Editorial Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Surg. May 27, 2024; 16(5): 1223-1230
Published online May 27, 2024. doi: 10.4240/wjgs.v16.i5.1223
Neoadjuvant treatment of pancreatic ductal adenocarcinoma: Whom, when and how
Nebojsa Manojlovic, Clinic for Gastroenterology and Hepatology, Military Medical Academy, Faculty of Medicine of the Military Medical Academy, University of Defence, Belgrade 11000, Serbia
Goran Savic, Military Medical Academy, Faculty of Medicine of the Military Medical Academy, University of Defence, Belgrade 11000, Serbia
Stevan Manojlovic, Faculty of Medicine, University of Belgrade, Belgrade 11000, Serbia
ORCID number: Nebojsa Manojlovic (0000-0002-3639-1328); Goran Savic (0000-0002-3810-2323); Stevan Manojlovic (0009-0001-2385-7096).
Author contributions: Manojlovic N, Savic G, and Manojlovic S contributed to this paper; Manojlovic N designed the overall concept and outline of the manuscript, and drafted and edited the manuscript; Savic G and Manojlovic S contributed to the discussion and design of the manuscript, and made critical revision of the manuscript and contributed to the review of literature.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Nebojsa Manojlovic, PhD, Associate Professor, Clinic for Gastroenterology and Hepatology, Military Medical Academy, Faculty of Medicine of the Military Medical Academy, University of Defence, Crnotravska 17, Belgrade 11000, Serbia. nebojsa.manojlovic1@gmail.com
Received: December 28, 2023
Revised: March 13, 2024
Accepted: April 22, 2024
Published online: May 27, 2024

Abstract

Pancreatic ductal adenocarcinoma (PDAC), which is notorious for its aggressiveness and poor prognosis, remains an area of great unmet medical need, with a 5-year survival rate of 10% - the lowest of all solid tumours. At diagnosis, only 20% of patients have resectable pancreatic cancer (RPC) or borderline RPC (BRPC) disease, while 80% of patients have unresectable tumours that are locally advanced pancreatic cancer (LAPC) or have distant metastases. Nearly 60% of patients who undergo upfront surgery for RPC are unable to receive adequate adjuvant chemotherapy (CHT) because of postoperative complications and early cancer recurrence. An important paradigm shift to achieve better outcomes has been the sequence of therapy, with neoadjuvant CHT preceding surgery. Three surgical stages have emerged for the preoperative assessment of nonmetastatic pancreatic cancers: RPC, BRPC, and LAPC. The main goal of neoadjuvant treatment (NAT) is to improve postoperative outcomes through enhanced selection of candidates for curative-intent surgery by identifying patients with aggressive or metastatic disease during initial CHT, reducing tumour volume before surgery to improve the rate of margin-negative resection (R0 resection, a microscopic margin-negative resection), reducing the rate of positive lymph node occurrence at surgery, providing early treatment of occult micrometastatic disease, and assessing tumour chemosensitivity and tolerance to treatment as potential surgical criteria. In this editorial, we summarize evidence concerning NAT of PDAC, providing insights into future practice and study design. Future research is needed to establish predictive biomarkers, measures of therapeutic response, and multidisciplinary strategies to improve patient-centered outcomes.

Key Words: Pancreatic adenocarcinoma, Neoadjuvant treatment, Chemotherapy, Upfront surgery, Radiotherapy, Response evaluation

Core Tip: Pancreatic ductal adenocarcinoma, which is notorious for its aggressiveness and poor prognosis, remains an area of great unmet medical need. The most important determinant of survival is surgical resection. Nearly 60% of patients who undergo upfront surgery for resectable tumours are unable to receive adequate adjuvant chemotherapy (CHT). An important paradigm shift to achieve better outcomes has been the sequence of therapy, with neoadjuvant CHT preceding surgery. Three surgical stages have emerged for the preoperative assessment of nonmetastatic pancreatic cancers: Resectable, borderline-resectable and unresectable locally advanced tumours. In the development of new neoadjuvant and induction strategies, the distinct molecular and biological characteristics of the various pancreatic cancer subgroups need to be integrated to optimize the selection and sequencing of both established and novel treatment modalities that may improve survival outcomes.



INTRODUCTION

Pancreatic ductal adenocarcinoma (PDAC), which is notorious for its aggressiveness and poor prognosis, remains an area of great unmet medical need, with a 5-year survival rate of 10% - the lowest of all solid tumours. Surgical resection is the most important predictor of survival (due to lower morbidity and death rates) along with postoperative adjuvant treatment. The sole potentially curative treatment is surgical resection; nevertheless, the 5- and 10-year overall survival (OS) rates for surgery by itself are quite low, at 10.4% and 7.7%, respectively. The actual rates are even lower, as these numbers represent the “best of the best” in clinical trial patients; real-world results are much worse[1].

Merely 20% of patients are diagnosed with resectable pancreatic cancer (RPC) or borderline RPC (BRPC) illness, whereas the remaining 80% have tumors that are unresectable, have distant metastases, or are locally advanced pancreatic cancer (LAPC)[2]. Single-agent adjuvant chemotherapy (CHT) increases the 5-year OS rate for upfront, locally resectable PDAC (about 20% of all patients) from 8% for surgery alone to 16%-21% with adjuvant therapy[3-6]. Gemcitabine plus capecitabine in unselected patients, including those older than 80 years, provided a median OS (mOS) of 28 months. Modified folinic acid, fluorouracil, irinotecan, and oxaliplatin (mFOLFIRINOX) in selected patients younger than 79 years of age increased 3-year OS to 63.4% and mOS to 54.4 months[7,8].

Nearly 60% of patients who undergo upfront surgery for RPC are unable to receive adequate adjuvant CHT due to postoperative complications and early cancer recurrence[9]. Just 7% of Medicare patients, for instance, were able to receive all prescribed adjuvant CHT, according to a recent study. Although it is not common to finish because of the previously described reasons, adjuvant CHT for pancreatic cancer patients who have received upfront surgical resection is associated with a greater chance of survival. Patients who undergo appropriate surgical resection for pancreatic cancer often experience cancer recurrence and are not cured, even with aggressive adjuvant management[10].

STAGING CRITERIA

Radiographically localized pancreatic adenocarcinoma is staged based on anatomical tumour-vessel interfaces into RPC, BRPC, and LAPC. Three fundamental ideas form the basis of these classifications: (1) Increasing vascular involvement may raise the risk of harboring radiographically occult metastatic disease; (2) Margin negative resection is more frequently linked to long-term survival; and (3) Margin positivity increases with increasing degree of vascular involvement[11]. The complexity and variability of classification based on these criteria is demonstrated by the fact that, even in high-volume institutions, approximately 30% of patients were misclassified anatomically in a prospective multicenter clinical trial[12]. Three subsets were developed by the MD Anderson Cancer Center Pancreatic Cancer Group (BR-A, BR-B, and BR-C) to evaluate patients with BR-PDAC and determine resectability using parameters other than anatomic criteria. The anatomic criteria for BR disease are met by BR-A disease, and it lacks any additional characteristics that would prevent surgical resection. The biological characteristics of extra-pancreatic disease, such as biopsy-proven lymph node metastases, elevated carbohydrate antigen 19-9 (CA 19-9) levels (1000 U/mL), and indeterminate liver lesions, are indicative of BR-B disease. Individuals diagnosed with BR-C illness form a conditional group that takes into account comorbidities such as advanced age (> 80 years) or an ECOG performance status of 2 or higher[13].

The systemic inflammatory response of cancer cells is involved in cancer progression and malignant transformation[14]. An elevated neutrophil-to-lymphocyte ratio is associated with poor OS and poor cancer-specific survival in patients with advanced-stage disease, and it is predictive of survival after resection of early-stage PDAC[15]. In PDAC patients receiving neoadjuvant treatment (NAT), the systemic inflammation response index (SIRI) can predict the survival of patients following resection and neoadjuvant chemotherapy (NACT), while the post-treatment systemic immune-inflammation index may be a useful prognostic marker[16]. Changes in the SIRI were associated with OS, but the preoperative SIRI was correlated with disease-free survival[17].

Treatment paradigms must be modified in light of advances in biomarkers, such as the discovery of circulating tumor DNA (ctDNA), based on staging criteria that take into consideration morphological restrictions, the biological predisposition for distant failure, and patient performance status[18]. Furthermore, circulating tumour cells can be isolated in the bloodstream of patients with early-stage pancreatic cancer, and their levels are correlated with the probability of survival[19].

SEQUENCE OF THERAPY

A significant paradigm change that has improved outcomes is the order in which therapies are administered, with NACT coming before surgery. Three surgical stages have emerged for the preoperative assessment of nonmetastatic pancreatic cancers: RPC, BRPC, and LAPC. Identifying patients with aggressive or metastatic disease during initial CHT, reducing tumour volume before surgery to improve the rate of margin-negative resection (R0 resection, a microscopic margin-negative resection), lowering the rate of positive lymph node occurrence at surgery, offering early treatment of occult micrometastatic disease, and evaluating tumour chemosensitivity and tolerance to treatment as potential surgical criteria are all part of the main objective of NAT, which aims to improve postoperative outcomes through an enhanced selection of candidates for curative-intent surgery. Selecting patients with a high chance of undergoing margin-negative resection based on response evaluation represents one of the most difficult scenarios with NAT[20]. The primary risk associated with neoadjuvant therapy is that about 30% of patients will develop metastatic disease while receiving treatment. Thus, a crucial unmet need is instruments to help physicians choose first-line CHT regimens that are more effective[21].

As such, it is critical to carefully weigh the various therapeutic options and customize them in light of clinically meaningful biomarkers. We can improve our capacity to choose patients who will most likely benefit from NACT by finding strong predictive markers. Additionally, the discovery of prognostic markers can help guide treatment choices and offer insightful information about the overall course of the disease.

CURRENT TREATMENT TRENDS IN NAT

Due to a lack of randomized phase III trials, NACT for RPC is not currently included in clinical guidelines. However, it may be an alternative to surgery first for patients with clearly resectable disease who only have suspicious imaging findings of metastasis, CA 19-9 levels suggestive of metastatic disease, or who require medical optimization before surgery. Some studies support the use of NAT for treating resectable tumours[22,23], and others have shown that NAT may be considered for patients with resectable tumours and high-risk features[24,25]. A retrospective study aimed at exploring patient subgroups with high-risk resectable PDAC for selecting candidates who may benefit from NAT reported that NAT is an effective treatment for patients with resectable PDAC, particularly when the tumour is in contact with the portal vein/superior mesenteric vein and has a CA 19-9 concentration ≥ 150 U/mL[26]. Propensity score matching analysis of retrospectively analyzed early-stage PDAC patients showed that in patients with early-stage resected pancreatic head adenocarcinoma, NAT followed by resection demonstrated a significant survival benefit when compared with upfront surgery, while the analysis, which represented an updated nationwide retrospective study, also supports multiagent NACT vs upfront surgery[27,28]. On the other hand, prospective randomized studies like NORPACT1 (neoadj mFOLFIRINOX 4 cycles and adj mFOLFIRINOX 8 cycles vs adj mFOLFIRINOX 12 cycles)[29], PANACHE01-PRODIGE48 (neoadj mFOLFIRINOX 4 cycles and adj chemo 8 cycles vs neoadj FOLFOX 4 cycles and adj chemo 8 cycles vs adj chemo 12 cycles)[30], and SWOG S1505 (neoadj mFOLFIRINOX 12 wk and adj mFOLFIRINOX 12 wk vs neoadj gem/nab-P 12 wk and adj gem/nab-P 12 wk) did not confirm the usefullness of NACT[31]. Additionally, there was no evidence supporting the use of NACT in the PREOPANC-2 study, which used mono-agent neoadjuvant chemoradiotherapy (NACRT)[32]. However, neoadjuvant therapy in these patients serves as a biological time-test and increases the delivery of multimodal therapy. Neoadjuvant therapy was not expected to be beneficial in patients with RPC, according to a meta-analysis of 111 cohort series neoadjuvant response rates and resection rates. However, roughly one-third of patients with nonresectable tumors could be anticipated to have a resectable tumor after neoadjuvant therapy, with a similar survival rate to those with primary resectable tumors[25]. Ongoing and future trials, such as Alliance A021806 (perioperative mFOLFIRINOX vs adjuvant mFOLFIRINOX)[33] and PREOPANC-3 (perioperative mFOLFIRINOX vs adjuvant FOLFIRINOX)[34], are intended to provide additional insight into the function of neoadjuvant therapy in patients with RPC.

A short course of neoadjuvant therapy, usually administered for two months before surgery, boosts the 12-month OS to roughly 77% in patients with borderline-resectable disease (with R0/R1 resection rates of 64%-85%), compared with 40% with upfront surgery (with a resection rate of 75%). An 18-month OS of 67% can result from a prolonged course of NACT, often lasting 4 months. Additionally, after 4-6 months of induction combination CHT with or without radiation therapy, 20% of patients with unresectable nonmetastatic PDAC may undergo resection, even in the absence of a discernible radiological response, improving their OS[35]. An optimal NAT regimen has not been agreed upon due to a lack of data from randomized clinical trials[36]. The two most efficient CHT regimens for PDAC are FOLFIRINOX and gemcitabine-nab-paclitaxel (GNP). For patients with pancreatic adenocarcinomas that were both resectable and borderline resectable, neoadjuvant FOLFIRINOX vs neoadjuvant gemcitabine-based CRT did not show any OS benefit, according to the first study’s phase III PREOPANC-2 trial data. Specifically, the mOS was 21.9% and 21.3% [hazard ratio (HR) = 0.87; 95% confidence interval (CI): 0.68-1.12; P = 0.28]. Between treatment arms, resection rates (77% vs 75%, respectively; P = 0.7) and serious adverse rates (49% vs 43%, respectively; P = 0.26) were likewise comparable[32]. The resection rate, R0 resection rate, and 2-year OS did not differ between the groups, according to the SWOG-1505 study, which assessed mFOLFIRINOX and GNP in the NAT setting of RPC. The toxicity profiles of the GNP and mFOLFIRINOX groups in this investigation did not significantly differ, which is noteworthy[31]. The NAPOLI 3 trial, which included liposomal irinotecan in the NALIRIFOX regimen, showed statistically significant and clinically meaningful improvements in OS and progression-free survival (PFS) compared with nab-paclitaxel and gemcitabine in patients with metastatic PDAC who had not previously received treatment in the metastatic setting. However, FOLFIRINOX is not superior in head-to-head comparisons[37]. According to the phase II nITRO trial, perioperative NALIRIFOX is safe and effective, and it should be further studied in randomized trials that contrast it with current CHT regimens for patients with BRPC and LAPC as well as standard surgery upfront followed by adjuvant therapy in patients with RPC[38].

Prospective randomized phase II and III trials exclusively enrolled patients with LAPC in the LAP-07 trial only, and other data supporting NAT in LAPC patients were derived from retrospective reports[39]. Apart from LAPC, which represents a true unresectable tumour, patients who suffer liver oligometastasis from PDAC may be candidates for surgery after NACT according to a meta-analysis based on small studies without randomized controlled trials and according to the opinions of one-third of the experts[40-42].

The role of radiation or chemoradiation during NAT for BRPC remains unclear. There is considerable debate on the best NAT regimen, and it is still uncertain whether is better for BRPC: NACT or NACRT[35]. CRT and maintenance CHT were examined in 449 patients in the LAP-07 study, who did not have progressing illness after CHT alone. After receiving either gemcitabine and erlotinib or gemcitabine alone for four months (the first randomization), 269 participants were randomly assigned to receive either CHT or CRT for an additional two months of treatment. Although CRT was linked to delayed locoregional development, the mOS in the CRT group did not improve[39]. When FOLFIRINOX was the primary induction therapy, the CONKO-007 trial yielded remarkably comparable outcomes. Without impacting OS or PFS, the addition of CRT raised the pathological complete response rate[43]. Some theoretical advantages of new radiotherapy techniques over conventional radiation include shorter treatment times, more focused treatment fields, higher biological effective doses, and the potential for better sparing of adjacent organs at risk. One such technique is stereotactic body radiotherapy, which targets primary tumors with minimal margins and high single doses in few fractions. The problem of delivering ablative doses while minimizing severe toxicity may potentially be solved by magnetic resonance-guided radiotherapy, or MRgRT[44].

RESPONSE EVALUATION

One of the greatest difficulties faced when dealing with NAT is identifying patients who are highly likely to undergo margin-negative resection based on response evaluation. It is widely recognized that conventional imaging has its limitations when it comes to restaging tumors[45]. PDAC is characterized by a significant presence of desmoplastic stroma, accounting for a substantial portion of the tumor volume, often estimated to be as high as 90%. This stroma is believed to primarily arise from cancer-associated fibroblasts[46]. The Response Evaluation Criteria in Solid Tumours have certain limitations when it comes to PDAC with a high presence of desmoplastic stroma. This is because commonly used imaging methods like computed tomography or magnetic resonance imaging cannot accurately distinguish between fibrosis caused by NACT and viable tumor tissue that remains. As a result, it becomes difficult to accurately determine the resectability of the tumor based solely on imaging studies conducted after neoadjuvant therapy. Numerous studies have also highlighted that the radiological appearance following neoadjuvant therapy does not necessarily reflect the patient’s actual response to the treatment[47].

Numerous institutions have made efforts to assess the biochemical response using CA 19-9, a widely recognized tumor marker in PDAC. Nevertheless, there has been a lack of consistency in the evaluation criteria employed by different institutions[48]. Moreover, the evaluation of treatment outcomes becomes complex when there are disparities between radiological and biochemical responses[49]. Yun et al[50] categorized patients into three groups based on their response to NACT and prognosis: Biochemical responders (BR+), BR-/radiology-only responders (RR+), and nonresponders (BR-/RR-). The research revealed that the 3-year survival rate differed significantly among the BR+ group (71.0%), the BR-/RR+ group (53.6%), and the BR-/RR- group (33.1%) (P < 0.001). Furthermore, the response to NACT was highlighted as a crucial factor in predicting recurrence risk, with HR of 2.15 (95%CI: 1.19-3.88; P = 0.011) for BR-/RR+ patients and 3.82 (95%CI: 2.41-6.08; P < 0.001) for BR-/RR- patients when compared to BR+ patients. Irrespective of the response to NACT, patients who underwent adjuvant CHT exhibited a significantly improved 3-year OS rate in comparison to those who did not[50]. Newhook et al[51] proposed innovative classification systems A (progressively decreasing to normal), B (fluctuating bidirectionally towards normal), C (consistently within normal range), D (decreasing but not normalizing), and E (increasing without normalization) based on the dynamics of CA 19-9. The study findings indicated that categories A and B, where CA 19-9 levels eventually normalize post NACT, are linked to favorable prognoses, while types D and E, where CA 19-9 levels remain elevated post NACT, are associated with poorer outcomes. Hence, apart from CA 19-9, investigations should be carried out to assess the efficacy of NACT by examining glucose metabolic activity or utilizing more sensitive biomarkers like ctDNA[52-54].

The efficacy of adjuvant CHT in patients who have undergone NACT remains a topic of debate. Drawing comparisons between the outcomes of adjuvant and neoadjuvant trials poses a challenge due to the fact that adjuvant trials typically involve only those patients with resected pancreatic cancer who are deemed suitable for adjuvant therapy without any signs of disease progression. On the other hand, neoadjuvant studies generally encompass all patients with RPC, as determined by diagnostic imaging, prior to the initiation of any treatment. Therefore, comparisons of the survival outcomes of patients in neoadjuvant and adjuvant trials are difficult if not impossible[55,56].

The development of cytotoxic treatments for various phases of pancreatic cancer has predominantly relied on empirical methods. A reductionist strategy aimed at creating therapies that target crucial genetic mutations has seen minimal achievements. Nevertheless, several elements of neoadjuvant therapy for PDAC, such as the most effective treatment plan, the incorporation of radiotherapy, and the identification of suitable candidates for neoadjuvant therapy, still require further clarification. Since other tumors with a much better prognosis have a greater number of average mutations, such as breast and colorectal cancers, and because the genetic spectra of colorectal, brain, and pancreatic tumors are similar, the average number of genetic alterations in PDAC tumors is insufficient to explain the abnormally poor prognosis and response to therapy[57,58].

There is accumulating research suggesting a relationship between pancreatic cancer subtype (classical vs basal-like) and treatment response[59]. Basal-like PDAC was resistant to FOLFIRINOX, paclitaxel, and tyrosine kinase inhibitors, whereas the classical transcriptional subtype was more susceptible to epidermal growth factor receptor suppression by erlotinib. These data significantly underscore the need for molecular subtyping in therapeutic decision-making in patients with PDAC[60].

CONCLUSION

The development of new neoadjuvant and induction strategies must take into account the distinct molecular and biological characteristics of the various pancreatic cancer subgroups, optimizing the selection and sequencing of both established and novel treatment modalities to improve survival rates. We can improve our capacity to choose patients who will benefit most from NACT by identifying strong prognostic markers. Moreover, prognostic markers can provide valuable insight into the overall disease course and illuminate treatment choices. Future studies are wanted to set up predictive biomarkers, measures of healing response, and multidisciplinary techniques to enhance patient-targeted outcomes.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: Serbia

Peer-review report’s classification

Scientific Quality: Grade B, Grade C, Grade C, Grade D

Novelty: Grade A, Grade B, Grade B, Grade C

Creativity or Innovation: Grade B, Grade B, Grade B, Grade C

Scientific Significance: Grade B, Grade B, Grade B, Grade C

P-Reviewer: Ding HG, China; Liu S, China; Maher H, China S-Editor: Wang JJ L-Editor: A P-Editor: Guo X

References
1.  Turner KM, Wilson GC, Patel SH, Ahmad SA. ASO Practice Guidelines Series: Management of Resectable, Borderline Resectable, and Locally Advanced Pancreas Cancer. Ann Surg Oncol. 2024;31:1884-1897.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
2.  Rawla P, Sunkara T, Gaduputi V. Epidemiology of Pancreatic Cancer: Global Trends, Etiology and Risk Factors. World J Oncol. 2019;10:10-27.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 893]  [Cited by in F6Publishing: 1262]  [Article Influence: 252.4]  [Reference Citation Analysis (0)]
3.  Neoptolemos JP, Dunn JA, Stocken DD, Almond J, Link K, Beger H, Bassi C, Falconi M, Pederzoli P, Dervenis C, Fernandez-Cruz L, Lacaine F, Pap A, Spooner D, Kerr DJ, Friess H, Büchler MW; European Study Group for Pancreatic Cancer. Adjuvant chemoradiotherapy and chemotherapy in resectable pancreatic cancer: a randomised controlled trial. Lancet. 2001;358:1576-1585.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 817]  [Cited by in F6Publishing: 724]  [Article Influence: 31.5]  [Reference Citation Analysis (0)]
4.  Neoptolemos JP, Stocken DD, Friess H, Bassi C, Dunn JA, Hickey H, Beger H, Fernandez-Cruz L, Dervenis C, Lacaine F, Falconi M, Pederzoli P, Pap A, Spooner D, Kerr DJ, Büchler MW; European Study Group for Pancreatic Cancer. A randomized trial of chemoradiotherapy and chemotherapy after resection of pancreatic cancer. N Engl J Med. 2004;350:1200-1210.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1945]  [Cited by in F6Publishing: 1828]  [Article Influence: 91.4]  [Reference Citation Analysis (0)]
5.  Neoptolemos JP, Stocken DD, Bassi C, Ghaneh P, Cunningham D, Goldstein D, Padbury R, Moore MJ, Gallinger S, Mariette C, Wente MN, Izbicki JR, Friess H, Lerch MM, Dervenis C, Oláh A, Butturini G, Doi R, Lind PA, Smith D, Valle JW, Palmer DH, Buckels JA, Thompson J, McKay CJ, Rawcliffe CL, Büchler MW; European Study Group for Pancreatic Cancer. Adjuvant chemotherapy with fluorouracil plus folinic acid vs gemcitabine following pancreatic cancer resection: a randomized controlled trial. JAMA. 2010;304:1073-1081.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1008]  [Cited by in F6Publishing: 943]  [Article Influence: 67.4]  [Reference Citation Analysis (0)]
6.  Oettle H, Neuhaus P, Hochhaus A, Hartmann JT, Gellert K, Ridwelski K, Niedergethmann M, Zülke C, Fahlke J, Arning MB, Sinn M, Hinke A, Riess H. Adjuvant chemotherapy with gemcitabine and long-term outcomes among patients with resected pancreatic cancer: the CONKO-001 randomized trial. JAMA. 2013;310:1473-1481.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1180]  [Cited by in F6Publishing: 1221]  [Article Influence: 111.0]  [Reference Citation Analysis (0)]
7.  Neoptolemos JP, Palmer DH, Ghaneh P, Psarelli EE, Valle JW, Halloran CM, Faluyi O, O'Reilly DA, Cunningham D, Wadsley J, Darby S, Meyer T, Gillmore R, Anthoney A, Lind P, Glimelius B, Falk S, Izbicki JR, Middleton GW, Cummins S, Ross PJ, Wasan H, McDonald A, Crosby T, Ma YT, Patel K, Sherriff D, Soomal R, Borg D, Sothi S, Hammel P, Hackert T, Jackson R, Büchler MW; European Study Group for Pancreatic Cancer. Comparison of adjuvant gemcitabine and capecitabine with gemcitabine monotherapy in patients with resected pancreatic cancer (ESPAC-4): a multicentre, open-label, randomised, phase 3 trial. Lancet. 2017;389:1011-1024.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1327]  [Cited by in F6Publishing: 1244]  [Article Influence: 177.7]  [Reference Citation Analysis (0)]
8.  Conroy T, Hammel P, Hebbar M, Ben Abdelghani M, Wei AC, Raoul JL, Choné L, Francois E, Artru P, Biagi JJ, Lecomte T, Assenat E, Faroux R, Ychou M, Volet J, Sauvanet A, Breysacher G, Di Fiore F, Cripps C, Kavan P, Texereau P, Bouhier-Leporrier K, Khemissa-Akouz F, Legoux JL, Juzyna B, Gourgou S, O'Callaghan CJ, Jouffroy-Zeller C, Rat P, Malka D, Castan F, Bachet JB; Canadian Cancer Trials Group and the Unicancer-GI–PRODIGE Group. FOLFIRINOX or Gemcitabine as Adjuvant Therapy for Pancreatic Cancer. N Engl J Med. 2018;379:2395-2406.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1427]  [Cited by in F6Publishing: 1654]  [Article Influence: 275.7]  [Reference Citation Analysis (0)]
9.  Merkow RP, Bilimoria KY, Tomlinson JS, Paruch JL, Fleming JB, Talamonti MS, Ko CY, Bentrem DJ. Postoperative complications reduce adjuvant chemotherapy use in resectable pancreatic cancer. Ann Surg. 2014;260:372-377.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 286]  [Cited by in F6Publishing: 294]  [Article Influence: 29.4]  [Reference Citation Analysis (0)]
10.  Altman AM, Wirth K, Marmor S, Lou E, Chang K, Hui JYC, Tuttle TM, Jensen EH, Denbo JW. Completion of Adjuvant Chemotherapy After Upfront Surgical Resection for Pancreatic Cancer Is Uncommon Yet Associated With Improved Survival. Ann Surg Oncol. 2019;26:4108-4116.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 73]  [Article Influence: 14.6]  [Reference Citation Analysis (0)]
11.  Nakao A, Kanzaki A, Fujii T, Kodera Y, Yamada S, Sugimoto H, Nomoto S, Nakamura S, Morita S, Takeda S. Correlation between radiographic classification and pathological grade of portal vein wall invasion in pancreatic head cancer. Ann Surg. 2012;255:103-108.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 136]  [Cited by in F6Publishing: 140]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
12.  Sohal DPS, Duong M, Ahmad SA, Gandhi NS, Beg MS, Wang-Gillam A, Wade JL 3rd, Chiorean EG, Guthrie KA, Lowy AM, Philip PA, Hochster HS. Efficacy of Perioperative Chemotherapy for Resectable Pancreatic Adenocarcinoma: A Phase 2 Randomized Clinical Trial. JAMA Oncol. 2021;7:421-427.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 156]  [Cited by in F6Publishing: 144]  [Article Influence: 48.0]  [Reference Citation Analysis (0)]
13.  Denbo JW, Fleming JB. Definition and Management of Borderline Resectable Pancreatic Cancer. Surg Clin North Am. 2016;96:1337-1350.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
14.  Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646-674.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39812]  [Cited by in F6Publishing: 43499]  [Article Influence: 3346.1]  [Reference Citation Analysis (4)]
15.  Pointer DT Jr, Roife D, Powers BD, Murimwa G, Elessawy S, Thompson ZJ, Schell MJ, Hodul PJ, Pimiento JM, Fleming JB, Malafa MP. Neutrophil to lymphocyte ratio, not platelet to lymphocyte or lymphocyte to monocyte ratio, is predictive of patient survival after resection of early-stage pancreatic ductal adenocarcinoma. BMC Cancer. 2020;20:750.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
16.  Murthy P, Zenati MS, Al Abbas AI, Rieser CJ, Bahary N, Lotze MT, Zeh HJ 3rd, Zureikat AH, Boone BA. Prognostic Value of the Systemic Immune-Inflammation Index (SII) After Neoadjuvant Therapy for Patients with Resected Pancreatic Cancer. Ann Surg Oncol. 2020;27:898-906.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 46]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
17.  Kim JS, Choi M, Kim SH, Hwang HK, Lee WJ, Kang CM. Systemic inflammation response index correlates with survival and predicts oncological outcome of resected pancreatic cancer following neoadjuvant chemotherapy. Pancreatology. 2022;22:987-993.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
18.  Bernard V, Kim DU, San Lucas FA, Castillo J, Allenson K, Mulu FC, Stephens BM, Huang J, Semaan A, Guerrero PA, Kamyabi N, Zhao J, Hurd MW, Koay EJ, Taniguchi CM, Herman JM, Javle M, Wolff R, Katz M, Varadhachary G, Maitra A, Alvarez HA. Circulating Nucleic Acids Are Associated With Outcomes of Patients With Pancreatic Cancer. Gastroenterology. 2019;156:108-118.e4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 205]  [Cited by in F6Publishing: 235]  [Article Influence: 47.0]  [Reference Citation Analysis (0)]
19.  Patel SH, Katz MHG, Ahmad SA. The Landmark Series: Preoperative Therapy for Pancreatic Cancer. Ann Surg Oncol. 2021;28:4104-4129.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
20.  Zins M, Matos C, Cassinotto C. Pancreatic Adenocarcinoma Staging in the Era of Preoperative Chemotherapy and Radiation Therapy. Radiology. 2018;287:374-390.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 101]  [Article Influence: 16.8]  [Reference Citation Analysis (0)]
21.  Tsai S, Borazanci E, Gulley M, Rashid N, Merker J, Khan AH, Chisholm P, Hunt B, Giorgadze T, Hall W, Kamgar M, Evans DB, Yeh JJ. Abstract PO-055: Phase II clinical trial of subtype directed neoadjuvant therapy in patients with localized pancreatic cancer. Cancer Res. 2021;81:PO-055.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
22.  Janssen QP, O'Reilly EM, van Eijck CHJ, Groot Koerkamp B. Neoadjuvant Treatment in Patients With Resectable and Borderline Resectable Pancreatic Cancer. Front Oncol. 2020;10:41.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 56]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
23.  Khorana AA, Mangu PB, Berlin J, Engebretson A, Hong TS, Maitra A, Mohile SG, Mumber M, Schulick R, Shapiro M, Urba S, Zeh HJ, Katz MHG. Potentially Curable Pancreatic Cancer: American Society of Clinical Oncology Clinical Practice Guideline Update. J Clin Oncol. 2017;35:2324-2328.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 133]  [Article Influence: 19.0]  [Reference Citation Analysis (0)]
24.  Kung HC, Yu J. Targeted therapy for pancreatic ductal adenocarcinoma: Mechanisms and clinical study. MedComm (2020). 2023;4:e216.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 6]  [Reference Citation Analysis (0)]
25.  Gillen S, Schuster T, Meyer Zum Büschenfelde C, Friess H, Kleeff J. Preoperative/neoadjuvant therapy in pancreatic cancer: a systematic review and meta-analysis of response and resection percentages. PLoS Med. 2010;7:e1000267.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1085]  [Cited by in F6Publishing: 1096]  [Article Influence: 78.3]  [Reference Citation Analysis (0)]
26.  Jung HS, Han Y, Yun WG, Cho YJ, Lee M, Lee DH, Kwon W, Jang JY. Examining neoadjuvant treatment candidates in resectable pancreatic cancer based on tumor-vessel interactions and CA 19-9 levels: A retrospective cohort study. Int J Surg. 2024;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
27.  Mokdad AA, Minter RM, Zhu H, Augustine MM, Porembka MR, Wang SC, Yopp AC, Mansour JC, Choti MA, Polanco PM. Neoadjuvant Therapy Followed by Resection Versus Upfront Resection for Resectable Pancreatic Cancer: A Propensity Score Matched Analysis. J Clin Oncol. 2017;35:515-522.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 234]  [Cited by in F6Publishing: 277]  [Article Influence: 34.6]  [Reference Citation Analysis (0)]
28.  Sugawara T, Rodriguez Franco S, Sherman S, Torphy RJ, Colborn K, Franklin O, Ishida J, Grandi S, Al-Musawi MH, Gleisner A, Schulick RD, Del Chiaro M. Neoadjuvant Chemotherapy Versus Upfront Surgery for Resectable Pancreatic Adenocarcinoma: An Updated Nationwide Study. Ann Surg. 2024;279:331-339.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Reference Citation Analysis (0)]
29.  Labori KJ, Bratlie SO, Andersson B, Angelsen JH, Biörserud C, Björnsson B, Bringeland EA, Elander N, Garresori H, Grønbech JE, Haux J, Hemmingsson O, Liljefors MG, Myklebust TÅ, Nymo LS, Peltola K, Pfeiffer P, Sallinen V, Sandström P, Sparrelid E, Stenvold H, Søreide K, Tingstedt B, Verbeke C, Öhlund D, Klint L, Dueland S, Lassen K; Nordic Pancreatic Cancer Trial-1 study group. Neoadjuvant FOLFIRINOX versus upfront surgery for resectable pancreatic head cancer (NORPACT-1): a multicentre, randomised, phase 2 trial. Lancet Gastroenterol Hepatol. 2024;9:205-217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 1]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
30.  Schwarz L, Vernerey D, Bachet JB, Tuech JJ, Portales F, Michel P, Cunha AS. Resectable pancreatic adenocarcinoma neo-adjuvant FOLF(IRIN)OX-based chemotherapy - a multicenter, non-comparative, randomized, phase II trial (PANACHE01-PRODIGE48 study). BMC Cancer. 2018;18:762.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 60]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
31.  Ahmad SA, Duong M, Sohal DPS, Gandhi NS, Beg MS, Wang-Gillam A, Wade JL 3rd, Chiorean EG, Guthrie KA, Lowy AM, Philip PA, Hochster HS. Surgical Outcome Results From SWOG S1505: A Randomized Clinical Trial of mFOLFIRINOX Versus Gemcitabine/Nab-paclitaxel for Perioperative Treatment of Resectable Pancreatic Ductal Adenocarcinoma. Ann Surg. 2020;272 481-486 [PMID:32740235 DOI: 10.1097/SLA.0000000000004155.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Versteijne E, van Dam JL, Suker M, Janssen QP, Groothuis K, Akkermans-Vogelaar JM, Besselink MG, Bonsing BA, Buijsen J, Busch OR, Creemers GM, van Dam RM, Eskens FALM, Festen S, de Groot JWB, Groot Koerkamp B, de Hingh IH, Homs MYV, van Hooft JE, Kerver ED, Luelmo SAC, Neelis KJ, Nuyttens J, Paardekooper GMRM, Patijn GA, van der Sangen MJC, de Vos-Geelen J, Wilmink JW, Zwinderman AH, Punt CJ, van Tienhoven G, van Eijck CHJ; Dutch Pancreatic Cancer Group. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 216]  [Cited by in F6Publishing: 244]  [Article Influence: 122.0]  [Reference Citation Analysis (0)]
33.  Chawla A, Shi Q, Ko AH, Beg S, Varghese AM, Behrman SW, Bloomston M, Ahmed FS, Frankel WL, Dixon JG, Carrero XW, Shergill A, Crawley J, Akin O, Renouf DJ, Zogopoulos G, Hubbard J M, Meyerhardt JA, Oreilly EM, Ferrone CR. Alliance A021806: A phase III trial evaluating perioperative versus adjuvant therapy for resectable pancreatic cancer. J Clin Oncol. 2023;41.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  van Dam JL, Verkolf EMM, Dekker EN, Bonsing BA, Bratlie SO, Brosens LAA, Busch OR, van Driel LMJW, van Eijck CHJ, Feshtali S, Ghorbani P, de Groot DJA, de Groot JWB, Haberkorn BCM, de Hingh IH, van der Holt B, Karsten TM, van der Kolk MB, Labori KJ, Liem MSL, Loosveld OJL, Molenaar IQ, Polée MB, van Santvoort HC, de Vos-Geelen J, Wumkes ML, van Tienhoven G, Homs MYV, Besselink MG, Wilmink JW, Groot Koerkamp B; Dutch Pancreatic Cancer Group. Perioperative or adjuvant mFOLFIRINOX for resectable pancreatic cancer (PREOPANC-3): study protocol for a multicenter randomized controlled trial. BMC Cancer. 2023;23:728.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Reference Citation Analysis (0)]
35.  Springfeld C, Ferrone CR, Katz MHG, Philip PA, Hong TS, Hackert T, Büchler MW, Neoptolemos J. Neoadjuvant therapy for pancreatic cancer. Nat Rev Clin Oncol. 2023;20:318-337.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 67]  [Reference Citation Analysis (0)]
36.  Wu HY, Li JW, Li JZ, Zhai QL, Ye JY, Zheng SY, Fang K. Comprehensive multimodal management of borderline resectable pancreatic cancer: Current status and progress. World J Gastrointest Surg. 2023;15:142-162.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (1)]
37.  Wainberg ZA, Melisi D, Macarulla T, Pazo Cid R, Chandana SR, De La Fouchardière C, Dean A, Kiss I, Lee WJ, Goetze TO, Van Cutsem E, Paulson AS, Bekaii-Saab T, Pant S, Hubner RA, Xiao Z, Chen H, Benzaghou F, O'Reilly EM. NALIRIFOX versus nab-paclitaxel and gemcitabine in treatment-naive patients with metastatic pancreatic ductal adenocarcinoma (NAPOLI 3): a randomised, open-label, phase 3 trial. Lancet. 2023;402:1272-1281.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 31]  [Article Influence: 31.0]  [Reference Citation Analysis (0)]
38.  Melisi D, Zecchetto C, Merz V, Malleo G, Landoni L, Quinzii A, Casalino S, Fazzini F, Gaule M, Pesoni C, Casetti L, Esposito A, Marchegiani G, Piazzola C, D'Onofrio M, de Robertis R, Gabbrielli A, Bernardoni L, Crino SF, Pietrobono S, Luchini C, Aliberti C, Martignoni G, Milleri S, Butturini G, Scarpa A, Salvia R, Bassi C. Perioperative NALIRIFOX in patients with resectable pancreatic ductal adenocarcinoma: The open-label, multicenter, phase II nITRO trial. Eur J Cancer. 2024;196:113430.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
39.  Hammel P, Huguet F, van Laethem JL, Goldstein D, Glimelius B, Artru P, Borbath I, Bouché O, Shannon J, André T, Mineur L, Chibaudel B, Bonnetain F, Louvet C; LAP07 Trial Group. Effect of Chemoradiotherapy vs Chemotherapy on Survival in Patients With Locally Advanced Pancreatic Cancer Controlled After 4 Months of Gemcitabine With or Without Erlotinib: The LAP07 Randomized Clinical Trial. JAMA. 2016;315:1844-1853.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 622]  [Cited by in F6Publishing: 667]  [Article Influence: 83.4]  [Reference Citation Analysis (0)]
40.  Kong Q, Teng F, Li H, Chen Z. Radical resection benefits patients suffering pancreatic ductal adenocarcinoma with liver oligometastases. Ann Surg Treat Res. 2024;106:51-60.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
41.  Boggi U, Kauffmann E, Napoli N, Barreto SG, Besselink MG, Fusai GK, Hackert T, Abu Hilal M, Marchegiani G, Salvia R, Shrikhande S, Truty M, Werner J, Wolfgang C, Bannone E, Capretti G, Cattelani A, Coppola A, Cucchetti A, De Sio D, Di Dato A, Di Meo G, Fiorillo C, Gianfaldoni C, Ginesini M, Hidalgo Salinas C, Lai Q, Miccoli M, Montorsi R, Pagnanelli M, Poli A, Ricci C, Sucameli F, Tamburrino D, Viti V, Addeo PF, Alfieri S, Bachellier P, Baiocchi G, Balzano G, Barbarello L, Brolese A, Busquets J, Butturini G, Caniglia F, Caputo D, Casadei R, Chunhua X, Colangelo E, Coratti A, Costa F, Crafa F, Dalla Valle R, De Carlis L, de Wilde RF, Del Chiaro M, Di Benedetto F, Di Sebastiano P, Dokmak S, Hogg M, Egorov VI, Ercolani G, Ettorre GM, Falconi M, Ferrari G, Ferrero A, Filauro M, Giardino A, Grazi GL, Gruttaduaria S, Izbicki JR, Jovine E, Katz M, Keck T, Khatkov I, Kiguchi G, Kooby D, Lang H, Lombardo C, Malleo G, Massani M, Mazzaferro V, Memeo R, Miao Y, Mishima K, Molino C, Nagakawa Y, Nakamura M, Nardo B, Panaro F, Pasquali C, Perrone V, Rangelova E, Riu L, Romagnoli R, Romito R, Rosso E, Schulick R, Siriwardena A, Spampinato M, Strobel O, Testini M, Troisi R, Uzunoglo FG, Valente R, Veneroni L, Zerbi A, Vicente E, Vistoli F, Vivarelli M, Wakabayashi G, Zanus G, Zureikat A, Zyromski NJ, Coppola R, D'Andrea V, Davide J, Dervenis C, Frigerio I, Konlon KC, Michelassi F, Montorsi M, Nealon W, Portolani N, Sousa Silva D, Bozzi G, Ferrari V, Trivella MG, Cameron J, Clavien PA, Asbun HJ; Multidisciplinary advisory board. REDISCOVER International Guidelines on the Perioperative Care of Surgical Patients With Borderline-resectable and Locally Advanced Pancreatic Cancer. Ann Surg. 2024;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Reference Citation Analysis (0)]
42.  Reames BN, Blair AB, Krell RW, Groot VP, Gemenetzis G, Padussis JC, Thayer SP, Falconi M, Wolfgang CL, Weiss MJ, Are C, He J. Management of Locally Advanced Pancreatic Cancer: Results of an International Survey of Current Practice. Ann Surg. 2021;273:1173-1181.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 37]  [Article Influence: 12.3]  [Reference Citation Analysis (1)]
43.  Fietkau R, Ghadimi M, Grützmann R, Wittel UA, Jacobasch L, Uhl W, Croner RS, Bechstein WO, Neumann UP, Waldschmidt D, Boeck SH, Moosmann N, Anke C. Reinacher-Schick, Henriette Golcher, Werner Adler, Sabine Semrau, Kallies A, Hecht M, Tannapfel A, Oettle H. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
44.  Shouman MA, Fuchs F, Walter F, Corradini S, Westphalen CB, Vornhülz M, Beyer G, Andrade D, Belka C, Niyazi M, Rogowski P. Stereotactic body radiotherapy for pancreatic cancer - A systematic review of prospective data. Clin Transl Radiat Oncol. 2024;45:100738.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
45.  Wang ZJ, Arif-Tiwari H, Zaheer A, Ameli S, Bhosale PR, Do RK, Goenka AH, Guimares AR, Sangster GP, Soloff EV, Tamm EP, Zins M, Kambadakone AR. Therapeutic response assessment in pancreatic ductal adenocarcinoma: society of abdominal radiology review paper on the role of morphological and functional imaging techniques. Abdom Radiol (NY). 2020;45:4273-4289.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 14]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
46.  Dougan SK. The Pancreatic Cancer Microenvironment. Cancer J. 2017;23:321-325.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 116]  [Article Influence: 19.3]  [Reference Citation Analysis (0)]
47.  Rezai P, Mulcahy MF, Tochetto SM, Berggruen S, Yaghmai V. Morphological analysis of pancreatic adenocarcinoma on multidetector row computed tomography: implications for treatment response evaluation. Pancreas. 2009;38:799-803.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
48.  Boone BA, Steve J, Zenati MS, Hogg ME, Singhi AD, Bartlett DL, Zureikat AH, Bahary N, Zeh HJ 3rd. Serum CA 19-9 response to neoadjuvant therapy is associated with outcome in pancreatic adenocarcinoma. Ann Surg Oncol. 2014;21:4351-4358.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 125]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
49.  Al Abbas AI, Zenati M, Reiser CJ, Hamad A, Jung JP, Zureikat AH, Zeh HJ 3rd, Hogg ME. Serum CA19-9 Response to Neoadjuvant Therapy Predicts Tumor Size Reduction and Survival in Pancreatic Adenocarcinoma. Ann Surg Oncol. 2020;27:2007-2014.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 35]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
50.  Yun WG, Han Y, Cho YJ, Jung HS, Lee M, Kwon W, Jang JY. In Neoadjuvant FOLFIRINOX Chemotherapy for Pancreatic Ductal Adenocarcinoma, Which Response is the More Reliable Indicator for Prognosis, Radiologic or Biochemical? Ann Surg Oncol. 2024;31:1336-1346.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
51.  Newhook TE, Vreeland TJ, Griffin JF, Tidwell RSS, Prakash LR, Koay EJ, Ludmir EB, Smaglo BG, Pant S, Overman M, Wolff RA, Ikoma N, Maxwell J, Kim MP, Lee JE, Katz MHG, Tzeng CD. Prognosis Associated With CA19-9 Response Dynamics and Normalization During Neoadjuvant Therapy in Resected Pancreatic Adenocarcinoma. Ann Surg. 2023;277:484-490.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 11]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
52.  Lee W, Oh M, Kim JS, Park Y, Kwon JW, Jun E, Song KB, Lee JH, Hwang DW, Yoo C, Kim KP, Jeong JH, Chang HM, Ryoo BY, Park SY, Kim SC. Metabolic activity by FDG-PET/CT after neoadjuvant chemotherapy in borderline resectable and locally advanced pancreatic cancer and association with survival. Br J Surg. 2021;109:61-70.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 13]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
53.  Abdelrahman AM, Goenka AH, Alva-Ruiz R, Yonkus JA, Leiting JL, Graham RP, Merrell KW, Thiels CA, Hallemeier CL, Warner SG, Haddock MG, Grotz TE, Tran NH, Smoot RL, Ma WW, Cleary SP, McWilliams RR, Nagorney DM, Halfdanarson TR, Kendrick ML, Truty MJ. FDG-PET Predicts Neoadjuvant Therapy Response and Survival in Borderline Resectable/Locally Advanced Pancreatic Adenocarcinoma. J Natl Compr Canc Netw. 2022;20:1023-1032.e3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 20]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
54.  Yin L, Pu N, Thompson E, Miao Y, Wolfgang C, Yu J. Improved Assessment of Response Status in Patients with Pancreatic Cancer Treated with Neoadjuvant Therapy using Somatic Mutations and Liquid Biopsy Analysis. Clin Cancer Res. 2021;27:740-748.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 17]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
55.  van Dam JL, Janssen QP, Besselink MG, Homs MYV, van Santvoort HC, van Tienhoven G, de Wilde RF, Wilmink JW, van Eijck CHJ, Groot Koerkamp B; Dutch Pancreatic Cancer Group. Neoadjuvant therapy or upfront surgery for resectable and borderline resectable pancreatic cancer: A meta-analysis of randomised controlled trials. Eur J Cancer. 2022;160:140-149.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 88]  [Article Influence: 29.3]  [Reference Citation Analysis (0)]
56.  Lee SH, Hwang DW, Yoo C, Kim KP, Kang S, Jeong JH, Oh D, Song TJ, Lee SS, Park DH, Seo DW, Park JH, Song KB, Lee JH, Lee W, Park Y, Kwak BJ, Chang HM, Ryoo BY, Kim SC. Survival Benefit of Adjuvant Chemotherapy in Patients with Pancreatic Ductal Adenocarcinoma Who Underwent Surgery Following Neoadjuvant FOLFIRINOX. Cancer Res Treat. 2023;55:956-968.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
57.  Bailey P, Zhou X, An J, Peccerella T, Hu K, Springfeld C, Büchler M, Neoptolemos JP. Refining the Treatment of Pancreatic Cancer From Big Data to Improved Individual Survival. Function (Oxf). 2023;4:zqad011.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
58.  Jones S, Zhang X, Parsons DW, Lin JC, Leary RJ, Angenendt P, Mankoo P, Carter H, Kamiyama H, Jimeno A, Hong SM, Fu B, Lin MT, Calhoun ES, Kamiyama M, Walter K, Nikolskaya T, Nikolsky Y, Hartigan J, Smith DR, Hidalgo M, Leach SD, Klein AP, Jaffee EM, Goggins M, Maitra A, Iacobuzio-Donahue C, Eshleman JR, Kern SE, Hruban RH, Karchin R, Papadopoulos N, Parmigiani G, Vogelstein B, Velculescu VE, Kinzler KW. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science. 2008;321:1801-1806.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3082]  [Cited by in F6Publishing: 2907]  [Article Influence: 181.7]  [Reference Citation Analysis (0)]
59.  Connor AA, Gallinger S. Next generation sequencing of pancreatic ductal adenocarcinoma: right or wrong? Expert Rev Gastroenterol Hepatol. 2017;11:683-694.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
60.  Rashid NU, Peng XL, Jin C, Moffitt RA, Volmar KE, Belt BA, Panni RZ, Nywening TM, Herrera SG, Moore KJ, Hennessey SG, Morrison AB, Kawalerski R, Nayyar A, Chang AE, Schmidt B, Kim HJ, Linehan DC, Yeh JJ. Purity Independent Subtyping of Tumors (PurIST), A Clinically Robust, Single-sample Classifier for Tumor Subtyping in Pancreatic Cancer. Clin Cancer Res. 2020;26:82-92.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 94]  [Article Influence: 18.8]  [Reference Citation Analysis (0)]