Skip to main content
Erschienen in: European Surgery 3/2019

Open Access 15.04.2019 | main topic

Surgical and local therapeutic concepts of oligometastatic pancreatic cancer in the era of effective chemotherapy

verfasst von: Willem Niesen, Florian Primavesi, Silvia Gasteiger, John Neoptolemos, Thilo Hackert, PD Dr. Stefan Stättner

Erschienen in: European Surgery | Ausgabe 3/2019

Summary

Background

Pancreatic cancer (PC) remains a disease characterized by an extremely poor prognosis, which is often limited by advanced tumor stage at diagnosis. As surgery remains the only option for long-term survival, indications for resection to achieve a complete tumor removal have been extended in recent years, including locally advanced as well as metastatic disease.

Methods

Here, we provide a literature overview of modern multimodal treatment concepts of metastatic PC focused on surgery and local interventions including neoadjuvant concepts, stratification of patients, prognostic parameters, and oncological outcomes.

Results

The current literature lacks level I evidence studies on surgery in stage IV PC. The available observational studies show that resection for liver metastasis has been increasingly performed in recent years, outcomes improve after neoadjuvant therapy, and certain prognostic parameters can identify patients who benefit from this approach. In addition, interventional or radio-oncological liver-directed therapies have been evaluated showing the possibility of some disease control. Resection of pulmonary metastases is rarely performed, although this patient subgroup may have a more favorable prognosis than patients with stage IV liver cancer. Surgery in the setting of peritoneal carcinomatosis remains experimental without any valid supporting data.

Conclusions

There are promising data to support resection of metastatic PC, presuming this approach is embedded in a multimodal oncological concept with modern and effective multi-agent chemotherapies and proper patient selection. Based on this, future studies should specify distinct groups of patients who benefit from extended surgical approaches including synchronous or staged metastasectomy.
Hinweise
Willem Niesen and Florian Primavesi contributed equally to the manuscript.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Novel aspects
  • This review summarizes the available literature on surgical and interventional treatment of metastatic pancreatic cancer in the light of modern, effective chemotherapies.
  • Risk factors derived from studies are highlighted and a possible clinical algorithm is proposed.
  • An outlook on future perspectives regarding multimodal treatment of metastatic pancreatic cancer is provided.

Introduction

Despite enormous efforts and improvements in terms of effective chemotherapy regimens as well as the perioperative and surgical management of patients with pancreatic cancer (PC; [14]), the prognosis for the whole cohort suffering from this disease remains poor, with a 5-year overall survival (OS) of approximately 10% in central European countries such as Germany or Austria according to national statistics [5]. Since most patients present with metastatic disease or with locally unresectable, advanced tumors, only a small proportion of patients of around 20% are primarily classified amendable for surgical resection according to accepted guidelines [6, 7]. Therefore, palliative systemic chemotherapy represents the therapeutic mainstay in most cases. However, in some patients with PC, the metastatic spread at the time of the first diagnosis is limited in terms of number of nodules and organs involved, commonly defined as oligometastatic cancer [6, 8, 9]. This subset of cancers potentially depicts a transitional state between localized and widespread systemic involvement and also exhibits specific biological behavior, e. g., by expression of typical micro Ribonucleid acid (mi-RNA) patterns. Insights from oligometastatic situations in other cancer types such as colorectal malignancies with improved outcomes achieved by aggressive surgical resection in an environment of modern perioperative chemotherapies [10, 11] have triggered a spark of hope also in PC. Several single-center reports showed potentially beneficial outcomes when progressive surgery for extra-regional lymph node or liver metastases (LM) was performed in PC [1214]. This has also been suggested by a retrospective European multicenter study [15] and a recent analysis of the surveillance, epidemiology and end results (SEER) database in the United States, showing a potentially improved survival in a subset of patients with isolated liver and lung metastases [16]. Furthermore, successes reported for poly-chemotherapy combined with progressive surgery in locally advanced or borderline-resectable PC [1, 1719] as well as adjuvant treatment [20, 21] have once more inspired interdisciplinary discussions on the selection criteria and usefulness of curative-intent surgical resection for oligometastatic PC in the era of effective, modern chemotherapies [22]. However, no high-level evidence via prospective trials or meta-analyses has been generated on this topic to date, as studies are ongoing [23]. Also, biomarkers or clinical factors guiding treatment algorithms are not well defined [24]. The present narrative literature review summarizes available oncosurgical reports on potentially resectable oligometastatic PC and compares these outcomes with other local treatments such as liver-directed stereotactic body radiotherapy (SBRT), selective internal radiation therapy (SIRT), or radiofrequency ablation (RFA; [25, 26]) to propose a treatment algorithm (Fig. 1) and also gives an outlook on potential factors and biomarkers for stratification.

The age of effective chemotherapies in pancreatic cancer

Influence of chemotherapy on resectable, borderline-resectable and unresectable PC

Presently, the use of neoadjuvant chemotherapy alone or in combination with radiotherapy in patients with resectable, borderline-resectable, and locally advanced PC is a field of intense studies. Before considering neoadjuvant therapy and surgery, the general and performance status of the patient has to be evaluated after accurate staging to assess the presence and volume of any metastatic disease as well as the local resectability status [27]. Contrast-enhanced computed tomography (CE-CT) is the most commonly used imaging modality for the basic examination, which may be supplemented by magnetic resonance imaging (MRI) if LM are suspected as well as 18fluorine-2-fluoro-2-deoxy-d-glucose positron emission tomography with computed tomography (18FDG-PET–CT) scanning, which is more sensitive for the detection of widespread metastatic disease [28, 29].
The consideration of neoadjuvant treatment in primary resectable PC is to treat micro-metastatic disease at an early stage and to increase the rate of complete microscopic tumor clearance. To date, there is no high-level evidence for this concept; however, a number of randomized controlled trials on this topic are ongoing. Among others these include the PANACHE01-PRODIGE48 trial (four cycles of modified 5‑FU, FA, irinotecan, and oxaliplatin [mFOLFIRINOX] or 5‑FU, FA, and oxaliplatin [FOLFOX] to upfront surgery followed by adjuvant chemotherapy; [30]) and the NEONAX trial (gemcitabine/nab-paclitaxel pre- and postoperatively vs. upfront resection and only adjuvant therapy; [31]).
Neoadjuvant therapy for borderline-resectable PC is currently practiced in many centers, although—similar to resectable PC—sufficient evidence is lacking. The most recent PREOPANC trial included borderline patients randomized to neoadjuvant therapy (n = 63) vs. upfront resection (n = 58; [32]). Regarding the results, there was a significant benefit for neoadjuvant therapy of 29.9 months vs. 16.8 months after upfront resection plus adjuvant chemotherapy. However, when compared with the results of the ESPAC 4 study (28.4 months median survival with upfront resection + adjuvant treatment; [21]), survival was not strikingly improved in the neoadjuvant PREOPANC arm, therefore raising the question regarding the true advantages of this concept. A multicenter randomized controlled phase II/III trial from Korea investigated neoadjuvant chemoradiotherapy and chemotherapy (with adjuvant chemoradiotherapy and chemotherapy) compared with adjuvant chemoradiotherapy and chemotherapy alone [33]. After 58 patients had been enrolled, an interim analysis was performed to show a statistically significant treatment efficacy for the neoadjuvant arm and the trial was terminated. Finally, there were only eight and six patients, respectively, who actually completed the per-protocol treatment, rendering the data highly unstable and no conclusion possible.
In contrast to resectable and borderline-resectable PC, locally advanced PC certainly represents a tumor stage requiring neoadjuvant therapy, although no high-quality randomized trials are published. However, there is considerable interest in this approach given remarkable findings in large single-center reports. A series of 575 consecutive patients with unresectable PC who had undergone neoadjuvant therapy found that 292 (50.8%) patients could undergo resection using advanced surgical techniques [1]. Neoadjuvant treatment protocols included FOLFIRINOX (n = 125), gemcitabine and radiotherapy (n = 322), and other regimens (n = 128). The highest resection rates were achieved after FOLFIRINOX (60.8%) and median OS and 3‑year survival rates were 15.3 months and 23.0% after resection compared with 8.5 months and 2.4% after exploration alone (p < 0.0001). With regard to metastatic PC (mPC) in this study, metastases were still present at the time of resection in 38% of patients in the FOLFIRINOX group, in 7% in the gemcitabine and radiotherapy group, and in 17% of patients receiving other regimens (p < 0.0001). Median survival after resection in the three groups was 16.0, 16.5, and 14.5 months, respectively. Multivariable analysis suggested that FOLFIRINOX was an independent prognostic factor compared with the other regimens. As survival after resection was not different, this underlines that resection itself is the crucial prognostic factor and that a stage of resectability can most effectively be achieved by FOLFIRINOX even in the metastatic setting. This is consistent with a recent individual patient data meta-analysis showing that patients with locally advanced PC treated with FOLFIRINOX had a median OS of 24.2 months that is far superior to previously reported median OS with gemcitabine in these patients [34].

Palliative setting studies

The history of palliative chemotherapy treatment for locally advanced and metastatic disease changed fundamentally in 1997 when Burris et al. published their randomized trial comparing gemcitabine with 5‑FU [35]. In this study of 126 patients, gemcitabine was the first agent demonstrating a significant survival benefit over 5‑FU, increasing the median survival from 4.4 to 5.7 months (p = 0.0025) and the dismal 1‑year survival of 2% after 5‑FU to 18% after gemcitabine. Furthermore, the generally good tolerability in patients resulted in gemcitabine becoming the standard agent of care in palliative treatment for almost 20 years. Meanwhile combining different agents like platinum-based therapies with gemcitabine did not show any superiority compared with gemcitabine mono use, although meta-analyses on that topic showed slightly improved survival for gemcitabine combined with platin-based agents [3638].
It was not until 2011 that the results of the ACCORD11/PRODIGE4 trial changed the general treatment for metastatic disease entirely. Conroy et al. recruited 342 patients with either irresectable or mPC, comparing FOLFIRINOX and gemcitabine. The median survival in the gemcitabine group was 6.8 months and thus comparable to the data of Burris et al. [35], whereas the use of FOLFIRINOX resulted in an increased survival of 11 months (p < 0.001). The superiority of FOLFIRINOX over gemcitabine was also seen in the response rate increase of 9.4% for gemcitabine to 31.6% for FOLFIRINOX. The study population, however, represented a (highly) selected fit of patients, i. e., only patients with limited cardiac comorbidities, age 76 or younger, and an Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1 were included. The price for the higher survival rates of FOLFIRINOX was increased toxicities, i. e., grade III/IV effects.
Several attempts to decrease the toxicities of FOLFIRINOX were made since Conroy’s trial was published. Therefore, the use of a modified FOLFIRINOX (mFOLFIRINOX) has been analyzed by various groups yielding similar results with regard to survival [39]. Nevertheless, there are no data available comparing the initially proposed regimen with the modified scheme, which is why FOLFIRINOX is still the standard of care for patients in a good health state who are competent to undergo an aggressive, yet potent therapy.
Besides FOLFIRINOX, the use of nanoparticle albumin-bound paclitaxel (Nab-paclitaxel) in combination with gemcitabine was compared with gemcitabine monotherapy in 861 patients with mPC in 2015 [40]. Combining Nab-paclitaxel with gemcitabine increased the OS from 6.7 months for gemcitabine monotherapy to 8.5 months (p < 0.001). The response rate was also increased from 7% for gemcitabine to 23% in the gemcitabine with Nab-paclitaxel group giving two potent options for mPC.

Surgery for oligometastatic pancreatic cancer

Liver metastases

Current guidelines do not recommend surgery for metastatic disease. This recommendation is derived from the generally poor prognosis and especially the lack of randomized controlled trials. Thus, the available data are limited to retrospective analyses, case series, and case reports [14]. However, high-volume centers tend to push boundaries and perform partial hepatectomy combined with pancreatic resections or metachronous liver resections in cases of isolated hepatic recurrence. A systematic review published in 2008 [41] included three case reports, and 21 retrospective case series, and studies that included up to 22 patients with heterogeneous results, showing that this approach was applied only in a very small number of patients.
However, a recently published European multicenter retrospective analysis included 69 patients who underwent pancreatic and simultaneous hepatic resection for mPC in six European pancreas centers [15]. These 69 patients were matched to patients who did not receive curative resection with regard to several factors, e. g., number of hepatic metastases, age, sex, and tumor stage. Unsurprisingly, the OS in the resection group was significantly better than in the control group with a median survival of 14.5 months for curative surgery while the control group had a median survival of 7.5 months (p < 0.001). The 5‑year survival rate was 0% for palliative patients and 5.8% for resected patients. Of note, 65% (n = 35) of patients underwent adjuvant treatment with gemcitabine, a regimen inferior to modern treatments such as (m)FOLFIRINOX. It can be assumed that the age of modern and more effective chemotherapy protocols will have a further effect on survival in the very near future.
A series published in 2016 analyzed 127 patients with PC LM treated with palliative chemotherapy [42]. A subgroup of 11 of those patients eventually showed a good response after neoadjuvant treatment making them eligible for curative resection. The chemotherapy protocols used varied, including FOLFIRINOX, GEMOX, and others. The OS of these 11 patients was 39 months while the palliative control group had a median OS of 11 months (p < 0.0001).
The largest monocenter analysis from Heidelberg published in 2017 included 85 patients with either synchronous or metachronous LM [12]. Resection types varied from atypical to extended hepatectomies with acceptable morbidity and mortality. The median survival for patients with synchronous LM was 10.6 months, for patients undergoing liver surgery for metachronous LM it was 14.8 months (p = 0.210). For all patients combined (n = 85), the median survival was 12.3 months, comparable to the above multicenter analysis. Although median survival data do not seem too encouraging at first glance, long-term survival was observed after metastasectomy with 8.1% 5‑year OS. By contrast, even modern chemotherapy treatment cannot achieve long-term survival in mPC.
A common feature of both the Heidelberg study and the European registry is that only a minority of patients underwent neoadjuvant therapy before LM resection was performed. By contrast, three other reports showed much longer survival rates when all of the analyzed patients received neoadjuvant therapy prior to undergoing resection [4244]. This observation is certainly explained by the selection effect of neoadjuvant treatment, which is exemplarily shown in the study by Crippa et al. [42]. Although only 11 patients eventually underwent metastasectomy in this collective, survival was impressively high when multiagent chemotherapy was used, the number of resected metastases did not exceed five lesions, and CA 19-9 levels showed a reduction of >50% during neoadjuvant therapy. Interestingly, in one study from Verona, patients with hepatic metastases had exploration laparotomy after preoperative chemotherapy and underwent isolated primary tumor resection only in cases with complete intraoperatively vanished liver lesions, and therefore did never receive hepatectomy [43]. Still, the resulting median OS in these 24 patients was 13 months after pancreatic resection and 56 months after the initial diagnosis of metastases. Table 1 summarizes the most recent studies on hepatic metastasectomy together with pancreatic resection in a synchronous or metachronous setting.
Table 1
Studies of patients undergoing resection for metastatic pancreatic cancer
Author
Year
Inclusion period
Center
N
Type of metastases
Mortality (%)/morbidity (%)
Max. size (cm)
Lesions (n)
Primary tumor resected?
Median follow-up (mo)
Median OS (mo) after resection
Median RFS (mo)
Recurrence (%)
(Independent) factors for poor survival
Zanini N et al. [14]
2015
2003–2014
Bologna
15
Liver
0/60
3.5
1–3
All
12.6
9.1
5.2
93
Timing (synchronous), >1LM
Crippa S et al. [42]
2016
2003–2013
Milan and Ancona
11
Liver
0/27
n.a.
1–5+
All
n.a.
39 (from diagnosis)
n.a.
91
>5LM, <50% CA 19-9 reduction, Gem monotherapy
Wright P et al. [44]
2016
2008–2013
Pittsburgh and Baltimore
23
Liver, Lung, Peritoneum
0/13 (major)
n.a.
1–5+
All
30
18.2
8.6
74
n.a.
Tachezy M et al. [15]
2016
1994–2014
European Multicenter
69
Liver
1/68
n.a.
1–11
All
n.a.
15
n.a.
n.a.
Tumor located in pancreatic tail
Frigerio I et al. [43]
2017
2007–2015
Verona
24
Liver (all complete hep. response)
No liver resection
n.a.
1–2+
All
31
13 (after resection)
56 (after diagnosis)
11 (after surgery)
21 (after diagnosis)
75
None
Hackert T et al. [17]
2017
2001–2014
Heidelberg
128
Liver (85), ILN (43)
2.9/45
4.3/22 (syn./met.)
75% ≤2 cm
1–3+
All
n.a.
12.3
n.a.
n.a.
None
CA 19-9 Carbohydrate Antigen 19-9, Gem gemcitabine, hep hepatic, ILN inter-aortocaval lymph nodes, LM liver metastases, N number of patients included in (sub)analysis, n.a. data not available, met. metachronous disease, mo months, OS overall survival, RFS recurrence-free survival, syn. synchronous disease

Pulmonary and peritoneal metastases

The available data on pulmonary metastases and surgical treatment are even more limited than those for LM. However, the few available studies suggest that patients who develop isolated pulmonary metastases represent a favorable subgroup in mPC over patients with distant metastases in the liver or peritoneum [45].
A recently published trial [46] analyzed 12 patients with isolated pulmonary metastases. While one patient underwent pulmonary resection despite local inoperable primary disease, the other 11 patient’s curative intent procedures varied from atypical wedge resections up to lobectomy. The median survival after diagnosis of pulmonary metastases was 47 months (6–66 months) with reported 3‑year and 5‑year survival rates of 62.3 and 31.2%, respectively. Taking the time span between primary pancreatic resection and survival after metastasectomy into consideration, the estimated 5‑year OS was 82.5%. Although these data are very limited with a small number of patients who are both biologically and medically selected, no similar data are available for palliative chemotherapy treatment.
Patients who develop peritoneal spread generally have a very dismal prognosis with survival rates ranging between weeks and several months. Furthermore, local complications such as jaundice, ascites, malnutrition, and eventually cachexia often obviate the administration of palliative chemotherapy. The location itself inside the peritoneum results in low chemotherapy effectiveness. The group of Satoi et al. conducted a multicenter trial including 33 patients with either macroscopic peritoneal spread or positive wash cytology in PC [47]. The chemotherapy agent administered intravenously was S‑1 while the protocol included intraperitoneal application of paclitaxel every 3 weeks for a median observational time of 8.8 months. Considering the devastating prognosis of peritoneal metastases in PC patients, the results of this trial were extremely encouraging with 16.3 months median survival and 62% 1‑year survival rate. The response rates were 36% partial response, 46% stable disease, 6% progressive disease, and in 12% response was not available. It has to be added that the efficacy of S‑1 is different between the Asian and Caucasian population due to differences in the cytochrome P‑450 2A6 activity eventually resulting in higher toxicity in Caucasians. Therefore, adjustments in the S‑1 protocol are needed for other ethnicities to evaluate the promising effects.

Nonsurgical, local treatment of oligometastases

Experience with nonsurgical, local treatment is limited in mPC compared with other malignancies such as metastatic colorectal cancer or primary liver cancers, where techniques like RFA, microwave ablation (MWA), or transarterial chemoembolization (TACE) have become widely accepted modalities in treatment algorithms [4850]. Furthermore, studies of stage IV PC have to be interpreted with caution, since most of these publications are single-arm, proof-of-principle studies and no randomized controlled trial exists comparing nonsurgical hepatic treatment vs. liver resection or systemic therapy alone. Also, in the majority of cases the primary tumor has not been surgically removed [6]. An early series from Johns Hopkins and the Mayo Clinic (USA) published in 2010 gives an overview of the utilization of different liver-directed treatments as an adjunct to pancreatic resection in mPC and other malignancies necessitating pancreatic head resections [51]. The authors state that pancreaticoduodenectomies plus liver-directed therapies are associated with considerable morbidity of >34% (more than 50% major complications) with an overall mortality of 2.4%. Intriguingly, hepatic abscesses are especially common after two-stage resection and local therapy (14.5% vs. 7%).
Table 2 gives a summary of studies evaluating specific locoregional treatments for mPC including data on postinterventional complications and oncological survival. In summary, most series are retrospective reports of far less than 100 patients with inhomogeneous inclusion criteria with details on hepatic tumor number and size often missing. Most commonly, median OS ranges from 5.5 to 14 months, only rarely surpassing 16–20 months in selected cohorts [52, 53], as principally all patients either experience local or distant recurrence after RFA/TACE or do not reach complete response after SIRT or local radiotherapy.
Table 2
Studies of patients undergoing locoregional treatment for pancreatic cancer metastases
Author
Year
Inclusion period
Centre
N
Type of intervention
Mortality (%)/morbidity (%)
Max. size (cm)
Lesions (n)
Primary tumor resected?
Median follow-up (mo/range)
Median OS (mo)
Median RFS (mo)
Recurrence (%)
(Independent) factors for poor survival
Park JB et al. [26]
2012
2002–2009
Seoul
34
RFA (liver)
0/12 (all mild)
3.2
1–4
Always
15 (3–65)
14
n.a.
>91
G3, >1LM, ≥2 cm LM
Hua Y-Q et al. [54]
2017
2012–2015
Shanghai
102
RFA (liver)
0/10 (all mild)
5.0
1–3+
Never
21 (4–43.8)
11.4
n.a.
82.4
Primary tumor location (head), ≥3 cm LM, NLR ≥2.5
Sun J-H et al. [55]
2017
2009–2015
Hangzhou
18
TACE (liver)
0/>85 (all mild)
n.a.
n.a.
<50% of cases
n.a.
9.2
n.a.
n.a.
Extrahepatic disease
Azizi et al. [52]
2011
2002–2007
Frankfurt
32
TACE (liver)
0/n.a.
n.a.
60% >5
Some (% n.a.)
n.a.
16 (10% 5y)
n.a. (10% 5y)
~90
Progressive disease during TACE, >5 LM
Huang Z‑M et al. [56]
2013
1998–2011
Guangzhou
71
TACE + RFA + seeds (pancreas + liver)
0/n.a.
n.a.
54% >3
Never
n.a.
11 (6% 3y)
n.a. (no CR)
n.a. (no CR)
>3 LM
Michl M et al. [57]
2014
2004–2011
Munich
19
SIRT (liver)
11/74
n.a.
n.a.
>70%
n.a.
9
2.6 mo systemic PFS
n.a. (no CR)
Increased CA 19-9, CRP, extrahepatic disease
Kim A et al. [53]
2016
2012–2015
Washington
16
SIRT (liver)
0/13 (all < grade 4)
n.a.
n.a.
1 patient
n.a.
20 after LM
12.5 after SIRT
3.4 mo systemic PFS
n.a.
n.a.
Gibbs P et al. [58]
2015
2006–2009
Melbourne
14
SIRT (liver)
7/57 (grade 3/4)
n.a.
1–5
29%
n.a.
5.5
4.4 mo systemic PFS
n.a. (no CR)
Primary tumor in situ
Mahadevan A et al. [25]
2018
2005–2017
25 centers (LM registry)
20
SBRT (liver)
0/n.a. (no severe)
n.a.
n.a.
n.a.
n.a.
6
n.a.
All had local progress
Tumor volume <40 cm3
Wieners G et al. [59]
2015
n.a.
Berlin
20
HDRBT
0/15 (severe)
7.3
1–3+
n.a.
14 (mean)
8.6
4.9 mo systemic PFS
10% local recurrence
No chemotherapy received after HDRBT
CA 19-9 Carbohydrate Antigen 19-9, CR complete response, CRP C-reactive protein, HDBRT high-dose-rate brachy-radiotherapy, LM liver metastases, N number of patients included in (sub)analysis, n.a. data not available, NLR neutrophil-to-lymphocyte ratio, mo months, OS overall survival, PFS progression-free survival, RFA radiofrequency ablation, RFS recurrence-free survival, TACE transarterial chemoembolization, SIRT selective internal radiation therapy

Liver-directed therapies

Radiofrequency ablation In a Korean series published 2012, a total of 34 patients underwent ultrasound-guided hepatic RFA either intraoperatively with simultaneous primary tumor resection or in a two-stage approach after pancreatic surgery [26]. The number of lesions treated ranged from one to four with a maximum diameter of 3.2 cm, resulting in a median survival after diagnosis of LM of 14 months. About half of the patients underwent re-ablation of recurring LM. Intriguingly, at the time of analysis, one patient was alive and free from recurrence for 44 months after RFA, showing that long-term survival is possibly in exceptional cases of mPC. This study identified the presence of a single <2-cm-diameter LM and good or moderate differentiation (G1 or G2) as factors associated with favorable OS. The major limitation in the study besides the single-arm design was the low number of pathological confirmations of LM of only 27% of patients. However, the authors state that concurrent CA 19-9 increment and post-ablational metastatic recurrence in almost all patients proves the presence of mPC.
The second, recently published study on RFA for hepatic oligometastatic PC comes from the Fudan University Shanghai Cancer Center in China including patients with synchronous LM between 2012 and 2015 [54]. In the study period a total of 469 patients with LM from mPC presented to the department, of whom 102 (22%) were amenable for treatment with RFA, showing that less than one fourth of LM patients do indeed have oligometastatic disease. The main endpoint in this study was analysis of safety and efficacy, through CT or MRI imaging after 1 month of treatment. In a total of 145 ultrasound-guided RFA sessions, in 95% of cases a radiologically complete tumor ablation was achieved. The overall morbidity rate reported was 10%, all manageable with conservative treatment, with no severe complications. After a median follow-up of 21 months, the median OS from diagnosis of LM was 11.4 months. Factors associated with poor survival analyzed with multivariable regression were a location of the primary tumor in the pancreatic head, a maximum diameter of LM of ≥3 cm, and a neutrophil-to-lymphocyte ratio (NLR) of ≥2.5. In this series, no patient underwent primary tumor resection, limiting comparability with the aforementioned publication from Korea.

Transarterial chemoembolization

In a study of 27 patients (18 with LM from PC and 9 cases with neuroendocrine tumors [NET]), authors from Hangzhou, China examined the efficacy and survival outcome after TACE [55]. In summary, 52 TACE sessions (1–7) were applied, including eight treatments with drug-eluting beads (DEB) after an average time of 4.5 months after diagnosis of LM. In less than half of the patients, the primary tumor was resected, 14 patients had extrahepatic lesions, and three patients had undergone partial liver resection before, four had previous RFA or HIFU (high intensity focused ultrasound). Only 12 patients did not receive any other treatment than TACE. Almost all patients experienced mild complications such as bone marrow suppression, epigastric pain, or lack of appetite. No severe complications or mortality occurred related to the procedures. The OS in the mPC group was 9.2 months (vs. 50.1 months in NET patients). The main risk factor for poor survival was extrahepatic involvement.
Another series from Frankfurt, Germany suggested that repetitive TACE may result in a relevant response in PC-LM [52]. In summary, 32 patients who had not undergone any other ablative therapies previously and had no extrahepatic disease, received a mean of 3.2 TACE sessions (range 2–4) and underwent retrospective evaluation. No major complications occurred, the treatment response according to RECIST criteria was stable disease (SD) in 72% and partial response (PR) in 9.4%, and no patient experienced complete response. The median OS calculated from the first TACE session was 16 months, and was significantly better for patients with SD or PR (20 months) during TACE treatment compared with those with progressive disease (5 months). Also, median OS was better in patients with oligonodular disease (≤5 LM; 20 months) compared with multinodular mPC (11 months).
In another study from Guangzhou, China, 71 patients with unresectable PC primary tumor with (n = 51) or without (n = 20) LM underwent primarily TACE (mean 3 sessions, range 2–6; [56]). In the case of residual lesions in the primary tumor site, the liver, or metastatic lymph nodes, this was followed by RFA (24 patients) or radioactive seed implantation (n = 24) or both (n = 31). The resulting median OS was 11 months after diagnosis of LM, and was significantly better for patients with oligonodular (≤3 LM) metastases (18 months) compared with multinodular disease.

Selective internal radiation therapy

In 2014, radioembolization with Yttrium-90 microspheres in PC-LM was evaluated regarding efficacy, safety, and prognostic factors in 19 patients by a German group [57]. Most patients had previously undergone systemic therapy with gemcitabine-based regimens. Objective response in the liver was 47%, the median local progression-free survival (PFS) in the liver was 3.4 months and median OS was 9.0 months. No severe adverse short-term events occurred; however, in the long-term, liver abscesses, gastroduodenal ulceration, cholangitis, ascites, and spleen infraction were observed. In particular, two patients later died due to liver abscess and radioembolization-induced liver disease, necessitating careful long-term observation. Increased CA 19-9 and CRP were associated with shorter OS.
Another retrospective single-center series from Washington, DC analyzed 16 patients with liver-dominant mPC (>50% had extrahepatic disease) undergoing SIRT, 15 of whom also had received systemic chemotherapy, while only one patient underwent primary tumor resection [53]. Of these patients, 69%had SD or PR in the liver. No treatment-related grade 4 or 5 toxicities were observed, and the resulting median OS was 22 months since diagnosis of LM and 12.5 months after SIRT therapy.
Most recently, a phase II study from Melbourne, Australia was published with 14 patients, of whom ten had a primary tumor in situ and eight had liver-limited metastases [58]. Median PFS in the liver was 5.2 months and 4.4 at any site, and was prolonged in those with resected primary tumor (7.8 months vs. 3.4 months). Median OS was 5.5 months in all patients (13.6 months with resected primary tumor). Grade 3/4 adverse events occurred in 57% of patients within 60 days, and there was one death related to posttreatment liver failure and another one with unclear underlying cause.

SBRT or CT-guided brachytherapy

In a recently published international, multi-institutional registry of SBRT for 427 patients with LM of a number of different entities, 20 patients with mPC were also included [25]. Median OS was 6 months in these patients compared to, for example, 27 months in colorectal or 21 months in breast cancer (p < 0.0001). Although local control after 12 months in mPC was comparable to other entities with an encouraging 70%, it rapidly decreased thereafter with all patients experiencing recurrence in the study period. In the whole cohort, small tumors (<40 cm3) had improved local control rates compared with larger-volume tumors. Although toxicity data were not available from all participating studies, there was no grade 3 or higher adverse event reported from any institution.
Computed tomography-guided high-dose-rate brachytherapy (CT-HDRBT) is an interventional technique applying radiation through a percutaneous brachycatheter into targeted lesions. A study with 20 consecutive patients with 49 PC-LM retrospectively evaluated local tumor control, safety, and efficacy of this technique applied with a single-fraction high dose (15–20 Gy; [59]). With a mean diameter of 29 mm (10–73) and mean radiation time of 20 min (7–42), a mean coverage of 98% of the clinical target volume was reached. Three major complications (20%) occurred in terms of abscesses, all in patients with biliodigestive anastomosis. After a mean follow-up of 13.7 months, the median PFS was 4.9 months, with a local recurrence in 10% of 49 metastases. The median OS was 8.6 months after treatment.

Other nonsurgical local treatments

Other techniques currently evaluated for non-resectable mPC include electrochemotherapy [60], high-intensity ultrasound [61], and irreversible electroporation [62]. So far, only small series exist with very heterogeneous inclusion criteria and varying utilization of concomitant surgical resection, resulting in a median OS between 13 and 16 months for these therapies in mPC.

Prognostic factors for patient selection

Clinical factors and blood markers in mPC patients

In an attempt to evaluate established classic blood parameters as prognostic factors for survival compared to classic tumor markers like CA 19-9 (carbohydrate antigen 19-9), a German multicenter analysis pooled data from two phase II chemotherapy trials and one prospective high-volume center registry [63]. In a total of 291 patients with locally advanced and metastatic PC, the authors confirmed high baseline CA 19-9 to be associated with time-to-progression (TTP) and increased bilirubin as well as pretreatment CRP with poor OS in multivariable analysis. In this study, advanced age (>64 years) and decreased performance index (Karnofsky index ≤80%) were also linked to shortened TTP and OS, respectively. The participating high-volume center in a separate publication also reported increased pretreatment cytokeratin 19-fragments (CYFRA 21-1) to be associated with poor OS and TTP in 78 prospectively recorded, advanced PC patients [64]. In a multivariable Cox model, CYFRA 21-1 was shown to be an independent prognostic factor in addition to CA 19-9 and Karnofsky index, also providing prognostic information in Lewis antigen-negative (about 5–10%) or cholestatic individuals, cases that are both associated with unreliable CA 19-9 results.
Markers of inflammation have been shown to be predictors of poor survival in many cancer types. In mPC, NLR (cutoff: ≥3.75) was confirmed to be associated with decreased OS and may serve as a useful combination marker to enhance the prognostic value of CA 19-9 (cutoff: ≥626 U/ml) alone, with an Area under the Curve (AUC) of 0.843 when these two values were combined [65]. The prognostic value of NLR has also been confirmed in a recent Italian study, with an NLR cutoff of >5 [66]. Regarding the prognostic value of preoperative C‑reactive protein (CRP), evidence for some time has been less clear since studies on resectable PC reported conflicting results, as summarized in a previous systematic review [67]. However, it was shown in a recent report from Shanghai of 386 patients with PC of whom 258 had TNM stage III or IV cancers, that a CRP/albumin ratio of ≥0.180 was significantly associated with poor OS in these advanced stages, whereas there was no difference in early-tumor stage patients. Also, in an earlier study from the year 2000, a prognostic index including CRP ≥5 mg/dl was associated with poor median OS [68].
In a large Chinese multicenter trial, a nomogram including decreased performance status, presence of LM, increased CA 19-9, high absolute neutrophil count, and decreased albumin was created to predict OS in mPC patients and validated in two cohorts of external Caucasian patient populations [69]. According to the nomogram score, the three resulting subgroups (high-, intermediate- and low-risk) showed significantly different median OS of approximately 4 vs. 7 vs. 11 months (p < 0.001) in both the training and validation set.
All of the aforementioned non-tumor marker factors (performance index ECOG ≥2, albumin <35 g/l, CRP >10 mg/l, or NLR >5) were also confirmed as prognostic for OS in a large Korean study of 343 patients with stage IV disease [70]. The authors furthermore found that the initial metastatic site was prognostic: While extrahepatic-limited disease had a median OS of 7.5 months, isolated LM showed inferior OS of 4.8 and simultaneous extrahepatic and hepatic involvement had the worst prognosis of 2.4 median OS.
Table 3 summarizes blood markers as well as patient and tumor factors associated with poor survival in mPC.
Table 3
Factors associated with worse overall survival or time-to-progression in metastatic pancreatic cancer patients
 
Examples of cutoffs evaluated in studies
Blood markers
CA 19-9
≥200 or ≥626 or ≥1000 U/ml
CEA
≥2.9 or ≥4.5 ng/ml
CYFRA 21-1
≥2.7 ng/ml
Neutrophil–lymphocyte ratio
≥3.75 or >5
CRP
≥1 mg/dl
Bilirubin
>1 mg/dl
Albumin
<35 g/L
Patient or tumor factors
Poor performance index
ECOG >0 or ECOG ≥2, Karnofsky index ≤80%
Metastatic spread
Hepatic involvement compared with isolated extrahepatic
CA 19-9 Carbohydrate Antigen 19-9, CEA carcinoembryonic antigen, CRP C-reactive protein, CYFRA 21-1 cytokeratin 19-fragments, ECOG Eastern Cooperative Oncology Group performance status

New biomarkers

Future diagnostic tools such as circulating tumor cells and serum micro-RNAs to predict resectability, prognosis, and response to chemotherapy are continuously evaluated in ongoing studies [7174]. Another review on “Molecular biology in pancreatic ductal adenocarcinoma” by Søreide K et al. in this journal gives further insight into this promising new field [75].

Conclusion

In conclusion, PC therapy has undergone substantial changes during the past decade with regard to the use of effective chemotherapy regimens in the neoadjuvant as well as the adjuvant setting. These approaches have been shown to improve patients’ prognosis in all situations. However, surgery still remains the key to long-term survival in PC, and with the standardization and centralization of pancreatic surgery, surgical outcomes have also improved. These developments have led to more extended surgical indications in PC, and patients with tumor stages that were historically defined as palliative are increasingly regarded as potential candidates for surgery in a multimodal therapy setting. Especially for locally advanced PC and oligometastatic disease, there are promising survival results if these patients undergo surgery after proper stratification and selection. Regarding the latter, this seems to be an essential task for future studies and research since currently available markers and imaging modalities still fail to predict tumor biology accurately and are therefore of only limited reliability in identifying patients who will benefit from extended surgical approaches with possible long-term survival.

Conflict of interest

W. Niesen, F. Primavesi, S. Gasteiger, J. Neoptolemos, T. Hackert, and S. Stättner declare that they have no competing interests.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

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 Hackert T, Sachsenmaier M, Hinz U, et al. Locally advanced pancreatic cancer: Neoadjuvant therapy with Folfirinox results in resectability in 60% of the patients. Ann Surg. 2016;264(3):457–63.CrossRef Hackert T, Sachsenmaier M, Hinz U, et al. Locally advanced pancreatic cancer: Neoadjuvant therapy with Folfirinox results in resectability in 60% of the patients. Ann Surg. 2016;264(3):457–63.CrossRef
2.
Zurück zum Zitat Hartwig W, Gluth A, Hinz U, et al. Outcomes after extended pancreatectomy in patients with borderline resectable and locally advanced pancreatic cancer. Br J Surg. 2016;103(12):1683–94.CrossRef Hartwig W, Gluth A, Hinz U, et al. Outcomes after extended pancreatectomy in patients with borderline resectable and locally advanced pancreatic cancer. Br J Surg. 2016;103(12):1683–94.CrossRef
3.
Zurück zum Zitat Gangl O, Sahora K, Kornprat P, et al. Preparing for prospective clinical trials: A national initiative of an excellence registry for consecutive pancreatic cancer resections. World J Surg. 2014;38(2):456–62.CrossRef Gangl O, Sahora K, Kornprat P, et al. Preparing for prospective clinical trials: A national initiative of an excellence registry for consecutive pancreatic cancer resections. World J Surg. 2014;38(2):456–62.CrossRef
4.
Zurück zum Zitat Schindl M, Függer R, Götzinger P, et al. Randomized clinical trial of the effect of a fibrin sealant patch on pancreatic fistula formation after pancreatoduodenectomy. Br J Surg. 2018;105(7):811–9.CrossRef Schindl M, Függer R, Götzinger P, et al. Randomized clinical trial of the effect of a fibrin sealant patch on pancreatic fistula formation after pancreatoduodenectomy. Br J Surg. 2018;105(7):811–9.CrossRef
6.
Zurück zum Zitat Gebauer F, Damanakis AI, Bruns C. Oligometastasis in pancreatic cancer: Current state of knowledge and spectrum of local therapy. Chirurg. 2018;89(7):510–5.CrossRef Gebauer F, Damanakis AI, Bruns C. Oligometastasis in pancreatic cancer: Current state of knowledge and spectrum of local therapy. Chirurg. 2018;89(7):510–5.CrossRef
7.
Zurück zum Zitat Ducreux M, Cuhna AS, Caramella C, et al. Cancer of the pancreas: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2015;26(Suppl 5):v56–v68.CrossRef Ducreux M, Cuhna AS, Caramella C, et al. Cancer of the pancreas: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2015;26(Suppl 5):v56–v68.CrossRef
8.
Zurück zum Zitat Reyes DK, Pienta KJ. The biology and treatment of oligometastatic cancer. Oncotarget. 2015;6(11):8491–524.CrossRef Reyes DK, Pienta KJ. The biology and treatment of oligometastatic cancer. Oncotarget. 2015;6(11):8491–524.CrossRef
9.
Zurück zum Zitat Lussier YA, Xing HR, Salama JK, et al. MicroRNA expression characterizes oligometastasis(es). PLoS ONE. 2011;6(12):e28650.CrossRef Lussier YA, Xing HR, Salama JK, et al. MicroRNA expression characterizes oligometastasis(es). PLoS ONE. 2011;6(12):e28650.CrossRef
10.
Zurück zum Zitat Evrard S, Poston G, Kissmeyer-Nielsen P, et al. Combined ablation and resection (CARe) as an effective parenchymal sparing treatment for extensive colorectal liver metastases. PLoS ONE. 2014;9(12):e114404.CrossRef Evrard S, Poston G, Kissmeyer-Nielsen P, et al. Combined ablation and resection (CARe) as an effective parenchymal sparing treatment for extensive colorectal liver metastases. PLoS ONE. 2014;9(12):e114404.CrossRef
11.
Zurück zum Zitat Primavesi F, Stättner S, Jäger T, et al. Progressive oncological surgery is associated with increased curative resection rates and improved survival in metastatic colorectal cancer. Cancers (Basel). 2019;11(2):218.CrossRef Primavesi F, Stättner S, Jäger T, et al. Progressive oncological surgery is associated with increased curative resection rates and improved survival in metastatic colorectal cancer. Cancers (Basel). 2019;11(2):218.CrossRef
12.
Zurück zum Zitat Hackert T, Niesen W, Hinz U, et al. Radical surgery of oligometastatic pancreatic cancer. Eur J Surg Oncol. 2017;43(2):358–63.CrossRef Hackert T, Niesen W, Hinz U, et al. Radical surgery of oligometastatic pancreatic cancer. Eur J Surg Oncol. 2017;43(2):358–63.CrossRef
13.
Zurück zum Zitat de Jong MC, Tsai S, Cameron JL, et al. Safety and efficacy of curative intent surgery for peri-ampullary liver metastasis. J Surg Oncol. 2010;102(3):256–63.CrossRef de Jong MC, Tsai S, Cameron JL, et al. Safety and efficacy of curative intent surgery for peri-ampullary liver metastasis. J Surg Oncol. 2010;102(3):256–63.CrossRef
14.
Zurück zum Zitat Zanini N, Lombardi R, Masetti M, Giordano M, Landolfo G, Jovine E. Surgery for isolated liver metastases from pancreatic cancer. Updates Surg. 2015;67(1):19–25.CrossRef Zanini N, Lombardi R, Masetti M, Giordano M, Landolfo G, Jovine E. Surgery for isolated liver metastases from pancreatic cancer. Updates Surg. 2015;67(1):19–25.CrossRef
15.
Zurück zum Zitat Tachezy M, Gebauer F, Janot 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.CrossRef Tachezy M, Gebauer F, Janot 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.CrossRef
16.
Zurück zum Zitat Oweira H, Petrausch U, Helbling D, et al. Prognostic value of site-specific metastases in pancreatic adenocarcinoma: A surveillance epidemiology and end results database analysis. World J Gastroenterol. 2017;23(10):1872–80.CrossRef Oweira H, Petrausch U, Helbling D, et al. Prognostic value of site-specific metastases in pancreatic adenocarcinoma: A surveillance epidemiology and end results database analysis. World J Gastroenterol. 2017;23(10):1872–80.CrossRef
17.
Zurück zum Zitat Hackert T. Surgery for pancreatic cancer after neoadjuvant treatment. Ann Gastroenterol Surg. 2018;2(6):413–8.CrossRef Hackert T. Surgery for pancreatic cancer after neoadjuvant treatment. Ann Gastroenterol Surg. 2018;2(6):413–8.CrossRef
18.
Zurück zum Zitat Klaiber U, Leonhardt CS, Strobel O, Tjaden C, Hackert T, Neoptolemos JP. Neoadjuvant and adjuvant chemotherapy in pancreatic cancer. Langenbecks Arch Surg. 2018;403(8):917–32.CrossRef Klaiber U, Leonhardt CS, Strobel O, Tjaden C, Hackert T, Neoptolemos JP. Neoadjuvant and adjuvant chemotherapy in pancreatic cancer. Langenbecks Arch Surg. 2018;403(8):917–32.CrossRef
19.
Zurück zum Zitat Hackert T, Ulrich A, Buchler MW. Can neoadjuvant therapy in pancreatic cancer increase the pool of patients eligible for pancreaticoduodenectomy? Adv Surg. 2017;51(1):1–10.CrossRef Hackert T, Ulrich A, Buchler MW. Can neoadjuvant therapy in pancreatic cancer increase the pool of patients eligible for pancreaticoduodenectomy? Adv Surg. 2017;51(1):1–10.CrossRef
20.
Zurück zum Zitat Conroy T, Hammel P, Hebbar M, et al. FOLFIRINOX or gemcitabine as adjuvant therapy for pancreatic cancer. N Engl J Med. 2018;379(25):2395–406.CrossRef Conroy T, Hammel P, Hebbar M, et al. FOLFIRINOX or gemcitabine as adjuvant therapy for pancreatic cancer. N Engl J Med. 2018;379(25):2395–406.CrossRef
21.
Zurück zum Zitat Neoptolemos JP, Palmer DH, Ghaneh P, et al. Comparison of adjuvant gemcitabine and capecitabine with gemcitabine monotherapy in patients with resected pancreatic cancer (ESPAC-4): A multicentre, open-label, randomised, phase 3 trial. Lancet. 2017;389(10073):1011–24.CrossRef Neoptolemos JP, Palmer DH, Ghaneh P, et al. Comparison of adjuvant gemcitabine and capecitabine with gemcitabine monotherapy in patients with resected pancreatic cancer (ESPAC-4): A multicentre, open-label, randomised, phase 3 trial. Lancet. 2017;389(10073):1011–24.CrossRef
22.
Zurück zum Zitat Shi S, Yu XJ. Time to think: Selecting patients who may benefit from synchronous resection of primary pancreatic cancer and liver metastases. World J Gastroenterol. 2018;24(33):3677–80.CrossRef Shi S, Yu XJ. Time to think: Selecting patients who may benefit from synchronous resection of primary pancreatic cancer and liver metastases. World J Gastroenterol. 2018;24(33):3677–80.CrossRef
24.
Zurück zum Zitat Shi HJ, Jin C, Fu DL. Preoperative evaluation of pancreatic ductal adenocarcinoma with synchronous liver metastasis: Diagnosis and assessment of unresectability. World J Gastroenterol. 2016;22(45):10024–37.CrossRef Shi HJ, Jin C, Fu DL. Preoperative evaluation of pancreatic ductal adenocarcinoma with synchronous liver metastasis: Diagnosis and assessment of unresectability. World J Gastroenterol. 2016;22(45):10024–37.CrossRef
25.
Zurück zum Zitat Mahadevan A, Blanck O, Lanciano R, et al. Stereotactic body radiotherapy (SBRT) for liver metastasis—Clinical outcomes from the international multi-institutional RSsearch(R) patient registry. Radiat Oncol. 2018;13(1):26.CrossRef Mahadevan A, Blanck O, Lanciano R, et al. Stereotactic body radiotherapy (SBRT) for liver metastasis—Clinical outcomes from the international multi-institutional RSsearch(R) patient registry. Radiat Oncol. 2018;13(1):26.CrossRef
26.
Zurück zum Zitat Park JB, Kim YH, Kim J, et al. Radiofrequency ablation of liver metastasis in patients with locally controlled pancreatic ductal adenocarcinoma. J Vasc Interv Radiol. 2012;23(5):635–41.CrossRef Park JB, Kim YH, Kim J, et al. Radiofrequency ablation of liver metastasis in patients with locally controlled pancreatic ductal adenocarcinoma. J Vasc Interv Radiol. 2012;23(5):635–41.CrossRef
27.
Zurück zum Zitat Kleeff J, Korc M, Apte M, et al. Pancreatic cancer. Nat Rev Dis Primers. 2016;2:16022.CrossRef Kleeff J, Korc M, Apte M, et al. Pancreatic cancer. Nat Rev Dis Primers. 2016;2:16022.CrossRef
28.
Zurück zum Zitat Somers I, Bipat S. Contrast-enhanced CT in determining resectability in patients with pancreatic carcinoma: A meta-analysis of the positive predictive values of CT. Eur Radiol. 2017;27(8):3408–35.CrossRef Somers I, Bipat S. Contrast-enhanced CT in determining resectability in patients with pancreatic carcinoma: A meta-analysis of the positive predictive values of CT. Eur Radiol. 2017;27(8):3408–35.CrossRef
29.
Zurück zum Zitat Tsurusaki M, Sofue K, Murakami T. Current evidence for the diagnostic value of gadoxetic acid-enhanced magnetic resonance imaging for liver metastasis. Hepatol Res. 2016;46(9):853–61.CrossRef Tsurusaki M, Sofue K, Murakami T. Current evidence for the diagnostic value of gadoxetic acid-enhanced magnetic resonance imaging for liver metastasis. Hepatol Res. 2016;46(9):853–61.CrossRef
30.
Zurück zum Zitat Schwarz L, Vernerey D, Bachet JB, et al. Resectable pancreatic adenocarcinoma neo-adjuvant FOLF(IRIN)OX-based chemotherapy—A multicenter, non-comparative, randomized, phase II trial (PANACHE01-PRODIGE48 study). BMC Cancer. 2018;18(1):762.CrossRef Schwarz L, Vernerey D, Bachet JB, et al. Resectable pancreatic adenocarcinoma neo-adjuvant FOLF(IRIN)OX-based chemotherapy—A multicenter, non-comparative, randomized, phase II trial (PANACHE01-PRODIGE48 study). BMC Cancer. 2018;18(1):762.CrossRef
31.
Zurück zum Zitat Ettrich TJ, Berger AW, Perkhofer L, et al. Neoadjuvant plus adjuvant or only adjuvant nab-paclitaxel plus gemcitabine for resectable pancreatic cancer—The NEONAX trial (AIO-PAK-0313), a prospective, randomized, controlled, phase II study of the AIO pancreatic cancer group. BMC Cancer. 2018;18(1):1298.CrossRef Ettrich TJ, Berger AW, Perkhofer L, et al. Neoadjuvant plus adjuvant or only adjuvant nab-paclitaxel plus gemcitabine for resectable pancreatic cancer—The NEONAX trial (AIO-PAK-0313), a prospective, randomized, controlled, phase II study of the AIO pancreatic cancer group. BMC Cancer. 2018;18(1):1298.CrossRef
33.
Zurück zum Zitat Jang JY, Han Y, Lee H, et al. Oncological benefits of neoadjuvant chemoradiation with gemcitabine versus upfront surgery in patients with borderline resectable pancreatic cancer: A prospective, randomized, open-label, multicenter phase 2/3 trial. Ann Surg. 2018;268(2):215–22.CrossRef Jang JY, Han Y, Lee H, et al. Oncological benefits of neoadjuvant chemoradiation with gemcitabine versus upfront surgery in patients with borderline resectable pancreatic cancer: A prospective, randomized, open-label, multicenter phase 2/3 trial. Ann Surg. 2018;268(2):215–22.CrossRef
34.
Zurück zum Zitat Suker M, Beumer BR, Sadot E, et al. FOLFIRINOX for locally advanced pancreatic cancer: A systematic review and patient-level meta-analysis. Lancet Oncol. 2016;17(6):801–10.CrossRef Suker M, Beumer BR, Sadot E, et al. FOLFIRINOX for locally advanced pancreatic cancer: A systematic review and patient-level meta-analysis. Lancet Oncol. 2016;17(6):801–10.CrossRef
35.
Zurück zum Zitat Burris HA 3rd, Moore MJ, Andersen J, et al. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: A randomized trial. J Clin Oncol. 1997;15(6):2403–13.CrossRef Burris HA 3rd, Moore MJ, Andersen J, et al. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: A randomized trial. J Clin Oncol. 1997;15(6):2403–13.CrossRef
36.
Zurück zum Zitat Sultana A, Smith CT, Cunningham D, Starling N, Neoptolemos JP, Ghaneh P. Meta-analyses of chemotherapy for locally advanced and metastatic pancreatic cancer. J Clin Oncol. 2007;25(18):2607–15.CrossRef Sultana A, Smith CT, Cunningham D, Starling N, Neoptolemos JP, Ghaneh P. Meta-analyses of chemotherapy for locally advanced and metastatic pancreatic cancer. J Clin Oncol. 2007;25(18):2607–15.CrossRef
37.
Zurück zum Zitat Sultana A, Tudur Smith C, Cunningham D, Starling N, Neoptolemos JP, Ghaneh P. Meta-analyses of chemotherapy for locally advanced and metastatic pancreatic cancer: Results of secondary end points analyses. Br J Cancer. 2008;99(1):6–13.CrossRef Sultana A, Tudur Smith C, Cunningham D, Starling N, Neoptolemos JP, Ghaneh P. Meta-analyses of chemotherapy for locally advanced and metastatic pancreatic cancer: Results of secondary end points analyses. Br J Cancer. 2008;99(1):6–13.CrossRef
38.
Zurück zum Zitat Heinemann V, Boeck S, Hinke A, Labianca R, Louvet C. Meta-analysis of randomized trials: evaluation of benefit from gemcitabine-based combination chemotherapy applied in advanced pancreatic cancer. BMC Cancer. 2008;8:82.CrossRef Heinemann V, Boeck S, Hinke A, Labianca R, Louvet C. Meta-analysis of randomized trials: evaluation of benefit from gemcitabine-based combination chemotherapy applied in advanced pancreatic cancer. BMC Cancer. 2008;8:82.CrossRef
39.
Zurück zum Zitat Ramaswamy A, Ostwal V, Goel A, et al. Treatment practices for metastatic pancreatic cancer: Can we deliver an appropriately efficacious and safe regimen in Indian patients? Indian J Cancer. 2018;55(2):138–43.CrossRef Ramaswamy A, Ostwal V, Goel A, et al. Treatment practices for metastatic pancreatic cancer: Can we deliver an appropriately efficacious and safe regimen in Indian patients? Indian J Cancer. 2018;55(2):138–43.CrossRef
40.
Zurück zum Zitat Von Hoff DD, Ervin T, Arena FP, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369(18):1691–703.CrossRef Von Hoff DD, Ervin T, Arena FP, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369(18):1691–703.CrossRef
41.
Zurück zum Zitat Michalski CW, Erkan M, Hüser N, et al. Resection of primary pancreatic cancer and liver metastasis: A systematic review. Dig Surg. 2008;25(6):473–80.CrossRef Michalski CW, Erkan M, Hüser N, et al. Resection of primary pancreatic cancer and liver metastasis: A systematic review. Dig Surg. 2008;25(6):473–80.CrossRef
42.
Zurück zum Zitat Crippa S, Bittoni A, Sebastiani E, et al. Is there a role for surgical resection in patients with pancreatic cancer with liver metastases responding to chemotherapy? Eur J Surg Oncol. 2016;42(10):1533–9.CrossRef Crippa S, Bittoni A, Sebastiani E, et al. Is there a role for surgical resection in patients with pancreatic cancer with liver metastases responding to chemotherapy? Eur J Surg Oncol. 2016;42(10):1533–9.CrossRef
43.
Zurück zum Zitat Frigerio I, Regi P, Giardino A, et al. Downstaging in stage IV pancreatic cancer: A new population eligible for surgery? Ann Surg Oncol. 2017;24(8):2397–403.CrossRef Frigerio I, Regi P, Giardino A, et al. Downstaging in stage IV pancreatic cancer: A new population eligible for surgery? Ann Surg Oncol. 2017;24(8):2397–403.CrossRef
44.
Zurück zum Zitat Wright GP, Poruk KE, Zenati MS, et al. Primary tumor resection following favorable response to systemic chemotherapy in stage IV pancreatic adenocarcinoma with synchronous metastases: A bi-institutional analysis. J Gastrointest Surg. 2016;20(11):1830–5.CrossRef Wright GP, Poruk KE, Zenati MS, et al. Primary tumor resection following favorable response to systemic chemotherapy in stage IV pancreatic adenocarcinoma with synchronous metastases: A bi-institutional analysis. J Gastrointest Surg. 2016;20(11):1830–5.CrossRef
45.
Zurück zum Zitat Kruger S, Haas M, Burger PJ, et al. Isolated pulmonary metastases define a favorable subgroup in metastatic pancreatic cancer. Pancreatology. 2016;16(4):593–8.CrossRef Kruger S, Haas M, Burger PJ, et al. Isolated pulmonary metastases define a favorable subgroup in metastatic pancreatic cancer. Pancreatology. 2016;16(4):593–8.CrossRef
46.
Zurück zum Zitat Yasukawa M, Kawaguchi T, Kawai N, Tojo T, Taniguchi S. Surgical treatment for pulmonary metastasis of pancreatic ductal adenocarcinoma: Study of 12 cases. Anticancer Res. 2017;37(10):5573–6.PubMed Yasukawa M, Kawaguchi T, Kawai N, Tojo T, Taniguchi S. Surgical treatment for pulmonary metastasis of pancreatic ductal adenocarcinoma: Study of 12 cases. Anticancer Res. 2017;37(10):5573–6.PubMed
47.
Zurück zum Zitat Satoi S, Fujii T, Yanagimoto H, et al. Multicenter phase II study of intravenous and Intraperitoneal paclitaxel with S‑1 for pancreatic ductal adenocarcinoma patients with peritoneal metastasis. Ann Surg. 2017;265(2):397–401.CrossRef Satoi S, Fujii T, Yanagimoto H, et al. Multicenter phase II study of intravenous and Intraperitoneal paclitaxel with S‑1 for pancreatic ductal adenocarcinoma patients with peritoneal metastasis. Ann Surg. 2017;265(2):397–401.CrossRef
48.
Zurück zum Zitat Putzer D, Schullian P, Braunwarth E, et al. Integrating interventional oncology in the treatment of liver tumors. Eur Surg. 2018;50(3):117–24.CrossRef Putzer D, Schullian P, Braunwarth E, et al. Integrating interventional oncology in the treatment of liver tumors. Eur Surg. 2018;50(3):117–24.CrossRef
50.
Zurück zum Zitat Stättner S, Primavesi F, Yip VS, et al. Evolution of surgical microwave ablation for the treatment of colorectal cancer liver metastasis: Review of the literature and a single centre experience. Surg Today. 2015;45(4):407–15.CrossRef Stättner S, Primavesi F, Yip VS, et al. Evolution of surgical microwave ablation for the treatment of colorectal cancer liver metastasis: Review of the literature and a single centre experience. Surg Today. 2015;45(4):407–15.CrossRef
51.
Zurück zum Zitat De Jong MC, Farnell MB, Sclabas G, et al. Liver-directed therapy for hepatic metastases in patients undergoing pancreaticoduodenectomy: A dual-center analysis. Ann Surg. 2010;252(1):142–8.CrossRef De Jong MC, Farnell MB, Sclabas G, et al. Liver-directed therapy for hepatic metastases in patients undergoing pancreaticoduodenectomy: A dual-center analysis. Ann Surg. 2010;252(1):142–8.CrossRef
52.
Zurück zum Zitat Azizi A, Naguib NN, Mbalisike E, Farshid P, Emami AH, Vogl TJ. Liver metastases of pancreatic cancer: Role of repetitive transarterial chemoembolization (TACE) on tumor response and survival. Pancreas. 2011;40(8):1271–5.CrossRef Azizi A, Naguib NN, Mbalisike E, Farshid P, Emami AH, Vogl TJ. Liver metastases of pancreatic cancer: Role of repetitive transarterial chemoembolization (TACE) on tumor response and survival. Pancreas. 2011;40(8):1271–5.CrossRef
53.
Zurück zum Zitat Kim AY, Unger K, Wang H, Pishvaian MJ. Incorporating Yttrium-90 trans-arterial radioembolization (TARE) in the treatment of metastatic pancreatic adenocarcioma: A single center experience. BMC Cancer. 2016;16:492.CrossRef Kim AY, Unger K, Wang H, Pishvaian MJ. Incorporating Yttrium-90 trans-arterial radioembolization (TARE) in the treatment of metastatic pancreatic adenocarcioma: A single center experience. BMC Cancer. 2016;16:492.CrossRef
54.
Zurück zum Zitat Hua YQ, Wang P, Zhu XY, et al. Radiofrequency ablation for hepatic oligometastatic pancreatic cancer: An analysis of safety and efficacy. Pancreatology. 2017;17(6):967–73.CrossRef Hua YQ, Wang P, Zhu XY, et al. Radiofrequency ablation for hepatic oligometastatic pancreatic cancer: An analysis of safety and efficacy. Pancreatology. 2017;17(6):967–73.CrossRef
55.
Zurück zum Zitat Sun JH, Zhou TY, Zhang YL, et al. Efficacy of transcatheter arterial chemoembolization for liver metastases arising from pancreatic cancer. Oncotarget. 2017;8(24):39746–55.PubMedPubMedCentral Sun JH, Zhou TY, Zhang YL, et al. Efficacy of transcatheter arterial chemoembolization for liver metastases arising from pancreatic cancer. Oncotarget. 2017;8(24):39746–55.PubMedPubMedCentral
56.
Zurück zum Zitat Huang ZM, Pan CC, Wu PH, et al. Efficacy of minimally invasive therapies on unresectable pancreatic cancer. Chin J Cancer. 2013;32(6):334–41.CrossRef Huang ZM, Pan CC, Wu PH, et al. Efficacy of minimally invasive therapies on unresectable pancreatic cancer. Chin J Cancer. 2013;32(6):334–41.CrossRef
57.
Zurück zum Zitat Michl M, Haug AR, Jakobs TF, et al. Radioembolization with yttrium-90 microspheres (SIRT) in pancreatic cancer patients with liver metastases: Efficacy, safety and prognostic factors. Oncology. 2014;86(1):24–32.CrossRef Michl M, Haug AR, Jakobs TF, et al. Radioembolization with yttrium-90 microspheres (SIRT) in pancreatic cancer patients with liver metastases: Efficacy, safety and prognostic factors. Oncology. 2014;86(1):24–32.CrossRef
58.
Zurück zum Zitat Gibbs P, Do C, Lipton L, et al. Phase II trial of selective internal radiation therapy and systemic chemotherapy for liver-predominant metastases from pancreatic adenocarcinoma. BMC Cancer. 2015;15:802.CrossRef Gibbs P, Do C, Lipton L, et al. Phase II trial of selective internal radiation therapy and systemic chemotherapy for liver-predominant metastases from pancreatic adenocarcinoma. BMC Cancer. 2015;15:802.CrossRef
59.
Zurück zum Zitat Wieners G, Schippers AC, Collettini F, et al. CT-guided high-dose-rate brachytherapy in the interdisciplinary treatment of patients with liver metastases of pancreatic cancer. HBPD INT. 2015;14(5):530–8.PubMed Wieners G, Schippers AC, Collettini F, et al. CT-guided high-dose-rate brachytherapy in the interdisciplinary treatment of patients with liver metastases of pancreatic cancer. HBPD INT. 2015;14(5):530–8.PubMed
60.
Zurück zum Zitat Tafuto S, von Arx C, De Divitiis C, et al. Electrochemotherapy as a new approach on pancreatic cancer and on liver metastases. Int J Surg. 2015;21(Suppl 1):S78–S82.CrossRef Tafuto S, von Arx C, De Divitiis C, et al. Electrochemotherapy as a new approach on pancreatic cancer and on liver metastases. Int J Surg. 2015;21(Suppl 1):S78–S82.CrossRef
61.
Zurück zum Zitat Vidal-Jove J, Perich E, Del Castillo MA. Ultrasound guided high intensity focused ultrasound for malignant tumors: The Spanish experience of survival advantage in stage III and IV pancreatic cancer. Ultrason Sonochem. 2015;27:703–6.CrossRef Vidal-Jove J, Perich E, Del Castillo MA. Ultrasound guided high intensity focused ultrasound for malignant tumors: The Spanish experience of survival advantage in stage III and IV pancreatic cancer. Ultrason Sonochem. 2015;27:703–6.CrossRef
62.
Zurück zum Zitat Hong Y, Rice J, Sharma D, Martin RCG 2nd. The use of IRE in multi-modality treatment for oligometastatic pancreatic cancer. Am J Surg. 2018;216(1):106–10.CrossRef Hong Y, Rice J, Sharma D, Martin RCG 2nd. The use of IRE in multi-modality treatment for oligometastatic pancreatic cancer. Am J Surg. 2018;216(1):106–10.CrossRef
63.
Zurück zum Zitat Haas M, Heinemann V, Kullmann F, et al. Prognostic value of CA 19-9, CEA, CRP, LDH and bilirubin levels in locally advanced and metastatic pancreatic cancer: Results from a multicenter, pooled analysis of patients receiving palliative chemotherapy. J Cancer Res Clin Oncol. 2013;139(4):681–9.CrossRef Haas M, Heinemann V, Kullmann F, et al. Prognostic value of CA 19-9, CEA, CRP, LDH and bilirubin levels in locally advanced and metastatic pancreatic cancer: Results from a multicenter, pooled analysis of patients receiving palliative chemotherapy. J Cancer Res Clin Oncol. 2013;139(4):681–9.CrossRef
64.
Zurück zum Zitat Boeck S, Wittwer C, Heinemann V, et al. Cytokeratin 19-fragments (CYFRA 21-1) as a novel serum biomarker for response and survival in patients with advanced pancreatic cancer. Br J Cancer. 2013;108(8):1684–94.CrossRef Boeck S, Wittwer C, Heinemann V, et al. Cytokeratin 19-fragments (CYFRA 21-1) as a novel serum biomarker for response and survival in patients with advanced pancreatic cancer. Br J Cancer. 2013;108(8):1684–94.CrossRef
65.
Zurück zum Zitat Song JY, Chen MQ, Guo JH, Lian SF, Xu BH. Combined pretreatment serum CA19-9 and neutrophil-to-lymphocyte ratio as a potential prognostic factor in metastatic pancreatic cancer patients. Medicine (Baltimore). 2018;97(4):e9707.CrossRef Song JY, Chen MQ, Guo JH, Lian SF, Xu BH. Combined pretreatment serum CA19-9 and neutrophil-to-lymphocyte ratio as a potential prognostic factor in metastatic pancreatic cancer patients. Medicine (Baltimore). 2018;97(4):e9707.CrossRef
66.
Zurück zum Zitat Piciucchi M, Stigliano S, Archibugi L, et al. The neutrophil/lymphocyte ratio at diagnosis is significantly associated with survival in metastatic pancreatic cancer patients. Int J Mol Sci. 2017;18(4):730.CrossRef Piciucchi M, Stigliano S, Archibugi L, et al. The neutrophil/lymphocyte ratio at diagnosis is significantly associated with survival in metastatic pancreatic cancer patients. Int J Mol Sci. 2017;18(4):730.CrossRef
67.
Zurück zum Zitat Stevens L, Pathak S, Nunes QM, et al. Prognostic significance of pre-operative C‑reactive protein and the neutrophil-lymphocyte ratio in resectable pancreatic cancer: A systematic review. HPB (Oxford). 2015;17(4):285–91.CrossRef Stevens L, Pathak S, Nunes QM, et al. Prognostic significance of pre-operative C‑reactive protein and the neutrophil-lymphocyte ratio in resectable pancreatic cancer: A systematic review. HPB (Oxford). 2015;17(4):285–91.CrossRef
68.
Zurück zum Zitat Ueno H, Okada S, Okusaka T, Ikeda M. Prognostic factors in patients with metastatic pancreatic adenocarcinoma receiving systemic chemotherapy. Oncology. 2000;59(4):296–301.CrossRef Ueno H, Okada S, Okusaka T, Ikeda M. Prognostic factors in patients with metastatic pancreatic adenocarcinoma receiving systemic chemotherapy. Oncology. 2000;59(4):296–301.CrossRef
69.
Zurück zum Zitat Hang J, Wu L, Zhu L, et al. Prediction of overall survival for metastatic pancreatic cancer: Development and validation of a prognostic nomogram with data from open clinical trial and real-world study. Cancer Med. 2018;7(7):2974–84.CrossRef Hang J, Wu L, Zhu L, et al. Prediction of overall survival for metastatic pancreatic cancer: Development and validation of a prognostic nomogram with data from open clinical trial and real-world study. Cancer Med. 2018;7(7):2974–84.CrossRef
70.
Zurück zum Zitat Kim HW, Lee JC, Paik KH, Lee YS, Hwang JH, Kim J. Initial metastatic site as a prognostic factor in patients with stage IV pancreatic ductal adenocarcinoma. Medicine (Baltimore). 2015;94(25):e1012.CrossRef Kim HW, Lee JC, Paik KH, Lee YS, Hwang JH, Kim J. Initial metastatic site as a prognostic factor in patients with stage IV pancreatic ductal adenocarcinoma. Medicine (Baltimore). 2015;94(25):e1012.CrossRef
71.
Zurück zum Zitat Court CM, Ankeny JS, Sho S, et al. Circulating tumor cells predict occult metastatic disease and prognosis in pancreatic cancer. Ann Surg Oncol. 2018;25(4):1000–8.CrossRef Court CM, Ankeny JS, Sho S, et al. Circulating tumor cells predict occult metastatic disease and prognosis in pancreatic cancer. Ann Surg Oncol. 2018;25(4):1000–8.CrossRef
72.
Zurück zum Zitat Effenberger KE, Schroeder C, Hanssen A, et al. Improved risk stratification by circulating tumor cell counts in pancreatic cancer. Clin Cancer Res. 2018;24(12):2844–50.CrossRef Effenberger KE, Schroeder C, Hanssen A, et al. Improved risk stratification by circulating tumor cell counts in pancreatic cancer. Clin Cancer Res. 2018;24(12):2844–50.CrossRef
73.
Zurück zum Zitat Bhat K, Wang F, Ma Q, et al. Advances in biomarker research for pancreatic cancer. Curr Pharm Des. 2012;18(17):2439–51.CrossRef Bhat K, Wang F, Ma Q, et al. Advances in biomarker research for pancreatic cancer. Curr Pharm Des. 2012;18(17):2439–51.CrossRef
74.
Zurück zum Zitat Canivet C, Gourgou-Bourgade S, Napoléon B, et al. A prospective clinical and biological database for pancreatic adenocarcinoma: The BACAP cohort. BMC Cancer. 2018;18(1):986.CrossRef Canivet C, Gourgou-Bourgade S, Napoléon B, et al. A prospective clinical and biological database for pancreatic adenocarcinoma: The BACAP cohort. BMC Cancer. 2018;18(1):986.CrossRef
Metadaten
Titel
Surgical and local therapeutic concepts of oligometastatic pancreatic cancer in the era of effective chemotherapy
verfasst von
Willem Niesen
Florian Primavesi
Silvia Gasteiger
John Neoptolemos
Thilo Hackert
PD Dr. Stefan Stättner
Publikationsdatum
15.04.2019
Verlag
Springer Vienna
Erschienen in
European Surgery / Ausgabe 3/2019
Print ISSN: 1682-8631
Elektronische ISSN: 1682-4016
DOI
https://doi.org/10.1007/s10353-019-0589-6

Weitere Artikel der Ausgabe 3/2019

European Surgery 3/2019 Zur Ausgabe