Skip to main content

Open Access 02.10.2023 | main topic

Personalized treatment in localized pancreatic cancer

verfasst von: Prof. Dr. med. John P. Neoptolemos, MA, MB, BChir, MD, FRCS, FMedSci, MAE, Kai Hu, MD, Peter Bailey, PhD, Christoph Springfeld, MD, PhD, Baobao Cai, MD, Yi Miao, MD, Christoph Michalski, MD, Carlos Carvalho, MD, Thilo Hackert, MD, Markus W. Büchler, MD

Erschienen in: European Surgery

Summary

The treatment elements used for pancreatic ductal adenocarcinoma (PDAC) include surgical resection, systemic cytotoxic agents, and targeted drugs. For second- and third-line therapies in PDAC, approximately 15% of patients have actionable mutations although only 2.5% receive matched targeted treatment but with a significant improvement in survival of around 16 months. For the majority of PDAC patients the current most effective strategy is surgical resection of the primary tumor and systemic combination chemotherapy. The chemotherapy regimens and the order of delivery relative to the resection reference point have been based to a large extent on randomized trials using a newly developed empirical staging (Em) system. Although the reductionist TNM based AJCC and UICC systems work well for pathology staging, they are less accurate and less manageable for treatment decision-making. This Em system defines locally resectable (EmR), borderline resectable (EmBR), and unresectable (EmUR) stages, plus the emerging entity of oligometastatic disease (EmOm). For EmR patients, 6 months of adjuvant chemotherapy achieves 5‑year survival rates of 30–50%. In EmBR short-course (2 months) neoadjuvant plus 6‑month adjuvant chemotherapy increases 12-month survival rates to around 77%, compared to 40% for upfront surgery, despite resection rates of 64–85% and 75%, respectively. Longer-course (4 months) neoadjuvant chemotherapy has also been shown to achieve an 18-month overall survival of 67%. In EmUR, induction therapy (3–6 months) may result in resections rates of 20–60% with significantly improved survival rates compared to no resection. For all stages including the polymetastatic (EmPm) setting, patients with good performance status receive combination chemotherapies based on either oxaliplatin (FOLFIRINOX or NALIRIFOX) or gemcitabine (GEM-CAP, or Gem-NabP). Molecular subtypes (Moffitt, Collisson, Bailey, and Cheng-Sen-Yue) are shown to be associated with treatment responses. Transcriptomic signatures have also been developed as classifiers for determining either oxaliplatin- or gemcitabine-based therapies (PurIST, Tiriac, GemPred+, and ESPAC) and are being evaluated in various studies. Most notably the ESPAC transcriptomic signature is being used as the treatment classifier in the experimental arms of the randomized ESPAC6 adjuvant trial in EmR patients and the ESPAC7 induction therapy trial in EmUR patients. Genomic and transcriptomic profiling at baseline and over time is an integral part of ESPAC6/7 to deepen our understanding of tumor plasticity during the course of therapy, identifying the intrinsic (persister cell) and acquired (genetic) tumor plasticity evolving over time and in reaction to different therapies in order to enable a scientific approach to overcoming clonal-resistance clades.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Pancreatic ductal adenocarcinoma (PDAC) remains one of the most lethal cancers with a 5-year relative survival of 11% [1]. The late stage of diagnosis, rapid progression, and resistance to systemic therapies are the most important factors that are attributed to the low survival rate. Nevertheless, the past two decades have witnessed momentous strides in combating this relentless disease. The advent of improved surgical techniques combined with multimodal systemic chemotherapy and the introduction of empirical staging systems, centered on the assessment of resectability, have emerged as pivotal tools in helping to improve survival [26]. By contrast, few patients benefit from targeted therapies and for only a limited survival period [7]. Primary surgical resection in localized PDAC is now achieved in 15–20% of patients as well as secondary resection after neoadjuvant or induction chemotherapy in many of the 30–35% of patients with locally advanced disease [3, 7]. The integration of adjuvant multiagent chemotherapy after primary resection has significantly improved prognosis from 10% or less with surgery ± chemoradiotherapy to 30–50% with combination regimens, depending on patient selection [813]. The pivotal proof-of-concept study was the first European Study Group for Pancreatic Cancer (ESPAC) trial using adjuvant 5‑fluorouracil (5-FU) monotherapy and the 5‑FU enhancer folinic acid (FA; [8, 9]). The ESPAC3 showed that while there was less toxicity with adjuvant gemcitabine, it did not improve the overall survival of 5‑FU/FA [11]. Combination regimens have increased survival even longer. The evolution in adjuvant chemotherapy regimens has been developed to more potent combination therapies such as gemcitabine plus capecitabine (GEMCAP) with a 5-year survival rate of 30% in unselected patients including those aged over 80 years, and in younger patients with more stringent criteria a 5-year survival rate of 50% has been achieved using modified FA, 5‑FU, irinotecan, and oxaliplatin (mFOLFIRINOX) albeit with greater toxicity [12, 13].
Despite significant strides in achieving higher resectability and improving survival outcomes through advanced surgical techniques and adjuvant chemotherapy, the prospect of long-term survival remains elusive for most patients with PDAC. While empirically driven systemic cytotoxic drugs and reductionist-driven targeted therapies show promise in patients with advanced-stage and metastatic PDAC, they have yet to yield major breakthroughs in extending survival [37]. More recently neoadjuvant therapies have garnered immense interest in the PDAC landscape [3]. The goal is to extend the survival of patients with resectable tumors and to increase resection rates and survival rates in those with borderline-resectable tumors, even exploring possibilities in selected patients with locally advanced tumors and/or oligometastatic disease [3].
With the development of sequencing technology, several molecular classifications of PDAC tumors based on transcriptomic or whole-genome sequencing have been identified in the past 12 years aiming to better understand treatment responses and hence treatment selection, which has been shown to be associated with treatment responses (Fig. 1; [1417]). While molecular subtypes enable a broad phenotypic classification, they are too broad at present to permit more direct treatment allocation, and several transcriptomic signatures have been developed, serving as classifiers to distinguish between oxaliplatin- or gemcitabine-based therapies, notably PurIST, Tiriac, GemPred+, and ESPAC ([1820]; NCT05314998). These signatures are currently under evaluation in various research studies and differ significantly in their mode of development: The PurIST signature derived from various types of tumor samples from 321 patients and has 16 genes defining FOLFIRINOX; the Tiriac signature has 138 genes defining gemcitabine and 98 genes defining oxaliplatin derived from 60 organoids; and the GemPred+ has 420 genes derived from 38 primary cell cultures defining gemcitabine treatment [1820]. The ESPAC signature is unique in using RNASeq data from standardized fresh frozen PDAC tumor samples that have undergone epithelial cell enrichment, defining gemcitabine-based and oxaliplatin-based therapies [NCT05314998].
The traditional view of basing the treatment of pancreatic cancer is to use a reductionist pathological staging systems such as the Union for International Cancer Control (UICC) American Joint Committee on Cancer (AJCC) TNM systems, avoiding surgery in patients in whom an R0 resection cannot be achieved, to consider the tumor as a fixed phenotype, and to give all patients what seems to be the most effective systemic chemotherapy—presently mFOLFIRINOX [21].

Empirical staging: assessment of resectability

The UICC and AJCC TNM staging systems are commonly used to classify the pathological disease stages of PDAC based on tumor, nodes, and metastasis criteria. While this system is effective for pathological and radiological assessments, it has limitations in accurately determining maximum tumor diameter, lymph node involvement, and small metastases [22, 23].
More importantly, PDACs are genetically and biologically heterogeneous, and even using the simplest dichotomized molecular classification subtypes (basal-like and classic-like) there are considerable differences across all of the TNM stages, as well as PDAC neighborhood subtypes and responses to therapy [1417, 24]. The TNM pathology systems used in PDAC have been based on general (assumed a priori and reductionist) oncological principals. There is a shortfall, however, in this reductionist approach, as in clinical practice the TNM does not completely match decision-making (resection being the treatment nodal point), nor the biological nature of PDAC at the diagnostic start point and the subsequent biological behavior in response to time (tumor evolution and acquired mutational resistance) and interventions including surgery and chemotherapy (enrichment of intrinsic persister cell resistance). Thus, an empirically derived staging (Em), based on practical (a posterior) experience, is an alternative approach, with an unbiased discovery scope [3].
Surgical resectability is determined practically using empirical staging criteria established by various organizations (Table 1), including the Americas Hepato-Pancreato-Biliary Association, the Society of Surgical Oncology, the Society for Surgery of the Alimentary Tract (AHPBA/SSO/SSAT), The University of Texas MD Anderson Cancer Center, the Alliance for Clinical Trials in Oncology, and the National Comprehensive Cancer Network (NCCN; Fig. 2a–c; [21, 23, 25, 26]). Each of these criteria sets characterizes the anatomy of the primary tumor and its relationships with crucial blood vessels such as the superior mesenteric vein, portal vein, superior mesenteric artery (SMA), common hepatic artery, its first-order branches, and the celiac trunk (Table 1). They can be applied to stage tumors located in the head, body, or tail of the pancreas. However, there are differences in how radiologists, surgeons, and multidisciplinary teams interpret and utilize each system for staging tumors. To ensure consistency and accuracy, it is essential to clearly report the staging criteria used and rigorously apply them as a condition for patient enrollment in clinical trials. Most of trials such as ESPAC‑5 and Alliance A021101, A021501, and A021806 have stricter enrollment criteria ([2628]; NCT04340141). They mandate prospective centralized specialized review of the staging images of each patient, ensuring that all enrolled patients meet the stringent staging requirements.
Table 1
Assessment of empirically based (Em) tumor resectability according to preoperative computed tomography
 
AHPBA/SSAT/SSO
[22]
MD Anderson
[25]
Alliance
[26]
NCCN
[21]
Superior mesenteric vein-portal vein
EmR: Resectable
No abutment, encasement or occlusion
Abutment or encasement without occlusion
Interface between tumor and vessel measuring < 180°f
No tumor contact or ≤ 180° contact without vein contour irregularity
EmBR: Borderline Resectable
Abutment, encasement, or occlusion
Occlusion
Interface between tumor and vessel measuring ≥ 180°, and/or reconstructable occlusion
Solid tumor contact measuring > 180°, or solid tumor contact ≤ 180° with contour irregularity or thrombosis
EmUR: Locally uresectable
Unreconstructable
Unreconstructable
Unreconstructable
Unreconstructable
Superior mesenteric artery
EmR: Resectable
No abutment
No abutment
No interface between tumor and vessel
No solid tumor contact
EmBR: Borderline Resectable
Abutment
Abutment
Interface between tumor and vessel measuring < 180°f
Solid tumor contact ≤ 180°
EmUR: Locally uresectable
Encasement
Encasement
Interface between tumor and vessel measuring ≥ 180°f
Solid tumor contact > 180°
Common hepatic artery or its first-order branches
EmR: Resectable
No abutment or encasement
No abutment or encasement
No interface between tumor and vessel
No solid tumor contact
EmBR: Borderline Resectable
Abutment or short-segment encasement
Abutment or short-segment encasement
Reconstructable a, short-segment interface between tumor and vessel of any degree
Solid tumor contact without extension to CA or hepatic artery bifurcation
EmUR: Locally uresectable
Unreconstructable
Unreconstructable
Unreconstructable
Unreconstructable
Celiac trunk
EmR: Resectable
No abutment or encasement
No abutment or encasement
No interface between tumor and vessel
No solid tumor contact
EmBR: Borderline Resectable
No abutment or encasement
Abutment
Interface between tumor and vessel measuring < 180°
Solid tumor contact ≤ 180°
EmUR: Locally uresectable
Abutment or encasement
Encasement
Interface between tumor and vessel measuring ≥ 180°
Solid tumor contact > 180°
AHPBA Americas Hepatopancreatobiliary Association, SSO Society of Surgical Oncology, SSAT Society for Surgery of the Alimentary Tract, Alliance Alliance for Clinical Trials in Oncology, NCCN National Comprehensive Cancer Network
To include other important prognostic factors, such as the inherent biological characteristics of the tumor, the physiological condition of the patient, or radiographically occult disease that might not be detectable with routine imaging protocols, the MD Anderson system takes a more comprehensive approach by incorporating additional factors such as serum carbohydrate antigen 19‑9 (CA19-9) levels and performance status, in addition to tumor anatomy [29, 30]. This system has been integrated into international consensus criteria to facilitate more nuanced prognosis assessments. Biological staging of potentially resectable PDACs focuses on a subset of tumors that are anatomically potentially resectable (EmBR, EmUR, EmOm) but may exhibit clinical features suggestive of occult distant polymetastatic disease (EmPm). These worrying features may include a serum CA19‑9 level > 500 kU/L or the presence of regional lymph node metastases diagnosed through biopsy sampling and/or positron emission tomography–computed tomography (PET-CT). Considerable further refinement of this concept is required as studies from France and Japan have shown that the biological aspect of these criteria has limited clinical impact [31, 32]. Therefore, the investigation into biological staging and its relationship to tumor plasticity and acquired and inherent resistance continues to be a critical focus of research [3].

Surgery and cytotoxic therapies in empirically staged tumors

Over the past 50 years numerous clinical trials have been conducted in pancreatic cancer. The majority of these studies have been phase I/II trials, evaluating the potential survival benefits of newer agents and various treatment approaches, including chemoradiation. Unfortunately, many of these trials have not yielded successful outcomes. Currently, clinicaltrials.gov lists over 3000 registered PDAC trials, with 1099 in phase I, 1441 in phase II, and 306 in phase III. Among them, 254 trials are neoadjuvant, of which 23 are in phase III. The EU Clinical Trials Register shows 487 PDAC trials, with 87 in phase III, including 10 neoadjuvant trials.
Given the vast amount of information available, the focus should be on well-conducted phase II/III randomized trials that include appropriate control arms. Combination chemotherapy remains the mainstay of systemic treatment for PDAC with no role for chemoradiation regarding survival, at least in the adjuvant setting. Interestingly, only a relatively small number of agents and combinations have demonstrated sufficient efficacy to gain regulatory approval or become standard-of-care options [3]. An overview of adjuvant, neoadjuvant, and induction chemotherapy based on empirical staging is illustrated in Fig. 3.

Empirical resectable PDAC

The standard of care for EmR PDAC remains upfront resection followed by 6 months of adjuvant chemotherapy but there is no survival advantage to using chemoradiotherapy, resulting only in additional toxicity (Table 2, 2.1). A ceiling in optimal survival has now been reached with combination chemotherapies and is unlikely to be improved without radically new effective therapies, and/or more intelligent utilization of existing cytotoxic drugs based on the actual biology of the tumor at the point of treatment. When comparing the overall survival outcomes of adjuvant trials it is very important to consider the different selection eligibility criteria as well as the toxicity of each regimen [50]. The APACT study (which failed its primary endpoint and hence adjuvant GEM-NabP is not approved by the FDA) only included patients with an Eastern Cooperative Oncology Group performance status of ≤ 1 and a serum 19‑9 level of < 100 kU/L and PRODIGE24 was restricted to patients ≤ 79 years, with a WHO performance score of 0/1, no significant cardiovascular disease, and a serum CA19‑9 level of < 180 kU/L, whereas ESPAC4 had none of these restrictions [12, 23, 44]. These discrepancies are reflected in the notable differences in overall survival for the same gemcitabine control arms used in these studies with overall median (95% CI) survival rates of 37.7 months (range: 31.1–40.5) for APACT, 35.0 months (range: 28.7–43.9) for PRODIGE24, and 25.5 months (range: 22.7–27.9) for ESPAC4 [12, 23, 44]. Thus, the APACT trial had the most favorable selection criteria leading to longer overall survival irrespective of the type of intervention. It is also noteworthy that both ESPAC4 and PRODIGE24 had an absolute increase in 5‑year overall survival in the experimental arms by 12% but only 7% in the APACT trial compared to the control gemcitabine arms [12, 23, 44].
Table 2
Selected randomized trials of adjuvant and neoadjuvant treatment for resectable pancreatic cancer
Trial
Recruitment period
Treatment arms
Number of patients
Median overall survival (months)
5‑year overall survival (%)
Comments
2.1 Adjuvant treatment for resectable pancreatic cancer
GITSG 9173 [33]
1974–1982
CRT + 5FU
Observation
21
22
21
10.9 (p = 0.03)
19
5
R0 only
Norway multi-center [34]
1984–1987
5FU/DOX/MMC
Observation
30
31
23
11
(p = 0.04)
4
8
Includes 14 patients with ampullary cancer
Japan multi-center [35]
1986–1992
MMC/Oral 5‑FU
Observation
81
77
17.1
12.6
(n. s.)
11.5
18
Includes patients with metastases
EORTC 40891 [36, 37]
1987–1995
CRT
Observation
60
54
24.5
19 (p = 0.099)
20
10
T1‑2, N0-1a, M0 pancreatic head cancer
ESPAC‑1 [8], All patients and early follow-up of 2 × 2 factorial
1994–2000
No CRT
CRT
178
175
16.1
15.5 (p = 0.24)
19.5
10.3
Significant for chemotherapy overall but not 2 × 2 factorial. Not significant
No chemotherapy 5FU/FA
235
238
14
19.7 (p = 0.0005)
9.9
23.3
ESPAC‑1 [9], 2 × 2 factorial, final follow-up
1994–2000
No CRT
CRT
144
145
17.9
15.9 (p = 0.05)
19.6
10.8
ECOG 0‑2, R0/R1
No chemotherapy vs. 5FU/FA
142
147
15.5
20.1 (p = 0.009)
8.4
21.1
Observation
69
16.9
10.7
CRT
73
13.9
7.3
5FU/FA
75
21.6
29
CRT + 5FU/FA
72
19.9
13.2
CONKO-001 [10, 38]
1998–2004
GEM
Observation
179
175
22.1
20.2 (p = 0.01)
22.5
11.5
RTOG 9704 [39, 40]
1998–2002
5FU/FA + 5FU-RT + 5FU/FA
GEM + 5FU-RT + GEM
230
221
p = 0.34
ESPAC‑3 [11]
2000–2007
5FU/FA
GEM
551
537
23
23.6 (p = 0.39)
15.9
17.5
ECOG 0‑2, R0/R1
JSAP-02 [41]
2002–2005
GEM (+ IORT in 27) Observation (+ IORT in 47)
58
60
22.3
18.4 (p = 0.19)
23.9
10.6
Karnofsky > 50
CapRI [42]
2004–2007
5FU + cisplatin + IFNα2b + RT + CI 5FU 5FU/FA
64
68
32.1
25.5 (p = 0.49)
25
25
ECOG 0‑2, R0/R1
JASPAC-01 [43]
2007–2010
GEM
S‑1
190
187
25.2
46.5 (p < 0.0001)
24.4
44.1
ECOG 0 = 69%, postop CA19-9 > 37 kU/L = 21, R1+ = 31%, LN+ = 63%
CONKO-005 [44]
2008–2013
GEM
GEM + erlotinib
217
219
26.2
24.5 (p = 0.061)
20
25
R0 only, Karnofsky PS ≥ 60%
CONKO-006 [45]
2008–2013
GEM
GEM + sorafenib
65
57
17.1
18.2 (p = 0.94)
R1 only, Karnofsky PS ≥ 60%
ESPAC‑4 [12]
2008–2014
GEM
GEM-CAP
366
365
25.5
28 (P = 0.032)
16.3
28.8
R0/R1, no restrictions
NRG Oncology/RTOG 0848 [46] NCT01013649
2009–2014
1st randomization
GEM
GEM + erlotinib
163
159
29.9
28.1
(p = 0.62)
Postop CA 19-9 < 180 kU/L, 2nd randomization to CRT reports Q4 2023
PRODIGE-24 [13, 47]
2012–2016
GEM vs. mFOLFIRINOX
246 vs. 247
35 vs. 54.4 (p = 0.003)
ECOG 0/1, post-OP CA 19-9 < 180 kU/L, < 80 years
APACT [48, 49]
2014–2018
GEM vs. GEM-NabP
434 vs. 432
37.7 vs. 41.8 (p = 0.0090). Not primary end point
31 vs. 38
ECOG 0‑1, post-OP CA 19-9 < 100 kU/L, primary endpoint (DFS) not met: 18.0 vs. 19.4 months (p = 0.18)
2.2 Neoadjuvant treatment for resectable pancreatic cancer
Germany multi-center [56]
2003–2009
CRT + surgery + GEM
Surgery + GEM
33
33
17.4
14.4 (p = 0.79)
Terminated due to slow recruitment
Bologna [57]
2007–2014
CRT + surgery Surgery
18
20
19.5
22.4
Terminated due to slow recruitment, not significant
PACT-15 [58]
2010–2015
PEXG + surg + PEXG Surgery + GEM Surgery + PEXG
26
30
32
≤ 75 years, stage I-II, protocol event-free at 1 year: 6 (23%, 95% CI: 7–39) of 30; 15 (50%, 32–68) of 30; 19 (66%, 49–83) of 29
Prep-02/JSAP-05 [59]
2013–2016
GEM + S1 + surg + S1
Surgery + S1
182 (not resected = 42) 180 (not resected = 51)
36.7
26.6 (p = 0.015)
ECOG = 0/1, < 80 years, cf. JASPAC-01 median survival for adjuvant S1 = 46.5 months vs. 26.6 months in JSAP-05
PREOPANC1 [60, 61]
2013–2017
CRT = GEM + surg + GEM
Surgery + GEM
65
68
14.6
15.6
(p = 0.83)
SWOG S1505, [62] NCT02562716
2015–2018
mFOLFIRINOX + surg + mFOLFIRINOX
GEM-NabP + surg + GEM-NabP
55
47
23.2
23.6
Primary endpoint > 2-year OS of 40%: 47% (95% CI: 31–61) for arm 1 and 48% (31–63) for arm 2, neither was significant
NEONAX-AIO-PAK-0313 [63]
2015–2021
GEM-NabP + surg + GEM-NabP
Surgery + GEM
63 (not resected = 18) 64 (not resected = 13)
25.2
16.7
(n. s.)
Planned 166 patients. Primary endpoint median DFS rate of 55% at 18 months double negative result: neoadjuvant 32.2%: adjuvant 41.4%
PANACHE01-PRODIGE48, [64] NCT02959879
2017–2020
mFOLFIRINOX + surg + CTX
FOLFOX + surg + CTX
Surgery + CTX
70
50
26
30.6
31.3
36
(n. s.)
CTX = Initially GEM, 5FU, and GEMCAP, but latterly and mostly mFOLFIRINOX
NORPACT‑1, [65]
NCT02919787
2017–2021
FOLFIRINOX + surg + mFOLFIRINOX
Surg + mFOLFIRINOX
77
63
25.1
38.5
(p = 0.096)
Alive at 1 year: neoadj = 60% vs. adj = 73%, upfront surgery had longer survival
CRT chemoradiotherapy, RT radiotherapy, CTX chemotherapy, CI continuous infusion, 5FU 5-fluorouracil, DOX doxorubicin, MMC mitomycin C, FA folinic acid, GEM gemcitabine, CAP capecitabine, IORT intraoperative radiotherapy, mFOLFIRINOX modified folinic acid (FA), 5‑fluorouracil (5FU), irinotecan (IR) and oxaliplatin (OX), NabP nab-paclitaxel, SBRT stereotactic body radiotherapy, DFS disease-free survival, OS overall survival
Although a considerable number of patients with pancreatic cancer initially have surgically resectable disease, a significant portion of them eventually experience distant recurrences [51, 52]. The promising outcomes seen with adjuvant chemotherapy have led to the investigation of the same drug regimens in the neoadjuvant and perioperative settings for EmR disease. Strong arguments for the use of neoadjuvant therapy have been put forward, but the randomized evidence clearly shows a lack of survival advantage in this setting (Table 2, 2.2; [3, 5355]).
The combination of resectable (EmR) and borderline resectable (EmBR) populations in several of the clinical trials created some confusion, at least at first, in understanding the outcome data. Some early trials, like those led by Golcher et al. and Casadei et al., had to be closed prematurely due to slow patient accrual during a period when neoadjuvant therapy was less accepted [56, 57]. The Prep-02/JSAP-05 trial claimed an improvement in median overall survival for the neoadjuvant group (neoadjuvant gemcitabine and S1 followed by resection then adjuvant S1) at 36.7 months compared to 26.6 months in the upfront resection group (adjuvant S1; [59]). This was a problematic study, however, (only ever published as an abstract), as in the phase III JASPAC-01 trial, patients who had received adjuvant S1 had a median overall survival of 46.5 months, surpassing the adjuvant gemcitabine group with an overall survival of 25.5 months [43, 59]. The trial, however, did not lead to a change in practice (in Japan) as recommended by the Clinical Practice Guidelines for Pancreatic Cancer 2019 of the Japan Pancreas Society [66].
The NORPACT‑1 is the latest multicenter trial to report the results of short-course neoadjuvant FOLFIRINOX versus upfront surgery for EmR pancreatic head cancer, which again does not support the use of neoadjuvant therapy for EmR [65]. In this study 140 patients were randomly assigned to neoadjuvant chemotherapy, (n = 77) or upfront surgery (n = 63) with a median (95% CI) survival that was actually less for the neoadjuvant group (25.1 [17.2–34.9] months) than for upfront surgery (38.5 months [27.6–not reached]) although not statistically significant (p = 0.096; [65]).

Empirical borderline-resectable PDAC

The PREOPANC1 and ESPAC5 trials now provide strong evidence for the use of neoadjuvant chemotherapies for EmBR PDAC while the Alliance A021501 trial suggests that stereotactic body radiotherapy added to neoadjuvant chemotherapy adds to toxicity and may also depress survival (Table 3; [27, 28, 60, 61]). In the PREOPANC1 trial, patients with EmBR had a significant survival impact with neoadjuvant therapy (chemoradiation plus gemcitabine and then adjuvant gemcitabine compared to upfront surgery and adjuvant gemcitabine; [60, 61]). The role of chemoradiation in this context remains unproven especially since survival rates were poor in both groups and the control arm employed mono-gemcitabine, which is no longer the standard of care. In the Alliance A021501 phase II study for EmBR, patients were randomized to receive neoadjuvant mFOLFIRINOX with or without 33–40 Gy hypofractionated radiation therapy, and both groups received adjuvant 5‑FU and oxaliplatin (mFOLFOX6) after resection. The median overall survival was 29.8 months for chemotherapy alone versus 17.1 months for chemotherapy plus radiotherapy [28]. Similarly, the ESPAC5 study for EmBR PDAC found that neoadjuvant chemotherapy with randomization to either FOLFIRINOX or GEMCAP resulted in superior survival compared to upfront surgery, while neoadjuvant 50.4-Gy capecitabine-based chemoradiotherapy did not show the same benefit [27]. The ESPAC5 trial is the only randomized trial that has a head-to-head comparison between FOLFIRINOX and GEMCAP, which demonstrated similar survival outcomes but with less toxicity for GEMCAP.
Table 3
Selected randomized trials of adjuvant and neoadjuvant regimens for borderline-resectable pancreatic cancer
Trial
Recruitment period
Treatment arms
Number of patients
Median overall survival (months)
5‑year overall survival (%)
Comments
Korea multicenter [67]
2012–2014
CRT + GEM + surg + GEM + CRT
Surgery + GEM + CRT
27 (8)
23 (6)
21
12
(1-sided p = 0.028)
Target = 110, premature as interim analysis plan at 50% enrolment, only 1‑sided α = 0.05; only 8 and 6, respectively, had protocol treatment, pre-planned p value not valid
PREOPANC1 [60, 61]
2013–2017
CRT + GEM + surg + GEM
Surg + GEM
54
59
15.7
14.3
(p = 0.029)
ESPAC5 [27]
2014–2018
Surgery + adj
GEM-CAP + surg + adj
FOLFIRINOX + surg + adj
CRT + surg + adj
32
20
20
16
1‑year OS:
42 (27, 64) %
79 (62, 100) %
84 (70, 100) %
64 (43, 95) %
(p = 0.002)
Neoadj GEMCAP and FOLFIRINOX similar, both superior to neoadj CRT and upfront surgery, no difference in resection rates
NUPAT-01 [68]
2015–2020
FOLFIRINOX + surg + CTX
GEM-NabP + surg + CTX
26
25
_
3‑year OS 55.3% vs. 54.4% (n. s.)
Primary endpoint R0 resection rate: 73.1% for FOLFIRINOX, 56.0%for Gem-NabP
Alliance A021501, [28] NCT02839343
2016–2019
mFOLFIRINOX + surg
mFOLFIRINOX + SBRT + surg
54
56
29.8
17.1
18 months OS: 66.7 vs. 47.3
CRT chemoradiotherapy, RT radiotherapy, CTX chemotherapy, CI continuous infusion, 5FU 5-fluorouracil, DOX doxorubicin, MMC mitomycin C, FA folinic acid, GEM gemcitabine, CAP capecitabine, IORT intraoperative radiotherapy, mFOLFIRINOX modified FA/5FU + IR + 5-FU/FA, NabP nab-paclitaxel, SBRT stereotactic body radiotherapy, DFS disease-free survival, OS overall survival, surg surgery, neoadj neoadjuvant
In the PREOPANC1 trial, the resection rates for EmBR were 52% for the neoadjuvant group and 64% for the upfront surgery group, while the R0 resection rates were 79% and 13%, respectively [60]. In the ESPAC5 trial, the resection rates in the three neoadjuvant arms were 65% after FOLFIRINOX, 85% after GEMCAP, and 80% after chemoradiotherapy, with a resection rate of 75% for upfront surgery. The R0 resection rates after neoadjuvant therapy were 12% with FOLFIRINOX, 15% with GEMCAP, and 30% with chemoradiotherapy, compared to 11% with upfront surgery [27]. In the Alliance A021501 trial, the resection rates were 35% with neoadjuvant chemotherapy followed by radiotherapy and 49% with neoadjuvant chemotherapy only (without radiotherapy; [28]). The R0 resection rates were 74% after neoadjuvant chemotherapy followed by radiotherapy and 88% after neoadjuvant chemotherapy only [28]. Thus, for EmBR, neoadjuvant chemotherapy favors overall survival but resection rates and R‑status do not predict survival, while the role of neoadjuvant radiation therapy is not proven.

Empirical locally advanced, unresectable PDAC

Induction chemotherapy

According to NCCN guidelines, selected patients without systemic metastases are recommended to receive 4–6 months of induction combination chemotherapy, followed by chemoradiation or stereotactic body radiation therapy. Surgical resection should be considered if feasible after induction therapy, with adjuvant chemotherapy as clinically indicated [21]. Among the various induction therapy regimens, FOLFIRINOX is the preferred choice for patients with favorable performance status, while gemcitabine with nab-paclitaxel (GEM-NabP) is an alternative (Table 4). The CONKO-007 included 495 patients who received 3 versus 6 months of either FOLFIRINOX or gemcitabine, with or without chemoradiation, followed by surgical exploration (if technically resectable; [70]). The primary endpoint of overall survival was later amended to R0 resection due to delayed patient accrual. The study showed no significant difference in overall survival between the groups, indicating that the addition of radiation during induction therapy did not provide a survival benefit. Importantly, however, this prospective study showed that a resection rate of 23.2% was achieved in EmUR PDAC after induction chemotherapy.
Table 4
Selected randomized trials of induction regimens for unresectable pancreatic cancer
Trial
Recruitment period
Treatment arms
Number of patients
Median overall survival (months)
5‑year overall survival (%)
Comments
LAP-07 [69]
2008–2011
1st randomization GEM vs. GEM + erlotinib
223 vs. 219
13.6 vs. 11.9 (p = 0.09)
Resection rate = 18/449 (4.0%)—not an endpoint
2nd randomization GEM vs. CRT
136 vs 133
16.5 vs. 15.2 (p = 0.83)
CONKO-007, [70]
NCT01827553
2013–2021
mFOLFIRINOX + surg vs.
GEM + surg vs.
140
27
CTX-resect = 60
CTX-surg =
15
4.3
Primary endpoint was altered to R0, 525: surg n = 122 (median OS 19 months, 5‑year OS = 17.5%), non-surg = 214 (median OS = 14 months, 5‑year OS = 0%, p < 0.001), resection 122/525 = 23.2%
mFOLFIRINOX + CRT + surg
vs. GEM + CRT + surg
147
22
CRT-resect = 62
CRT-surg = 15 (p = 0.71)
9.6
NEOLAP, [71] NCT02125136
2014–2018
FOLFIRINOX + surgery GEM-NabP + surg
85
85
20.7
18.5 (p = 0.53)
Primary endpoint resection rate, 168 registered, 165 induction GEM-NabP, 130 randomized to further induction CTX. Median OS with resection (52) = 27.5 months vs. no-resection (113) = 13.9 months, p < 0.0001. Overall resection rate 52/168 = 31%
NEOPAN, [72]
2015–2027
FOLFIRINOX
GEM
85
86
Primary endpoint was PFS
PFS 9.7 vs. 7.5 (p = 0.03), OS 15.1 vs. 15.6 (p = 0.5)
CRT chemoradiotherapy, RT radiotherapy, CTX chemotherapy, CI continuous infusion, 5FU 5-fluorouracil, DOX doxorubicin, MMC mitomycin C, FA folinic acid, GEM gemcitabine, CAP capecitabine, IORT intraoperative radiotherapy, mFOLFIRINOX modified FA/5FU + IR + 5-FU/FA, NabP nab-paclitaxel, SBRT stereotactic body radiotherapy, DFS disease-free survival, OS overall survival, surg surgery
The NEOLAP trial treated EmUR patients with two cycles of induction GEM-NabP, and those without progression were randomized to receive four cycles of FOLFIRINOX or two additional cycles of GEM-NabP [71]. The study found no statistically significant difference in resection rates (the primary endpoint) between the two groups, with both showing overall median survival rates of around 20 months, but importantly again demonstrated in this prospective study an overall reaction rate of 31% [71]. A decade ago, none of these patients would have even been considered for surgical resection.

Surgical exploration after induction chemotherapy

Determining resectability is based on the expertise of the center and surgeon’s experience. Surgical exploration should be undertaken if resection and vascular reconstruction seem feasible, as conventional cross-sectional imaging may not accurately reflect response to induction therapy or the likelihood of resection [2, 3, 73]. A decrease in serum CA19‑9 levels after induction therapy is associated with an increase in successful resections [7477]. Different surgical techniques, such as “artery first,” “triangle resection,” and “arterial divestment,” should be considered to optimize resection rates [7882]. A clean dissection of lymphatic and neural tissue structures in the anatomical triangle bordered by the SMA, celiac axis, and portal vein is crucial for a successful operation (Fig. 2d,e; [81]). The periarterial divestment technique aims to achieve radical tumor clearance without the need for arterial dissection (Fig. 2f; [78, 79, 82, 83]). In a retrospective analysis of EmUR patients who had R0 portal venous resections, the median overall survival was 24 months with a 5-year overall survival of 20% [84].

Empirical oligometastatic disease PDAC

Patients with limited metastases can be considered for resection of the primary tumor and simultaneous metastasectomy achieving median overall survival of 12.3–14.5 months in selected patients [3, 84, 85]. In the largest recent series, in patients with a good pathological response at metastatic sites (ypM0) a median overall survival of 25.5 months was achieved, which increased to 29.0 months with further adjuvant chemotherapy [86]. Patients with lung metastases generally seem to survive longer than those with liver metastases, and moreover pulmonary resection can result in a median survival of 29.2 months from the time of diagnosis of disease recurrence compared with 19.6 months in those with unresected lung metastases [87, 88].

Adjuvant therapies after neoadjuvant treatment

It remains unclear whether patients who have undergone pancreatectomy for localized PDAC after neoadjuvant treatment still benefit from adjuvant chemotherapy. In a retrospective study, adjuvant chemotherapy was found to offer no survival benefit in this setting [89]. By contrast, adjuvant chemotherapy has been found to be associated with improved survival after adjustment for treatment and tumor characteristics in multivariable analyses in a comprehensive analysis of the National Cancer Database [90].
In a multicenter, retrospective study, patients with localized PDAC (EmR, EmBR und EmUR) who underwent pancreatic surgery after at least two cycles of neoadjuvant FOLFIRINOX chemotherapy were retrospectively analyzed. In patients with node-positive disease, adjuvant chemotherapy after neoadjuvant FOLFIRINOX treatment and resection was associated with significantly improved median overall survival of 26 vs. 13 months, respectively [91]. However, adjuvant chemotherapy did not show a significant survival benefit in patients with node-negative disease with a median overall survival of 38 vs. 54 months, respectively [91]. A National Cancer Database study showed that the associated survival was significantly improved in patients who has received adjuvant chemotherapy after neoadjuvant treatment and resection compared with neoadjuvant treatment alone [92]. Furthermore, the survival benefit of additional adjuvant chemotherapy remained significant for those with node-negative disease, a lymph node ratio of < 0.15, low-grade tumor histology, and negative resection margin status [92]. A separate National Cancer Database analysis revealed that additional adjuvant chemotherapy was significantly associated with substantially better median overall survival of 26.6 vs. 21.2 months, with a varied benefit by age, tumor stage, and tumor differentiation [93]. These findings suggest that patients with localized PDAC may benefit from additional adjuvant chemotherapy to achieve prolonged survival after multiagent neoadjuvant or induction chemotherapy and surgical resection. Patients receiving FOLFIRINOX are more prone to greater cumulative toxicity than with other regimens; thus, for example, GEMCAP followed by, for instance, gemcitabine monotherapy could be given for many more cycles, but what is really needed right now is more evidence from well-designed randomized trials.

Targeted therapies

The development of cytotoxic therapies for different stages of pancreatic cancer has primarily relied on empirical approaches. Reductionist attempts to develop treatments by targeting key pathogenic gene alterations have seen limited success, with most targets having a prevalence ranging from 0.1% to 5%, and the survival advantage provided by these agents being only a few months, despite the frequency of altered mutational pathways [94, 95]. The Know Your Tumor Registry represents the largest pancreatic cancer targeted therapy program. Out of 1856 referred patients, 282 (15.2%) had actionable mutations, but only 46 (2.5%) received matched therapy [7]. Survival since diagnosis varied among patients, with a median overall survival of 1.3 years for those with no actionable alteration, 1.5 years for patients with actionable mutations who received unmatched therapy, and 2.6 years for those who had matched therapy [7]. Maintenance therapy with the poly(adenosine diphosphate-ribose) polymerase (PARP) inhibitor olaparib improved progression-free survival from 3.8 to 7.4 months in patients with metastatic pancreatic cancer and germline mutations in the BRCA1/2 genes, provided they had not progressed after at least 4 months of platinum-based first-line chemotherapy [96]. The potential fraction of PDAC patients with druggable driver alterations may significantly increase if novel KRAS inhibitors, targeting the G12D mutation present in about 40% of PDAC patients, prove successful in clinical trials [97].

Drug-resistant persisters in PDAC

In the evolution of PDAC, acquired genomic mutations accumulate in pancreatic exocrine cells leading to increasing dysplastic pancreatic intraepithelial neoplasia, PanIN1 and PanIN2, then carcinoma in situ PanIN3 before progressing to invasion [98]. Low-grade PanIN1A consists of a flat to papillary ductal epithelium with abundant supranuclear mucin and loss of polarity while high-grade PanIN3 is characterized by cytonuclear atypia, dysplasia, or carcinoma in situ with a more complex papillary architecture, nuclear hyperchromatism, and pleomorphism. Telomere shortening and oncogenic KRAS activation are initiating events followed by hypermethylation and/or intragenic mutations and/or loss of heterozygosity of CDKN2A/p16, and mutational activation and or/loss of TP53, as well as homozygous loss or intragenic mutation and second allele loss of DPC4/SMAD4 [99].
Loss of TP53 enables a deterministic pattern of genome evolution in PDAC, suggesting that TP53 mutations are associated with heterogeneous gains in KRAS, MYC, and GATA6, which can further drive metastasis and/or influence PDAC subtypes [100]. The development of PDAC toward greater genomic diversity offers proof that subclonal heterogeneity probably contributes to the unsatisfactory reactions to therapy in PDAC. Within this framework, the simultaneous development of distinct subclones in the identical tumor and at varying stages along a set pathway might clarify the prevalent incidence of varied therapy responses in PDAC. Although these findings propose a traditional model of step-by-step genomic evolution in therapy resistance, mounting proof proposes that non-genetic priming or adjustment significantly adds to therapy resistance [101].
Emerging findings from our research laboratory indicate that intrinsic drug-tolerant cells, often referred to as “persisters,” have the capacity to arise from a preexisting subset of cancerous cells subsequent to neoadjuvant chemotherapy (Fig. 4; [95, 101103]). These persister-like cells adapt to the impact of chemotherapy by enhancing the expression of genes such as CYP3A5 and other co-expressed drug-metabolizing genes. These genes are responsible for metabolizing irinotecan, a constituent of FOLFIRINOX, into forms that lack therapeutic activity. Cancer persister cells can enter a state of dormancy or quiescence, rendering them impervious to the effects of chemotherapy and/or chemoradiotherapy. Consequently, while chemotherapy might effectively eliminate the majority of cancer cells, it could falter in eradicating a minor subset of persister cells, which manage to endure and subsequently repopulate the tumor. Within this conceptual framework, the persister cell state mirrors the notion of minimal residual disease, potentially leading to relapse if treatment discontinues. Accordingly, the identification and comprehensive understanding of persister cells could potentially pave the way for innovative strategies to combat drug-resistant forms of cancer [103].

Molecular classifications and personalized therapy of PDAC

Pancreatic ductal adenocarcinomas exhibit genetic and biological heterogeneity. The simplest molecular classification includes basal-like and classic-like subtypes, which vary across different disease stages and respond differently to various treatments (Fig. 1; [1417, 95, 104]). Classic-like subtypes define well-differentiated tumors that tend to be associated with better outcomes and expression of the key pancreatic-specific transcription factors GATA6, HNF1A, and PDX. Basal-like subtypes are less differentiated tumors with mesenchymal characteristics, including upregulated expression of ∆NP63 and TGFβ-signaling and tend to be associated with poor prognosis [104]. While current subtyping schemas can identify prognostic subgroups among patients with resectable tumors, they are less effective in those with advanced-stage disease.
Different molecular subtypes also respond differently to chemotherapies [1417, 95, 104]. Basal-like PDACs show poor response to chemotherapy in locally advanced or metastatic PDAC [14, 15]. The utility of the classic and basal-like subtypes for predicting survival and response to two major first-line schemes—mFOLFIRINOX and gemcitabine nab-paclitaxel—in advanced PDAC was assessed in the COMPASS study (NCT02750657; [105]). There was a nearly 4‑month longer overall survival for patients with classic-like than for those with a basal-like phenotype [105]. Additionally, patients with high expression of the classic-like marker GATA6 had 2 months longer survival than those with GATA6 low expression. Patients with the classic-like subtype who had mFOLFIRINOX had longer survival than those with the basal-like subtype [105]. Collectively, these findings suggest that GATA6-low and basal-like subtype may be associated with poor response to mFOLFIRINOX.
Intratumor heterogeneity is linked to stromal heterogeneity, leading to various tumor microenvironment programs, including “reactive,” “intermediate,” and “deserted” cellular and transcriptomic subtypes [24]. A deserted-like microenvironment is found in untreated tumors and has been associated with poor treatment response in patients. Drug-tolerant persister cells, arising from different tumor cell lineages during first-line and/or second-line therapy, contribute to disease relapse. Both FOLFIRINOX and chemoradiation therapy have been reported to induce phenotype switching from classic-like to basal-like subtypes, resulting in greater chemoresistance and reduced survival [95, 103105]. What may be of more importance is the acquisition of hybrid molecular phenotypes with both classic- and basal-like characteristics and persister cell type features. Understanding and addressing post-therapy cellular plasticity and persister cell enrichment will require well-designed clinical trials with multidimensional omics analysis approaches.

New trials

Several studies are exploring the use of transcriptomic profiling in the clinical trial setting. The PANCREAS study (PurIST Classification-Guided Adaptive Neoadjuvant Chemotherapy by RNA Expression Profiling of EUS Aspiration Samples) aims to enroll 41 patients, determining the PurIST molecular subtypes in tumor samples obtained by endoscopic ultrasound fine-needle aspiration (EUS/FNA) to establish the pancreatic cancer subtype (NCT04683315). Therapy is directed on the basis of molecular subtype (classic vs. basal). Patients with the classic subtype will receive mFOLFIRINOX and patients with the basal subtype will receive neoadjuvant Gem-NabP. The primary outcome measure is the number of patients who receive PurIST classification-directed therapy and have a treatment response following 12 weeks of therapy—but the stage of disease is not specified.
PASS-01 (Pancreatic Adenocarcinoma Signature Stratification for Treatment) is a randomized phase II trial that will enroll 150 patients with PDAC of any stage to receive either mFOLFIRINOX or Gem-NabP (NCT04469556). The primary outcome measure is progression-free survival, and it is the first randomized trial to have a head-to-head comparison of these two regimens. Important secondary outcome measures include overall survival associated with treatment-specific signatures, and GATA6 concordance between organoid transcriptomic profiles and patient transcriptomic profiles.
The ESPAC6 trial (NCT05314998) will randomize resectable (EmR) patients 1:1 to an experimental arm in which patients will receive either adjuvant mFOLFIRINOX or GEMCAP based on the ESPAC transcriptomic treatment-specific signature or to the control arm in which all patients will receive adjuvant mFOLFIRINOX. The primary endpoint is disease-free survival, with multiple secondary endpoints in transcriptomic and genomic analyses. The ESPAC7 is similarly designed for patients with EmUR locally advanced PDAC tumors, with the primary endpoint in this case being the rate of resection.
Several standard clinical trials are comparing neoadjuvant and adjuvant alternatives in EmR and EmBR settings (Table 5). The Phase III Alliance 21806 trial is evaluating perioperative mFOLFIRINOX (eight cycles neoadjuvant and four cycles adjuvant) compared to adjuvant mFOLFIRINOX (12 cycles). The PREOPANC-3 trial in The Netherlands is evaluating the same treatment regimens. PREOPANC‑2 is also evaluating neoadjuvant mFOLFIRINOX (eight cycles) compared to neoadjuvant gemcitabine-based chemoradiotherapy, with both groups receiving four cycles of adjuvant gemcitabine in the borderline and resectable PDAC populations.
Table 5
Selected ongoing perioperative and molecular studies for localized pancreatic cancer and oligometastatic disease
Trial Name
Identifier
Population
Number
Phase
Interventions (number of chemotherapy cycles)
Primary endpoint
PREOPANC‑2
EudraCT 2017-002036-17
Resectable and borderline resectable
368
III
Neoadjuvant FOLFIRINOX (8) vs. neoadjuvant gemcitabine-based chemoradiotherapy plus adjuvant gemcitabine (4)
Overall survival
PREOPANC‑3
NCT04927780
Resectable
378
III
Neoadjuvant (8) plus adjuvant (4) mFOLFIRINOX vs. adjuvant FOLFIRINOX (12)
Overall survival
ALLIANCE A021806
NCT04340141
Resectable
352
III
Neoadjuvant (8) plus adjuvant mFOLFIRINOX (4) vs. adjuvant mFOLFIRINOX (12)
Overall survival
HOLIPANC
NCT04617457
Oligo-metastatic
150
II
Induction NAPOX (4) stage + (4) restage. Adjuvant chemotherapy discretionary.
Non-randomized
Overall survival
METAPANC
AIO-PAK-0219
Oligo-metastatic
272
III
Induction mFOLFIRINOX (8) + NO SURGERY + maintenance FOLFIRI or capecitabine (3 months) vs. mFOLFIRINOX (8) + surgery + adjuvant FOLFIRI or capecitabine (3 months)
Overall survival
ESPAC‑6
NCT05314998
Resected
394
III
Adjuvant mFOLFIRINOX (12) or GEM-CAP (6) based on transcriptomic signature vs. adjuvant mFOLFIRINOX (12)
Disease-free survival
ESPAC‑7
n.a.
Locally advanced
196
II
Induction mFOLFIRINOX or GEM + NabP based on transcriptomic signature vs. mFOLFIRINOX
Resection rate
mFOLFIRINOX modified FA/5FU + IR + 5-FU/FA, NAPOX liposomal IR + OX + 5-FU/FA, FOLFIRI 5-FU/FA, IR (1 cycle = 14 days), n.a. not available
Further evidence on the role of surgical resection in patients with EmOm is expected from several ongoing studies. The non-randomized single-arm, phase II HOLIPANC study will enroll 150 patients with hepatic oligometastatic PDAC to receive induction therapy with liposomal irinotecan, oxaliplatin, and 5‑FU/FA (NAPOX) followed by surgical exploration and synchronous resection of both primary and metastatic lesions if feasible.
The METAPANC study will randomize 272 patients with EmOm to receive either FOLFIRINOX until disease progression or surgery after at least eight cycles of FOLFIRINOX.

Conclusion

The personalized treatment in localized pancreatic cancer is now firmly established based on the empirical system of staging. For locally resectable (EmR), this requires primary resection followed by adjuvant combination chemotherapy, utilizing either mFOLFIRINOX for patients aged 79 or younger with good performance status and no significant cardiovascular issues, or GEMCAP in other cases. For borderline resectable (EmBR), a neoadjuvant chemotherapy regimen followed by adjuvant therapy, particularly FOLFIRINOX or GEMCAP, is favored. For individuals initially diagnosed with unresectable (EmUR) disease, a 3–6-month induction course of combination chemotherapy, preferably mFOLFIRINOX, is recommended. A similar approach may be taken in selected patients with EmOm. The total number of cycles and dosing when both pre- and postoperative chemotherapy is administered needs to be evaluated in future trials. Chemoradiotherapy in the adjuvant setting is not supported, while its role in the neoadjuvant and induction settings is questioned by a number of studies (Lap-07, Alliance A021501, and ESPAC5). Surrogate markers in outcome evaluation in neoadjuvant and induction strategies are not supported by the evidence, requiring disease-free and overall survival as the primary endpoints. Real progress is now dependent on integrating the empirical staging systems with a deeper understanding of tumor plasticity according to tumor evolution with resistance mechanism based on acquired mutations and the selection of cell persister populations with intrinsic resistance mechanisms. Several key studies that are ongoing are designed to do precisely this, notably PASS-01, ESPAC6, and ESPAC7.

Conflict of interest

J.P. Neoptolemos, K. Hu, P. Bailey, C. Springfeld, B. Cai, Y. Miao, C. Michalski, C. Carvalho, T. Hackert and M.W. Büchler declare that they have no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

Abo für kostenpflichtige Inhalte

Literatur
1.
Zurück zum Zitat Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA Cancer J Clin. 2023;73(1):17–48PubMedCrossRef Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA Cancer J Clin. 2023;73(1):17–48PubMedCrossRef
2.
Zurück zum Zitat Strobel O, Neoptolemos J, Jager D, Buchler MW. Optimizing the outcomes of pancreatic cancer surgery. Nat Rev Clin Oncol. 2019;16(1):11–26.PubMedCrossRef Strobel O, Neoptolemos J, Jager D, Buchler MW. Optimizing the outcomes of pancreatic cancer surgery. Nat Rev Clin Oncol. 2019;16(1):11–26.PubMedCrossRef
3.
Zurück zum Zitat Springfeld C, Ferrone CR, Katz MHG, Philip PA, Hong TS, Hackert T, et al. Neoadjuvant therapy for pancreatic cancer. Nat Rev Clin Oncol. 2023;20(5):318–37.PubMedCrossRef Springfeld C, Ferrone CR, Katz MHG, Philip PA, Hong TS, Hackert T, et al. Neoadjuvant therapy for pancreatic cancer. Nat Rev Clin Oncol. 2023;20(5):318–37.PubMedCrossRef
4.
Zurück zum Zitat Neoptolemos JP, Kleeff J, Michl P, Costello E, Greenhalf W, Palmer DH. Therapeutic developments in pancreatic cancer: current and future perspectives. Nat Rev Gastroenterol Hepatol. 2018;15(6):333–48.PubMedCrossRef Neoptolemos JP, Kleeff J, Michl P, Costello E, Greenhalf W, Palmer DH. Therapeutic developments in pancreatic cancer: current and future perspectives. Nat Rev Gastroenterol Hepatol. 2018;15(6):333–48.PubMedCrossRef
5.
Zurück zum Zitat Kleeff J, Korc M, Apte M, La Vecchia C, Johnson CD, Biankin AV, et al. Pancreatic cancer. Nat Rev Dis Primers. 2016;2:16022.PubMedCrossRef Kleeff J, Korc M, Apte M, La Vecchia C, Johnson CD, Biankin AV, et al. Pancreatic cancer. Nat Rev Dis Primers. 2016;2:16022.PubMedCrossRef
6.
Zurück zum Zitat Kamisawa T, Wood LD, Itoi T, Takaori K. Pancreatic cancer. Lancet. 2016;388(10039):73–85.PubMedCrossRef Kamisawa T, Wood LD, Itoi T, Takaori K. Pancreatic cancer. Lancet. 2016;388(10039):73–85.PubMedCrossRef
7.
Zurück zum Zitat Pishvaian MJ, Wang H, He AR, Hwang JJ, Smaglo BG, Kim SS, et al. A Phase I/II Study of Veliparib (ABT-888) in Combination with 5‑Fluorouracil and Oxaliplatin in Patients with Metastatic Pancreatic Cancer. Clin Cancer Res. 2020;26(19):5092–101.PubMedPubMedCentralCrossRef Pishvaian MJ, Wang H, He AR, Hwang JJ, Smaglo BG, Kim SS, et al. A Phase I/II Study of Veliparib (ABT-888) in Combination with 5‑Fluorouracil and Oxaliplatin in Patients with Metastatic Pancreatic Cancer. Clin Cancer Res. 2020;26(19):5092–101.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Neoptolemos JP, Dunn JA, Stocken DD, Almond J, Link K, Beger H, et al. Adjuvant chemoradiotherapy and chemotherapy in resectable pancreatic cancer: a randomised controlled trial. Lancet. 2001;358(9293):1576–85.PubMedCrossRef Neoptolemos JP, Dunn JA, Stocken DD, Almond J, Link K, Beger H, et al. Adjuvant chemoradiotherapy and chemotherapy in resectable pancreatic cancer: a randomised controlled trial. Lancet. 2001;358(9293):1576–85.PubMedCrossRef
9.
Zurück zum Zitat Neoptolemos JP, Stocken DD, Friess H, Bassi C, Dunn JA, Hickey H, et al. A randomized trial of chemoradiotherapy and chemotherapy after resection of pancreatic cancer. N Engl J Med. 2004;350(12):1200–10.PubMedCrossRef Neoptolemos JP, Stocken DD, Friess H, Bassi C, Dunn JA, Hickey H, et al. A randomized trial of chemoradiotherapy and chemotherapy after resection of pancreatic cancer. N Engl J Med. 2004;350(12):1200–10.PubMedCrossRef
10.
Zurück zum Zitat Oettle H, Post S, Neuhaus P, Gellert K, Langrehr J, Ridwelski K, et al. Adjuvant chemotherapy with gemcitabine vs observation in patients undergoing curative-intent resection of pancreatic cancer: a randomized controlled trial. JAMA. 2007;297(3):267–77.PubMedCrossRef Oettle H, Post S, Neuhaus P, Gellert K, Langrehr J, Ridwelski K, et al. Adjuvant chemotherapy with gemcitabine vs observation in patients undergoing curative-intent resection of pancreatic cancer: a randomized controlled trial. JAMA. 2007;297(3):267–77.PubMedCrossRef
11.
Zurück zum Zitat Neoptolemos JP, Stocken DD, Bassi C, Ghaneh P, Cunningham D, Goldstein D, et al. Adjuvant chemotherapy with fluorouracil plus folinic acid vs gemcitabine following pancreatic cancer resection: a randomized controlled trial. JAMA. 2010;304(10):1073–81.PubMedCrossRef Neoptolemos JP, Stocken DD, Bassi C, Ghaneh P, Cunningham D, Goldstein D, et al. Adjuvant chemotherapy with fluorouracil plus folinic acid vs gemcitabine following pancreatic cancer resection: a randomized controlled trial. JAMA. 2010;304(10):1073–81.PubMedCrossRef
12.
Zurück zum Zitat Neoptolemos JP, Palmer DH, Ghaneh P, Psarelli EE, Valle JW, Halloran CM, 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(10073):1011–24.PubMedCrossRef Neoptolemos JP, Palmer DH, Ghaneh P, Psarelli EE, Valle JW, Halloran CM, 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(10073):1011–24.PubMedCrossRef
13.
Zurück zum Zitat Conroy T, Hammel P, Hebbar M, Abdelghani BM, Wei AC, Raoul JL, et al. FOLFIRINOX or gemcitabine as adjuvant therapy for pancreatic cancer. N Engl J Med. 2018;379(25):2395–406.PubMedCrossRef Conroy T, Hammel P, Hebbar M, Abdelghani BM, Wei AC, Raoul JL, et al. FOLFIRINOX or gemcitabine as adjuvant therapy for pancreatic cancer. N Engl J Med. 2018;379(25):2395–406.PubMedCrossRef
14.
Zurück zum Zitat Collisson EA, Sadanandam A, Olson P, Gibb WJ, Truitt M, Gu S, et al. Subtypes of pancreatic ductal adenocarcinoma and their differing responses to therapy. Nat Med. 2011;17(4):500–3.PubMedPubMedCentralCrossRef Collisson EA, Sadanandam A, Olson P, Gibb WJ, Truitt M, Gu S, et al. Subtypes of pancreatic ductal adenocarcinoma and their differing responses to therapy. Nat Med. 2011;17(4):500–3.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Moffitt RA, Marayati R, Flate EL, Volmar KE, Loeza SG, Hoadley KA, et al. Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma. Nat Genet. 2015;47(10):1168–78.PubMedPubMedCentralCrossRef Moffitt RA, Marayati R, Flate EL, Volmar KE, Loeza SG, Hoadley KA, et al. Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma. Nat Genet. 2015;47(10):1168–78.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Bailey P, Chang DK, Nones K, Johns AL, Patch AM, Gingras MC, et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature. 2016;531(7592):47–52.PubMedCrossRef Bailey P, Chang DK, Nones K, Johns AL, Patch AM, Gingras MC, et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature. 2016;531(7592):47–52.PubMedCrossRef
17.
Zurück zum Zitat Chan-Seng-Yue M, Kim JC, Wilson GW, Ng K, Figueroa EF, O’Kane GM, et al. Transcription phenotypes of pancreatic cancer are driven by genomic events during tumor evolution. Nat Genet. 2020;52(2):231–40.PubMedCrossRef Chan-Seng-Yue M, Kim JC, Wilson GW, Ng K, Figueroa EF, O’Kane GM, et al. Transcription phenotypes of pancreatic cancer are driven by genomic events during tumor evolution. Nat Genet. 2020;52(2):231–40.PubMedCrossRef
18.
Zurück zum Zitat Rashid NU, Peng XL, Jin C, Moffitt RA, Volmar KE, Belt BA, et al. Purity independent subtyping of tumors (PurIST), a clinically robust, single-sample classifier for tumor subtyping in pancreatic cancer. Clin Cancer Res. 2020;26(1):82–92.PubMedCrossRef Rashid NU, Peng XL, Jin C, Moffitt RA, Volmar KE, Belt BA, et al. Purity independent subtyping of tumors (PurIST), a clinically robust, single-sample classifier for tumor subtyping in pancreatic cancer. Clin Cancer Res. 2020;26(1):82–92.PubMedCrossRef
19.
Zurück zum Zitat Tiriac H, Belleau P, Engle DD, Plenker D, Deschenes A, Somerville TDD, et al. Organoid profiling identifies common responders to chemotherapy in pancreatic cancer. Cancer Discov. 2018;8(9):1112–29.PubMedPubMedCentralCrossRef Tiriac H, Belleau P, Engle DD, Plenker D, Deschenes A, Somerville TDD, et al. Organoid profiling identifies common responders to chemotherapy in pancreatic cancer. Cancer Discov. 2018;8(9):1112–29.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Nicolle R, Gayet O, Duconseil P, Vanbrugghe C, Roques J, Bigonnet M, et al. A transcriptomic signature to predict adjuvant gemcitabine sensitivity in pancreatic adenocarcinoma. Ann Oncol. 2021;32(2):250–60.PubMedCrossRef Nicolle R, Gayet O, Duconseil P, Vanbrugghe C, Roques J, Bigonnet M, et al. A transcriptomic signature to predict adjuvant gemcitabine sensitivity in pancreatic adenocarcinoma. Ann Oncol. 2021;32(2):250–60.PubMedCrossRef
22.
Zurück zum Zitat Amin MB, Greene FL, Edge SB, Compton CC, Gershenwald JE, Brookland RK, Meyer L, Gress DM, Byrd DR, Winchester DP. The Eighth Edition AJCC Cancer Staging Manual: Continuing to build a bridge from a population-based to a more “personalized” approach to cancer staging. CA Cancer J Clin. 2017;67(2):93–9. https://doi.org/10.3322/caac.21388. Amin MB, Greene FL, Edge SB, Compton CC, Gershenwald JE, Brookland RK, Meyer L, Gress DM, Byrd DR, Winchester DP. The Eighth Edition AJCC Cancer Staging Manual: Continuing to build a bridge from a population-based to a more “personalized” approach to cancer staging. CA Cancer J Clin. 2017;67(2):93–9. https://​doi.​org/​10.​3322/​caac.​21388.
23.
Zurück zum Zitat Callery MP, Chang KJ, Fishman EK, Talamonti MS, Traverso WL, Linehan DC. Pretreatment assessment of resectable and borderline resectable pancreatic cancer: expert consensus statement. Ann Surg Oncol. 2009;16(7):1727–33.PubMedCrossRef Callery MP, Chang KJ, Fishman EK, Talamonti MS, Traverso WL, Linehan DC. Pretreatment assessment of resectable and borderline resectable pancreatic cancer: expert consensus statement. Ann Surg Oncol. 2009;16(7):1727–33.PubMedCrossRef
24.
Zurück zum Zitat Grunwald BT, Devisme A, Andrieux G, Vyas F, Aliar K, McCloskey CW, et al. Spatially confined sub-tumor microenvironments in pancreatic cancer. Cell. 2021;184(22):5577–5592.e18.PubMedCrossRef Grunwald BT, Devisme A, Andrieux G, Vyas F, Aliar K, McCloskey CW, et al. Spatially confined sub-tumor microenvironments in pancreatic cancer. Cell. 2021;184(22):5577–5592.e18.PubMedCrossRef
25.
Zurück zum Zitat Varadhachary GR, Tamm EP, Abbruzzese JL, Xiong HQ, Crane CH, Wang H, et al. Borderline resectable pancreatic cancer: definitions, management, and role of preoperative therapy. Ann Surg Oncol. 2006;13(8):1035–46.PubMedCrossRef Varadhachary GR, Tamm EP, Abbruzzese JL, Xiong HQ, Crane CH, Wang H, et al. Borderline resectable pancreatic cancer: definitions, management, and role of preoperative therapy. Ann Surg Oncol. 2006;13(8):1035–46.PubMedCrossRef
26.
Zurück zum Zitat Katz MH, Shi Q, Ahmad SA, Herman JM, Marsh Rde W, Collisson E, et al. Preoperative modified FOLFIRINOX treatment followed by capecitabine-based chemoradiation for borderline resectable pancreatic cancer: alliance for clinical trials in oncology trial A021101. JAMA Surg. 2016;151(8):e161137.PubMedPubMedCentralCrossRef Katz MH, Shi Q, Ahmad SA, Herman JM, Marsh Rde W, Collisson E, et al. Preoperative modified FOLFIRINOX treatment followed by capecitabine-based chemoradiation for borderline resectable pancreatic cancer: alliance for clinical trials in oncology trial A021101. JAMA Surg. 2016;151(8):e161137.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Ghaneh P, Palmer D, Cicconi S, Jackson R, Halloran CM, Rawcliffe C, et al. Immediate surgery compared with short-course neoadjuvant gemcitabine plus capecitabine, FOLFIRINOX, or chemoradiotherapy in patients with borderline resectable pancreatic cancer (ESPAC5): a four-arm, multicentre, randomised, phase 2 trial. Lancet Gastroenterol Hepatol. 2023;8(2):157–68.PubMedCrossRef Ghaneh P, Palmer D, Cicconi S, Jackson R, Halloran CM, Rawcliffe C, et al. Immediate surgery compared with short-course neoadjuvant gemcitabine plus capecitabine, FOLFIRINOX, or chemoradiotherapy in patients with borderline resectable pancreatic cancer (ESPAC5): a four-arm, multicentre, randomised, phase 2 trial. Lancet Gastroenterol Hepatol. 2023;8(2):157–68.PubMedCrossRef
29.
Zurück zum Zitat Katz MH, Pisters PW, Evans DB, Sun CC, Lee JE, Fleming JB, et al. Borderline resectable pancreatic cancer: the importance of this emerging stage of disease. J Am Coll Surg. 2008;206(5):833–46. discussion 46–8.PubMedPubMedCentralCrossRef Katz MH, Pisters PW, Evans DB, Sun CC, Lee JE, Fleming JB, et al. Borderline resectable pancreatic cancer: the importance of this emerging stage of disease. J Am Coll Surg. 2008;206(5):833–46. discussion 46–8.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Isaji S, Mizuno S, Windsor JA, Bassi C, Fernandez-Del CC, Hackert T, et al. International consensus on definition and criteria of borderline resectable pancreatic ductal adenocarcinoma 2017. Pancreatology. 2018;18(1):2–11.PubMedCrossRef Isaji S, Mizuno S, Windsor JA, Bassi C, Fernandez-Del CC, Hackert T, et al. International consensus on definition and criteria of borderline resectable pancreatic ductal adenocarcinoma 2017. Pancreatology. 2018;18(1):2–11.PubMedCrossRef
31.
Zurück zum Zitat Hayasaki A, Isaji S, Kishiwada M, Fujii T, Iizawa Y, Kato H, et al. Survival analysis in patients with pancreatic ductal adenocarcinoma undergoing chemoradiotherapy followed by surgery according to the international consensus on the 2017 definition of borderline Resectable cancer. Cancers. 2018;10(3):65.PubMedPubMedCentralCrossRef Hayasaki A, Isaji S, Kishiwada M, Fujii T, Iizawa Y, Kato H, et al. Survival analysis in patients with pancreatic ductal adenocarcinoma undergoing chemoradiotherapy followed by surgery according to the international consensus on the 2017 definition of borderline Resectable cancer. Cancers. 2018;10(3):65.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Medrano J, Garnier J, Ewald J, Marchese U, Gilabert M, Launay S, et al. Patient outcome according to the 2017 international consensus on the definition of borderline resectable pancreatic ductal adenocarcinoma. Pancreatology. 2020;20(2):223–8.PubMedCrossRef Medrano J, Garnier J, Ewald J, Marchese U, Gilabert M, Launay S, et al. Patient outcome according to the 2017 international consensus on the definition of borderline resectable pancreatic ductal adenocarcinoma. Pancreatology. 2020;20(2):223–8.PubMedCrossRef
33.
Zurück zum Zitat Kalser MH, Ellenberg SS. Pancreatic cancer. Adjuvant combined radiation and chemotherapy following curative resection. Arch Surg. 1985;120(8):899–903.PubMedCrossRef Kalser MH, Ellenberg SS. Pancreatic cancer. Adjuvant combined radiation and chemotherapy following curative resection. Arch Surg. 1985;120(8):899–903.PubMedCrossRef
34.
Zurück zum Zitat Bakkevold KE, Arnesjo 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(5):698–703.PubMedCrossRef Bakkevold KE, Arnesjo 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(5):698–703.PubMedCrossRef
35.
Zurück zum Zitat Takada T, Amano H, Yasuda H, Nimura Y, Matsushiro T, Kato H, et al. Is postoperative adjuvant chemotherapy useful for gallbladder carcinoma? A phase III multicenter prospective randomized controlled trial in patients with resected pancreaticobiliary carcinoma. Cancer. 2002;95(8):1685–95.PubMedCrossRef Takada T, Amano H, Yasuda H, Nimura Y, Matsushiro T, Kato H, et al. Is postoperative adjuvant chemotherapy useful for gallbladder carcinoma? A phase III multicenter prospective randomized controlled trial in patients with resected pancreaticobiliary carcinoma. Cancer. 2002;95(8):1685–95.PubMedCrossRef
36.
Zurück zum Zitat Klinkenbijl JH, Jeekel J, Sahmoud T, van Pel R, Couvreur ML, Veenhof CH, 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(6):776–82. discussion 82–4.PubMedPubMedCentralCrossRef Klinkenbijl JH, Jeekel J, Sahmoud T, van Pel R, Couvreur ML, Veenhof CH, 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(6):776–82. discussion 82–4.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Smeenk HG, Incrocci L, Kazemier G, van Dekken H, Tran KT, Jeekel J, van Eijck CH. Adjuvant 5‑FU-based chemoradiotherapy for patients undergoing R‑1/R‑2 resections for pancreatic cancer. Dig Surg. 2005;22(5):321–8.PubMedCrossRef Smeenk HG, Incrocci L, Kazemier G, van Dekken H, Tran KT, Jeekel J, van Eijck CH. Adjuvant 5‑FU-based chemoradiotherapy for patients undergoing R‑1/R‑2 resections for pancreatic cancer. Dig Surg. 2005;22(5):321–8.PubMedCrossRef
38.
Zurück zum Zitat Oettle H, Neuhaus P, Hochhaus A, Hartmann JT, Gellert K, Ridwelski K, et al. Adjuvant chemotherapy with gemcitabine and long-term outcomes among patients with resected pancreatic cancer: the CONKO-001 randomized trial. JAMA. 2013;310(14):1473–81.PubMedCrossRef Oettle H, Neuhaus P, Hochhaus A, Hartmann JT, Gellert K, Ridwelski K, et al. Adjuvant chemotherapy with gemcitabine and long-term outcomes among patients with resected pancreatic cancer: the CONKO-001 randomized trial. JAMA. 2013;310(14):1473–81.PubMedCrossRef
39.
Zurück zum Zitat Regine WF, Winter KA, Abrams R, Safran H, Hoffman JP, Konski A, et al. Fluorouracil-based chemoradiation with either gemcitabine or fluorouracil chemotherapy after resection of pancreatic adenocarcinoma: 5‑year analysis of the U.S. Intergroup/RTOG 9704 phase III trial. Ann Surg Oncol. 2011;18(5):1319–26.PubMedPubMedCentralCrossRef Regine WF, Winter KA, Abrams R, Safran H, Hoffman JP, Konski A, et al. Fluorouracil-based chemoradiation with either gemcitabine or fluorouracil chemotherapy after resection of pancreatic adenocarcinoma: 5‑year analysis of the U.S. Intergroup/RTOG 9704 phase III trial. Ann Surg Oncol. 2011;18(5):1319–26.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Regine WF, Winter KA, Abrams RA, Safran H, Hoffman JP, Konski A, et al. Fluorouracil vs gemcitabine chemotherapy before and after fluorouracil-based chemoradiation following resection of pancreatic adenocarcinoma: a randomized controlled trial. JAMA. 2008;299(9):1019–26.PubMedCrossRef Regine WF, Winter KA, Abrams RA, Safran H, Hoffman JP, Konski A, et al. Fluorouracil vs gemcitabine chemotherapy before and after fluorouracil-based chemoradiation following resection of pancreatic adenocarcinoma: a randomized controlled trial. JAMA. 2008;299(9):1019–26.PubMedCrossRef
41.
Zurück zum Zitat Ueno H, Kosuge T, Matsuyama Y, Yamamoto J, Nakao A, Egawa S, 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(6):908–15.PubMedPubMedCentralCrossRef Ueno H, Kosuge T, Matsuyama Y, Yamamoto J, Nakao A, Egawa S, 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(6):908–15.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Schmidt J, Abel U, Debus J, Harig S, Hoffmann K, Herrmann T, et al. Open-label, multicenter, randomized phase III trial of adjuvant chemoradiation plus interferon Alfa-2b versus fluorouracil and folinic acid for patients with resected pancreatic adenocarcinoma. J Clin Oncol. 2012;30(33):4077–83.PubMedCrossRef Schmidt J, Abel U, Debus J, Harig S, Hoffmann K, Herrmann T, et al. Open-label, multicenter, randomized phase III trial of adjuvant chemoradiation plus interferon Alfa-2b versus fluorouracil and folinic acid for patients with resected pancreatic adenocarcinoma. J Clin Oncol. 2012;30(33):4077–83.PubMedCrossRef
43.
Zurück zum Zitat Uesaka K, Boku N, Fukutomi A, Okamura Y, Konishi M, Matsumoto I, 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(10041):248–57.PubMedCrossRef Uesaka K, Boku N, Fukutomi A, Okamura Y, Konishi M, Matsumoto I, 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(10041):248–57.PubMedCrossRef
44.
Zurück zum Zitat Sinn M, Bahra M, Liersch T, Gellert K, Messmann H, Bechstein W, 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(29):3330–7.PubMedCrossRef Sinn M, Bahra M, Liersch T, Gellert K, Messmann H, Bechstein W, 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(29):3330–7.PubMedCrossRef
45.
Zurück zum Zitat Sinn M, Liersch T, Riess H, Gellert K, Stubs P, Waldschmidt D, et al. CONKO-006: A randomised double-blinded phase IIb-study of additive therapy with gemcitabine + sorafenib/placebo in patients with R1 resection of pancreatic cancer—Final results. Eur J Cancer. 2020;138:172–81.PubMedCrossRef Sinn M, Liersch T, Riess H, Gellert K, Stubs P, Waldschmidt D, et al. CONKO-006: A randomised double-blinded phase IIb-study of additive therapy with gemcitabine + sorafenib/placebo in patients with R1 resection of pancreatic cancer—Final results. Eur J Cancer. 2020;138:172–81.PubMedCrossRef
46.
Zurück zum Zitat Abrams RA, Winter KA, Safran H, Goodman KA, Regine WF, Berger AC, 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(3):173–9.PubMedPubMedCentralCrossRef Abrams RA, Winter KA, Safran H, Goodman KA, Regine WF, Berger AC, 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(3):173–9.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Conroy T, Castan F, Lopez A, Turpin A, Abdelghani BM, Wei AC, et al. Five-year outcomes of FOLFIRINOX vs gemcitabine as adjuvant therapy for pancreatic cancer: a randomized clinical trial. JAMA Oncol. 2022;8(11):1571–8.PubMedPubMedCentralCrossRef Conroy T, Castan F, Lopez A, Turpin A, Abdelghani BM, Wei AC, et al. Five-year outcomes of FOLFIRINOX vs gemcitabine as adjuvant therapy for pancreatic cancer: a randomized clinical trial. JAMA Oncol. 2022;8(11):1571–8.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Tempero M, O’Reilly E, Van Cutsem E, Berlin J, Philip P, Goldstein D, et al. LBA-1 Phase 3 APACT trial of adjuvant nab-paclitaxel plus gemcitabine (nab‑P + Gem) vs gemcitabine (Gem) alone in patients with resected pancreatic cancer (PC): Updated 5‑year overall survival. Ann Oncol. 2021;32:S226.CrossRef Tempero M, O’Reilly E, Van Cutsem E, Berlin J, Philip P, Goldstein D, et al. LBA-1 Phase 3 APACT trial of adjuvant nab-paclitaxel plus gemcitabine (nab‑P + Gem) vs gemcitabine (Gem) alone in patients with resected pancreatic cancer (PC): Updated 5‑year overall survival. Ann Oncol. 2021;32:S226.CrossRef
49.
Zurück zum Zitat Tempero MA, Reni M, Riess H, Pelzer U, O’Reilly EM, Winter JM, et al. APACT: phase III, multicenter, international, open-label, randomized trial of adjuvant nab-paclitaxel plus gemcitabine (nab-P/G) vs gemcitabine (G) for surgically resected pancreatic adenocarcinoma. J Clin Oncol. 2019;37(15_suppl):4000.CrossRef Tempero MA, Reni M, Riess H, Pelzer U, O’Reilly EM, Winter JM, et al. APACT: phase III, multicenter, international, open-label, randomized trial of adjuvant nab-paclitaxel plus gemcitabine (nab-P/G) vs gemcitabine (G) for surgically resected pancreatic adenocarcinoma. J Clin Oncol. 2019;37(15_suppl):4000.CrossRef
51.
Zurück zum Zitat Groot VP, Rezaee N, Wu W, Cameron JL, Fishman EK, Hruban RH, et al. Patterns, timing, and predictors of recurrence following pancreatectomy for pancreatic ductal adenocarcinoma. Ann Surg. 2018;267(5):936–45.PubMedCrossRef Groot VP, Rezaee N, Wu W, Cameron JL, Fishman EK, Hruban RH, et al. Patterns, timing, and predictors of recurrence following pancreatectomy for pancreatic ductal adenocarcinoma. Ann Surg. 2018;267(5):936–45.PubMedCrossRef
53.
Zurück zum Zitat Neoptolemos JP, Springfeld C, Hackert T. A review of pancreatic cancer. JAMA. 2021;326(23):2436.PubMedCrossRef Neoptolemos JP, Springfeld C, Hackert T. A review of pancreatic cancer. JAMA. 2021;326(23):2436.PubMedCrossRef
54.
Zurück zum Zitat Springfeld C, Neoptolemos JP. The role of neoadjuvant therapy for resectable pancreatic cancer remains uncertain. Nat Rev Clin Oncol. 2022;19(5):285–6.PubMedCrossRef Springfeld C, Neoptolemos JP. The role of neoadjuvant therapy for resectable pancreatic cancer remains uncertain. Nat Rev Clin Oncol. 2022;19(5):285–6.PubMedCrossRef
55.
56.
Zurück zum Zitat Golcher H, Brunner TB, Witzigmann H, Marti L, Bechstein WO, Bruns C, 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(1):7–16.PubMedCrossRef Golcher H, Brunner TB, Witzigmann H, Marti L, Bechstein WO, Bruns C, 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(1):7–16.PubMedCrossRef
57.
Zurück zum Zitat Casadei R, Di Marco M, Ricci C, Santini D, Serra C, Calculli L, 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(10):1802–12.PubMedCrossRef Casadei R, Di Marco M, Ricci C, Santini D, Serra C, Calculli L, 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(10):1802–12.PubMedCrossRef
58.
Zurück zum Zitat Reni M, Balzano G, Zanon S, Zerbi A, Rimassa L, Castoldi R, et al. Safety and efficacy of preoperative or postoperative chemotherapy for resectable pancreatic adenocarcinoma (PACT-15): a randomised, open-label, phase 2–3 trial. Lancet Gastroenterol Hepatol. 2018;3(6):413–23.PubMedCrossRef Reni M, Balzano G, Zanon S, Zerbi A, Rimassa L, Castoldi R, et al. Safety and efficacy of preoperative or postoperative chemotherapy for resectable pancreatic adenocarcinoma (PACT-15): a randomised, open-label, phase 2–3 trial. Lancet Gastroenterol Hepatol. 2018;3(6):413–23.PubMedCrossRef
59.
Zurück zum Zitat Unno M, Motoi F, Matsuyama Y, Satoi S, Matsumoto I, Aosasa S, 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). JCO. 2019;37(4_suppl):189.CrossRef Unno M, Motoi F, Matsuyama Y, Satoi S, Matsumoto I, Aosasa S, 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). JCO. 2019;37(4_suppl):189.CrossRef
60.
Zurück zum Zitat Versteijne E, van Dam JL, Suker M, Janssen QP, Groothuis K, Akkermans-Vogelaar JM, 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(11):1220–30.PubMedCrossRef Versteijne E, van Dam JL, Suker M, Janssen QP, Groothuis K, Akkermans-Vogelaar JM, 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(11):1220–30.PubMedCrossRef
61.
Zurück zum Zitat Versteijne E, Suker M, Groothuis K, Akkermans-Vogelaar JM, Besselink MG, Bonsing BA, 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(16):1763–73.PubMedPubMedCentralCrossRef Versteijne E, Suker M, Groothuis K, Akkermans-Vogelaar JM, Besselink MG, Bonsing BA, 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(16):1763–73.PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Sohal DPS, Duong M, Ahmad SA, Gandhi NS, Beg MS, Wang-Gillam A, et al. Efficacy of perioperative chemotherapy for resectable pancreatic adenocarcinoma: a phase 2 randomized clinical trial. JAMA Oncol. 2021;7(3):421–7.PubMedCrossRef Sohal DPS, Duong M, Ahmad SA, Gandhi NS, Beg MS, Wang-Gillam A, et al. Efficacy of perioperative chemotherapy for resectable pancreatic adenocarcinoma: a phase 2 randomized clinical trial. JAMA Oncol. 2021;7(3):421–7.PubMedCrossRef
63.
Zurück zum Zitat Seufferlein T, Uhl W, Kornmann M, Algul H, Friess H, Konig A, et al. Perioperative or only adjuvant gemcitabine plus nab-paclitaxel for resectable pancreatic cancer (NEONAX)—a randomized phase II trial of the AIO pancreatic cancer group. Ann Oncol. 2023;34(1):91–100.PubMedCrossRef Seufferlein T, Uhl W, Kornmann M, Algul H, Friess H, Konig A, et al. Perioperative or only adjuvant gemcitabine plus nab-paclitaxel for resectable pancreatic cancer (NEONAX)—a randomized phase II trial of the AIO pancreatic cancer group. Ann Oncol. 2023;34(1):91–100.PubMedCrossRef
64.
Zurück zum Zitat Schwarz L, Bachet J‑B, Meurisse A, Bouché O, Assenat E, Piessen G, et al. Resectable pancreatic adenocarcinoma neo-adjuvant FOLF(IRIN)OX-based chemotherapy: A multicenter, non-comparative, randomized, phase II trial (PANACHE01-PRODIGE48 study). J Clin Oncol. 2022;40(16_suppl):4134.CrossRef Schwarz L, Bachet J‑B, Meurisse A, Bouché O, Assenat E, Piessen G, et al. Resectable pancreatic adenocarcinoma neo-adjuvant FOLF(IRIN)OX-based chemotherapy: A multicenter, non-comparative, randomized, phase II trial (PANACHE01-PRODIGE48 study). J Clin Oncol. 2022;40(16_suppl):4134.CrossRef
65.
Zurück zum Zitat Labori KJ, Bratlie SO, Biörserud C, Björnsson B, Bringeland EN, et al. Short-course neoadjuvant FOLFIRINOX versus upfront surgery for resectable pancreatic head cancer: A multicenter randomized phase-II trial (NORPACT-1). J Clin Oncol. 2023;41:LBA4005–LBA4005. Labori KJ, Bratlie SO, Biörserud C, Björnsson B, Bringeland EN, et al. Short-course neoadjuvant FOLFIRINOX versus upfront surgery for resectable pancreatic head cancer: A multicenter randomized phase-II trial (NORPACT-1). J Clin Oncol. 2023;41:LBA4005–LBA4005.
66.
Zurück zum Zitat Okusaka T, Nakamura M, Yoshida M, Kitano M, Uesaka K, Ito Y, et al. Clinical practice guidelines for pancreatic cancer 2019 from the Japan pancreas society: a synopsis. Pancreas. 2020;49(3):326–35.PubMedCrossRef Okusaka T, Nakamura M, Yoshida M, Kitano M, Uesaka K, Ito Y, et al. Clinical practice guidelines for pancreatic cancer 2019 from the Japan pancreas society: a synopsis. Pancreas. 2020;49(3):326–35.PubMedCrossRef
67.
Zurück zum Zitat Jang JY, Han Y, Lee H, Kim SW, Kwon W, Lee KH, 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(2):215–22.PubMedCrossRef Jang JY, Han Y, Lee H, Kim SW, Kwon W, Lee KH, 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(2):215–22.PubMedCrossRef
68.
Zurück zum Zitat Yamaguchi J, Yokoyama Y, Fujii T, Yamada S, Takami H, Kawashima H, 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(6):1043–9.PubMedCrossRef Yamaguchi J, Yokoyama Y, Fujii T, Yamada S, Takami H, Kawashima H, 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(6):1043–9.PubMedCrossRef
69.
Zurück zum Zitat Hammel P, Huguet F, van Laethem JL, Goldstein D, Glimelius B, Artru P, et al. Effect of chemoradiotherapy vs chemotherapy on survival in patients with locally advanced pancreatic cancer controlled after 4 months of gemcitabine with or without erlotinib: the LAP07 randomized clinical trial. JAMA. 2016;315(17):1844–53.PubMedCrossRef Hammel P, Huguet F, van Laethem JL, Goldstein D, Glimelius B, Artru P, et al. Effect of chemoradiotherapy vs chemotherapy on survival in patients with locally advanced pancreatic cancer controlled after 4 months of gemcitabine with or without erlotinib: the LAP07 randomized clinical trial. JAMA. 2016;315(17):1844–53.PubMedCrossRef
70.
Zurück zum Zitat Fietkau R, Ghadimi M, Grützmann R, Wittel UA, Jacobasch L, Uhl W, et al. Randomized phase III trial of induction chemotherapy followed by chemoradiotherapy or chemotherapy alone for nonresectable locally advanced pancreatic cancer: First results of the CONKO-007 trial. J Clin Oncol. 2022;40(16_suppl):4008.CrossRef Fietkau R, Ghadimi M, Grützmann R, Wittel UA, Jacobasch L, Uhl W, et al. Randomized phase III trial of induction chemotherapy followed by chemoradiotherapy or chemotherapy alone for nonresectable locally advanced pancreatic cancer: First results of the CONKO-007 trial. J Clin Oncol. 2022;40(16_suppl):4008.CrossRef
71.
Zurück zum Zitat Kunzmann V, Siveke JT, Algul H, Goekkurt E, Siegler G, Martens U, et al. Nab-paclitaxel plus gemcitabine versus nab-paclitaxel plus gemcitabine followed by FOLFIRINOX induction chemotherapy in locally advanced pancreatic cancer (NEOLAP-AIO-PAK-0113): a multicentre, randomised, phase 2 trial. Lancet Gastroenterol Hepatol. 2021;6(2):128–38.PubMedCrossRef Kunzmann V, Siveke JT, Algul H, Goekkurt E, Siegler G, Martens U, et al. Nab-paclitaxel plus gemcitabine versus nab-paclitaxel plus gemcitabine followed by FOLFIRINOX induction chemotherapy in locally advanced pancreatic cancer (NEOLAP-AIO-PAK-0113): a multicentre, randomised, phase 2 trial. Lancet Gastroenterol Hepatol. 2021;6(2):128–38.PubMedCrossRef
72.
Zurück zum Zitat Ducreux MP, Desgrippes R, Rinaldi Y, Fiore F, Guimbaud R, Follana P, et al. 1296MO PRODIGE 29-UCGI 26(NEOPAN): A phase III randomised trial comparing chemotherapy with folfirinox or gemcitabine in locally advanced pancreatic carcinoma (LAPC). Ann Oncol. 2022;33:S1136.CrossRef Ducreux MP, Desgrippes R, Rinaldi Y, Fiore F, Guimbaud R, Follana P, et al. 1296MO PRODIGE 29-UCGI 26(NEOPAN): A phase III randomised trial comparing chemotherapy with folfirinox or gemcitabine in locally advanced pancreatic carcinoma (LAPC). Ann Oncol. 2022;33:S1136.CrossRef
73.
Zurück zum Zitat Ferrone CR, Marchegiani G, Hong TS, Ryan DP, Deshpande V, McDonnell EI, et al. Radiological and surgical implications of neoadjuvant treatment with FOLFIRINOX for locally advanced and borderline resectable pancreatic cancer. Ann Surg. 2015;261(1):12–7.PubMedCrossRef Ferrone CR, Marchegiani G, Hong TS, Ryan DP, Deshpande V, McDonnell EI, et al. Radiological and surgical implications of neoadjuvant treatment with FOLFIRINOX for locally advanced and borderline resectable pancreatic cancer. Ann Surg. 2015;261(1):12–7.PubMedCrossRef
74.
Zurück zum Zitat Heger U, Sun H, Hinz U, Klaiber U, Tanaka M, Liu B, et al. Induction chemotherapy in pancreatic cancer: CA 19‑9 may predict resectability and survival. HPB. 2020;22(2):224–32.PubMedCrossRef Heger U, Sun H, Hinz U, Klaiber U, Tanaka M, Liu B, et al. Induction chemotherapy in pancreatic cancer: CA 19‑9 may predict resectability and survival. HPB. 2020;22(2):224–32.PubMedCrossRef
75.
Zurück zum Zitat Springfeld C, Jager D, Buchler MW, Strobel O, Hackert T, Palmer DH, Neoptolemos JP. Chemotherapy for pancreatic cancer. Presse Med. 2019;48(3 Pt 2):e159–e74.PubMedCrossRef Springfeld C, Jager D, Buchler MW, Strobel O, Hackert T, Palmer DH, Neoptolemos JP. Chemotherapy for pancreatic cancer. Presse Med. 2019;48(3 Pt 2):e159–e74.PubMedCrossRef
76.
Zurück zum Zitat Philip PA, Lacy J, Portales F, Sobrero A, Pazo-Cid R, Manzano Mozo JL, et al. Nab-paclitaxel plus gemcitabine in patients with locally advanced pancreatic cancer (LAPACT): a multicentre, open-label phase 2 study. Lancet Gastroenterol Hepatol. 2020;5(3):285–94.PubMedCrossRef Philip PA, Lacy J, Portales F, Sobrero A, Pazo-Cid R, Manzano Mozo JL, et al. Nab-paclitaxel plus gemcitabine in patients with locally advanced pancreatic cancer (LAPACT): a multicentre, open-label phase 2 study. Lancet Gastroenterol Hepatol. 2020;5(3):285–94.PubMedCrossRef
77.
Zurück zum Zitat Hartwig W, Strobel O, Hinz U, Fritz S, Hackert T, Roth C, et al. CA19‑9 in potentially resectable pancreatic cancer: perspective to adjust surgical and perioperative therapy. Ann Surg Oncol. 2013;20(7):2188–96.PubMedCrossRef Hartwig W, Strobel O, Hinz U, Fritz S, Hackert T, Roth C, et al. CA19‑9 in potentially resectable pancreatic cancer: perspective to adjust surgical and perioperative therapy. Ann Surg Oncol. 2013;20(7):2188–96.PubMedCrossRef
78.
Zurück zum Zitat Cai B, Lu Z, Neoptolemos JP, Diener MK, Li M, Yin L, et al. Sub-adventitial divestment technique for resecting artery-involved pancreatic cancer: a retrospective cohort study. Langenbecks Arch Surg. 2021;406(3):691–701.PubMedCrossRef Cai B, Lu Z, Neoptolemos JP, Diener MK, Li M, Yin L, et al. Sub-adventitial divestment technique for resecting artery-involved pancreatic cancer: a retrospective cohort study. Langenbecks Arch Surg. 2021;406(3):691–701.PubMedCrossRef
79.
Zurück zum Zitat Diener MK, Mihaljevic AL, Strobel O, Loos M, Schmidt T, Schneider M, et al. Periarterial divestment in pancreatic cancer surgery. Surgery. 2021;169(5):1019–25.PubMedCrossRef Diener MK, Mihaljevic AL, Strobel O, Loos M, Schmidt T, Schneider M, et al. Periarterial divestment in pancreatic cancer surgery. Surgery. 2021;169(5):1019–25.PubMedCrossRef
80.
Zurück zum Zitat Klompmaker S, Boggi U, Hackert T, Salvia R, Weiss M, Yamaue H, et al. Distal pancreatectomy with celiac axis resection (DP-CAR) for pancreatic cancer. How I do it. J Gastrointest Surg. 2018;22(10):1804–10.PubMedPubMedCentralCrossRef Klompmaker S, Boggi U, Hackert T, Salvia R, Weiss M, Yamaue H, et al. Distal pancreatectomy with celiac axis resection (DP-CAR) for pancreatic cancer. How I do it. J Gastrointest Surg. 2018;22(10):1804–10.PubMedPubMedCentralCrossRef
81.
Zurück zum Zitat Klotz R, Hackert T, Heger P, Probst P, Hinz U, Loos M, et al. The TRIANGLE operation for pancreatic head and body cancers: early postoperative outcomes. HPB. 2022;24(3):332–41.PubMedCrossRef Klotz R, Hackert T, Heger P, Probst P, Hinz U, Loos M, et al. The TRIANGLE operation for pancreatic head and body cancers: early postoperative outcomes. HPB. 2022;24(3):332–41.PubMedCrossRef
82.
Zurück zum Zitat Schneider M, Hackert T, Strobel O, Buchler MW. Technical advances in surgery for pancreatic cancer. Br J Surg. 2021;108(7):777–85.PubMedCrossRef Schneider M, Hackert T, Strobel O, Buchler MW. Technical advances in surgery for pancreatic cancer. Br J Surg. 2021;108(7):777–85.PubMedCrossRef
83.
Zurück zum Zitat Habib JR, Kinny-Koster B, van Oosten F, Javed AA, Cameron JL, Lafaro KJ, et al. Periadventitial dissection of the superior mesenteric artery for locally advanced pancreatic cancer: Surgical planning with the “halo sign” and “string sign”. Surgery. 2021;169(5):1026–31.PubMedCrossRef Habib JR, Kinny-Koster B, van Oosten F, Javed AA, Cameron JL, Lafaro KJ, et al. Periadventitial dissection of the superior mesenteric artery for locally advanced pancreatic cancer: Surgical planning with the “halo sign” and “string sign”. Surgery. 2021;169(5):1026–31.PubMedCrossRef
84.
Zurück zum Zitat Hackert T, Niesen W, Hinz U, Tjaden C, Strobel O, Ulrich A, et al. Radical surgery of oligometastatic pancreatic cancer. Eur J Surg Oncol. 2017;43(2):358–63.PubMedCrossRef Hackert T, Niesen W, Hinz U, Tjaden C, Strobel O, Ulrich A, et al. Radical surgery of oligometastatic pancreatic cancer. Eur J Surg Oncol. 2017;43(2):358–63.PubMedCrossRef
85.
Zurück zum Zitat Tachezy M, Gebauer F, Janot M, Uhl W, Zerbi A, Montorsi M, et al. Synchronous resections of hepatic oligometastatic pancreatic cancer: disputing a principle in a time of safe pancreatic operations in a retrospective multicenter analysis. Surgery. 2016;160(1):136–44.PubMedCrossRef Tachezy M, Gebauer F, Janot M, Uhl W, Zerbi A, Montorsi M, et al. Synchronous resections of hepatic oligometastatic pancreatic cancer: disputing a principle in a time of safe pancreatic operations in a retrospective multicenter analysis. Surgery. 2016;160(1):136–44.PubMedCrossRef
87.
Zurück zum Zitat Jones RP, Psarelli EE, Jackson R, Ghaneh P, Halloran CM, Palmer DH, et al. Patterns of recurrence after resection of pancreatic ductal adenocarcinoma: a secondary analysis of the ESPAC‑4 randomized adjuvant chemotherapy trial. JAMA Surg. 2019;154(11):1038–48.PubMedPubMedCentralCrossRef Jones RP, Psarelli EE, Jackson R, Ghaneh P, Halloran CM, Palmer DH, et al. Patterns of recurrence after resection of pancreatic ductal adenocarcinoma: a secondary analysis of the ESPAC‑4 randomized adjuvant chemotherapy trial. JAMA Surg. 2019;154(11):1038–48.PubMedPubMedCentralCrossRef
88.
Zurück zum Zitat Homma Y, Endo I, Matsuyama R, Sho M, Mizuno S, Seyama Y, et al. Outcomes of lung metastasis from pancreatic cancer: a nationwide multicenter analysis. J Hepato Biliary Pancreat. 2022;29(5):552–61.CrossRef Homma Y, Endo I, Matsuyama R, Sho M, Mizuno S, Seyama Y, et al. Outcomes of lung metastasis from pancreatic cancer: a nationwide multicenter analysis. J Hepato Biliary Pancreat. 2022;29(5):552–61.CrossRef
89.
Zurück zum Zitat Truty MJ, Kendrick ML, Nagorney DM, Smoot RL, Cleary SP, Graham RP, et al. Factors predicting response, perioperative outcomes, and survival following total neoadjuvant therapy for borderline/locally advanced pancreatic cancer. Ann Surg. 2021;273(2):341–9.PubMedCrossRef Truty MJ, Kendrick ML, Nagorney DM, Smoot RL, Cleary SP, Graham RP, et al. Factors predicting response, perioperative outcomes, and survival following total neoadjuvant therapy for borderline/locally advanced pancreatic cancer. Ann Surg. 2021;273(2):341–9.PubMedCrossRef
90.
Zurück zum Zitat Mokdad AA, Minter RM, Zhu H, Augustine MM, Porembka MR, Wang SC, et al. Neoadjuvant therapy followed by resection versus Upfront resection for resectable pancreatic cancer: a propensity score matched analysis. J Clin Oncol. 2017;35(5):515–22.PubMedCrossRef Mokdad AA, Minter RM, Zhu H, Augustine MM, Porembka MR, Wang SC, et al. Neoadjuvant therapy followed by resection versus Upfront resection for resectable pancreatic cancer: a propensity score matched analysis. J Clin Oncol. 2017;35(5):515–22.PubMedCrossRef
91.
Zurück zum Zitat van Roessel S, van Veldhuisen E, Klompmaker S, Janssen QP, Hilal AM, Alseidi A, et al. Evaluation of adjuvant chemotherapy in patients with Resected pancreatic cancer after neoadjuvant FOLFIRINOX treatment. JAMA Oncol. 2020;6(11):1733–40.PubMedCrossRef van Roessel S, van Veldhuisen E, Klompmaker S, Janssen QP, Hilal AM, Alseidi A, et al. Evaluation of adjuvant chemotherapy in patients with Resected pancreatic cancer after neoadjuvant FOLFIRINOX treatment. JAMA Oncol. 2020;6(11):1733–40.PubMedCrossRef
92.
Zurück zum Zitat Olecki EJ, Stahl KA, Torres MB, Peng JS, Dixon M, Shen C, Gusani NJ. Adjuvant chemotherapy after neoadjuvant chemotherapy for pancreatic cancer is associated with improved survival for patients with low-risk pathology. Ann Surg Oncol. 2021;28(6):3111–22.PubMedCrossRef Olecki EJ, Stahl KA, Torres MB, Peng JS, Dixon M, Shen C, Gusani NJ. Adjuvant chemotherapy after neoadjuvant chemotherapy for pancreatic cancer is associated with improved survival for patients with low-risk pathology. Ann Surg Oncol. 2021;28(6):3111–22.PubMedCrossRef
93.
Zurück zum Zitat Sugawara T, Rodriguez Franco S, Sherman S, Kirsch MJ, Colborn K, Ishida J, et al. Association of Adjuvant chemotherapy in patients with Resected pancreatic adenocarcinoma after multiagent neoadjuvant chemotherapy. JAMA Oncol. 2023;9(3):316–23.PubMedCrossRef Sugawara T, Rodriguez Franco S, Sherman S, Kirsch MJ, Colborn K, Ishida J, et al. Association of Adjuvant chemotherapy in patients with Resected pancreatic adenocarcinoma after multiagent neoadjuvant chemotherapy. JAMA Oncol. 2023;9(3):316–23.PubMedCrossRef
94.
Zurück zum Zitat Jones S, Zhang X, Parsons DW, Lin JC, Leary RJ, Angenendt P, et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science. 2008;321(5897):1801–6.PubMedPubMedCentralCrossRef Jones S, Zhang X, Parsons DW, Lin JC, Leary RJ, Angenendt P, et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science. 2008;321(5897):1801–6.PubMedPubMedCentralCrossRef
95.
Zurück zum Zitat Bailey P, Zhou X, An J, Peccerella T, Hu K, Springfeld C, et al. Refining the treatment of pancreatic cancer from big data to improved individual survival. Function. 2023;4(3):zqad11.PubMedPubMedCentralCrossRef Bailey P, Zhou X, An J, Peccerella T, Hu K, Springfeld C, et al. Refining the treatment of pancreatic cancer from big data to improved individual survival. Function. 2023;4(3):zqad11.PubMedPubMedCentralCrossRef
96.
Zurück zum Zitat Golan T, Hammel P, Reni M, Van Cutsem E, Macarulla T, Hall MJ, et al. Maintenance olaparib for germline BRCA-mutated metastatic pancreatic cancer. N Engl J Med. 2019;381(4):317–27.PubMedPubMedCentralCrossRef Golan T, Hammel P, Reni M, Van Cutsem E, Macarulla T, Hall MJ, et al. Maintenance olaparib for germline BRCA-mutated metastatic pancreatic cancer. N Engl J Med. 2019;381(4):317–27.PubMedPubMedCentralCrossRef
97.
98.
Zurück zum Zitat Perez-Mancera PA, Guerra C, Barbacid M, Tuveson DA. What we have learned about pancreatic cancer from mouse models. Gastroenterology. 2012;142(5):1079–92.PubMedCrossRef Perez-Mancera PA, Guerra C, Barbacid M, Tuveson DA. What we have learned about pancreatic cancer from mouse models. Gastroenterology. 2012;142(5):1079–92.PubMedCrossRef
99.
Zurück zum Zitat Kanda M, Matthaei H, Wu J, Hong SM, Yu J, Borges M, et al. Presence of somatic mutations in most early-stage pancreatic intraepithelial neoplasia. Gastroenterology. 2012;142(4):730–733.e9.PubMedCrossRef Kanda M, Matthaei H, Wu J, Hong SM, Yu J, Borges M, et al. Presence of somatic mutations in most early-stage pancreatic intraepithelial neoplasia. Gastroenterology. 2012;142(4):730–733.e9.PubMedCrossRef
100.
Zurück zum Zitat Baslan T, Morris JP, Zhao Z, Reyes J, Ho YJ, Tsanov KM, et al. Ordered and deterministic cancer genome evolution after p53 loss. Nature. 2022;608(7924):795–802.PubMedPubMedCentralCrossRef Baslan T, Morris JP, Zhao Z, Reyes J, Ho YJ, Tsanov KM, et al. Ordered and deterministic cancer genome evolution after p53 loss. Nature. 2022;608(7924):795–802.PubMedPubMedCentralCrossRef
101.
Zurück zum Zitat Shen S, Vagner S, Robert C. Persistent cancer cells: the deadly survivors. Cell. 2020;183(4):860–74.PubMedCrossRef Shen S, Vagner S, Robert C. Persistent cancer cells: the deadly survivors. Cell. 2020;183(4):860–74.PubMedCrossRef
104.
Zurück zum Zitat Xu Z, Hu K, Bailey P, Springfeld C, Roth S, Kurilov R, et al. Clinical impact of molecular subtyping of pancreatic cancer. Front Cell Dev Biol. 2021;9:743908.CrossRef Xu Z, Hu K, Bailey P, Springfeld C, Roth S, Kurilov R, et al. Clinical impact of molecular subtyping of pancreatic cancer. Front Cell Dev Biol. 2021;9:743908.CrossRef
105.
Zurück zum Zitat O’Kane GM, Grunwald BT, Jang GH, Masoomian M, Picardo S, Grant RC, et al. GATA6 expression distinguishes classical and basal-like subtypes in advanced pancreatic cancer. Clin Cancer Res. 2020;26(18):4901–10.PubMedCrossRef O’Kane GM, Grunwald BT, Jang GH, Masoomian M, Picardo S, Grant RC, et al. GATA6 expression distinguishes classical and basal-like subtypes in advanced pancreatic cancer. Clin Cancer Res. 2020;26(18):4901–10.PubMedCrossRef
Metadaten
Titel
Personalized treatment in localized pancreatic cancer
verfasst von
Prof. Dr. med. John P. Neoptolemos, MA, MB, BChir, MD, FRCS, FMedSci, MAE
Kai Hu, MD
Peter Bailey, PhD
Christoph Springfeld, MD, PhD
Baobao Cai, MD
Yi Miao, MD
Christoph Michalski, MD
Carlos Carvalho, MD
Thilo Hackert, MD
Markus W. Büchler, MD
Publikationsdatum
02.10.2023
Verlag
Springer Vienna
Erschienen in
European Surgery
Print ISSN: 1682-8631
Elektronische ISSN: 1682-4016
DOI
https://doi.org/10.1007/s10353-023-00814-x