Article Search
검색
검색 팝업 닫기

Metrics

Help

  • 1. Aims and Scope

    Gut and Liver is an international journal of gastroenterology, focusing on the gastrointestinal tract, liver, biliary tree, pancreas, motility, and neurogastroenterology. Gut atnd Liver delivers up-to-date, authoritative papers on both clinical and research-based topics in gastroenterology. The Journal publishes original articles, case reports, brief communications, letters to the editor and invited review articles in the field of gastroenterology. The Journal is operated by internationally renowned editorial boards and designed to provide a global opportunity to promote academic developments in the field of gastroenterology and hepatology. +MORE

  • 2. Editorial Board

    Editor-in-Chief + MORE

    Editor-in-Chief
    Yong Chan Lee Professor of Medicine
    Director, Gastrointestinal Research Laboratory
    Veterans Affairs Medical Center, Univ. California San Francisco
    San Francisco, USA

    Deputy Editor

    Deputy Editor
    Jong Pil Im Seoul National University College of Medicine, Seoul, Korea
    Robert S. Bresalier University of Texas M. D. Anderson Cancer Center, Houston, USA
    Steven H. Itzkowitz Mount Sinai Medical Center, NY, USA
  • 3. Editorial Office
  • 4. Articles
  • 5. Instructions for Authors
  • 6. File Download (PDF version)
  • 7. Ethical Standards
  • 8. Peer Review

    All papers submitted to Gut and Liver are reviewed by the editorial team before being sent out for an external peer review to rule out papers that have low priority, insufficient originality, scientific flaws, or the absence of a message of importance to the readers of the Journal. A decision about these papers will usually be made within two or three weeks.
    The remaining articles are usually sent to two reviewers. It would be very helpful if you could suggest a selection of reviewers and include their contact details. We may not always use the reviewers you recommend, but suggesting reviewers will make our reviewer database much richer; in the end, everyone will benefit. We reserve the right to return manuscripts in which no reviewers are suggested.

    The final responsibility for the decision to accept or reject lies with the editors. In many cases, papers may be rejected despite favorable reviews because of editorial policy or a lack of space. The editor retains the right to determine publication priorities, the style of the paper, and to request, if necessary, that the material submitted be shortened for publication.

Search

Search

Year

to

Article Type

Review Article

Split Viewer

Neoadjuvant and Adjuvant Treatments for Resectable and Borderline Resectable Pancreatic Ductal Adenocarcinoma: The Current Status of Pancreatic Ductal Adenocarcinoma Treatment in Japan

Shigenori Ei , Shinichiro Takahashi , Toshihito Ogasawara , Taro Mashiko , Yoshihito Masuoka , Toshio Nakagohri

Department of Gastroenterological Surgery, Tokai University School of Medicine, Isehara, Japan

Correspondence to: Shinichiro Takahashi
ORCID https://orcid.org/0000-0002-4828-3965
E-mail shtakaha@tsc.u-tokai.ac.jp

Received: July 18, 2022; Revised: October 30, 2022; Accepted: October 30, 2022

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Gut Liver 2023;17(5):698-710. https://doi.org/10.5009/gnl220311

Published online February 27, 2023, Published date September 15, 2023

Copyright © Gut and Liver.

Resection is the only curative treatment for pancreatic ductal adenocarcinoma (PDAC). Although the outcome of technically resectable PDAC has improved with advances in surgery and adjuvant therapy, the 5-year survival rate remains low at 20% to 40%. More effective therapy is needed. Almost 15 years ago, the National Comprehensive Cancer Network guidelines proposed a resectability classification of PDAC based on preoperative imaging. Since then, treatment strategies for PDAC have been devised based on resectability. The standard of care for resectable PDAC is adjuvant chemotherapy after R0 resection, as shown by the results of pivotal clinical trials. With regard to neoadjuvant treatment, several recent clinical trials comparing neoadjuvant treatment with upfront resection have been conducted on resectable PDAC and borderline resectable PDAC, and the benefits and efficacy of neoadjuvant treatment for pancreatic cancer has become clearer. The significance of neoadjuvant treatment for resectable PDAC remains controversial, but in borderline resectable PDAC the efficacy of neoadjuvant treatment has been further recognised, although the standard of care has not yet been established. Several promising clinical trials for PDAC are ongoing. This review presents previous and ongoing trials of perioperative treatment for resectable and borderline resectable PDAC, focusing on the difference between Asian and Western countries.

Keywords: Pancreatic neoplasms, Neoadjuvant therapy, Adjuvant chemotherapy, Chemoradiotherapy, Clinical trial

Pancreatic ductal adenocarcinoma (PDAC) frequently recurs after resection even if radical resection is performed. Perioperative treatment such as neoadjuvant or adjuvant therapy is essential to improve the prognosis of PDAC. The 5-year survival rate after pancreatic resection without any perioperative treatment for PDAC is approximately 10%,1,2 whereas that with adjuvant therapy has improved to 20% to 40% in the last two decades. This study reviewed previous pivotal studies of perioperative treatment for PDAC to reconfirm the evolution of the standard treatment to date and also highlighted the interesting ongoing trials to discuss future development trends of perioperative treatment for PDAC.

When planning treatment strategies for PDAC, the resectability status is critically important alongside the staging classification (TNM classification) to determine the possibility of curative resection, prognosis, and tumor progression patterns. The National Comprehensive Cancer Network guidelines3 have classified resectability since 2006, and the Japanese Clinical Practice Guidelines for Pancreatic Cancer (7th edition)4 also defines resectability classification (Table 1). Both classifications define the resectability status as resectable (R), borderline resectable (BR), or unresectable (UR). Either R-PDAC or BR-PDAC is technically resectable; however, the tumor stage including local extension and lymph node metastasis is more advanced in BR-PDAC than that in R-PDAC. The risk of local and distant BR-PDAC progression is higher than that of R-PDAC.5-7 Thus, perioperative treatment strategies for BR-PDAC and R-PDAC should be different. Therefore, this review separately examined the perioperative treatment development for R-PDAC and BR-PDAC.

Table 1 Criteria Defining Resectability Status in Japan Pancreas Society Classification 7th Edition (2016)

Resectable: R
  • SMV/PV: no tumor contact or contact of <180° without occlusion

  • SMA, CA, CHA: no tumor contact/invasion

Borderline resectable: BR (subclassified according to SMV/PV invasion alone or arterial invasion)
BR-PV (SMV/PV invasion alone)
  • SMV/PV: tumor contact/invasion of ≥180°/occlusion, not exceeding the inferior border of the duodenum

  • SMA, CA, CHA: no tumor contact/invasion

BR-A (arterial invasion)
  • SMA, CA: tumor contact/invasion of <180° without deformity or narrowing

  • CHA: tumor contact/invasion without showing tumor contact/invasion of the PHA and/or CA (cases of contact/invasion of both PV and peripancreatic arteries are considered BR-A)

Unresectable: UR (subclassified according to the presence or absence of distant metastases)
UR-LA (locally advanced)
  • SMV/PV: tumor contact/invasion of ≥180°/occlusion, exceeding the inferior border of the duodenum

  • SMA, CA: tumor contact/invasion of ≥180°

  • CHA: tumor contact/invasion showing tumor contact/invasion of the PHA and/or CA

  • AO: tumor contact or invasion

UR-M (metastasis)
  • Distant metastasis including non-regional lymph node metastasis.

SMV, superior mesenteric vein; PV, portal vein; SMA, superior mesenteric artery; CA, celiac artery; CHA, common hepatic artery; PHA, proper hepatic artery; AO, aorta.



This review also focuses on the uniqueness of perioperative treatment development in Asia, including Japan, and compares it with that in Europe and United States.

1. Establishment of adjuvant gemcitabine therapy

Randomized controlled trials (RCTs) comparing postoperative adjuvant chemotherapy with resection alone for PDAC have been conducted since the 1990s; however, the development of effective treatments had not yet been established until the ESPAC-1 trial8 in 2004, which demonstrated a significant survival benefit of postoperative 5-fluorouracil+leucovorin (5-FU+LV) therapy over resection alone, and results of this trial had made postoperative 5-FU+LV therapy the standard of care for R-PDAC in Europe. However, the complex study design using a 2×2 factorial design was controversial and had not been implemented as the standard therapy in the United States and Japan.

Subsequently, the CONKO-001 trial2,9 was conducted mainly in Germany, in which 354 patients after PDAC resection were randomized to the adjuvant chemotherapy group with gemcitabine (GEM) and resection-alone group, showing a significant prolongation of recurrence-free survival with the adjuvant chemotherapy group (13.4 months vs 6.9 months, p<0.001). Initially, adjuvant chemotherapy did not show a significant increase in the overall survival (OS) (p=0.06); however, current long-term follow-up studies reported a significant increase not only in recurrence-free survival but also in the median OS (22.8 months vs 20.2 months, p=0.01).2

At about the same time, the JSAP-02 trial10 was conducted in Japan, which randomized 118 patients after PDAC resection to adjuvant chemotherapy with GEM and resection alone, and reported a significantly prolonged recurrence-free survival with postoperative GEM, a finding similar to the initial report of the CONKO-001 trial. As a result, GEM therapy was considered one of the standard treatments in addition to 5-FU+LV therapy.

European Study Group for Pancreatic Cancer (ESPAC) subsequently conducted the ESPAC-3 trial,11 in which 1,088 patients with resected PDAC were randomly allocated to the postoperative GEM or 5-FU+LV treatment groups, and reported no significant difference in the OS between the two groups (23.6 months vs 23.0 months: hazard ratio [HR], 0.94; 95% confidential interval [CI], 0.81 to 1.08; p=0.39). However, serious adverse events were significantly lower in the GEM group than in the 5-FU+LV group, and due to its safety profile, GEM therapy was preferentially selected as the standard of care not only in Japan but also worldwide. Subsequently, postoperative GEM became the standard regimen for the control group in many phase III trials for R-PDAC (Table 2).2,8-16

Table 2 Pivotal Trial on Adjuvant Chemotherapy for Pancreatic Cancer after Radical Resection

TrialYearAdjuvant treatmentNo. of patientsPrimary outcomeMedianHR (95% CI)p-value
ESPAC-1820045-FU+LV treated arm in 2×2 factorial design1472-yr survival rate40%0.71 (0.55–0.92)<0.01
5-FU+LV non-treated arm in 2×2 factorial design14230%
CONKO-0012,92007GEM186DFS13.4 mo0.55 (0.44–0.69)<0.01
Surgery alone1826.7 mo
JSAP-02102009GEM58OS22.3 mo0.77 (0.51–1.14)0.19
Surgery alone6018.4 mo
ESPAC-3112010GEM478OS23.6 mo0.94 (0.81–1.08)0.39
5-FU+LV48623.0 mo
JASPAC-01122013S-1187OS46.5 mo0.57 (0.44–0.72)<0.01
GEM19025.5 mo
ESPAC-4132016GEM+capecitabine364OS28.0 mo0.82 (0.68–0.98)0.03
GEM36625.5 mo
PRODIGE24/
ACCORD2414
2018mFOLFIRINOX247DFS21.6 mo0.58 (0.46–0.73)<0.01
GEM24612.8 mo
APACT15
(ASCO)
2019GEM+nab-PTX432DFS19.4 mo0.88 (0.73–1.06)0.18
GEM43418.8 mo
JSAP-0416On goingGEM+S-1-OS/DFS---
GEM--

HR, hazard ratio; CI, confidence interval; 5-FU+LV, 5-fluorouracil+leucovorin; GEM, gemcitabine; DFS, disease-free survival; OS, overall survival; mFOLFIRINOX, modified FOLFIRINOX; nab-PTX, nab-paclitaxel.



The use of 5-FU+LV therapy was recommended for patients who could not be safely administered with GEM, such as those with a history of interstitial pneumonia17,18 or hemolytic uremic syndrome19,20 in the Japanese Clinical Practice Guidelines for Pancreatic Cancer.

2. Development of postoperative adjuvant therapy superior to adjuvant GEM

In Europe, ESPAC conducted the ESPAC-4 trial13 involving 730 patients who underwent PDAC resection randomly allocated to the postoperative GEM and GEM+capecitabine (Cape) groups. The results showed that the OS was more significantly improved in the postoperative GEM+Cape group than in the GEM group (28.0 months vs 25.5 months: HR, 0.82; 95% CI, 0.68 to 0.98; p=0.032), and the study results led to the replacement of GEM therapy with GEM+Cape therapy as the standard postoperative adjuvant treatment.

Studies in France and Canada reported the results of a multicenter RCT (PRODIGE 24-ACCORD 24/CCTG PA 6)14 comparing postoperative modified FOLFIRINOX (mFOLFIRINOX) with GEM alone. This study used mFOLFIRINOX therapy considering the toxicity of FOLFIRINOX therapy, in which rapid administration of 5-FU was omitted and the irinotecan dose was reduced. The median disease-free survival, the primary endpoint, was 21.6 and 12.8 months in the mFOLFIRINOX and GEM-alone groups, respectively, showing the superiority in the mFOLFIRINOX group (HR, 0.58; 95% CI, 0.46 to 0.73; p<0.0001). The mFOLFIRINOX group also showed a significantly prolonged OS (54.4 months vs 35.0 months: HR, 0.64; 95% CI, 0.48 to 0.86; p=0.003). Grade 3/4 adverse events occurred in 75.9% and 52.9% of the mFOLFIRINOX and GEM-alone groups, respectively, and the treatment completion rates were 66.4% and 79.0% for mFOLFIRINOX and GEM alone, respectively (p=0.002). Based on the study results, postoperative mFOLFIRINOX therapy has become one of the standard treatments for resected PDAC. Because of its high toxicity, postoperative mFOLFIRINOX therapy is recommended for patients with good performance status (PS).

GEM plus erlotinib, an epidermal growth factor receptor tyrosine kinase inhibitor (CONKO-005 trial, RTOG-0848 trial),21,22 and GEM+nab-paclitaxel (GnP), one of the standard treatments for UR-PDAC (APACT trial),15 were also compared with adjuvant GEM in the global phase III trial. However, none of these therapies were recommended as the standard treatment for PDAC due to a lack of statistically significant efficacy by comparing with adjuvant GEM.

3. Different trends in Japan: postoperative S-1 therapy

The Japan Adjuvant Study Group of Pancreatic Cancer (JASPAC) conducted a phase III comparative study (JASPAC 01)12 to examine the non-inferiority of S-1 to GEM in adjuvant chemotherapy after the PDAC resection. The study enrolled 385 patients and showed not only non-inferiority of the S-1 group to the GEM group in the primary endpoint of OS, but also demonstrated superiority (median OS S-1 vs GEM, 46.5 months vs 25.5 months: HR, 0.57; 95% CI, 0.44 to 0.72; p<0.0001). The pre-completion discontinuation rate of the adjuvant treatment was 42% and 28% in the GEM and S-1 groups (p=0.0050), respectively, mainly due to adverse events (25% and 21%, respectively) and recurrence (13% and 5%, respectively). The study results clearly changed the standard postoperative adjuvant therapy in Japan from GEM therapy to S-1 therapy due to its high safety and convenience.

However, S-1 is not widely used for treatment of the pancreatic cancer including adjuvant therapy in Europe and the United States. The major reason is that the maximum tolerated dose of S-1 for Caucasians is lower than that for Mongolians due to gastrointestinal toxicity,23 mainly diarrhea, making it difficult to reproduce in the West the favorable antitumor effects of chemotherapy with S-1 reported in Asian clinical trials. The conversion efficiency of tegafur to 5-FU is higher in Caucasians due to polymorphisms in CYP2A6, resulting in higher gastrointestinal toxicity, especially diarrhea than that in Mongolians.24,25 In Europe, S-1 is approved for the treatment of advanced gastric cancer in combination with cisplatin,26 but the dose of S-1 was approved at about 60% of the dose under the original Japanese regimen. Additionally, S-1 is not approved for pancreatic cancer in Europe and United States. Due to the corporate strategy, development to expand the indication of S-1 for pancreatic cancer is not currently underway in the United States or Europe. Thus, S-1 has not become the standard of care for R-PDAC in Western countries.

Later, ESPAC-4 and PRODIGE24/ACCORD24 considered GEM+Cape and mFOLFIRINOX as the standard of care for R-PDAC in Western countries. In Japan, neither GEM+Cape nor mFOLFIRINOX became the standard of care mainly because the HR of the S-1 to GEM in the JASPAC-01 study (HR=0.57) was better than that of GEM+Cape to GEM in ESPAC-4 (HR=0.82) or that of mFOLFIRINOX to GEM in the PRODIGE24/ACCORD24 trial (HR=0.64). Then, adjuvant S-1 has been the standard postoperative adjuvant therapy to date.

4. Adjuvant chemoradiotherapy

A large number of case-control studies have suggested that adjuvant chemoradiotherapy (CRT) may prolong the prognosis;27-35 postoperative CRT analyses with novel anticancer agents including GEM and erlotinib are still insufficient;36-40 and meta-analyses have reported the benefits for patients with R1 resection compared to R0 (p=0.04).41 Postoperative adjuvant CRT has been one of the promising treatment modalities in the perioperative treatment of pancreatic cancer and remains a topic for further investigation.

The 2019 Clinical Practice Guidelines for Pancreatic Cancer committee42 conducted a meta-analysis of six RCTs comparing fluorouracil-based or GEM-based chemoradiation to surgery alone8,36,43-46 to evaluate the efficacy of adjuvant CRT. Therefore, the risk ratio of adjuvant CRT compared to surgery alone was 1.01 (95% CI, 0.93 to 1.10). The benefit of adjuvant CRT on the prognosis is unclear, and then adjuvant CRT is not recommended based on the Japanese guidelines at present.

1. Neoadjuvant therapy for R-PDAC

Although the aforementioned adjuvant chemotherapies are the current standard of care for patients with R-PDAC, 30% to 50% of patients who undergo laparotomy are unable to receive adjuvant therapy due to intraoperatively detected metastases, early postoperative recurrence, surgical complications, or poor PS. Conversely, neoadjuvant treatment has several theoretical advantages over adjuvant treatment, such as early treatment for micrometastasis under better physical condition, determination of tumor response for the treatment, and selection of patients with poor prognosis.47 Then, several clinical trials have been conducted to develop preoperative treatments not only for BR-PDAC but also for R-PDAC (Table 3).48-56

Table 3 Prospective Study on Neoadjuvant Chemotherapy for Pancreatic Cancer before Radical Resection

Author/trialYearResectabilityTreatmentNo. of patientsPrimary outcomeMedianHR (95% CI)p-value
Jang et al.482018BRCRT (54 Gy, GEM) → surgery → GEM272-yr survival40.7%0.51 (0.27–0.93)0.028
Surgery → GEM2326.1%
Unno et al. Prep-02/JSAP0549-512019R/BR-PVGEM + S-1 → surgery → S-1182OS36.7 mo0.72 (0.55–0.94)0.02
Surgery → S-118026.6 mo
Versteijne et al. PREOPANC-152,532020R/BRCRT (36 Gy, GEM) → surgery → GEM119OS15.7 mo0.73 (0.56–0.96)0.025
Surgery → GEM12714.3 mo
Takahashi et al. JASPAC05542020R/BRCRT (54 Gy, S-1) → surgery52R0 (OS)52% (30.8 mo)--
Yamaguchi et al. NUPAT-01552022BRmFOLFIRINOX → surgery26R073.1%-0.202
GEM + nab-PTX → surgery2556.0%
Katz et al. A021501562022BRmFOLFIRINOX → surgery → FOLFOX66518-mo survival66.7%--
mFOLFIRINOX → SBRT (33-40 Gy) or HIGRT (25 Gy) → surgery → FOLFOX65547.3%

HR, hazard ratio; CI, confidence interval; BR, Borderline Resectable; CRT, chemoradiotherapy; GEM, gemcitabine; R, resectable; PV, portal vein; OS, overall survival; mFOLFIRINOX, modified FOLFIRINOX; nab-PTX, nab-paclitaxel; SBRT, stereotactic body radiation therapy; HIGRT, hypofractionated image-guided radiation therapy.



In the 2010s, several clinical studies of preoperative treatment for R-PDAC were initiated in Europe. In 2015, randomized phase II trials of GEM+cisplatin+radiotherapy57 and GEM+radiotherapy58 were conducted; however, those trials could not be fully evaluated due to poor patient accrual. Evidence of preoperative treatment in R-PDAC is still lacking;59-62 however, important results were recently reported in two phase III trials comparing preoperative treatment for upfront resection in patients with R-PDAC or BR-PDAC.

A randomized multicenter phase III study (PREOPANC-1) was conducted in the Netherlands to examine the significance of neoadjuvant chemoradiation therapy (NACRT) for R-PDAC and BR-PDAC. The NACRT group (36 Gy/15 F + GEM 3 cycles → surgery → postoperative GEM for 4 cycles) showed a better tendency in the OS than the surgery first group. Although the difference was not significant in the initial report (modified OS 16.0 months vs 14.3 months: HR, 0.78; 95% CI, 0.58 to 1.05; p=0.096),52 long-term follow-up analysis showed significantly better OS in the NACRT group (modified OS 15.7 months vs 14.3 months: HR, 0.73; 95% CI, 0.56 to 0.96; p=0.025).53

A randomized phase III trial (Prep-02/JSAP05 trial)50 was performed in Japan to investigate the clinical significance of neoadjuvant chemotherapy (NAC) with GEM+S-1 (GS) for T1-T3 PDAC without invading the peripancreatic major arteries (hepatic artery, celiac artery, and superior mesenteric artery). This trial confirmed that NAC-GS safely and effectively improved the prognosis compared with upfront surgery (modified OS 36.7 months vs 26.6 months: HR, 0.72; 95% CI, 0.55 to 0.94; p=0.015). Furthermore, subsequent resection could be safely performed without significant differences in the operative time, intraoperative blood loss, and postoperative complication rates.49,51 Due to the following several limitations, (1) inclusion of 20% of BR-PDAC, (2) only tumors that could be biopsied and pathologically confirmed, and (3) age limit of 20 to 79 years, the study results just fell short by making neoadjuvant GS as the standard of care for R-PDAC. However, subgroup analysis in R-PDAC consisted of 80% of the enrolled patients revealed similar efficacy of the neoadjuvant GS. Then, using the results of the Prep-02/JSAP05 trial, the Japanese Clinical Practice Guidelines for Pancreatic Cancer were revised, and neoadjuvant GS therapy was recommended for R-PDAC.

The current most effective regimens, mFOLFIRINOX and GnP, have also being tried as preoperative treatments for R-PDAC. The randomized phase II SWOG S1505 compared perioperative mFOLFIRINOX and perioperative GnP in patients with R-PDAC.63,64 The primary outcome was 2-year survival. Investigators planned to compare the two arms to determine the better treatment if the 2-year survival reached 58% or more. However, the 2-year OSs of 41.6% with mFOLFIRINOX and 48.8% with GnP missed the target rate of 58% at 2 years. Furthermore, neither regimen showed a statistical improvement in 2-year survival over historical control, and the results were negative. However, these regimens remain the most promising at present, and the results of the ongoing phase III trial of perioperative mFOLFIRINOX therapy are awaited.

2. Neoadjuvant therapy for BR-PDAC

Compared to R-PDAC, BR-PDAC is more locally progressing and is at a higher tumor stage, with a higher risk of positive microscopic margins and early recurrence. Therefore, the significance of preoperative treatment in BR-PDAC is clearly greater than that in R-PDAC.

In BR-PDAC, several retrospective studies65-72 comparing neoadjuvant therapy with upfront surgery reported that neoadjuvant therapy improved the R0 resection rate and survival. Afterward, the efficacy of neoadjuvant therapy for BR-PDAC was evidently demonstrated in the phase II/III study reported from South Korea in 2018.48 A total of 58 patients were randomized to neoadjuvant chemoradiation (54 Gy) with GEM or upfront resection. The intention-to-treat analysis demonstrated that the primary endpoint of the 2-year survival was significantly better in the NACRT group (40.7%) than that in the upfront surgery group (26.1%) (HR, 1.97; 95% CI, 1.07 to 3.62; p=0.028).

Although the efficacy of neoadjuvant treatment was verified, the standard treatment has not yet been established in BR-PDAC. Several promising clinical trials of the neoadjuvant treatment are ongoing in this field (Table 4).73-81

Table 4 Prospective Ongoing Study on Neoadjuvant Chemotherapy for Pancreatic Cancer before Radical Resection

Author/trialPhaseStart yearResectabilityTreatmentScheduled number of patientsPrimary outcome
Labori et al. NorPACT-173II/III2017RmFOLFIRINOX → surgery → GEM + capecitabine140OS
Surgery → GEM + capecitabine
Schwarz et al. PANACHE01-PRODIGE4874II2017RmFOLFIRINOX → surgery → adjuvant therapy160OS
FOLFOX → surgery → adjuvant therapy
Surgery → adjuvant therapy
Ferrone Alliance A02180675III2020RmFOLFIRINOX → surgery →mFOLFIRINOX352OS
Surgery →mFOLFIRINOX
Koerkamp PREOPANC-376III2021RmFOLFIRINOX → surgery →mFOLFIRINOX378OS
Surgery → mFOLFIRINOX
Janssen et al. PREOPANC-277III2018R/BRmFOLFIRINOX → surgery368OS
GEM → CRT (36 Gy, GEM) → GEM → surgery → GEM
Grose et al. PRIMUS-00278II2019R/BRFOLFOX + nab-PTX → CRT → surgery278PFS
GEM + nab-PTX → CRT → surgery
Unno Prep-03/NS014-179I/II2014BRGEM + S-1 + concurrent RT (50 Gy) → surgery12–24/40R0
Okada et al. NAC-GA80II2017BRGEM + nab-PTX → surgery60OS
Takahashi GABARNANCE81II/III2017BRGEM + nab-PTX → surgery110R0/OS
CRT (54 Gy, S-1) → surgery

R, resectable; mFOLFIRINOX, modified FOLFIRINOX; GEM, gemcitabine; OS, overall survival; BR, borderline resectable; CRT, chemoradiotherapy; nab-PTX, nab-paclitaxel; RT, radiotherapy; PFS, progression-free survival.



In clinical practice of the United States or Europe, mFOLFIRINOX alone or mFOLFIRINOX followed by CRT or radiation therapy are the most common preoperative treatment for BR-PDAC. The A021501 phase II randomized clinical trial,56 which was conducted by the Alliance for Clinical Trials in Oncology, compared those promising treatments, namely, eight cycles of mFOLFIRINOX and seven cycles of mFOLFIRINOX followed by stereotactic body radiotherapy at 33 to 40 Gy in five fractions or hypofractionated image-guided radiotherapy at 25 Gy in five fractions as preoperative treatment for BR-PDAC. Each treatment arm’s 18-month OS rate was compared with a historical control rate of 50%. In the interim analysis, mFOLFIRINOX with hypofractionated radiotherapy arm was closed because R0 resection rate (33%) was proven to be under the threshold value (40%). The 18-month OS rate of chemotherapy arm was 66.7% and that of chemotherapy plus radiation arm was 47.3%. The results of the trial suggest that mFOLFIRINOX could be a reference neoadjuvant treatment regimen for BR-PDAC. However, the role of radiotherapy in the preoperative treatment for BR-PDAC remains undefined because hypofractionated radiotherapy instead of a more common conventional CRT was applied for chemotherapy plus radiation arm in the trial. Further studies are needed to clarify the significance of preoperative radiation therapy in BR-PDAC.

The perioperative treatment in Japan is unique due to the following two reasons. First, several perioperative treatments using S-1 have been developed in Japan and showed good results. Second, the toxic regimen tolerability, such as FOLFIRINOX in the Japanese population, might not be equivalent to that of the Western population.

As previously mentioned, adjuvant S-1 and neoadjuvant GS are recommended for R-PDAC in Japanese guidelines for the treatment of PDAC (Fig. 1A). S-1, oral fluorouracil, is a key medication for gastrointestinal malignancies (the stomach, colorectal, biliary tract, and PDAC)24,82,83 and is approved in many Asian countries. In Europe, S-1 is also approved by the European Medicines Agency and has been used for the treatment of advanced gastric cancer combined with cisplatin.26,84 However, European people tend to have more severe gastrointestinal side effects of S-1, such as diarrhea. S-1 is more toxic to the digestive tract in Caucasians than in Mongoloids due to metabolic and pharmacological differences. Genetic polymorphism differences, such as the higher activity of CYP2A6, the enzyme that converts tegafur in S-1 to 5-FU, in Caucasians, may be the cause of different toxicity profiles.25,82,85,86 The aforementioned perioperative treatments using S-1 are not recommended as the standard of care in Europe and the United States.

Figure 1.The strategy of PDAC treatment in Japan. (A) Current treatment strategy for R-PDAC in Japan. Perioperative treatment for R-PDAC is two cycles of neoadjuvant chemotherapy-GS (GEM=1,000 mg/m2, days 1 and 8; S-1=80 mg/m2, days 1–14) based on the results of the Prep-02/JSAP05 study and postoperative S-1 (S-1=80 mg/m2, days 1–28) in four cycles based on the results of the JASPAC-01 study. (B) Current treatment strategy for BR-PDAC in Japan. For preoperative treatment for BR-PDAC, GS (GEM=1,000 mg/m2, days 1 and 8; S-1=80 mg/m2, days 1–14) may be administered for two cycles based on the results of the Prep-02/JSAP05 study. However, several clinical trials have sought a stronger and more effective regimen, and no fixed strategy has been established yet. For postoperative treatment, S-1 (S-1=80 mg/m2, days 1–28) is administered for four cycles based on the results of the JASPAC-01 study.
PDAC, pancreatic ductal adenocarcinoma; R-PDCA, resectable PDAC; GEM, gemcitabine; GS, GEM+S-1; BR-PDAC, borderline resectable PDAC; mFOLFIRINOX, modified FOLFIRINOX; nab-PTX, nab-paclitaxel; CRT, chemoradiotherapy.

Conversely, postoperative therapy with mFOLFIRINOX has become the standard of care for R-PDAC in Europe and the United States, and FOLFIRINOX as preoperative treatment is also being developed. The postoperative toxicity of FOLFIRINOX is severe, especially in elderly people and in patients with poor PS, which increases the question of tolerability. Because of the high proportion of elderly patients in Japan, toxicity profiles are particularly important. When considering the efficacy and toxicity, both postoperative therapy with S-1 and preoperative GS therapy with S-1 balance between toxicity and efficacy seems suitable for Japanese patients with R-PDAC. Also, in East Asia with its large Mongoloid population, S-1 might be a key drug for R-PDAC.

S-1 is currently approved for the treatment of pancreatic cancer with locally advanced or metastatic disease in East Asian countries such as South Korea, Singapore, Taiwan, Thailand, and Malaysia. Moreover, adjuvant S-1 is recommended for R-PDAC as one of the treatments of grade I recommendations according to the clinical guidelines for the diagnosis and treatment of pancreatic cancer by the Chinese Society of Clinical Oncology.87 Several clinical trials using S-1 in the perioperative treatment of pancreatic cancer are currently being conducted or planned in China (NCT03278015, NCT03777462) and Taiwan (NCT04070313). However, at this time, it has not been verified whether perioperative chemotherapy with S-1 shows the same excellent efficacy and safety in East Asian countries other than China as observed in the Japanese clinical trial. Then, the results of the future clinical trials from East Asia other than Japan are expected to elucidate significance of perioperative treatment using S-1 for pancreatic cancer in East Asia.

Meanwhile, perioperative treatment with FOLFIRINOX, GnP, or chemoradiation is being developed in Japan for BR-PDAC, requiring more intensive therapy, and for younger patients who are more tolerable to toxicity (Table 4, Fig. 1B). The Prep-03/NS014-1 trial79 is a phase I/II study evaluating the efficacy of a GS+concurrent radiotherapy (50 Gy) regimen as NACRT. The NAC-GA trial80 is a phase II trial using GnP as NAC. Results of NUPAT-01 trial,55 a phase II trial of mFOLFIRINOX and GnP as NAC for BR-PDAC with a primary endpoint of R0 resection rate, were reported, showing that both groups had good R0 resection rates (73.1% vs 56.0%, respectively, p=0.202) and good 3-year survival rates (55.3% and 54.4%, respectively, p=0.389), demonstrating the NAC feasibility and tolerability with FOLFIRINOX or GnP. Moreover, the GABARNANCE trial,81 comparing the GnP therapy with S-1-based CRT (54 Gy) with the R0 resection rate and OS as primary endpoints, is awaiting results.

Currently, not a few clinical trials to evaluate two effective regimens, FOLFIRINOX88 and GnP89 which are standard therapies for UR-PDAC, as neoadjuvant therapy for R-/BR-PDAC are conducted and attracted attention (Table 4). The PREOPANC-1 and Prep-02/JSAP05 trials reported favorable outcomes for the preoperative treatment in R-PDAC; however, both of them were subgroup analyses and have not been established as evidence. Therefore, at present, neoadjuvant therapy is not yet recommended for R-PDAC in Western guidelines, except in high-risk patients.3 The major ongoing phase III trials for R-PDAC include Alliance A021806 in the United States75 and PREOPANC-3 in the Netherlands.76 These are trials to verify the significance of preoperative treatment, using mFOLFIRINOX for preoperative treatment and postoperative adjuvant therapy. The design of the two trials is quite similar: only resectable pancreatic cancer is included, eight courses of mFOLFIRINOX are performed preoperatively, followed by pancreatectomy and four courses of mFOLFIRINOX postoperatively, and the primary endpoint is OS. The results of the two large phase III trials examining the significance of preoperative treatment with mFOLFIRINOX, the most promising perioperative treatment regimen, will provide important insights into the efficacy of preoperative treatment.

The necessity for radiation therapy in the preoperative treatment of R- or BR-PDAC is an important issue. At present, preoperative chemoradiation remains controversial because pancreatic cancer has the aspect of systemic disease with a high incidence of recurrence in distant organs, whereas local control is necessary preoperatively because negative microscopic margins are important for prolonged survival. Several clinical trials are ongoing to investigate the significance of adding CRT to preoperative systemic chemotherapy. As previously mentioned, no clear benefit was found from adding hypofractionated radiotherapy to mFOLFIRINOX therapy as preoperative treatment for BR-PDAC in A021501.56 On the other hand, significance of adding conventional CRT to preoperative mFOLFIRINOX is presently examined in PANDAS-PRODIGE 44 phase II randomized clinical trial (NCT02676349). The results of the trial will provide important insights into preoperative radiation therapy for BR-PDAC.

Pancreatic cancer is conventionally known as a cold tumor as a target for immunotherapy.90 Then, immune activation at multiple levels of the cancer immune cycle may be required in tumors with low immunogenicity such as pancreatic cancer.91 It has recently been suggested that preoperative radiotherapy may enhance antitumor immunity via immunogenic cell death and may be useful in controlling subclinical PDAC micro-metastases. When tumor cells succumb to radiation therapy, they also emit a specific combination of signals that elicits tumor-specific cytotoxic T lymphocyte responses. Not a few clinical trials are investigating the combination of radiotherapy with immunotherapy for pancreas cancer, including perioperative therapy for R-PDAC or BR-PDAC (NCT02305186, NCT03161379, NCT01959672, and NCT02648282).

When considering the merits to patients and health economics, the appropriate target should be selected and appropriate treatment provided. Patients with R-PDAC include a certain number of patients with a low risk of recurrence who does not necessarily require multimodality treatment with preoperative therapy, and thus, it may be possible to omit preoperative therapy for such patients. Several biomarkers such as carbohydrate antigen 19-9 value92-94 and circulating tumor DNA95-97 are being investigated to determine whether they can select patients at high or low risk of recurrence.

BR-PDAC can be classified into BR-PV with the portal vein invasion only and BR-A with arterial invasion. Although some reports suggest that the BR-A PDAC prognosis is equivalent to that of BR-PV,67,98 BR-A PDAC is considered to have a significantly poorer prognosis than BR-PV PDAC.5,65,99 The median survival of patients with resected BR-A PDAC has been reported to be approximately 10.0 to 18.1 months,5,65,67,98,100 whereas the median survival for nonoperative treatment of UR-PDAC was 8.5 to 18.6 months.72,88,89,98,100 Thus, the BR-PV and BR-A prognoses are very different, and then, it may be necessary to determine and distinguish the treatment strategy (optimal regimen and number of courses) for each population. More intensive and prolonged treatment may be necessary for BR-A than BR-PV. A multicenter single-arm phase II trial101 was performed to evaluate the NAC-GAS clinical efficacy and safety (including GEM, nab-paclitaxel, and S-1) in 47 eligible patients with BR-A in 2021. The grade III/IV toxicity rates occurred in 30% of patients during the GAS regimen, whereas R0 resection was 86%, and primary endpoints of the 2-year OS rate and median OS duration were 70.1% and 41.0 months, respectively, indicating good efficacy.

While clinical trials for preoperative treatment of pancreatic cancer require large patient enrollment, the study results should be rapidly published. In addition, as represented by S-1, drugs with different efficacy and toxicity among different ethnic groups will be developed in the future, and thereby, a cooperative framework should be constructed in East Asia, where people have the similar genetic background to quickly and accurately evaluate the efficacy of preoperative treatment.

  1. Neoptolemos JP, Dunn JA, Stocken DD, et al. Adjuvant chemoradiotherapy and chemotherapy in resectable pancreatic cancer: a randomised controlled trial. Lancet 2001;358:1576-1585.
    Pubmed CrossRef
  2. Oettle H, Neuhaus P, Hochhaus A, et al. 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 CrossRef
  3. National Comprehensive Cancer Network (NCCN). NCCN guidelines: pancreatic adenocarcinoma [Internet]. Plymouth Meeting: NCCN; c2021 [cited 2022 Oct 30].
    Available from: https://www.nccn.org/guidelines/guidelines-detail?category=1&id=1455.
  4. Japan Pancreas Society. General rules for the study of pancreatic cancer. Tokyo: Kanehara & Co., Ltd., 2016.
  5. Murakami Y, Satoi S, Sho M, et al. National Comprehensive Cancer Network resectability status for pancreatic carcinoma predicts overall survival. World J Surg 2015;39:2306-2314.
    Pubmed CrossRef
  6. Kubo H, Ohgi K, Sugiura T, et al. The association between neoadjuvant therapy and pathological outcomes in pancreatic cancer patients after resection: prognostic significance of microscopic venous invasion. Ann Surg Oncol 2022;29:4992-5002.
    Pubmed CrossRef
  7. Russo S, Ammori J, Eads J, Dorth J. The role of neoadjuvant therapy in pancreatic cancer: a review. Future Oncol 2016;12:669-685.
    Pubmed KoreaMed CrossRef
  8. Neoptolemos JP, Stocken DD, Friess H, et al. A randomized trial of chemoradiotherapy and chemotherapy after resection of pancreatic cancer. N Engl J Med 2004;350:1200-1210.
    Pubmed CrossRef
  9. Oettle H, Post S, Neuhaus P, et al. Adjuvant chemotherapy with gemcitabine vs observation in patients undergoing curative-intent resection of pancreatic cancer: a randomized controlled trial. JAMA 2007;297:267-277.
    Pubmed CrossRef
  10. Ueno H, Kosuge T, Matsuyama Y, et al. A randomised phase III trial comparing gemcitabine with surgery-only in patients with resected pancreatic cancer: Japanese Study Group of Adjuvant Therapy for Pancreatic Cancer. Br J Cancer 2009;101:908-915.
    Pubmed KoreaMed CrossRef
  11. Neoptolemos JP, Stocken DD, Bassi C, et al. Adjuvant chemotherapy with fluorouracil plus folinic acid vs gemcitabine following pancreatic cancer resection: a randomized controlled trial. JAMA 2010;304:1073-1081.
    Pubmed CrossRef
  12. Uesaka K, Boku N, Fukutomi A, et al. Adjuvant chemotherapy of S-1 versus gemcitabine for resected pancreatic cancer: a phase 3, open-label, randomised, non-inferiority trial (JASPAC 01). Lancet 2016;388:248-257.
    Pubmed CrossRef
  13. Neoptolemos JP, Palmer DH, Ghaneh P, et al. 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 CrossRef
  14. Conroy T, Hammel P, Hebbar M, et al. FOLFIRINOX or gemcitabine as adjuvant therapy for pancreatic cancer. N Engl J Med 2018;379:2395-2406.
    Pubmed CrossRef
  15. Tempero MA, Reni M, Riess H, et al. Phase III, international, multicenter, randomized, open-label trial of adjuvant nab-paclitaxel plus gemcitabine (nab-P/G) vs gemcitabine (G) alone for surgically resected pancreatic adenocarcinoma (APACT): subgroup analyses. Ann Oncol 2019;30:v259-v260.
    CrossRef
  16. Kosuge T. Randomized phase III study of adjuvant chemotherapy with combination therapy of gemcitabine and S-1 vs. gemcitabine alone in patients with resected pancreatic cancer (JSAP-04) [Internet]. Tokyo: University Hospital Medical Information Network; c2010 [cited 2022 Oct 30].
    Available from: https://center6.umin.ac.jp/cgi-open-bin/ctr/ctr_view.cgi?recptno=R000005242.
  17. Hamada T, Yasunaga H, Nakai Y, et al. Interstitial lung disease associated with gemcitabine: a Japanese retrospective cohort study. Respirology 2016;21:338-343.
    Pubmed CrossRef
  18. Ioka T, Katayama K, Tanaka S, et al. Safety and effectiveness of gemcitabine in 855 patients with pancreatic cancer under Japanese clinical practice based on post-marketing surveillance in Japan. Jpn J Clin Oncol 2013;43:139-145.
    Pubmed CrossRef
  19. Müller S, Schütt P, Bojko P, et al. Hemolytic uremic syndrome following prolonged gemcitabine therapy: report of four cases from a single institution. Ann Hematol 2005;84:110-114.
    Pubmed CrossRef
  20. Fung MC, Storniolo AM, Nguyen B, Arning M, Brookfield W, Vigil J. A review of hemolytic uremic syndrome in patients treated with gemcitabine therapy. Cancer 1999;85:2023-2032.
    Pubmed CrossRef
  21. Abrams RA, Winter KA, Safran H, et al. Results of the NRG oncology/RTOG 0848 adjuvant chemotherapy question-erlotinib+gemcitabine for resected cancer of the pancreatic head: a phase II randomized clinical trial. Am J Clin Oncol 2020;43:173-179.
    Pubmed KoreaMed CrossRef
  22. Sinn M, Bahra M, Liersch T, et al. CONKO-005: adjuvant chemotherapy with gemcitabine plus erlotinib versus gemcitabine alone in patients after R0 resection of pancreatic cancer: a multicenter randomized phase III trial. J Clin Oncol 2017;35:3330-3337.
    Pubmed CrossRef
  23. van Groeningen CJ, Peters GJ, Schornagel JH, et al. Phase I clinical and pharmacokinetic study of oral S-1 in patients with advanced solid tumors. J Clin Oncol 2000;18:2772-2779.
    Pubmed CrossRef
  24. Chuah B, Goh BC, Lee SC, et al. Comparison of the pharmacokinetics and pharmacodynamics of S-1 between Caucasian and East Asian patients. Cancer Sci 2011;102:478-483.
    Pubmed CrossRef
  25. Ajani JA, Faust J, Ikeda K, et al. Phase I pharmacokinetic study of S-1 plus cisplatin in patients with advanced gastric carcinoma. J Clin Oncol 2005;23:6957-6965.
    Pubmed CrossRef
  26. Ajani JA, Rodriguez W, Bodoky G, et al. Multicenter phase III comparison of cisplatin/S-1 with cisplatin/infusional fluorouracil in advanced gastric or gastroesophageal adenocarcinoma study: the FLAGS trial. J Clin Oncol 2010;28:1547-1553.
    Pubmed CrossRef
  27. Smeenk HG, Incrocci L, Kazemier G, et al. Adjuvant 5-FU-based chemoradiotherapy for patients undergoing R-1/R-2 resections for pancreatic cancer. Dig Surg 2005;22:321-328.
    Pubmed CrossRef
  28. Hsu CC, Herman JM, Corsini MM, et al. Adjuvant chemoradiation for pancreatic adenocarcinoma: the Johns Hopkins Hospital-Mayo Clinic collaborative study. Ann Surg Oncol 2010;17:981-990.
    Pubmed KoreaMed CrossRef
  29. Merchant NB, Rymer J, Koehler EA, et al. Adjuvant chemoradiation therapy for pancreatic adenocarcinoma: who really benefits? J Am Coll Surg 2009;208:829-841.
    Pubmed KoreaMed CrossRef
  30. Corsini MM, Miller RC, Haddock MG, et al. Adjuvant radiotherapy and chemotherapy for pancreatic carcinoma: the Mayo Clinic experience (1975-2005). J Clin Oncol 2008;26:3511-3516.
    Pubmed CrossRef
  31. Gastrointestinal Tumor Study Group. Further evidence of effective adjuvant combined radiation and chemotherapy following curative resection of pancreatic cancer. Cancer 1987;59:2006-2010.
    Pubmed CrossRef
  32. The Gastrointestinal Tumor Study Group. A multi-institutional comparative trial of radiation therapy alone and in combination with 5-fluorouracil for locally unresectable pancreatic carcinoma. Ann Surg 1979;189:205-208.
    Pubmed KoreaMed CrossRef
  33. Yeo CJ, Abrams RA, Grochow LB, et al. Pancreaticoduodenectomy for pancreatic adenocarcinoma: postoperative adjuvant chemoradiation improves survival: a prospective, single-institution experience. Ann Surg 1997;225:621-636.
    Pubmed KoreaMed CrossRef
  34. You DD, Lee HG, Heo JS, Choi SH, Choi DW. Prognostic factors and adjuvant chemoradiation therapy after pancreaticoduodenectomy for pancreatic adenocarcinoma. J Gastrointest Surg 2009;13:1699-1706.
    Pubmed CrossRef
  35. Yang R, Cheung MC, Byrne MM, et al. Survival effects of adjuvant chemoradiotherapy after resection for pancreatic carcinoma. Arch Surg 2010;145:49-56.
    Pubmed CrossRef
  36. Van Laethem JL, Hammel P, Mornex F, et al. Adjuvant gemcitabine alone versus gemcitabine-based chemoradiotherapy after curative resection for pancreatic cancer: a randomized EORTC-40013-22012/FFCD-9203/GERCOR phase II study. J Clin Oncol 2010;28:4450-4456.
    Pubmed KoreaMed CrossRef
  37. Sainato A, Montrone S, Pasqualetti F, et al. Adjuvant chemoradiotherapy (gemcitabine-based) in pancreatic adenocarcinoma: the Pisa University experience. Tumori 2017;103:577-582.
    Pubmed CrossRef
  38. Abrams RA, Winter KA, Regine WF, et al. Failure to adhere to protocol specified radiation therapy guidelines was associated with decreased survival in RTOG 9704: a phase III trial of adjuvant chemotherapy and chemoradiotherapy for patients with resected adenocarcinoma of the pancreas. Int J Radiat Oncol Biol Phys 2012;82:809-816.
    Pubmed KoreaMed CrossRef
  39. Ozkok S, Demirci S, Yalman D, et al. Postoperative gemcitabine alone and concurrent with radiation therapy in locally advanced pancreatic carcinoma. Tumori 2010;96:560-567.
    Pubmed CrossRef
  40. Herman JM, Fan KY, Wild AT, et al. Phase 2 study of erlotinib combined with adjuvant chemoradiation and chemotherapy in patients with resectable pancreatic cancer. Int J Radiat Oncol Biol Phys 2013;86:678-685.
    Pubmed KoreaMed CrossRef
  41. Stocken DD, Büchler MW, Dervenis C, et al. Meta-analysis of randomised adjuvant therapy trials for pancreatic cancer. Br J Cancer 2005;92:1372-1381.
    Pubmed KoreaMed CrossRef
  42. Japan Pancreas Society. Clinical Practice Guidelines for Pancreatic Cancer 2019. Tokyo: Kanahara & Co, Ltd., 2019.
    CrossRef
  43. Bakkevold KE, Arnesjø B, Dahl O, Kambestad B. Adjuvant combination chemotherapy (AMF) following radical resection of carcinoma of the pancreas and papilla of Vater: results of a controlled, prospective, randomised multicentre study. Eur J Cancer 1993;29A:698-703.
    Pubmed CrossRef
  44. Morak MJ, van der Gaast A, Incrocci L, et al. Adjuvant intra-arterial chemotherapy and radiotherapy versus surgery alone in resectable pancreatic and periampullary cancer: a prospective randomized controlled trial. Ann Surg 2008;248:1031-1041.
    Pubmed CrossRef
  45. Klinkenbijl JH, Jeekel J, Sahmoud T, et al. Adjuvant radiotherapy and 5-fluorouracil after curative resection of cancer of the pancreas and periampullary region: phase III trial of the EORTC gastrointestinal tract cancer cooperative group. Ann Surg 1999;230:776-784.
    Pubmed KoreaMed CrossRef
  46. Kalser MH, Ellenberg SS. Pancreatic cancer: adjuvant combined radiation and chemotherapy following curative resection. Arch Surg 1985;120:899-903.
    Pubmed CrossRef
  47. Takadate T, Morikawa T, Ishida M, et al. Staging laparoscopy is mandatory for the treatment of pancreatic cancer to avoid missing radiologically negative metastases. Surg Today 2021;51:686-694.
    Pubmed CrossRef
  48. Jang JY, Han Y, Lee H, et al. Oncological benefits of neoadjuvant chemoradiation with gemcitabine versus upfront surgery in patients with borderline resectable pancreatic cancer: a prospective, randomized, open-label, multicenter phase 2/3 trial. Ann Surg 2018;268:215-222.
    Pubmed CrossRef
  49. Unno M, Motoi F, Matsuyama Y, et al. Randomized phase II/III trial of neoadjuvant chemotherapy with gemcitabine and S-1 versus upfront surgery for resectable pancreatic cancer (Prep-02/JSAP-05). J Clin Oncol 2019;37(4_suppl):189.
    Pubmed CrossRef
  50. Motoi F, Kosuge T, Ueno H, et al. Randomized phase II/III trial of neoadjuvant chemotherapy with gemcitabine and S-1 versus upfront surgery for resectable pancreatic cancer (Prep-02/JSAP05). Jpn J Clin Oncol 2019;49:190-194.
    Pubmed CrossRef
  51. Satoi S, Unno M, Motoi F, et al. The effect of neoadjuvant chemotherapy with gemcitabine and S-1 for resectable pancreatic cancer (randomized phase II/III trial; Prep-02/JSAP-05). J Clin Oncol 2019;37(15_suppl):4126.
    CrossRef
  52. Versteijne E, Suker M, Groothuis K, et al. Preoperative chemoradiotherapy versus immediate surgery for resectable and borderline resectable pancreatic cancer: results of the Dutch Randomized Phase III PREOPANC Trial. J Clin Oncol 2020;38:1763-1773.
    Pubmed KoreaMed CrossRef
  53. Versteijne E, van Dam JL, Suker M, 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.
    Pubmed CrossRef
  54. Takahashi S, Ohno I, Ikeda M, et al. Neoadjuvant S-1 with concurrent radiotherapy followed by surgery for borderline resectable pancreatic cancer: a phase II open-label multicenter prospective trial (JASPAC05). Ann Surg 2022;276:e510-e517.
    Pubmed CrossRef
  55. Yamaguchi J, Yokoyama Y, Fujii T, et al. Results of a phase II study on the use of neoadjuvant chemotherapy (FOLFIRINOX or GEM/nab-PTX) for borderline-resectable pancreatic cancer (NUPAT-01). Ann Surg 2022;275:1043-1049.
    Pubmed CrossRef
  56. Katz MH, Shi Q, Meyers J, et al. Efficacy of preoperative mFOLFIRINOX vs mFOLFIRINOX plus hypofractionated radiotherapy for borderline resectable adenocarcinoma of the pancreas: the A021501 phase 2 randomized clinical trial. JAMA Oncol 2022;8:1263-1270.
    Pubmed KoreaMed CrossRef
  57. Golcher H, Brunner TB, Witzigmann H, et al. Neoadjuvant chemoradiation therapy with gemcitabine/cisplatin and surgery versus immediate surgery in resectable pancreatic cancer: results of the first prospective randomized phase II trial. Strahlenther Onkol 2015;191:7-16.
    Pubmed KoreaMed CrossRef
  58. Casadei R, Di Marco M, Ricci C, et al. Neoadjuvant chemoradiotherapy and surgery versus surgery alone in resectable pancreatic cancer: a single-center prospective, randomized, controlled trial which failed to achieve accrual targets. J Gastrointest Surg 2015;19:1802-1812.
    Pubmed CrossRef
  59. Heinrich S, Pestalozzi B, Lesurtel M, et al. Adjuvant gemcitabine versus NEOadjuvant gemcitabine/oxaliplatin plus adjuvant gemcitabine in resectable pancreatic cancer: a randomized multicenter phase III study (NEOPAC Study). BMC Cancer 2011;11:346.
    Pubmed KoreaMed CrossRef
  60. Tachezy M, Gebauer F, Petersen C, et al. Sequential neoadjuvant chemoradiotherapy (CRT) followed by curative surgery vs. primary surgery alone for resectable, non-metastasized pancreatic adenocarcinoma: NEOPA: a randomized multicenter phase III study (NCT01900327, DRKS00003893, ISRCTN82191749). BMC Cancer 2014;14:411.
    Pubmed KoreaMed CrossRef
  61. Al-Batran SE, Reichart A, Bankstahl US, et al. Randomized multicenter phase II/III study with adjuvant gemcitabine versus neoadjuvant/adjuvant FOLFIRINOX in resectable pancreatic cancer: the NEPAFOX trial. J Clin Oncol 2021;39(3_suppl):406.
    CrossRef
  62. Hozaeel W, Pauligk C, Homann N, et al. Randomized multicenter phase II/III study with adjuvant gemcitabine versus neoadjuvant/adjuvant FOLFIRINOX in resectable pancreatic cancer: the NEPAFOX trial. J Clin Oncol 2015;33(15_suppl):TPS4152.
    CrossRef
  63. Ahmad SA, Duong M, Sohal DP, et al. 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.
    Pubmed KoreaMed CrossRef
  64. Sohal D, Duong MT, Ahmad SA, et al. SWOG S1505: results of perioperative chemotherapy (peri-op CTx) with mfolfirinox versus gemcitabine/nab-paclitaxel (Gem/nabP) for resectable pancreatic ductal adenocarcinoma (PDA). J Clin Oncol 2020;38(15_suppl):4504.
    CrossRef
  65. Kato H, Usui M, Isaji S, et al. Clinical features and treatment outcome of borderline resectable pancreatic head/body cancer: a multi-institutional survey by the Japanese Society of Pancreatic Surgery. J Hepatobiliary Pancreat Sci 2013;20:601-610.
    Pubmed CrossRef
  66. Fujii T, Yamada S, Murotani K, et al. Inverse probability of treatment weighting analysis of upfront surgery versus neoadjuvant chemoradiotherapy followed by surgery for pancreatic adenocarcinoma with arterial abutment. Medicine (Baltimore) 2015;94:e1647.
    Pubmed KoreaMed CrossRef
  67. Hirono S, Kawai M, Okada KI, et al. Treatment strategy for borderline resectable pancreatic cancer with radiographic artery involvement. Pancreas 2016;45:1438-1446.
    Pubmed CrossRef
  68. Ielpo B, Caruso R, Duran H, et al. A comparative study of neoadjuvant treatment with gemcitabine plus nab-paclitaxel versus surgery first for pancreatic adenocarcinoma. Surg Oncol 2017;26:402-410.
    Pubmed CrossRef
  69. Murakami Y, Uemura K, Sudo T, et al. Survival impact of neoadjuvant gemcitabine plus S-1 chemotherapy for patients with borderline resectable pancreatic carcinoma with arterial contact. Cancer Chemother Pharmacol 2017;79:37-47.
    Pubmed CrossRef
  70. Nagakawa Y, Sahara Y, Hosokawa Y, et al. Clinical impact of neoadjuvant chemotherapy and chemoradiotherapy in borderline resectable pancreatic cancer: analysis of 884 patients at facilities specializing in pancreatic surgery. Ann Surg Oncol 2019;26:1629-1636.
    Pubmed CrossRef
  71. Inoue Y, Saiura A, Oba A, et al. Neoadjuvant gemcitabine and nab-paclitaxel for borderline resectable pancreatic cancers: intention-to-treat analysis compared with upfront surgery. J Hepatobiliary Pancreat Sci 2021;28:143-155.
    Pubmed CrossRef
  72. Michelakos T, Pergolini I, Castillo CF, 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.
    Pubmed CrossRef
  73. Labori KJ, Lassen K, Hoem D, et al. Neoadjuvant chemotherapy versus surgery first for resectable pancreatic cancer (Norwegian Pancreatic Cancer Trial-1 (NorPACT-1)): study protocol for a national multicentre randomized controlled trial. BMC Surg 2017;17:94.
    Pubmed KoreaMed CrossRef
  74. Schwarz L, Vernerey D, Bachet JB, et al. 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 KoreaMed CrossRef
  75. Ferrone CR. Testing the use of the usual chemotherapy before and after surgery for removable pancreatic cancer. Identifier No. NCT04340141
    Available from: https://clinicaltrials.gov/ct2/show/NCT04340141.
  76. Koerkamp BG. Perioperative or Adjuvant mFOLFIRINOX for Resectable Pancreatic Cancer (PREOPANC-3). Identifier No. NCT04927780
    Available from: https://www.clinicaltrials.gov/ct2/show/NCT04927780.
  77. Janssen QP, van Dam JL, Bonsing BA, et al. Total neoadjuvant FOLFIRINOX versus neoadjuvant gemcitabine-based chemoradiotherapy and adjuvant gemcitabine for resectable and borderline resectable pancreatic cancer (PREOPANC-2 Trial): study protocol for a nationwide multicenter randomized controlled trial. BMC Cancer 2021;21:300.
    Pubmed KoreaMed CrossRef
  78. Grose DB, McKay CJ, Cooke S, et al. PRIMUS-002: a multicentre, open-label, phase II study examining FOLFOX and nab-paclitaxel (FA) and nab-paclitaxel and gemcitabine (AG) as neoadjuvant therapy for (borderline) resectable pancreatic cancer (PC), focusing on biomarker and liquid biopsy development. J Clin Oncol 2019;37(15_suppl):TPS4166.
    CrossRef
  79. Unno M. Phase I/II study of gemcitabine and S-1 (GS therapy) as neoadjuvant chemoradiationtherapy for borderline resectable pancreatic cancer (Prep-03/NS014-1) [Internet]. Tokyo: University Hospital Medical Information Network; c2014 [cited 2022 Oct 30].
    Available from: https://center6.umin.ac.jp/cgi-open-bin/ctr/ctr.cgi?function=brows&action=brows&recptno=R000016075&type=summary&language=J.
  80. Okada KI, Shimokawa T, Hirono S, et al. Effect of neoadjuvant nab-paclitaxel plus gemcitabine therapy on overall survival in patients with borderline resectable pancreatic cancer: a prospective multicenter phase II trial (NAC-GA Trial). Oncology 2017;93:343-346.
    Pubmed CrossRef
  81. Takahashi S. Randomized phase II/III study of gemcitabine and nab-paclitaxel therapy versus S-1 and concurrent radiotherapy as neoadjuvant treatment for Borderline resectable pancreatic cancer (GABARNANCE Trial) [Internet]. Wako: National Institute of Public Health; c2019 [cited 2022 Oct 30].
    Available from: https://jrct.niph.go.jp/en-latest-detail/jRCTs031180319.
  82. Sanford M. S-1 (TeysunoⓇ): a review of its use in advanced gastric cancer in non-Asian populations. Drugs 2013;73:845-855.
    Pubmed CrossRef
  83. Franck C, Canbay A, Malfertheiner P, Venerito M. Neoadjuvant treatment with cisplatin and S-1 in elderly patients with oesophagogastric adenocarcinoma and locoregional disease: two case reports and review of the literature. Mol Clin Oncol 2017;7:1069-1072.
    Pubmed KoreaMed CrossRef
  84. Lenz HJ, Lee FC, Haller DG, et al. Extended safety and efficacy data on S-1 plus cisplatin in patients with untreated, advanced gastric carcinoma in a multicenter phase II study. Cancer 2007;109:33-40.
    Pubmed CrossRef
  85. Schultheis B, Strumberg D, Bergmann L, et al. Results of a phase II trial of S-1 as first-line treatment of metastatic pancreatic cancer (CESAR-study group). Invest New Drugs 2012;30:1184-1192.
    Pubmed CrossRef
  86. Winther SB, Bjerregaard JK, Schonnemann KR, et al. S-1 (Teysuno) and gemcitabine in Caucasian patients with unresectable pancreatic adenocarcinoma. Cancer Chemother Pharmacol 2018;81:573-578.
    Pubmed CrossRef
  87. Cui J, Jiao F, Li Q, et al. Chinese Society of Clinical Oncology (CSCO): clinical guidelines for the diagnosis and treatment of pancreatic cancer. J Natl Cancer Cent 2022;2:205-215.
    CrossRef
  88. Conroy T, Desseigne F, Ychou M, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med 2011;364:1817-1825.
    Pubmed CrossRef
  89. Von Hoff DD, Ervin T, Arena FP, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med 2013;369:1691-1703.
    Pubmed KoreaMed CrossRef
  90. Teng MW, Ngiow SF, Ribas A, Smyth MJ. Classifying cancers based on T-cell infiltration and PD-L1. Cancer Res 2015;75:2139-2145.
    Pubmed KoreaMed CrossRef
  91. Gajiwala S, Torgeson A, Garrido-Laguna I, Kinsey C, Lloyd S. Combination immunotherapy and radiation therapy strategies for pancreatic cancer-targeting multiple steps in the cancer immunity cycle. J Gastrointest Oncol 2018;9:1014-1026.
    Pubmed KoreaMed CrossRef
  92. Bockhorn M, Uzunoglu FG, Adham M, et al. Borderline resectable pancreatic cancer: a consensus statement by the International Study Group of Pancreatic Surgery (ISGPS). Surgery 2014;155:977-988.
    Pubmed CrossRef
  93. Tjensvoll K, Nordgård O, Smaaland R. Circulating tumor cells in pancreatic cancer patients: methods of detection and clinical implications. Int J Cancer 2014;134:1-8.
    Pubmed CrossRef
  94. Chen J, Wang H, Zhou L, Liu Z, Tan X. A combination of circulating tumor cells and CA199 improves the diagnosis of pancreatic cancer. J Clin Lab Anal 2022;36:e24341.
    Pubmed KoreaMed CrossRef
  95. Isaji S, Mizuno S, Windsor JA, et al. International consensus on definition and criteria of borderline resectable pancreatic ductal adenocarcinoma 2017. Pancreatology 2018;18:2-11.
    Pubmed CrossRef
  96. Macgregor-Das A, Yu J, Tamura K, et al. Detection of circulating tumor DNA in patients with pancreatic cancer using digital next-generation sequencing. J Mol Diagn 2020;22:748-756.
    Pubmed KoreaMed CrossRef
  97. Affolter KE, Hellwig S, Nix DA, et al. Detection of circulating tumor DNA without a tumor-informed search using next-generation sequencing is a prognostic biomarker in pancreatic ductal adenocarcinoma. Neoplasia 2021;23:859-869.
    Pubmed KoreaMed CrossRef
  98. Yamada S, Fujii T, Takami H, et al. Evaluation and proposal of novel resectability criteria for pancreatic cancer established by the Japan Pancreas Society. Surgery 2017;162:784-791.
    Pubmed CrossRef
  99. Fujii T, Satoi S, Yamada S, et al. Clinical benefits of neoadjuvant chemoradiotherapy for adenocarcinoma of the pancreatic head: an observational study using inverse probability of treatment weighting. J Gastroenterol 2017;52:81-93.
    Pubmed CrossRef
  100. Yoshida K, Iwashita T, Uemura S, et al. A multicenter prospective phase II study of first-line modified FOLFIRINOX for unresectable advanced pancreatic cancer. Oncotarget 2017;8:111346-111355.
    Pubmed KoreaMed CrossRef
  101. Kondo N, Uemura K, Sudo T, et al. A phase II study of gemcitabine/nab-paclitaxel/S-1 combination neoadjuvant chemotherapy for patients with borderline resectable pancreatic cancer with arterial contact. Eur J Cancer 2021;159:215-223.
    Pubmed CrossRef

Article

Review Article

Gut and Liver 2023; 17(5): 698-710

Published online September 15, 2023 https://doi.org/10.5009/gnl220311

Copyright © Gut and Liver.

Neoadjuvant and Adjuvant Treatments for Resectable and Borderline Resectable Pancreatic Ductal Adenocarcinoma: The Current Status of Pancreatic Ductal Adenocarcinoma Treatment in Japan

Shigenori Ei , Shinichiro Takahashi , Toshihito Ogasawara , Taro Mashiko , Yoshihito Masuoka , Toshio Nakagohri

Department of Gastroenterological Surgery, Tokai University School of Medicine, Isehara, Japan

Correspondence to:Shinichiro Takahashi
ORCID https://orcid.org/0000-0002-4828-3965
E-mail shtakaha@tsc.u-tokai.ac.jp

Received: July 18, 2022; Revised: October 30, 2022; Accepted: October 30, 2022

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Resection is the only curative treatment for pancreatic ductal adenocarcinoma (PDAC). Although the outcome of technically resectable PDAC has improved with advances in surgery and adjuvant therapy, the 5-year survival rate remains low at 20% to 40%. More effective therapy is needed. Almost 15 years ago, the National Comprehensive Cancer Network guidelines proposed a resectability classification of PDAC based on preoperative imaging. Since then, treatment strategies for PDAC have been devised based on resectability. The standard of care for resectable PDAC is adjuvant chemotherapy after R0 resection, as shown by the results of pivotal clinical trials. With regard to neoadjuvant treatment, several recent clinical trials comparing neoadjuvant treatment with upfront resection have been conducted on resectable PDAC and borderline resectable PDAC, and the benefits and efficacy of neoadjuvant treatment for pancreatic cancer has become clearer. The significance of neoadjuvant treatment for resectable PDAC remains controversial, but in borderline resectable PDAC the efficacy of neoadjuvant treatment has been further recognised, although the standard of care has not yet been established. Several promising clinical trials for PDAC are ongoing. This review presents previous and ongoing trials of perioperative treatment for resectable and borderline resectable PDAC, focusing on the difference between Asian and Western countries.

Keywords: Pancreatic neoplasms, Neoadjuvant therapy, Adjuvant chemotherapy, Chemoradiotherapy, Clinical trial

INTRODUCTION

Pancreatic ductal adenocarcinoma (PDAC) frequently recurs after resection even if radical resection is performed. Perioperative treatment such as neoadjuvant or adjuvant therapy is essential to improve the prognosis of PDAC. The 5-year survival rate after pancreatic resection without any perioperative treatment for PDAC is approximately 10%,1,2 whereas that with adjuvant therapy has improved to 20% to 40% in the last two decades. This study reviewed previous pivotal studies of perioperative treatment for PDAC to reconfirm the evolution of the standard treatment to date and also highlighted the interesting ongoing trials to discuss future development trends of perioperative treatment for PDAC.

When planning treatment strategies for PDAC, the resectability status is critically important alongside the staging classification (TNM classification) to determine the possibility of curative resection, prognosis, and tumor progression patterns. The National Comprehensive Cancer Network guidelines3 have classified resectability since 2006, and the Japanese Clinical Practice Guidelines for Pancreatic Cancer (7th edition)4 also defines resectability classification (Table 1). Both classifications define the resectability status as resectable (R), borderline resectable (BR), or unresectable (UR). Either R-PDAC or BR-PDAC is technically resectable; however, the tumor stage including local extension and lymph node metastasis is more advanced in BR-PDAC than that in R-PDAC. The risk of local and distant BR-PDAC progression is higher than that of R-PDAC.5-7 Thus, perioperative treatment strategies for BR-PDAC and R-PDAC should be different. Therefore, this review separately examined the perioperative treatment development for R-PDAC and BR-PDAC.

Table 1 . Criteria Defining Resectability Status in Japan Pancreas Society Classification 7th Edition (2016).

Resectable: R
  • SMV/PV: no tumor contact or contact of <180° without occlusion.

  • SMA, CA, CHA: no tumor contact/invasion.

Borderline resectable: BR (subclassified according to SMV/PV invasion alone or arterial invasion)
BR-PV (SMV/PV invasion alone)
  • SMV/PV: tumor contact/invasion of ≥180°/occlusion, not exceeding the inferior border of the duodenum.

  • SMA, CA, CHA: no tumor contact/invasion.

BR-A (arterial invasion)
  • SMA, CA: tumor contact/invasion of <180° without deformity or narrowing.

  • CHA: tumor contact/invasion without showing tumor contact/invasion of the PHA and/or CA (cases of contact/invasion of both PV and peripancreatic arteries are considered BR-A).

Unresectable: UR (subclassified according to the presence or absence of distant metastases)
UR-LA (locally advanced)
  • SMV/PV: tumor contact/invasion of ≥180°/occlusion, exceeding the inferior border of the duodenum.

  • SMA, CA: tumor contact/invasion of ≥180°.

  • CHA: tumor contact/invasion showing tumor contact/invasion of the PHA and/or CA.

  • AO: tumor contact or invasion.

UR-M (metastasis)
  • Distant metastasis including non-regional lymph node metastasis..

SMV, superior mesenteric vein; PV, portal vein; SMA, superior mesenteric artery; CA, celiac artery; CHA, common hepatic artery; PHA, proper hepatic artery; AO, aorta..



This review also focuses on the uniqueness of perioperative treatment development in Asia, including Japan, and compares it with that in Europe and United States.

ADJUVANT THERAPY FOR R-PDAC

1. Establishment of adjuvant gemcitabine therapy

Randomized controlled trials (RCTs) comparing postoperative adjuvant chemotherapy with resection alone for PDAC have been conducted since the 1990s; however, the development of effective treatments had not yet been established until the ESPAC-1 trial8 in 2004, which demonstrated a significant survival benefit of postoperative 5-fluorouracil+leucovorin (5-FU+LV) therapy over resection alone, and results of this trial had made postoperative 5-FU+LV therapy the standard of care for R-PDAC in Europe. However, the complex study design using a 2×2 factorial design was controversial and had not been implemented as the standard therapy in the United States and Japan.

Subsequently, the CONKO-001 trial2,9 was conducted mainly in Germany, in which 354 patients after PDAC resection were randomized to the adjuvant chemotherapy group with gemcitabine (GEM) and resection-alone group, showing a significant prolongation of recurrence-free survival with the adjuvant chemotherapy group (13.4 months vs 6.9 months, p<0.001). Initially, adjuvant chemotherapy did not show a significant increase in the overall survival (OS) (p=0.06); however, current long-term follow-up studies reported a significant increase not only in recurrence-free survival but also in the median OS (22.8 months vs 20.2 months, p=0.01).2

At about the same time, the JSAP-02 trial10 was conducted in Japan, which randomized 118 patients after PDAC resection to adjuvant chemotherapy with GEM and resection alone, and reported a significantly prolonged recurrence-free survival with postoperative GEM, a finding similar to the initial report of the CONKO-001 trial. As a result, GEM therapy was considered one of the standard treatments in addition to 5-FU+LV therapy.

European Study Group for Pancreatic Cancer (ESPAC) subsequently conducted the ESPAC-3 trial,11 in which 1,088 patients with resected PDAC were randomly allocated to the postoperative GEM or 5-FU+LV treatment groups, and reported no significant difference in the OS between the two groups (23.6 months vs 23.0 months: hazard ratio [HR], 0.94; 95% confidential interval [CI], 0.81 to 1.08; p=0.39). However, serious adverse events were significantly lower in the GEM group than in the 5-FU+LV group, and due to its safety profile, GEM therapy was preferentially selected as the standard of care not only in Japan but also worldwide. Subsequently, postoperative GEM became the standard regimen for the control group in many phase III trials for R-PDAC (Table 2).2,8-16

Table 2 . Pivotal Trial on Adjuvant Chemotherapy for Pancreatic Cancer after Radical Resection.

TrialYearAdjuvant treatmentNo. of patientsPrimary outcomeMedianHR (95% CI)p-value
ESPAC-1820045-FU+LV treated arm in 2×2 factorial design1472-yr survival rate40%0.71 (0.55–0.92)<0.01
5-FU+LV non-treated arm in 2×2 factorial design14230%
CONKO-0012,92007GEM186DFS13.4 mo0.55 (0.44–0.69)<0.01
Surgery alone1826.7 mo
JSAP-02102009GEM58OS22.3 mo0.77 (0.51–1.14)0.19
Surgery alone6018.4 mo
ESPAC-3112010GEM478OS23.6 mo0.94 (0.81–1.08)0.39
5-FU+LV48623.0 mo
JASPAC-01122013S-1187OS46.5 mo0.57 (0.44–0.72)<0.01
GEM19025.5 mo
ESPAC-4132016GEM+capecitabine364OS28.0 mo0.82 (0.68–0.98)0.03
GEM36625.5 mo
PRODIGE24/
ACCORD2414
2018mFOLFIRINOX247DFS21.6 mo0.58 (0.46–0.73)<0.01
GEM24612.8 mo
APACT15
(ASCO)
2019GEM+nab-PTX432DFS19.4 mo0.88 (0.73–1.06)0.18
GEM43418.8 mo
JSAP-0416On goingGEM+S-1-OS/DFS---
GEM--

HR, hazard ratio; CI, confidence interval; 5-FU+LV, 5-fluorouracil+leucovorin; GEM, gemcitabine; DFS, disease-free survival; OS, overall survival; mFOLFIRINOX, modified FOLFIRINOX; nab-PTX, nab-paclitaxel..



The use of 5-FU+LV therapy was recommended for patients who could not be safely administered with GEM, such as those with a history of interstitial pneumonia17,18 or hemolytic uremic syndrome19,20 in the Japanese Clinical Practice Guidelines for Pancreatic Cancer.

2. Development of postoperative adjuvant therapy superior to adjuvant GEM

In Europe, ESPAC conducted the ESPAC-4 trial13 involving 730 patients who underwent PDAC resection randomly allocated to the postoperative GEM and GEM+capecitabine (Cape) groups. The results showed that the OS was more significantly improved in the postoperative GEM+Cape group than in the GEM group (28.0 months vs 25.5 months: HR, 0.82; 95% CI, 0.68 to 0.98; p=0.032), and the study results led to the replacement of GEM therapy with GEM+Cape therapy as the standard postoperative adjuvant treatment.

Studies in France and Canada reported the results of a multicenter RCT (PRODIGE 24-ACCORD 24/CCTG PA 6)14 comparing postoperative modified FOLFIRINOX (mFOLFIRINOX) with GEM alone. This study used mFOLFIRINOX therapy considering the toxicity of FOLFIRINOX therapy, in which rapid administration of 5-FU was omitted and the irinotecan dose was reduced. The median disease-free survival, the primary endpoint, was 21.6 and 12.8 months in the mFOLFIRINOX and GEM-alone groups, respectively, showing the superiority in the mFOLFIRINOX group (HR, 0.58; 95% CI, 0.46 to 0.73; p<0.0001). The mFOLFIRINOX group also showed a significantly prolonged OS (54.4 months vs 35.0 months: HR, 0.64; 95% CI, 0.48 to 0.86; p=0.003). Grade 3/4 adverse events occurred in 75.9% and 52.9% of the mFOLFIRINOX and GEM-alone groups, respectively, and the treatment completion rates were 66.4% and 79.0% for mFOLFIRINOX and GEM alone, respectively (p=0.002). Based on the study results, postoperative mFOLFIRINOX therapy has become one of the standard treatments for resected PDAC. Because of its high toxicity, postoperative mFOLFIRINOX therapy is recommended for patients with good performance status (PS).

GEM plus erlotinib, an epidermal growth factor receptor tyrosine kinase inhibitor (CONKO-005 trial, RTOG-0848 trial),21,22 and GEM+nab-paclitaxel (GnP), one of the standard treatments for UR-PDAC (APACT trial),15 were also compared with adjuvant GEM in the global phase III trial. However, none of these therapies were recommended as the standard treatment for PDAC due to a lack of statistically significant efficacy by comparing with adjuvant GEM.

3. Different trends in Japan: postoperative S-1 therapy

The Japan Adjuvant Study Group of Pancreatic Cancer (JASPAC) conducted a phase III comparative study (JASPAC 01)12 to examine the non-inferiority of S-1 to GEM in adjuvant chemotherapy after the PDAC resection. The study enrolled 385 patients and showed not only non-inferiority of the S-1 group to the GEM group in the primary endpoint of OS, but also demonstrated superiority (median OS S-1 vs GEM, 46.5 months vs 25.5 months: HR, 0.57; 95% CI, 0.44 to 0.72; p<0.0001). The pre-completion discontinuation rate of the adjuvant treatment was 42% and 28% in the GEM and S-1 groups (p=0.0050), respectively, mainly due to adverse events (25% and 21%, respectively) and recurrence (13% and 5%, respectively). The study results clearly changed the standard postoperative adjuvant therapy in Japan from GEM therapy to S-1 therapy due to its high safety and convenience.

However, S-1 is not widely used for treatment of the pancreatic cancer including adjuvant therapy in Europe and the United States. The major reason is that the maximum tolerated dose of S-1 for Caucasians is lower than that for Mongolians due to gastrointestinal toxicity,23 mainly diarrhea, making it difficult to reproduce in the West the favorable antitumor effects of chemotherapy with S-1 reported in Asian clinical trials. The conversion efficiency of tegafur to 5-FU is higher in Caucasians due to polymorphisms in CYP2A6, resulting in higher gastrointestinal toxicity, especially diarrhea than that in Mongolians.24,25 In Europe, S-1 is approved for the treatment of advanced gastric cancer in combination with cisplatin,26 but the dose of S-1 was approved at about 60% of the dose under the original Japanese regimen. Additionally, S-1 is not approved for pancreatic cancer in Europe and United States. Due to the corporate strategy, development to expand the indication of S-1 for pancreatic cancer is not currently underway in the United States or Europe. Thus, S-1 has not become the standard of care for R-PDAC in Western countries.

Later, ESPAC-4 and PRODIGE24/ACCORD24 considered GEM+Cape and mFOLFIRINOX as the standard of care for R-PDAC in Western countries. In Japan, neither GEM+Cape nor mFOLFIRINOX became the standard of care mainly because the HR of the S-1 to GEM in the JASPAC-01 study (HR=0.57) was better than that of GEM+Cape to GEM in ESPAC-4 (HR=0.82) or that of mFOLFIRINOX to GEM in the PRODIGE24/ACCORD24 trial (HR=0.64). Then, adjuvant S-1 has been the standard postoperative adjuvant therapy to date.

4. Adjuvant chemoradiotherapy

A large number of case-control studies have suggested that adjuvant chemoradiotherapy (CRT) may prolong the prognosis;27-35 postoperative CRT analyses with novel anticancer agents including GEM and erlotinib are still insufficient;36-40 and meta-analyses have reported the benefits for patients with R1 resection compared to R0 (p=0.04).41 Postoperative adjuvant CRT has been one of the promising treatment modalities in the perioperative treatment of pancreatic cancer and remains a topic for further investigation.

The 2019 Clinical Practice Guidelines for Pancreatic Cancer committee42 conducted a meta-analysis of six RCTs comparing fluorouracil-based or GEM-based chemoradiation to surgery alone8,36,43-46 to evaluate the efficacy of adjuvant CRT. Therefore, the risk ratio of adjuvant CRT compared to surgery alone was 1.01 (95% CI, 0.93 to 1.10). The benefit of adjuvant CRT on the prognosis is unclear, and then adjuvant CRT is not recommended based on the Japanese guidelines at present.

NEOADJUVANT THERAPY

1. Neoadjuvant therapy for R-PDAC

Although the aforementioned adjuvant chemotherapies are the current standard of care for patients with R-PDAC, 30% to 50% of patients who undergo laparotomy are unable to receive adjuvant therapy due to intraoperatively detected metastases, early postoperative recurrence, surgical complications, or poor PS. Conversely, neoadjuvant treatment has several theoretical advantages over adjuvant treatment, such as early treatment for micrometastasis under better physical condition, determination of tumor response for the treatment, and selection of patients with poor prognosis.47 Then, several clinical trials have been conducted to develop preoperative treatments not only for BR-PDAC but also for R-PDAC (Table 3).48-56

Table 3 . Prospective Study on Neoadjuvant Chemotherapy for Pancreatic Cancer before Radical Resection.

Author/trialYearResectabilityTreatmentNo. of patientsPrimary outcomeMedianHR (95% CI)p-value
Jang et al.482018BRCRT (54 Gy, GEM) → surgery → GEM272-yr survival40.7%0.51 (0.27–0.93)0.028
Surgery → GEM2326.1%
Unno et al. Prep-02/JSAP0549-512019R/BR-PVGEM + S-1 → surgery → S-1182OS36.7 mo0.72 (0.55–0.94)0.02
Surgery → S-118026.6 mo
Versteijne et al. PREOPANC-152,532020R/BRCRT (36 Gy, GEM) → surgery → GEM119OS15.7 mo0.73 (0.56–0.96)0.025
Surgery → GEM12714.3 mo
Takahashi et al. JASPAC05542020R/BRCRT (54 Gy, S-1) → surgery52R0 (OS)52% (30.8 mo)--
Yamaguchi et al. NUPAT-01552022BRmFOLFIRINOX → surgery26R073.1%-0.202
GEM + nab-PTX → surgery2556.0%
Katz et al. A021501562022BRmFOLFIRINOX → surgery → FOLFOX66518-mo survival66.7%--
mFOLFIRINOX → SBRT (33-40 Gy) or HIGRT (25 Gy) → surgery → FOLFOX65547.3%

HR, hazard ratio; CI, confidence interval; BR, Borderline Resectable; CRT, chemoradiotherapy; GEM, gemcitabine; R, resectable; PV, portal vein; OS, overall survival; mFOLFIRINOX, modified FOLFIRINOX; nab-PTX, nab-paclitaxel; SBRT, stereotactic body radiation therapy; HIGRT, hypofractionated image-guided radiation therapy..



In the 2010s, several clinical studies of preoperative treatment for R-PDAC were initiated in Europe. In 2015, randomized phase II trials of GEM+cisplatin+radiotherapy57 and GEM+radiotherapy58 were conducted; however, those trials could not be fully evaluated due to poor patient accrual. Evidence of preoperative treatment in R-PDAC is still lacking;59-62 however, important results were recently reported in two phase III trials comparing preoperative treatment for upfront resection in patients with R-PDAC or BR-PDAC.

A randomized multicenter phase III study (PREOPANC-1) was conducted in the Netherlands to examine the significance of neoadjuvant chemoradiation therapy (NACRT) for R-PDAC and BR-PDAC. The NACRT group (36 Gy/15 F + GEM 3 cycles → surgery → postoperative GEM for 4 cycles) showed a better tendency in the OS than the surgery first group. Although the difference was not significant in the initial report (modified OS 16.0 months vs 14.3 months: HR, 0.78; 95% CI, 0.58 to 1.05; p=0.096),52 long-term follow-up analysis showed significantly better OS in the NACRT group (modified OS 15.7 months vs 14.3 months: HR, 0.73; 95% CI, 0.56 to 0.96; p=0.025).53

A randomized phase III trial (Prep-02/JSAP05 trial)50 was performed in Japan to investigate the clinical significance of neoadjuvant chemotherapy (NAC) with GEM+S-1 (GS) for T1-T3 PDAC without invading the peripancreatic major arteries (hepatic artery, celiac artery, and superior mesenteric artery). This trial confirmed that NAC-GS safely and effectively improved the prognosis compared with upfront surgery (modified OS 36.7 months vs 26.6 months: HR, 0.72; 95% CI, 0.55 to 0.94; p=0.015). Furthermore, subsequent resection could be safely performed without significant differences in the operative time, intraoperative blood loss, and postoperative complication rates.49,51 Due to the following several limitations, (1) inclusion of 20% of BR-PDAC, (2) only tumors that could be biopsied and pathologically confirmed, and (3) age limit of 20 to 79 years, the study results just fell short by making neoadjuvant GS as the standard of care for R-PDAC. However, subgroup analysis in R-PDAC consisted of 80% of the enrolled patients revealed similar efficacy of the neoadjuvant GS. Then, using the results of the Prep-02/JSAP05 trial, the Japanese Clinical Practice Guidelines for Pancreatic Cancer were revised, and neoadjuvant GS therapy was recommended for R-PDAC.

The current most effective regimens, mFOLFIRINOX and GnP, have also being tried as preoperative treatments for R-PDAC. The randomized phase II SWOG S1505 compared perioperative mFOLFIRINOX and perioperative GnP in patients with R-PDAC.63,64 The primary outcome was 2-year survival. Investigators planned to compare the two arms to determine the better treatment if the 2-year survival reached 58% or more. However, the 2-year OSs of 41.6% with mFOLFIRINOX and 48.8% with GnP missed the target rate of 58% at 2 years. Furthermore, neither regimen showed a statistical improvement in 2-year survival over historical control, and the results were negative. However, these regimens remain the most promising at present, and the results of the ongoing phase III trial of perioperative mFOLFIRINOX therapy are awaited.

2. Neoadjuvant therapy for BR-PDAC

Compared to R-PDAC, BR-PDAC is more locally progressing and is at a higher tumor stage, with a higher risk of positive microscopic margins and early recurrence. Therefore, the significance of preoperative treatment in BR-PDAC is clearly greater than that in R-PDAC.

In BR-PDAC, several retrospective studies65-72 comparing neoadjuvant therapy with upfront surgery reported that neoadjuvant therapy improved the R0 resection rate and survival. Afterward, the efficacy of neoadjuvant therapy for BR-PDAC was evidently demonstrated in the phase II/III study reported from South Korea in 2018.48 A total of 58 patients were randomized to neoadjuvant chemoradiation (54 Gy) with GEM or upfront resection. The intention-to-treat analysis demonstrated that the primary endpoint of the 2-year survival was significantly better in the NACRT group (40.7%) than that in the upfront surgery group (26.1%) (HR, 1.97; 95% CI, 1.07 to 3.62; p=0.028).

Although the efficacy of neoadjuvant treatment was verified, the standard treatment has not yet been established in BR-PDAC. Several promising clinical trials of the neoadjuvant treatment are ongoing in this field (Table 4).73-81

Table 4 . Prospective Ongoing Study on Neoadjuvant Chemotherapy for Pancreatic Cancer before Radical Resection.

Author/trialPhaseStart yearResectabilityTreatmentScheduled number of patientsPrimary outcome
Labori et al. NorPACT-173II/III2017RmFOLFIRINOX → surgery → GEM + capecitabine140OS
Surgery → GEM + capecitabine
Schwarz et al. PANACHE01-PRODIGE4874II2017RmFOLFIRINOX → surgery → adjuvant therapy160OS
FOLFOX → surgery → adjuvant therapy
Surgery → adjuvant therapy
Ferrone Alliance A02180675III2020RmFOLFIRINOX → surgery →mFOLFIRINOX352OS
Surgery →mFOLFIRINOX
Koerkamp PREOPANC-376III2021RmFOLFIRINOX → surgery →mFOLFIRINOX378OS
Surgery → mFOLFIRINOX
Janssen et al. PREOPANC-277III2018R/BRmFOLFIRINOX → surgery368OS
GEM → CRT (36 Gy, GEM) → GEM → surgery → GEM
Grose et al. PRIMUS-00278II2019R/BRFOLFOX + nab-PTX → CRT → surgery278PFS
GEM + nab-PTX → CRT → surgery
Unno Prep-03/NS014-179I/II2014BRGEM + S-1 + concurrent RT (50 Gy) → surgery12–24/40R0
Okada et al. NAC-GA80II2017BRGEM + nab-PTX → surgery60OS
Takahashi GABARNANCE81II/III2017BRGEM + nab-PTX → surgery110R0/OS
CRT (54 Gy, S-1) → surgery

R, resectable; mFOLFIRINOX, modified FOLFIRINOX; GEM, gemcitabine; OS, overall survival; BR, borderline resectable; CRT, chemoradiotherapy; nab-PTX, nab-paclitaxel; RT, radiotherapy; PFS, progression-free survival..



In clinical practice of the United States or Europe, mFOLFIRINOX alone or mFOLFIRINOX followed by CRT or radiation therapy are the most common preoperative treatment for BR-PDAC. The A021501 phase II randomized clinical trial,56 which was conducted by the Alliance for Clinical Trials in Oncology, compared those promising treatments, namely, eight cycles of mFOLFIRINOX and seven cycles of mFOLFIRINOX followed by stereotactic body radiotherapy at 33 to 40 Gy in five fractions or hypofractionated image-guided radiotherapy at 25 Gy in five fractions as preoperative treatment for BR-PDAC. Each treatment arm’s 18-month OS rate was compared with a historical control rate of 50%. In the interim analysis, mFOLFIRINOX with hypofractionated radiotherapy arm was closed because R0 resection rate (33%) was proven to be under the threshold value (40%). The 18-month OS rate of chemotherapy arm was 66.7% and that of chemotherapy plus radiation arm was 47.3%. The results of the trial suggest that mFOLFIRINOX could be a reference neoadjuvant treatment regimen for BR-PDAC. However, the role of radiotherapy in the preoperative treatment for BR-PDAC remains undefined because hypofractionated radiotherapy instead of a more common conventional CRT was applied for chemotherapy plus radiation arm in the trial. Further studies are needed to clarify the significance of preoperative radiation therapy in BR-PDAC.

PRACTICE AND DEVELOPMENT OF PERIOPERATIVE TREATMENT FOR PDAC IN JAPAN

The perioperative treatment in Japan is unique due to the following two reasons. First, several perioperative treatments using S-1 have been developed in Japan and showed good results. Second, the toxic regimen tolerability, such as FOLFIRINOX in the Japanese population, might not be equivalent to that of the Western population.

As previously mentioned, adjuvant S-1 and neoadjuvant GS are recommended for R-PDAC in Japanese guidelines for the treatment of PDAC (Fig. 1A). S-1, oral fluorouracil, is a key medication for gastrointestinal malignancies (the stomach, colorectal, biliary tract, and PDAC)24,82,83 and is approved in many Asian countries. In Europe, S-1 is also approved by the European Medicines Agency and has been used for the treatment of advanced gastric cancer combined with cisplatin.26,84 However, European people tend to have more severe gastrointestinal side effects of S-1, such as diarrhea. S-1 is more toxic to the digestive tract in Caucasians than in Mongoloids due to metabolic and pharmacological differences. Genetic polymorphism differences, such as the higher activity of CYP2A6, the enzyme that converts tegafur in S-1 to 5-FU, in Caucasians, may be the cause of different toxicity profiles.25,82,85,86 The aforementioned perioperative treatments using S-1 are not recommended as the standard of care in Europe and the United States.

Figure 1. The strategy of PDAC treatment in Japan. (A) Current treatment strategy for R-PDAC in Japan. Perioperative treatment for R-PDAC is two cycles of neoadjuvant chemotherapy-GS (GEM=1,000 mg/m2, days 1 and 8; S-1=80 mg/m2, days 1–14) based on the results of the Prep-02/JSAP05 study and postoperative S-1 (S-1=80 mg/m2, days 1–28) in four cycles based on the results of the JASPAC-01 study. (B) Current treatment strategy for BR-PDAC in Japan. For preoperative treatment for BR-PDAC, GS (GEM=1,000 mg/m2, days 1 and 8; S-1=80 mg/m2, days 1–14) may be administered for two cycles based on the results of the Prep-02/JSAP05 study. However, several clinical trials have sought a stronger and more effective regimen, and no fixed strategy has been established yet. For postoperative treatment, S-1 (S-1=80 mg/m2, days 1–28) is administered for four cycles based on the results of the JASPAC-01 study.
PDAC, pancreatic ductal adenocarcinoma; R-PDCA, resectable PDAC; GEM, gemcitabine; GS, GEM+S-1; BR-PDAC, borderline resectable PDAC; mFOLFIRINOX, modified FOLFIRINOX; nab-PTX, nab-paclitaxel; CRT, chemoradiotherapy.

Conversely, postoperative therapy with mFOLFIRINOX has become the standard of care for R-PDAC in Europe and the United States, and FOLFIRINOX as preoperative treatment is also being developed. The postoperative toxicity of FOLFIRINOX is severe, especially in elderly people and in patients with poor PS, which increases the question of tolerability. Because of the high proportion of elderly patients in Japan, toxicity profiles are particularly important. When considering the efficacy and toxicity, both postoperative therapy with S-1 and preoperative GS therapy with S-1 balance between toxicity and efficacy seems suitable for Japanese patients with R-PDAC. Also, in East Asia with its large Mongoloid population, S-1 might be a key drug for R-PDAC.

S-1 is currently approved for the treatment of pancreatic cancer with locally advanced or metastatic disease in East Asian countries such as South Korea, Singapore, Taiwan, Thailand, and Malaysia. Moreover, adjuvant S-1 is recommended for R-PDAC as one of the treatments of grade I recommendations according to the clinical guidelines for the diagnosis and treatment of pancreatic cancer by the Chinese Society of Clinical Oncology.87 Several clinical trials using S-1 in the perioperative treatment of pancreatic cancer are currently being conducted or planned in China (NCT03278015, NCT03777462) and Taiwan (NCT04070313). However, at this time, it has not been verified whether perioperative chemotherapy with S-1 shows the same excellent efficacy and safety in East Asian countries other than China as observed in the Japanese clinical trial. Then, the results of the future clinical trials from East Asia other than Japan are expected to elucidate significance of perioperative treatment using S-1 for pancreatic cancer in East Asia.

Meanwhile, perioperative treatment with FOLFIRINOX, GnP, or chemoradiation is being developed in Japan for BR-PDAC, requiring more intensive therapy, and for younger patients who are more tolerable to toxicity (Table 4, Fig. 1B). The Prep-03/NS014-1 trial79 is a phase I/II study evaluating the efficacy of a GS+concurrent radiotherapy (50 Gy) regimen as NACRT. The NAC-GA trial80 is a phase II trial using GnP as NAC. Results of NUPAT-01 trial,55 a phase II trial of mFOLFIRINOX and GnP as NAC for BR-PDAC with a primary endpoint of R0 resection rate, were reported, showing that both groups had good R0 resection rates (73.1% vs 56.0%, respectively, p=0.202) and good 3-year survival rates (55.3% and 54.4%, respectively, p=0.389), demonstrating the NAC feasibility and tolerability with FOLFIRINOX or GnP. Moreover, the GABARNANCE trial,81 comparing the GnP therapy with S-1-based CRT (54 Gy) with the R0 resection rate and OS as primary endpoints, is awaiting results.

FUTURE PROSPECTS

Currently, not a few clinical trials to evaluate two effective regimens, FOLFIRINOX88 and GnP89 which are standard therapies for UR-PDAC, as neoadjuvant therapy for R-/BR-PDAC are conducted and attracted attention (Table 4). The PREOPANC-1 and Prep-02/JSAP05 trials reported favorable outcomes for the preoperative treatment in R-PDAC; however, both of them were subgroup analyses and have not been established as evidence. Therefore, at present, neoadjuvant therapy is not yet recommended for R-PDAC in Western guidelines, except in high-risk patients.3 The major ongoing phase III trials for R-PDAC include Alliance A021806 in the United States75 and PREOPANC-3 in the Netherlands.76 These are trials to verify the significance of preoperative treatment, using mFOLFIRINOX for preoperative treatment and postoperative adjuvant therapy. The design of the two trials is quite similar: only resectable pancreatic cancer is included, eight courses of mFOLFIRINOX are performed preoperatively, followed by pancreatectomy and four courses of mFOLFIRINOX postoperatively, and the primary endpoint is OS. The results of the two large phase III trials examining the significance of preoperative treatment with mFOLFIRINOX, the most promising perioperative treatment regimen, will provide important insights into the efficacy of preoperative treatment.

The necessity for radiation therapy in the preoperative treatment of R- or BR-PDAC is an important issue. At present, preoperative chemoradiation remains controversial because pancreatic cancer has the aspect of systemic disease with a high incidence of recurrence in distant organs, whereas local control is necessary preoperatively because negative microscopic margins are important for prolonged survival. Several clinical trials are ongoing to investigate the significance of adding CRT to preoperative systemic chemotherapy. As previously mentioned, no clear benefit was found from adding hypofractionated radiotherapy to mFOLFIRINOX therapy as preoperative treatment for BR-PDAC in A021501.56 On the other hand, significance of adding conventional CRT to preoperative mFOLFIRINOX is presently examined in PANDAS-PRODIGE 44 phase II randomized clinical trial (NCT02676349). The results of the trial will provide important insights into preoperative radiation therapy for BR-PDAC.

Pancreatic cancer is conventionally known as a cold tumor as a target for immunotherapy.90 Then, immune activation at multiple levels of the cancer immune cycle may be required in tumors with low immunogenicity such as pancreatic cancer.91 It has recently been suggested that preoperative radiotherapy may enhance antitumor immunity via immunogenic cell death and may be useful in controlling subclinical PDAC micro-metastases. When tumor cells succumb to radiation therapy, they also emit a specific combination of signals that elicits tumor-specific cytotoxic T lymphocyte responses. Not a few clinical trials are investigating the combination of radiotherapy with immunotherapy for pancreas cancer, including perioperative therapy for R-PDAC or BR-PDAC (NCT02305186, NCT03161379, NCT01959672, and NCT02648282).

When considering the merits to patients and health economics, the appropriate target should be selected and appropriate treatment provided. Patients with R-PDAC include a certain number of patients with a low risk of recurrence who does not necessarily require multimodality treatment with preoperative therapy, and thus, it may be possible to omit preoperative therapy for such patients. Several biomarkers such as carbohydrate antigen 19-9 value92-94 and circulating tumor DNA95-97 are being investigated to determine whether they can select patients at high or low risk of recurrence.

BR-PDAC can be classified into BR-PV with the portal vein invasion only and BR-A with arterial invasion. Although some reports suggest that the BR-A PDAC prognosis is equivalent to that of BR-PV,67,98 BR-A PDAC is considered to have a significantly poorer prognosis than BR-PV PDAC.5,65,99 The median survival of patients with resected BR-A PDAC has been reported to be approximately 10.0 to 18.1 months,5,65,67,98,100 whereas the median survival for nonoperative treatment of UR-PDAC was 8.5 to 18.6 months.72,88,89,98,100 Thus, the BR-PV and BR-A prognoses are very different, and then, it may be necessary to determine and distinguish the treatment strategy (optimal regimen and number of courses) for each population. More intensive and prolonged treatment may be necessary for BR-A than BR-PV. A multicenter single-arm phase II trial101 was performed to evaluate the NAC-GAS clinical efficacy and safety (including GEM, nab-paclitaxel, and S-1) in 47 eligible patients with BR-A in 2021. The grade III/IV toxicity rates occurred in 30% of patients during the GAS regimen, whereas R0 resection was 86%, and primary endpoints of the 2-year OS rate and median OS duration were 70.1% and 41.0 months, respectively, indicating good efficacy.

While clinical trials for preoperative treatment of pancreatic cancer require large patient enrollment, the study results should be rapidly published. In addition, as represented by S-1, drugs with different efficacy and toxicity among different ethnic groups will be developed in the future, and thereby, a cooperative framework should be constructed in East Asia, where people have the similar genetic background to quickly and accurately evaluate the efficacy of preoperative treatment.

CONFLICTS OF INTEREST

No potential conflict of interest relevant to this article was reported.

Fig 1.

Figure 1.The strategy of PDAC treatment in Japan. (A) Current treatment strategy for R-PDAC in Japan. Perioperative treatment for R-PDAC is two cycles of neoadjuvant chemotherapy-GS (GEM=1,000 mg/m2, days 1 and 8; S-1=80 mg/m2, days 1–14) based on the results of the Prep-02/JSAP05 study and postoperative S-1 (S-1=80 mg/m2, days 1–28) in four cycles based on the results of the JASPAC-01 study. (B) Current treatment strategy for BR-PDAC in Japan. For preoperative treatment for BR-PDAC, GS (GEM=1,000 mg/m2, days 1 and 8; S-1=80 mg/m2, days 1–14) may be administered for two cycles based on the results of the Prep-02/JSAP05 study. However, several clinical trials have sought a stronger and more effective regimen, and no fixed strategy has been established yet. For postoperative treatment, S-1 (S-1=80 mg/m2, days 1–28) is administered for four cycles based on the results of the JASPAC-01 study.
PDAC, pancreatic ductal adenocarcinoma; R-PDCA, resectable PDAC; GEM, gemcitabine; GS, GEM+S-1; BR-PDAC, borderline resectable PDAC; mFOLFIRINOX, modified FOLFIRINOX; nab-PTX, nab-paclitaxel; CRT, chemoradiotherapy.
Gut and Liver 2023; 17: 698-710https://doi.org/10.5009/gnl220311

Table 1 Criteria Defining Resectability Status in Japan Pancreas Society Classification 7th Edition (2016)

Resectable: R

SMV/PV: no tumor contact or contact of <180° without occlusion

SMA, CA, CHA: no tumor contact/invasion

Borderline resectable: BR (subclassified according to SMV/PV invasion alone or arterial invasion)
BR-PV (SMV/PV invasion alone)

SMV/PV: tumor contact/invasion of ≥180°/occlusion, not exceeding the inferior border of the duodenum

SMA, CA, CHA: no tumor contact/invasion

BR-A (arterial invasion)

SMA, CA: tumor contact/invasion of <180° without deformity or narrowing

CHA: tumor contact/invasion without showing tumor contact/invasion of the PHA and/or CA (cases of contact/invasion of both PV and peripancreatic arteries are considered BR-A)

Unresectable: UR (subclassified according to the presence or absence of distant metastases)
UR-LA (locally advanced)

SMV/PV: tumor contact/invasion of ≥180°/occlusion, exceeding the inferior border of the duodenum

SMA, CA: tumor contact/invasion of ≥180°

CHA: tumor contact/invasion showing tumor contact/invasion of the PHA and/or CA

AO: tumor contact or invasion

UR-M (metastasis)

Distant metastasis including non-regional lymph node metastasis.

SMV, superior mesenteric vein; PV, portal vein; SMA, superior mesenteric artery; CA, celiac artery; CHA, common hepatic artery; PHA, proper hepatic artery; AO, aorta.


Table 2 Pivotal Trial on Adjuvant Chemotherapy for Pancreatic Cancer after Radical Resection

TrialYearAdjuvant treatmentNo. of patientsPrimary outcomeMedianHR (95% CI)p-value
ESPAC-1820045-FU+LV treated arm in 2×2 factorial design1472-yr survival rate40%0.71 (0.55–0.92)<0.01
5-FU+LV non-treated arm in 2×2 factorial design14230%
CONKO-0012,92007GEM186DFS13.4 mo0.55 (0.44–0.69)<0.01
Surgery alone1826.7 mo
JSAP-02102009GEM58OS22.3 mo0.77 (0.51–1.14)0.19
Surgery alone6018.4 mo
ESPAC-3112010GEM478OS23.6 mo0.94 (0.81–1.08)0.39
5-FU+LV48623.0 mo
JASPAC-01122013S-1187OS46.5 mo0.57 (0.44–0.72)<0.01
GEM19025.5 mo
ESPAC-4132016GEM+capecitabine364OS28.0 mo0.82 (0.68–0.98)0.03
GEM36625.5 mo
PRODIGE24/
ACCORD2414
2018mFOLFIRINOX247DFS21.6 mo0.58 (0.46–0.73)<0.01
GEM24612.8 mo
APACT15
(ASCO)
2019GEM+nab-PTX432DFS19.4 mo0.88 (0.73–1.06)0.18
GEM43418.8 mo
JSAP-0416On goingGEM+S-1-OS/DFS---
GEM--

HR, hazard ratio; CI, confidence interval; 5-FU+LV, 5-fluorouracil+leucovorin; GEM, gemcitabine; DFS, disease-free survival; OS, overall survival; mFOLFIRINOX, modified FOLFIRINOX; nab-PTX, nab-paclitaxel.


Table 3 Prospective Study on Neoadjuvant Chemotherapy for Pancreatic Cancer before Radical Resection

Author/trialYearResectabilityTreatmentNo. of patientsPrimary outcomeMedianHR (95% CI)p-value
Jang et al.482018BRCRT (54 Gy, GEM) → surgery → GEM272-yr survival40.7%0.51 (0.27–0.93)0.028
Surgery → GEM2326.1%
Unno et al. Prep-02/JSAP0549-512019R/BR-PVGEM + S-1 → surgery → S-1182OS36.7 mo0.72 (0.55–0.94)0.02
Surgery → S-118026.6 mo
Versteijne et al. PREOPANC-152,532020R/BRCRT (36 Gy, GEM) → surgery → GEM119OS15.7 mo0.73 (0.56–0.96)0.025
Surgery → GEM12714.3 mo
Takahashi et al. JASPAC05542020R/BRCRT (54 Gy, S-1) → surgery52R0 (OS)52% (30.8 mo)--
Yamaguchi et al. NUPAT-01552022BRmFOLFIRINOX → surgery26R073.1%-0.202
GEM + nab-PTX → surgery2556.0%
Katz et al. A021501562022BRmFOLFIRINOX → surgery → FOLFOX66518-mo survival66.7%--
mFOLFIRINOX → SBRT (33-40 Gy) or HIGRT (25 Gy) → surgery → FOLFOX65547.3%

HR, hazard ratio; CI, confidence interval; BR, Borderline Resectable; CRT, chemoradiotherapy; GEM, gemcitabine; R, resectable; PV, portal vein; OS, overall survival; mFOLFIRINOX, modified FOLFIRINOX; nab-PTX, nab-paclitaxel; SBRT, stereotactic body radiation therapy; HIGRT, hypofractionated image-guided radiation therapy.


Table 4 Prospective Ongoing Study on Neoadjuvant Chemotherapy for Pancreatic Cancer before Radical Resection

Author/trialPhaseStart yearResectabilityTreatmentScheduled number of patientsPrimary outcome
Labori et al. NorPACT-173II/III2017RmFOLFIRINOX → surgery → GEM + capecitabine140OS
Surgery → GEM + capecitabine
Schwarz et al. PANACHE01-PRODIGE4874II2017RmFOLFIRINOX → surgery → adjuvant therapy160OS
FOLFOX → surgery → adjuvant therapy
Surgery → adjuvant therapy
Ferrone Alliance A02180675III2020RmFOLFIRINOX → surgery →mFOLFIRINOX352OS
Surgery →mFOLFIRINOX
Koerkamp PREOPANC-376III2021RmFOLFIRINOX → surgery →mFOLFIRINOX378OS
Surgery → mFOLFIRINOX
Janssen et al. PREOPANC-277III2018R/BRmFOLFIRINOX → surgery368OS
GEM → CRT (36 Gy, GEM) → GEM → surgery → GEM
Grose et al. PRIMUS-00278II2019R/BRFOLFOX + nab-PTX → CRT → surgery278PFS
GEM + nab-PTX → CRT → surgery
Unno Prep-03/NS014-179I/II2014BRGEM + S-1 + concurrent RT (50 Gy) → surgery12–24/40R0
Okada et al. NAC-GA80II2017BRGEM + nab-PTX → surgery60OS
Takahashi GABARNANCE81II/III2017BRGEM + nab-PTX → surgery110R0/OS
CRT (54 Gy, S-1) → surgery

R, resectable; mFOLFIRINOX, modified FOLFIRINOX; GEM, gemcitabine; OS, overall survival; BR, borderline resectable; CRT, chemoradiotherapy; nab-PTX, nab-paclitaxel; RT, radiotherapy; PFS, progression-free survival.


References

  1. Neoptolemos JP, Dunn JA, Stocken DD, et al. Adjuvant chemoradiotherapy and chemotherapy in resectable pancreatic cancer: a randomised controlled trial. Lancet 2001;358:1576-1585.
    Pubmed CrossRef
  2. Oettle H, Neuhaus P, Hochhaus A, et al. 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 CrossRef
  3. National Comprehensive Cancer Network (NCCN). NCCN guidelines: pancreatic adenocarcinoma [Internet]. Plymouth Meeting: NCCN; c2021 [cited 2022 Oct 30]. Available from: https://www.nccn.org/guidelines/guidelines-detail?category=1&id=1455.
  4. Japan Pancreas Society. General rules for the study of pancreatic cancer. Tokyo: Kanehara & Co., Ltd., 2016.
  5. Murakami Y, Satoi S, Sho M, et al. National Comprehensive Cancer Network resectability status for pancreatic carcinoma predicts overall survival. World J Surg 2015;39:2306-2314.
    Pubmed CrossRef
  6. Kubo H, Ohgi K, Sugiura T, et al. The association between neoadjuvant therapy and pathological outcomes in pancreatic cancer patients after resection: prognostic significance of microscopic venous invasion. Ann Surg Oncol 2022;29:4992-5002.
    Pubmed CrossRef
  7. Russo S, Ammori J, Eads J, Dorth J. The role of neoadjuvant therapy in pancreatic cancer: a review. Future Oncol 2016;12:669-685.
    Pubmed KoreaMed CrossRef
  8. Neoptolemos JP, Stocken DD, Friess H, et al. A randomized trial of chemoradiotherapy and chemotherapy after resection of pancreatic cancer. N Engl J Med 2004;350:1200-1210.
    Pubmed CrossRef
  9. Oettle H, Post S, Neuhaus P, et al. Adjuvant chemotherapy with gemcitabine vs observation in patients undergoing curative-intent resection of pancreatic cancer: a randomized controlled trial. JAMA 2007;297:267-277.
    Pubmed CrossRef
  10. Ueno H, Kosuge T, Matsuyama Y, et al. A randomised phase III trial comparing gemcitabine with surgery-only in patients with resected pancreatic cancer: Japanese Study Group of Adjuvant Therapy for Pancreatic Cancer. Br J Cancer 2009;101:908-915.
    Pubmed KoreaMed CrossRef
  11. Neoptolemos JP, Stocken DD, Bassi C, et al. Adjuvant chemotherapy with fluorouracil plus folinic acid vs gemcitabine following pancreatic cancer resection: a randomized controlled trial. JAMA 2010;304:1073-1081.
    Pubmed CrossRef
  12. Uesaka K, Boku N, Fukutomi A, et al. Adjuvant chemotherapy of S-1 versus gemcitabine for resected pancreatic cancer: a phase 3, open-label, randomised, non-inferiority trial (JASPAC 01). Lancet 2016;388:248-257.
    Pubmed CrossRef
  13. Neoptolemos JP, Palmer DH, Ghaneh P, et al. 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 CrossRef
  14. Conroy T, Hammel P, Hebbar M, et al. FOLFIRINOX or gemcitabine as adjuvant therapy for pancreatic cancer. N Engl J Med 2018;379:2395-2406.
    Pubmed CrossRef
  15. Tempero MA, Reni M, Riess H, et al. Phase III, international, multicenter, randomized, open-label trial of adjuvant nab-paclitaxel plus gemcitabine (nab-P/G) vs gemcitabine (G) alone for surgically resected pancreatic adenocarcinoma (APACT): subgroup analyses. Ann Oncol 2019;30:v259-v260.
    CrossRef
  16. Kosuge T. Randomized phase III study of adjuvant chemotherapy with combination therapy of gemcitabine and S-1 vs. gemcitabine alone in patients with resected pancreatic cancer (JSAP-04) [Internet]. Tokyo: University Hospital Medical Information Network; c2010 [cited 2022 Oct 30]. Available from: https://center6.umin.ac.jp/cgi-open-bin/ctr/ctr_view.cgi?recptno=R000005242.
  17. Hamada T, Yasunaga H, Nakai Y, et al. Interstitial lung disease associated with gemcitabine: a Japanese retrospective cohort study. Respirology 2016;21:338-343.
    Pubmed CrossRef
  18. Ioka T, Katayama K, Tanaka S, et al. Safety and effectiveness of gemcitabine in 855 patients with pancreatic cancer under Japanese clinical practice based on post-marketing surveillance in Japan. Jpn J Clin Oncol 2013;43:139-145.
    Pubmed CrossRef
  19. Müller S, Schütt P, Bojko P, et al. Hemolytic uremic syndrome following prolonged gemcitabine therapy: report of four cases from a single institution. Ann Hematol 2005;84:110-114.
    Pubmed CrossRef
  20. Fung MC, Storniolo AM, Nguyen B, Arning M, Brookfield W, Vigil J. A review of hemolytic uremic syndrome in patients treated with gemcitabine therapy. Cancer 1999;85:2023-2032.
    Pubmed CrossRef
  21. Abrams RA, Winter KA, Safran H, et al. Results of the NRG oncology/RTOG 0848 adjuvant chemotherapy question-erlotinib+gemcitabine for resected cancer of the pancreatic head: a phase II randomized clinical trial. Am J Clin Oncol 2020;43:173-179.
    Pubmed KoreaMed CrossRef
  22. Sinn M, Bahra M, Liersch T, et al. CONKO-005: adjuvant chemotherapy with gemcitabine plus erlotinib versus gemcitabine alone in patients after R0 resection of pancreatic cancer: a multicenter randomized phase III trial. J Clin Oncol 2017;35:3330-3337.
    Pubmed CrossRef
  23. van Groeningen CJ, Peters GJ, Schornagel JH, et al. Phase I clinical and pharmacokinetic study of oral S-1 in patients with advanced solid tumors. J Clin Oncol 2000;18:2772-2779.
    Pubmed CrossRef
  24. Chuah B, Goh BC, Lee SC, et al. Comparison of the pharmacokinetics and pharmacodynamics of S-1 between Caucasian and East Asian patients. Cancer Sci 2011;102:478-483.
    Pubmed CrossRef
  25. Ajani JA, Faust J, Ikeda K, et al. Phase I pharmacokinetic study of S-1 plus cisplatin in patients with advanced gastric carcinoma. J Clin Oncol 2005;23:6957-6965.
    Pubmed CrossRef
  26. Ajani JA, Rodriguez W, Bodoky G, et al. Multicenter phase III comparison of cisplatin/S-1 with cisplatin/infusional fluorouracil in advanced gastric or gastroesophageal adenocarcinoma study: the FLAGS trial. J Clin Oncol 2010;28:1547-1553.
    Pubmed CrossRef
  27. Smeenk HG, Incrocci L, Kazemier G, et al. Adjuvant 5-FU-based chemoradiotherapy for patients undergoing R-1/R-2 resections for pancreatic cancer. Dig Surg 2005;22:321-328.
    Pubmed CrossRef
  28. Hsu CC, Herman JM, Corsini MM, et al. Adjuvant chemoradiation for pancreatic adenocarcinoma: the Johns Hopkins Hospital-Mayo Clinic collaborative study. Ann Surg Oncol 2010;17:981-990.
    Pubmed KoreaMed CrossRef
  29. Merchant NB, Rymer J, Koehler EA, et al. Adjuvant chemoradiation therapy for pancreatic adenocarcinoma: who really benefits? J Am Coll Surg 2009;208:829-841.
    Pubmed KoreaMed CrossRef
  30. Corsini MM, Miller RC, Haddock MG, et al. Adjuvant radiotherapy and chemotherapy for pancreatic carcinoma: the Mayo Clinic experience (1975-2005). J Clin Oncol 2008;26:3511-3516.
    Pubmed CrossRef
  31. Gastrointestinal Tumor Study Group. Further evidence of effective adjuvant combined radiation and chemotherapy following curative resection of pancreatic cancer. Cancer 1987;59:2006-2010.
    Pubmed CrossRef
  32. The Gastrointestinal Tumor Study Group. A multi-institutional comparative trial of radiation therapy alone and in combination with 5-fluorouracil for locally unresectable pancreatic carcinoma. Ann Surg 1979;189:205-208.
    Pubmed KoreaMed CrossRef
  33. Yeo CJ, Abrams RA, Grochow LB, et al. Pancreaticoduodenectomy for pancreatic adenocarcinoma: postoperative adjuvant chemoradiation improves survival: a prospective, single-institution experience. Ann Surg 1997;225:621-636.
    Pubmed KoreaMed CrossRef
  34. You DD, Lee HG, Heo JS, Choi SH, Choi DW. Prognostic factors and adjuvant chemoradiation therapy after pancreaticoduodenectomy for pancreatic adenocarcinoma. J Gastrointest Surg 2009;13:1699-1706.
    Pubmed CrossRef
  35. Yang R, Cheung MC, Byrne MM, et al. Survival effects of adjuvant chemoradiotherapy after resection for pancreatic carcinoma. Arch Surg 2010;145:49-56.
    Pubmed CrossRef
  36. Van Laethem JL, Hammel P, Mornex F, et al. Adjuvant gemcitabine alone versus gemcitabine-based chemoradiotherapy after curative resection for pancreatic cancer: a randomized EORTC-40013-22012/FFCD-9203/GERCOR phase II study. J Clin Oncol 2010;28:4450-4456.
    Pubmed KoreaMed CrossRef
  37. Sainato A, Montrone S, Pasqualetti F, et al. Adjuvant chemoradiotherapy (gemcitabine-based) in pancreatic adenocarcinoma: the Pisa University experience. Tumori 2017;103:577-582.
    Pubmed CrossRef
  38. Abrams RA, Winter KA, Regine WF, et al. Failure to adhere to protocol specified radiation therapy guidelines was associated with decreased survival in RTOG 9704: a phase III trial of adjuvant chemotherapy and chemoradiotherapy for patients with resected adenocarcinoma of the pancreas. Int J Radiat Oncol Biol Phys 2012;82:809-816.
    Pubmed KoreaMed CrossRef
  39. Ozkok S, Demirci S, Yalman D, et al. Postoperative gemcitabine alone and concurrent with radiation therapy in locally advanced pancreatic carcinoma. Tumori 2010;96:560-567.
    Pubmed CrossRef
  40. Herman JM, Fan KY, Wild AT, et al. Phase 2 study of erlotinib combined with adjuvant chemoradiation and chemotherapy in patients with resectable pancreatic cancer. Int J Radiat Oncol Biol Phys 2013;86:678-685.
    Pubmed KoreaMed CrossRef
  41. Stocken DD, Büchler MW, Dervenis C, et al. Meta-analysis of randomised adjuvant therapy trials for pancreatic cancer. Br J Cancer 2005;92:1372-1381.
    Pubmed KoreaMed CrossRef
  42. Japan Pancreas Society. Clinical Practice Guidelines for Pancreatic Cancer 2019. Tokyo: Kanahara & Co, Ltd., 2019.
    CrossRef
  43. Bakkevold KE, Arnesjø B, Dahl O, Kambestad B. Adjuvant combination chemotherapy (AMF) following radical resection of carcinoma of the pancreas and papilla of Vater: results of a controlled, prospective, randomised multicentre study. Eur J Cancer 1993;29A:698-703.
    Pubmed CrossRef
  44. Morak MJ, van der Gaast A, Incrocci L, et al. Adjuvant intra-arterial chemotherapy and radiotherapy versus surgery alone in resectable pancreatic and periampullary cancer: a prospective randomized controlled trial. Ann Surg 2008;248:1031-1041.
    Pubmed CrossRef
  45. Klinkenbijl JH, Jeekel J, Sahmoud T, et al. Adjuvant radiotherapy and 5-fluorouracil after curative resection of cancer of the pancreas and periampullary region: phase III trial of the EORTC gastrointestinal tract cancer cooperative group. Ann Surg 1999;230:776-784.
    Pubmed KoreaMed CrossRef
  46. Kalser MH, Ellenberg SS. Pancreatic cancer: adjuvant combined radiation and chemotherapy following curative resection. Arch Surg 1985;120:899-903.
    Pubmed CrossRef
  47. Takadate T, Morikawa T, Ishida M, et al. Staging laparoscopy is mandatory for the treatment of pancreatic cancer to avoid missing radiologically negative metastases. Surg Today 2021;51:686-694.
    Pubmed CrossRef
  48. Jang JY, Han Y, Lee H, et al. Oncological benefits of neoadjuvant chemoradiation with gemcitabine versus upfront surgery in patients with borderline resectable pancreatic cancer: a prospective, randomized, open-label, multicenter phase 2/3 trial. Ann Surg 2018;268:215-222.
    Pubmed CrossRef
  49. Unno M, Motoi F, Matsuyama Y, et al. Randomized phase II/III trial of neoadjuvant chemotherapy with gemcitabine and S-1 versus upfront surgery for resectable pancreatic cancer (Prep-02/JSAP-05). J Clin Oncol 2019;37(4_suppl):189.
    Pubmed CrossRef
  50. Motoi F, Kosuge T, Ueno H, et al. Randomized phase II/III trial of neoadjuvant chemotherapy with gemcitabine and S-1 versus upfront surgery for resectable pancreatic cancer (Prep-02/JSAP05). Jpn J Clin Oncol 2019;49:190-194.
    Pubmed CrossRef
  51. Satoi S, Unno M, Motoi F, et al. The effect of neoadjuvant chemotherapy with gemcitabine and S-1 for resectable pancreatic cancer (randomized phase II/III trial; Prep-02/JSAP-05). J Clin Oncol 2019;37(15_suppl):4126.
    CrossRef
  52. Versteijne E, Suker M, Groothuis K, et al. Preoperative chemoradiotherapy versus immediate surgery for resectable and borderline resectable pancreatic cancer: results of the Dutch Randomized Phase III PREOPANC Trial. J Clin Oncol 2020;38:1763-1773.
    Pubmed KoreaMed CrossRef
  53. Versteijne E, van Dam JL, Suker M, 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.
    Pubmed CrossRef
  54. Takahashi S, Ohno I, Ikeda M, et al. Neoadjuvant S-1 with concurrent radiotherapy followed by surgery for borderline resectable pancreatic cancer: a phase II open-label multicenter prospective trial (JASPAC05). Ann Surg 2022;276:e510-e517.
    Pubmed CrossRef
  55. Yamaguchi J, Yokoyama Y, Fujii T, et al. Results of a phase II study on the use of neoadjuvant chemotherapy (FOLFIRINOX or GEM/nab-PTX) for borderline-resectable pancreatic cancer (NUPAT-01). Ann Surg 2022;275:1043-1049.
    Pubmed CrossRef
  56. Katz MH, Shi Q, Meyers J, et al. Efficacy of preoperative mFOLFIRINOX vs mFOLFIRINOX plus hypofractionated radiotherapy for borderline resectable adenocarcinoma of the pancreas: the A021501 phase 2 randomized clinical trial. JAMA Oncol 2022;8:1263-1270.
    Pubmed KoreaMed CrossRef
  57. Golcher H, Brunner TB, Witzigmann H, et al. Neoadjuvant chemoradiation therapy with gemcitabine/cisplatin and surgery versus immediate surgery in resectable pancreatic cancer: results of the first prospective randomized phase II trial. Strahlenther Onkol 2015;191:7-16.
    Pubmed KoreaMed CrossRef
  58. Casadei R, Di Marco M, Ricci C, et al. Neoadjuvant chemoradiotherapy and surgery versus surgery alone in resectable pancreatic cancer: a single-center prospective, randomized, controlled trial which failed to achieve accrual targets. J Gastrointest Surg 2015;19:1802-1812.
    Pubmed CrossRef
  59. Heinrich S, Pestalozzi B, Lesurtel M, et al. Adjuvant gemcitabine versus NEOadjuvant gemcitabine/oxaliplatin plus adjuvant gemcitabine in resectable pancreatic cancer: a randomized multicenter phase III study (NEOPAC Study). BMC Cancer 2011;11:346.
    Pubmed KoreaMed CrossRef
  60. Tachezy M, Gebauer F, Petersen C, et al. Sequential neoadjuvant chemoradiotherapy (CRT) followed by curative surgery vs. primary surgery alone for resectable, non-metastasized pancreatic adenocarcinoma: NEOPA: a randomized multicenter phase III study (NCT01900327, DRKS00003893, ISRCTN82191749). BMC Cancer 2014;14:411.
    Pubmed KoreaMed CrossRef
  61. Al-Batran SE, Reichart A, Bankstahl US, et al. Randomized multicenter phase II/III study with adjuvant gemcitabine versus neoadjuvant/adjuvant FOLFIRINOX in resectable pancreatic cancer: the NEPAFOX trial. J Clin Oncol 2021;39(3_suppl):406.
    CrossRef
  62. Hozaeel W, Pauligk C, Homann N, et al. Randomized multicenter phase II/III study with adjuvant gemcitabine versus neoadjuvant/adjuvant FOLFIRINOX in resectable pancreatic cancer: the NEPAFOX trial. J Clin Oncol 2015;33(15_suppl):TPS4152.
    CrossRef
  63. Ahmad SA, Duong M, Sohal DP, et al. 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.
    Pubmed KoreaMed CrossRef
  64. Sohal D, Duong MT, Ahmad SA, et al. SWOG S1505: results of perioperative chemotherapy (peri-op CTx) with mfolfirinox versus gemcitabine/nab-paclitaxel (Gem/nabP) for resectable pancreatic ductal adenocarcinoma (PDA). J Clin Oncol 2020;38(15_suppl):4504.
    CrossRef
  65. Kato H, Usui M, Isaji S, et al. Clinical features and treatment outcome of borderline resectable pancreatic head/body cancer: a multi-institutional survey by the Japanese Society of Pancreatic Surgery. J Hepatobiliary Pancreat Sci 2013;20:601-610.
    Pubmed CrossRef
  66. Fujii T, Yamada S, Murotani K, et al. Inverse probability of treatment weighting analysis of upfront surgery versus neoadjuvant chemoradiotherapy followed by surgery for pancreatic adenocarcinoma with arterial abutment. Medicine (Baltimore) 2015;94:e1647.
    Pubmed KoreaMed CrossRef
  67. Hirono S, Kawai M, Okada KI, et al. Treatment strategy for borderline resectable pancreatic cancer with radiographic artery involvement. Pancreas 2016;45:1438-1446.
    Pubmed CrossRef
  68. Ielpo B, Caruso R, Duran H, et al. A comparative study of neoadjuvant treatment with gemcitabine plus nab-paclitaxel versus surgery first for pancreatic adenocarcinoma. Surg Oncol 2017;26:402-410.
    Pubmed CrossRef
  69. Murakami Y, Uemura K, Sudo T, et al. Survival impact of neoadjuvant gemcitabine plus S-1 chemotherapy for patients with borderline resectable pancreatic carcinoma with arterial contact. Cancer Chemother Pharmacol 2017;79:37-47.
    Pubmed CrossRef
  70. Nagakawa Y, Sahara Y, Hosokawa Y, et al. Clinical impact of neoadjuvant chemotherapy and chemoradiotherapy in borderline resectable pancreatic cancer: analysis of 884 patients at facilities specializing in pancreatic surgery. Ann Surg Oncol 2019;26:1629-1636.
    Pubmed CrossRef
  71. Inoue Y, Saiura A, Oba A, et al. Neoadjuvant gemcitabine and nab-paclitaxel for borderline resectable pancreatic cancers: intention-to-treat analysis compared with upfront surgery. J Hepatobiliary Pancreat Sci 2021;28:143-155.
    Pubmed CrossRef
  72. Michelakos T, Pergolini I, Castillo CF, 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.
    Pubmed CrossRef
  73. Labori KJ, Lassen K, Hoem D, et al. Neoadjuvant chemotherapy versus surgery first for resectable pancreatic cancer (Norwegian Pancreatic Cancer Trial-1 (NorPACT-1)): study protocol for a national multicentre randomized controlled trial. BMC Surg 2017;17:94.
    Pubmed KoreaMed CrossRef
  74. Schwarz L, Vernerey D, Bachet JB, et al. 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 KoreaMed CrossRef
  75. Ferrone CR. Testing the use of the usual chemotherapy before and after surgery for removable pancreatic cancer. Identifier No. NCT04340141Available from: https://clinicaltrials.gov/ct2/show/NCT04340141.
  76. Koerkamp BG. Perioperative or Adjuvant mFOLFIRINOX for Resectable Pancreatic Cancer (PREOPANC-3). Identifier No. NCT04927780Available from: https://www.clinicaltrials.gov/ct2/show/NCT04927780.
  77. Janssen QP, van Dam JL, Bonsing BA, et al. Total neoadjuvant FOLFIRINOX versus neoadjuvant gemcitabine-based chemoradiotherapy and adjuvant gemcitabine for resectable and borderline resectable pancreatic cancer (PREOPANC-2 Trial): study protocol for a nationwide multicenter randomized controlled trial. BMC Cancer 2021;21:300.
    Pubmed KoreaMed CrossRef
  78. Grose DB, McKay CJ, Cooke S, et al. PRIMUS-002: a multicentre, open-label, phase II study examining FOLFOX and nab-paclitaxel (FA) and nab-paclitaxel and gemcitabine (AG) as neoadjuvant therapy for (borderline) resectable pancreatic cancer (PC), focusing on biomarker and liquid biopsy development. J Clin Oncol 2019;37(15_suppl):TPS4166.
    CrossRef
  79. Unno M. Phase I/II study of gemcitabine and S-1 (GS therapy) as neoadjuvant chemoradiationtherapy for borderline resectable pancreatic cancer (Prep-03/NS014-1) [Internet]. Tokyo: University Hospital Medical Information Network; c2014 [cited 2022 Oct 30]. Available from: https://center6.umin.ac.jp/cgi-open-bin/ctr/ctr.cgi?function=brows&action=brows&recptno=R000016075&type=summary&language=J.
  80. Okada KI, Shimokawa T, Hirono S, et al. Effect of neoadjuvant nab-paclitaxel plus gemcitabine therapy on overall survival in patients with borderline resectable pancreatic cancer: a prospective multicenter phase II trial (NAC-GA Trial). Oncology 2017;93:343-346.
    Pubmed CrossRef
  81. Takahashi S. Randomized phase II/III study of gemcitabine and nab-paclitaxel therapy versus S-1 and concurrent radiotherapy as neoadjuvant treatment for Borderline resectable pancreatic cancer (GABARNANCE Trial) [Internet]. Wako: National Institute of Public Health; c2019 [cited 2022 Oct 30]. Available from: https://jrct.niph.go.jp/en-latest-detail/jRCTs031180319.
  82. Sanford M. S-1 (TeysunoⓇ): a review of its use in advanced gastric cancer in non-Asian populations. Drugs 2013;73:845-855.
    Pubmed CrossRef
  83. Franck C, Canbay A, Malfertheiner P, Venerito M. Neoadjuvant treatment with cisplatin and S-1 in elderly patients with oesophagogastric adenocarcinoma and locoregional disease: two case reports and review of the literature. Mol Clin Oncol 2017;7:1069-1072.
    Pubmed KoreaMed CrossRef
  84. Lenz HJ, Lee FC, Haller DG, et al. Extended safety and efficacy data on S-1 plus cisplatin in patients with untreated, advanced gastric carcinoma in a multicenter phase II study. Cancer 2007;109:33-40.
    Pubmed CrossRef
  85. Schultheis B, Strumberg D, Bergmann L, et al. Results of a phase II trial of S-1 as first-line treatment of metastatic pancreatic cancer (CESAR-study group). Invest New Drugs 2012;30:1184-1192.
    Pubmed CrossRef
  86. Winther SB, Bjerregaard JK, Schonnemann KR, et al. S-1 (Teysuno) and gemcitabine in Caucasian patients with unresectable pancreatic adenocarcinoma. Cancer Chemother Pharmacol 2018;81:573-578.
    Pubmed CrossRef
  87. Cui J, Jiao F, Li Q, et al. Chinese Society of Clinical Oncology (CSCO): clinical guidelines for the diagnosis and treatment of pancreatic cancer. J Natl Cancer Cent 2022;2:205-215.
    CrossRef
  88. Conroy T, Desseigne F, Ychou M, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med 2011;364:1817-1825.
    Pubmed CrossRef
  89. Von Hoff DD, Ervin T, Arena FP, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med 2013;369:1691-1703.
    Pubmed KoreaMed CrossRef
  90. Teng MW, Ngiow SF, Ribas A, Smyth MJ. Classifying cancers based on T-cell infiltration and PD-L1. Cancer Res 2015;75:2139-2145.
    Pubmed KoreaMed CrossRef
  91. Gajiwala S, Torgeson A, Garrido-Laguna I, Kinsey C, Lloyd S. Combination immunotherapy and radiation therapy strategies for pancreatic cancer-targeting multiple steps in the cancer immunity cycle. J Gastrointest Oncol 2018;9:1014-1026.
    Pubmed KoreaMed CrossRef
  92. Bockhorn M, Uzunoglu FG, Adham M, et al. Borderline resectable pancreatic cancer: a consensus statement by the International Study Group of Pancreatic Surgery (ISGPS). Surgery 2014;155:977-988.
    Pubmed CrossRef
  93. Tjensvoll K, Nordgård O, Smaaland R. Circulating tumor cells in pancreatic cancer patients: methods of detection and clinical implications. Int J Cancer 2014;134:1-8.
    Pubmed CrossRef
  94. Chen J, Wang H, Zhou L, Liu Z, Tan X. A combination of circulating tumor cells and CA199 improves the diagnosis of pancreatic cancer. J Clin Lab Anal 2022;36:e24341.
    Pubmed KoreaMed CrossRef
  95. Isaji S, Mizuno S, Windsor JA, et al. International consensus on definition and criteria of borderline resectable pancreatic ductal adenocarcinoma 2017. Pancreatology 2018;18:2-11.
    Pubmed CrossRef
  96. Macgregor-Das A, Yu J, Tamura K, et al. Detection of circulating tumor DNA in patients with pancreatic cancer using digital next-generation sequencing. J Mol Diagn 2020;22:748-756.
    Pubmed KoreaMed CrossRef
  97. Affolter KE, Hellwig S, Nix DA, et al. Detection of circulating tumor DNA without a tumor-informed search using next-generation sequencing is a prognostic biomarker in pancreatic ductal adenocarcinoma. Neoplasia 2021;23:859-869.
    Pubmed KoreaMed CrossRef
  98. Yamada S, Fujii T, Takami H, et al. Evaluation and proposal of novel resectability criteria for pancreatic cancer established by the Japan Pancreas Society. Surgery 2017;162:784-791.
    Pubmed CrossRef
  99. Fujii T, Satoi S, Yamada S, et al. Clinical benefits of neoadjuvant chemoradiotherapy for adenocarcinoma of the pancreatic head: an observational study using inverse probability of treatment weighting. J Gastroenterol 2017;52:81-93.
    Pubmed CrossRef
  100. Yoshida K, Iwashita T, Uemura S, et al. A multicenter prospective phase II study of first-line modified FOLFIRINOX for unresectable advanced pancreatic cancer. Oncotarget 2017;8:111346-111355.
    Pubmed KoreaMed CrossRef
  101. Kondo N, Uemura K, Sudo T, et al. A phase II study of gemcitabine/nab-paclitaxel/S-1 combination neoadjuvant chemotherapy for patients with borderline resectable pancreatic cancer with arterial contact. Eur J Cancer 2021;159:215-223.
    Pubmed CrossRef
Gut and Liver

Vol.18 No.5
September, 2024

pISSN 1976-2283
eISSN 2005-1212

qrcode
qrcode

Share this article on :

  • line

Popular Keywords

Gut and LiverQR code Download
qr-code

Editorial Office