrossRef] [PubMed]
  77. Kawa, S.; Oguchi, H.; Kobayashi, T.; Tokoo, M.; Furuta, S.; Kanai, M.; Homma, T. Elevated serum levels of Dupan-2 in pancreatic cancer patients negative for Lewis blood group phenotype. Br. J. Cancer 1991, 64, 899–902. [Google Scholar] [CrossRef] [PubMed]
  78. Luo, G.; Liu, C.; Guo, M.; Cheng, H.; Lu, Y.; Jin, K.; Liu, L.; Long, J.; Xu, J.; Lu, R.; et al. Potential Biomarkers in Lewis Negative Patients with Pancreatic Cancer. Ann. Surg. 2017, 265, 800–805. [Google Scholar] [CrossRef]
  79. Doppenberg, D.; Stoop, T.F.; van Dieren, S.; Katz, M.H.G.; Janssen, Q.P.; Nasar, N.; Prakash, L.R.; Theijse, R.T.; Tzeng, C.D.; Wei, A.C.; et al. Serum CEA as a Prognostic Marker for Overall Survival in Patients with Localized Pancreatic Adenocarcinoma and Non-Elevated CA19-9 Levels Treated with FOLFIRINOX as Initial Treatment: A TAPS Consortium Study. Ann. Surg. Oncol. 2024, 1–14. [Google Scholar] [CrossRef]
  80. Cheng, H.; Long, F.; Jaiswar, M.; Yang, L.; Wang, C.; Zhou, Z. Prognostic role of the neutrophil-to-lymphocyte ratio in pancreatic cancer: A meta-analysis. Sci. Rep. 2015, 5, 11026. [Google Scholar] [CrossRef]
  81. Shrotriya, S.; Walsh, D.; Bennani-Baiti, N.; Thomas, S.; Lorton, C. C-Reactive Protein Is an Important Biomarker for Prognosis Tumor Recurrence and Treatment Response in Adult Solid Tumors: A Systematic Review. PLoS ONE 2015, 10, e0143080. [Google Scholar] [CrossRef] [PubMed]
  82. Recio-Boiles, A.; Nallagangula, A.; Veeravelli, S.; Vondrak, J.; Saboda, K.; Roe, D.; Elquza, E.; McBride, A.; Babiker, H.M. Neutrophil-to-lymphocyte and platelet-to-lymphocyte ratios inversely correlate to clinical and pathologic stage in patients with resectable pancreatic ductal adenocarcinoma. Ann. Pancreat. Cancer 2019, 2, 8. [Google Scholar] [CrossRef]
  83. Mowbray, N.G.; Griffith, D.; Hammoda, M.; Shingler, G.; Kambal, A.; Al-Sarireh, B. A meta-analysis of the utility of the neutrophil-to-lymphocyte ratio in predicting survival after pancreatic cancer resection. HPB 2018, 20, 379–384. [Google Scholar] [CrossRef] [PubMed]
  84. Cerami, E.; Gao, J.; Dogrusoz, U.; Gross, B.E.; Sumer, S.O.; Aksoy, B.A.; Jacobsen, A.; Byrne, C.J.; Heuer, M.L.; Larsson, E.; et al. The cBio cancer genomics portal: An open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012, 2, 401–404. [Google Scholar] [CrossRef]
  85. Yao, W.; Maitra, A.; Ying, H. Recent insights into the biology of pancreatic cancer. EBioMedicine 2020, 53, 102655. [Google Scholar] [CrossRef] [PubMed]
  86. Helal, C.; Valery, M.; Ducreux, M.; Hollebecque, A.; Smolenschi, C. FGFR2 fusion in metastatic pancreatic ductal adenocarcinoma: Is there hope? Eur. J. Cancer 2022, 176, 168–170. [Google Scholar] [CrossRef] [PubMed]
  87. Ecker, B.L.; Court, C.M.; Janssen, Q.P.; Tao, A.J.; D’Angelica, M.I.; Drebin, J.A.; Gonen, M.; O’Reilly, E.M.; Jarnagin, W.R.; Wei, A.C. Alterations in Somatic Driver Genes Are Associated with Response to Neoadjuvant FOLFIRINOX in Patients with Localized Pancreatic Ductal Adenocarcinoma. J. Am. Coll. Surg. 2022, 235, 342–349. [Google Scholar] [CrossRef] [PubMed]
  88. Bailey, P.; Chang, D.K.; Nones, K.; Johns, A.L.; Patch, A.M.; Gingras, M.C.; Miller, D.K.; Christ, A.N.; Bruxner, T.J.; Quinn, M.C.; et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature 2016, 531, 47–52. [Google Scholar] [CrossRef]
  89. Collisson, E.A.; Sadanandam, A.; Olson, P.; Gibb, W.J.; Truitt, M.; Gu, S.; Cooc, J.; Weinkle, J.; Kim, G.E.; Jakkula, L.; et al. Subtypes of pancreatic ductal adenocarcinoma and their differing responses to therapy. Nat. Med. 2011, 17, 500–503. [Google Scholar] [CrossRef]
  90. 90. Pranzini, E.; Raugei, G.; Taddei, M.L. Metabolic Features of Tumor Dormancy: Possible Therapeutic Strategies. Cancers 2022, 14, 547. [Google Scholar] [CrossRef]
  91. Dreyer, S.B.; Rae, S.; Bisset, K.; Upstill-Goddard, R.; Gemenetzis, G.; Johns, A.L.; Dickson, E.J.; Mittal, A.; Gill, A.J.; Duthie, F.; et al. The Impact of Molecular Subtyping on Pathological Staging of Pancreatic Cancer. Ann. Surg. 2023, 277, e396–e405. [Google Scholar] [CrossRef] [PubMed]
  92. Patel, H.; Okamura, R.; Fanta, P.; Patel, C.; Lanman, R.B.; Raymond, V.M.; Kato, S.; Kurzrock, R. Clinical correlates of blood-derived circulating tumor DNA in pancreatic cancer. J. Hematol. Oncol. 2019, 12, 130. [Google Scholar] [CrossRef] [PubMed]
  93. Zhu, Y.; Zhang, H.; Chen, N.; Hao, J.; Jin, H.; Ma, X. Diagnostic value of various liquid biopsy methods for pancreatic cancer: A systematic review and meta-analysis. Medicine 2020, 99, e18581. [Google Scholar] [CrossRef] [PubMed]
  94. Grunvald, M.W.; Jacobson, R.A.; Kuzel, T.M.; Pappas, S.G.; Masood, A. Current Status of Circulating Tumor DNA Liquid Biopsy in Pancreatic Cancer. Int. J. Mol. Sci. 2020, 21, 7651. [Google Scholar] [CrossRef]
  95. Dawson, S.J.; Tsui, D.W.; Murtaza, M.; Biggs, H.; Rueda, O.M.; Chin, S.F.; Dunning, M.J.; Gale, D.; Forshew, T.; Mahler-Araujo, B.; et al. Analysis of circulating tumor DNA to monitor metastatic breast cancer. N. Engl. J. Med. 2013, 368, 1199–1209. [Google Scholar] [CrossRef] [PubMed]
  96. Vidal, J.; Muinelo, L.; Dalmases, A.; Jones, F.; Edelstein, D.; Iglesias, M.; Orrillo, M.; Abalo, A.; Rodríguez, C.; Brozos, E.; et al. Plasma ctDNA RAS mutation analysis for the diagnosis and treatment monitoring of metastatic colorectal cancer patients. Ann. Oncol. 2017, 28, 1325–1332. [Google Scholar] [CrossRef]
  97. Schraa, S.J.; van Rooijen, K.L.; Koopman, M.; Vink, G.R.; Fijneman, R.J.A. Cell-Free Circulating (Tumor) DNA before Surgery as a Prognostic Factor in Non-Metastatic Colorectal Cancer: A Systematic Review. Cancers 2022, 14, 2218. [Google Scholar] [CrossRef]
  98. Lapin, M.; Edland, K.H.; Tjensvoll, K.; Oltedal, S.; Austdal, M.; Garresori, H.; Rozenholc, Y.; Gilje, B.; Nordgård, O. Comprehensive ctDNA Measurements Improve Prediction of Clinical Outcomes and Enable Dynamic Tracking of Disease Progression in Advanced Pancreatic Cancer. Clin. Cancer Res. 2023, 29, 1267–1278. [Google Scholar] [CrossRef]
  99. 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]
  100. McDuff, S.G.R.; Hardiman, K.M.; Ulintz, P.J.; Parikh, A.R.; Zheng, H.; Kim, D.W.; Lennerz, J.K.; Hazar-Rethinam, M.; Van Seventer, E.E.; Fetter, I.J.; et al. Circulating Tumor DNA Predicts Pathologic and Clinical Outcomes Following Neoadjuvant Chemoradiation and Surgery for Patients With Locally Advanced Rectal Cancer. JCO Precis. Oncol. 2021, 5, 123–132. [Google Scholar] [CrossRef]
  101. 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] [PubMed]
  102. Rhim, A.D.; Mirek, E.T.; Aiello, N.M.; Maitra, A.; Bailey, J.M.; McAllister, F.; Reichert, M.; Beatty, G.L.; Rustgi, A.K.; Vonderheide, R.H.; et al. EMT and Dissemination Precede Pancreatic Tumor Formation. Cell 2012, 148, 349–361. [Google Scholar] [CrossRef] [PubMed]
  103. Klein, C.A. Cancer. The metastasis cascade. Science 2008, 321, 1785–1787. [Google Scholar] [CrossRef] [PubMed]
  104. Deng, Z.; Wu, S.; Wang, Y.; Shi, D. Circulating tumor cell isolation for cancer diagnosis and prognosis. EBioMedicine 2022, 83, 104237. [Google Scholar] [CrossRef] [PubMed]
  105. Gemenetzis, G.; Groot, V.P.; Yu, J.; Ding, D.; Teinor, J.A.; Javed, A.A.; Wood, L.D.; Burkhart, R.A.; Cameron, J.L.; Makary, M.A.; et al. Circulating Tumor Cells Dynamics in Pancreatic Adenocarcinoma Correlate with Disease Status: Results of the Prospective CLUSTER Study. Ann. Surg. 2018, 268, 408–420. [Google Scholar] [CrossRef] [PubMed]
  106. Javed, A.A.; Ding, D.; Hasanain, A.; van Oosten, F.; Yu, J.; Cameron, J.L.; Burkhart, R.A.; Zheng, L.; He, J.; Wolfgang, C.L. Persistent Circulating Tumor Cells at One Year after Oncologic Resection Predict Late Recurrence in Pancreatic Cancer. Ann. Surg. 2022, 277, 859–865. [Google Scholar] [CrossRef]
  107. Schwartz, L.H.; Litière, S.; de Vries, E.; Ford, R.; Gwyther, S.; Mandrekar, S.; Shankar, L.; Bogaerts, J.; Chen, A.; Dancey, J.; et al. RECIST 1.1-Update and clarification: From the RECIST committee. Eur. J. Cancer 2016, 62, 132–137. [Google Scholar] [CrossRef]
  108. Perri, G.; Prakash, L.; Qiao, W.; Varadhachary, G.R.; Wolff, R.; Fogelman, D.; Overman, M.; Pant, S.; Javle, M.; Koay, E.J.; et al. Response and Survival Associated with First-line FOLFIRINOX vs Gemcitabine and nab-Paclitaxel Chemotherapy for Localized Pancreatic Ductal Adenocarcinoma. JAMA Surg. 2020, 155, 832–839. [Google Scholar] [CrossRef]
  109. Ferrone, C.R.; Marchegiani, G.; Hong, T.S.; Ryan, D.P.; Deshpande, V.; McDonnell, E.I.; Sabbatino, F.; Santos, D.D.; Allen, J.N.; Blaszkowsky, L.S.; et al. Radiological and surgical implications of neoadjuvant treatment with FOLFIRINOX for locally advanced and borderline resectable pancreatic cancer. Ann. Surg. 2015, 261, 12–17. [Google Scholar] [CrossRef]
  110. Welsh, J.L.; Bodeker, K.; Fallon, E.; Bhatia, S.K.; Buatti, J.M.; Cullen, J.J. Comparison of response evaluation criteria in solid tumors with volumetric measurements for estimation of tumor burden in pancreatic adenocarcinoma and hepatocellular carcinoma. Am. J. Surg. 2012, 204, 580–585. [Google Scholar] [CrossRef]
  111. Michelakos, T.; Pergolini, I.; Castillo, C.F.; Honselmann, K.C.; Cai, L.; Deshpande, V.; Wo, J.Y.; Ryan, D.P.; Allen, J.N.; Blaszkowsky, L.S.; et al. Predictors of Resectability and Survival in Patients With Borderline and Locally Advanced Pancreatic Cancer who Underwent Neoadjuvant Treatment with FOLFIRINOX. Ann. Surg. 2019, 269, 733–740. [Google Scholar] [CrossRef] [PubMed]
  112. Castellanos, J.A.; Merchant, N.B. Intensity of follow-up after pancreatic cancer resection. Ann. Surg. Oncol. 2014, 21, 747–751. [Google Scholar] [CrossRef] [PubMed]
  113. Meijer, L.L.; Garajová, I.; Caparello, C.; Le Large, T.Y.S.; Frampton, A.E.; Vasile, E.; Funel, N.; Kazemier, G.; Giovannetti, E. Plasma miR-181a-5p Downregulation Predicts Response and Improved Survival After FOLFIRINOX in Pancreatic Ductal Adenocarcinoma. Ann. Surg. 2020, 271, 1137–1147. [Google Scholar] [CrossRef] [PubMed]
  114. Golan, T.; Barenboim, A.; Lahat, G.; Nachmany, I.; Goykhman, Y.; Shacham-Shmueli, E.; Halpern, N.; Brazowski, E.; Geva, R.; Wolf, I.; et al. Increased Rate of Complete Pathologic Response After Neoadjuvant FOLFIRINOX for BRCA Mutation Carriers with Borderline Resectable Pancreatic Cancer. Ann. Surg. Oncol. 2020, 27, 3963–3970. [Google Scholar] [CrossRef]
Figure 1. Determination of anatomical and biological resectability. Considerations for anatomical and biological resectability of disease. Assessment of disease is a dynamic process that involves the evaluation of anatomical and biological characteristics for each patient. For anatomical resectability, the disease can vary from resectable to oligometastatic disease, while biological resectability can be determined using biomarkers. Currently CA19-9 is used clinically, and in the future, additional biomarkers such as ctDNA, CTCs, and circulating proteins can be incorporated into this approach. At diagnosis, a patient can be categorized into one of the four quadrants on the figure, and management decisions be made accordingly. However, tumor biology is dynamic, and a serial assessment of tumor biology can lead to changes in the therapeutic approach as indicated by the arrows. For example, a patient with locally advanced pancreatic cancer and favorable tumor biology at the time of diagnosis (left upper quadrant) could progress to biologically unresectable disease during induction chemotherapy (right upper quadrant) and should, therefore, not be an appropriate surgical candidate. On the other hand, if the same patient were to remain biological resectable after induction therapy, surgical resection could be warranted even with anatomically challenging disease. Contrastingly, a patient with anatomically resectable disease that is biologically unresectable at diagnosis (right lower quadrant) could be offered neoadjuvant therapy as opposed to upfront surgery.
Figure 1. Determination of anatomical and biological resectability. Considerations for anatomical and biological resectability of disease. Assessment of disease is a dynamic process that involves the evaluation of anatomical and biological characteristics for each patient. For anatomical resectability, the disease can vary from resectable to oligometastatic disease, while biological resectability can be determined using biomarkers. Currently CA19-9 is used clinically, and in the future, additional biomarkers such as ctDNA, CTCs, and circulating proteins can be incorporated into this approach. At diagnosis, a patient can be categorized into one of the four quadrants on the figure, and management decisions be made accordingly. However, tumor biology is dynamic, and a serial assessment of tumor biology can lead to changes in the therapeutic approach as indicated by the arrows. For example, a patient with locally advanced pancreatic cancer and favorable tumor biology at the time of diagnosis (left upper quadrant) could progress to biologically unresectable disease during induction chemotherapy (right upper quadrant) and should, therefore, not be an appropriate surgical candidate. On the other hand, if the same patient were to remain biological resectable after induction therapy, surgical resection could be warranted even with anatomically challenging disease. Contrastingly, a patient with anatomically resectable disease that is biologically unresectable at diagnosis (right lower quadrant) could be offered neoadjuvant therapy as opposed to upfront surgery.
Cancers 16 00489 g001
Figure 2. Serial Assessment of Tumor Biology: Serial assessment of anatomical and biological resectability can aid in the appropriate treatment for pancreatic cancer patients. (1) Determining the need for neoadjuvant therapy (anatomically resectable) and the mode of neoadjuvant/induction therapy (all anatomic stages). (2) Serial assessment of tumor response to determine surgical candidacy and the timing of resection. Local progression of disease can indicate the need for a change of regimen while systemic progression indicates the need for non-surgical management with investigational therapies or best supportive care. (3) Determining the need and mode of adjuvant treatment. Figure designed with Bio Render.
Figure 2. Serial Assessment of Tumor Biology: Serial assessment of anatomical and biological resectability can aid in the appropriate treatment for pancreatic cancer patients. (1) Determining the need for neoadjuvant therapy (anatomically resectable) and the mode of neoadjuvant/induction therapy (all anatomic stages). (2) Serial assessment of tumor response to determine surgical candidacy and the timing of resection. Local progression of disease can indicate the need for a change of regimen while systemic progression indicates the need for non-surgical management with investigational therapies or best supportive care. (3) Determining the need and mode of adjuvant treatment. Figure designed with Bio Render.
Cancers 16 00489 g002
Table 1. Different definitions of borderline resectable pancreatic cancer.
Table 1. Different definitions of borderline resectable pancreatic cancer.
Borderline ResectableNCCNAHBPAAllianceIAPMD Anderson
Superior mesenteric Vein/Portal VeinReconstructable involvement (distortion, narrowing, occlusion, thrombosis) Reconstructable abutment, encasement or occlusionSolid contact >180° or reconstructable occlusionSMV/PV: tumor contact 180° or greater or bilateral narrowing/occlusion, not exceeding the inferior border of the duodenumShort-segment occlusion with suitable vessel for reconstruction
Superior mesenteric arterySolid tumor contact ≤180°AbutmentInterface between tumor and vessel measuring < 180°Tumor contact of less than 180° without showing deformity/stenosisAbutment ≤ 180°
Common hepatic arterySolid tumor contact without extension to the coeliac artery or hepatic artery bifurcationAbutment or short-segment encasementReconstructable, short-segment interface between tumor and vessel of any degreeTumor contact without showing tumor contact of the PHA and/or CAShort-segment encasement/abutment
Celiac trunkSolid tumor contact <180°No abutment or encasementInterface between tumor and vessel measuring <180°Tumor contact of less than 180° without showing deformity/stenosisAbutment ≤ 180°
Biological- Suspicion for distant metastasis, including CA19-9 > 500 U/mL, or regional lymph node metastasis diagnosed by biopsy or PET-CTCT findings suspicious of metastatic disease; nodal-positive disease.
Conditional- ECOG performance status of 2 or more Performance status ≥ 3 or severe preexisting medical comorbidity
Legend: Anatomic resectability as defined by tumor contact to abdominal vessels by the National Comprehensive Cancer Network (NCCN), Americas Hepato-Pancreato-Biliary Association (AHBPA), Alliance, International Association of Pancreatology (IAP), and MD Anderson Cancer Center. Abbreviations: SMV, superior mesenteric vein; PV, portal vein; PHA, proper hepatic artery; CA, common hepatic artery; ECOG, Eastern Cooperative Oncology Group.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Rompen, I.F.; Habib, J.R.; Wolfgang, C.L.; Javed, A.A. Anatomical and Biological Considerations to Determine Resectability in Pancreatic Cancer. Cancers 2024, 16, 489. https://doi.org/10.3390/cancers16030489

AMA Style

Rompen IF, Habib JR, Wolfgang CL, Javed AA. Anatomical and Biological Considerations to Determine Resectability in Pancreatic Cancer. Cancers. 2024; 16(3):489. https://doi.org/10.3390/cancers16030489

Chicago/Turabian Style

Rompen, Ingmar F., Joseph R. Habib, Christopher L. Wolfgang, and Ammar A. Javed. 2024. "Anatomical and Biological Considerations to Determine Resectability in Pancreatic Cancer" Cancers 16, no. 3: 489. https://doi.org/10.3390/cancers16030489

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