Skip to main content
Erschienen in: Current Treatment Options in Oncology 11/2021

Open Access 01.11.2021 | Upper Gastrointestinal Cancers (JD Berlin, Section Editor)

Systemic Therapy for Metastatic Pancreatic Cancer

verfasst von: Thomas J. Ettrich, M.D., Thomas Seufferlein, M.D.

Erschienen in: Current Treatment Options in Oncology | Ausgabe 11/2021

Opinion statement

Pancreatic cancer is mainly diagnosed at an advanced, often metastatic stage and still has a poor prognosis. Over the last decades, chemotherapy of metastatic pancreatic cancer (mPDAC) has proven to be superior to a mere supportive treatment with respect to both survival and quality of life. Recently, even sequential treatment of mPDAC could be established. Options for first-line treatment are combination chemotherapy regimens such as FOLFIRINOX and gemcitabine plus nab-paclitaxel when the performance status of the patient is good. For patients with poorer performance status, gemcitabine single-agent treatment is a valid option. Recently, the PARP inhibitor olaparib has been demonstrated to improve progression-free survival when used as a maintenance treatment in the subgroup of patients with mPDAC and a BRCA1/-2 germ line mutation having received at least 16 weeks of platinum-based chemotherapy. This group of patients also benefits from platinum-based chemotherapy combinations. Therefore, the BRCA1/-2 stats should be examined early in patients with mPDAC even when the occurrence of these mutations is only about 5% in the general Caucasian population. After the failure of first-line treatment, patients should be offered a second-line treatment if their ECOG permits further treatment. Here, the combination of 5-FU/FA plus nanoliposomal irinotecan has shown to be superior to 5-FU/FA alone with respect to overall survival. Immune checkpoint inhibitors like PD1/PD-L1 mAbs are particularly efficacious in tumors with high microsatellite instability (MSI-h). Limited data in mPDACs shows that only a part of the already small subgroup of MSI-H mPDACs (frequency about 1%) appears to benefit substantially from a checkpoint inhibitor treatment. The identification of further subgroups, e.g., tumors with DNA damage repair deficiency, gene fusions, as well as novel approaches such as tumor-organoid-informed treatment decisions, may further improve therapeutic efficacy.
Hinweise
This article is part of the Topical Collection on Upper Gastrointestinal Cancers

Publisher's Note

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

Introduction

Pancreatic cancer still has a dismal prognosis mainly due to the fact that the majority of patients are only diagnosed at an advanced or even metastatic state. Surgery is only appropriate if there is a chance of cure. In contrast to other tumors, surgery of metastasis is not appropriate in pancreatic cancer outside of a clinical trial even in the case of isolated liver metastasis since there is no proof that surgery improves overall survival in this situation [15]. Also, radiation therapy plays mainly a supportive role in metastatic disease, e.g., in case of bone metastasis and as a measure to treat tumor-related pain. Therefore, systemic treatment plays an important role in the treatment of metastatic pancreatic cancer (mPDAC). All patients with mPDAC and an ECOG performance status of 0–2 should be offered systemic treatment since it improves both overall survival and quality of life [511]. Systemic treatment can also lead to a reduced requirement of pain medication, delay weight loss, as well as the time to the definitive deterioration of quality of life [12]. Systemic treatment should be started immediately after the detection of metastasis.
The performance status of a patient with mPDAC is prognostic. In the case of an ECOG > 2, patients only derive benefit from systemic treatment if the poor performance status is due to the tumor disease itself and is likely to improve if there is tumor remission in response to the treatment. There is also no optimal duration of systemic treatment in mPDAC, and maintenance strategies are only established for specific subgroups of patients. Thus, the duration of treatment in mPDAC depends on its efficacy as well as tolerability and, of course, on the individual patient requirements. Here, we present the current state of systemic treatment of mPDAC.

First-Line Treatment of mPDAC

There are several well-established options for first-line treatment of mPDAC (see Table 1): The FOLFIRINOX protocol, the combination of gemcitabine plus nab-paclitaxel, and gemcitabine monotherapy. The choice of the respective treatment depends on the patient’s ECOG performance status, comorbidity, and patient’s preferences.
Table 1
Metastatic pancreatic ductal adenocarcinoma: palliative first-line regimens
Regiment
Phase
n
mPFS (months)
mOS (months)
DCR
(%)
References
First-line therapy
 FOLFIRINOX (Prodige-4 Intergroup trial)
III
342
6.4
11.1
70
[6]
 Gemcitabine
3.3
6.8
51
 Gemcitabine/Nab-Paclitaxel (MPACT trial)
III
861
5.5
8.7
48
[15]
 Gemcitabine
3.7
6.6
33
 Gemcitabine/erlotinib
III
569
3.8
6.2 (RASH 2 + : 10.5)
58
[29, 30]
 Gemcitabine
3.6
5.9
49
 Gemcitabine
III
126
2.3
5.7
n.a
[7]
 5-FU
0.9
4.4
Maintenance therapy after first-line therapy
 Olaparib (POLO trial),
  only pat. with germline BRCA 1 or BRCA 2 mutation and
  disease control after at least 16 weeks of platinum-based induction therapy
III
154
7.4
19.0
n.a
[31, 32]
Placebo
3.8
19.2
mPFS median progression-free survival, mOS median overall survival, DCR disease control rate, FOLFIRINOX 5-FU, leucovorin, irinotecan, oxaliplatin

Role of the Performance Status for Treatment Selection

Patients with an ECOG 0–1 benefit from combination chemotherapy, while patients with an ECOG of ≥ 2 and marked comorbidities should preferably receive single-agent treatment. This statement is based on data from a meta-analysis examining gemcitabine or gemcitabine-based combination treatments. Patients with a good performance status (ECOG 0–1) benefitted with respect to survival from combination treatment (HR 0.76). However, when the ECOG was > 1, patients did not benefit from combination treatment (HR 1.08) [13]. There is a limitation to this statement: If tumor-related symptoms are mainly responsible for the poor ECOG and if the ECOG may be improved by tumor remission, combination treatment, e.g., gemcitabine plus nab-paclitaxel, can be given. In this case, lower doses of nab-paclitaxel have been employed [14].
Disregarding the ECOG status, it is paramount that supportive measures such as appropriate pain management and nutritional support after analysis of the nutritional status are also initiated as early as possible.

Does Age Limit the Treatment Intensity of mPDAC?

There is no sufficient data supporting chronological age as a criterion for the choice of systemic treatment of mPDAC. The biological age of a respective patient appears far more important to decide on the appropriate treatment in the metastatic setting. However, some studies did have an age limit for inclusion of patients into the trial (e.g., the PRODIGE 4 Intergroup study examining FOLFIRINOX vs. gemcitabine only included patients up 75 years of age [6]), and in general, there are few data on patients with advanced age.

Specific Treatment Protocols

Gemcitabine Monotherapy

Patients with an ECOG of ≥ 2 or comorbidities that prevent combination treatment should receive a single-agent treatment. Gemcitabine is preferred over 5-FU in this case [7]. There are numerous phase III trials that demonstrate the efficacy of gemcitabine with 1-year overall survival rates of 18–20% and a median OS of about 6 months [6, 15]. Gemcitabine also exhibited a clinical benefit response compared to 5-FU (albeit the latter was used at a suboptimal dose in the trial) [7]. Gemcitabine is generally well tolerated. Its most frequent grade 3/4 side effect is hematotoxicity with leuko-/neutropenia, thrombopenia, and anemia. Other effects such as interstitial pneumonitis are rare, but physicians should be aware of this adverse event to initiate prompt treatment.

Gemcitabine-Based Combinations

The phase III MPACT trial examined the combination of gemcitabine plus albumin-nanoparticle bound paclitaxel (nab-paclitaxel) compared to gemcitabine single agent. The trial showed a significantly improved mOS compared to the monotherapy (8.5 months compared to 6.7 months, HR 0.72; p < 0.001). The combination also significantly improved mPFS (5.5 months vs. 3.7 months, HR 0.69, p < 0.001) and response rate (23% vs. 7%, p < 0.001). The combination has a higher rate of grade 3/4 side effects with more neutropenia, neuropathy, and diarrhea. The trial recruited patients with a Karnofsky performance status of ≥ 70 so that patients with an ECOG of 0–2 can be treated with this protocol [8, 15, 16]. A phase 2 trial could confirm the safety and efficacy of gemcitabine plus nab-paclitaxel in patients with mPDAC and an ECOG of 2 [14].
Thus, the combination of gemcitabine and nab-paclitaxel can be employed in a larger population of patients with mPDAC (ECOG 0–2) compared to FOLFIRINOX (see below). In addition, the study included elderly patients up to the age of 88 years [15]. The subgroup analysis showed that patients ≥ 65 years of age also benefit from the combination. There is no separate evaluation of the group of patients above 75 years or even 80 years of age from this trial.
MPACT was a study that recruited centers worldwide. The results from the trial could be confirmed in a real-world setting with even better outcome data when only Western countries were considered. For example, mOS was 10.9 months with gemcitabine plus nab-paclitaxel in a retrospective Swedish study [17].
In more recent clinical trials, gemcitabine plus nab-paclitaxel was used as a control arm and also showed good efficiency, for example, an mOS of 11.5 months in phase III HALO-301 trial (gem/nab-paclitaxel vs. gem/nab-paclitaxel + PegPH20 for patients with hyaluronan-high mPDAC) [18]. This could be related to the therapy in further lines, although, especially in this study, patient selection by the inclusion criterion “hyaluronan-high mPDAC” should be considered.
Gemcitabine has been combined with many other drugs such as irinotecan, capecitabine, oxaliplatin, cisplatin, cisplatin/epirubicin, and 5-FU plus docetaxel or exatecan. None of these combinations could show a significant improvement in OS compared to gemcitabine alone in phase III trials in the overall study populations. However, there may be subgroups of patients benefitting from some of these combinations, e.g., patients with an ECOG of 0–1. A significant survival benefit, e.g., of the combination of gemcitabine plus oxaliplatin or cisplatin, could only be demonstrated in meta-analyses. However, the combination of gemcitabine plus cisplatin may be an interesting option for patients with germline BRCA1/2 mutations (see below).

5-FU-Based Combinations: FOLFIRINOX

The combination of 5-FU, irinotecan, and oxaliplatin in the FOLFIRINOX protocol is a landmark in the treatment of mPDAC. In the PRODIGE 4 Intergroup trial, this combination treatment achieved a median OS (mOS) of 11.1 months and a median PFS (mPFS) of 6.4 months compared to gemcitabine alone with an mOS of 6.8 months (HR 0.57; p < 0.001) and an mPFS of 3.3 months, respectively (HR 0.47; p < 0.001) [6]. The data of the initial study could be confirmed in numerous subsequent phase II trials and cohort studies [19].
FOLFIRINOX should be offered to patients with an ECOG of 0–1, a Bilirubin value of ≤ 1.5 × ULN, and a favorable comorbidity profile. This definition comprises about 30% of patients with mPDAC [20]. The toxicity of the FOLFIRINOX protocol is higher than that of gemcitabine with more grade 3/4 neutropenia (45.7% vs. 21%), more febrile neutropenia (5.4% vs. 1.2%), and more diarrhea (12.7% vs. 1.8%). About 42% of patients receiving FOLFIRINOX also received G-CSF compared to only 5.3% in the gemcitabine group. This higher toxicity rate is the reason why this protocol is often modified including dropping the 5-FU bolus, reducing irinotecan and the 5-FU bolus, or reducing oxaliplatin. Interestingly, despite lower toxicity, survival benefits are comparable with the original FOLFIRINOX protocol in a meta-analysis [21, 22]. In addition, modified FOLFIRINOX (here, modified by omitting the 5-FU bolus) showed an impressive mOS of 14.4 months in the control arm of the SWOG 1313 trial (mFOLFIRINOX vs. mFOLFIRINOX + PEGPH20) [23]. This could be related to the therapy in further lines.
Of note, despite the higher rate of toxicity of the FOLFIRINOX protocol, deterioration of quality of life was significantly delayed in the FOLFIRINOX group of the phase III PRODIGE 4 Intergroup trial [12].
The PRODIGE 4 Intergroup trial only included patients up to the age of 75 years. Thus, there are no data from this prospective randomized trial for this group of patients regarding FOLFIRINOX efficacy and tolerability. Retrospective data suggest that modified FOLFIRINOX protocols have similar toxicity but also similar efficacy when compared to younger patients [22].

Other 5-FU-Based Combination Treatments

Several phase III trials examined the effect of other 5-FU-based combination chemotherapies. Neither 5-FU plus mitomycin C [24] nor the combination of 5-FU, gemcitabine, epirubicin, and cisplatin [25, 26] has shown sufficient efficacy to qualify as a therapeutic standard.
The FIRGEM regime examined FOLFIRI.3 alternating with fixed-dose-rate gemcitabine compared to fixed-dose-rate gemcitabine alone in a randomized phase II trial [27]. The combination was superior to gemcitabine alone with respect to mOS (11 months vs. 8.2 months; HR = 0.71). However, due to a lack of phase III data, this combination cannot be regarded as a clinical standard. Another phase II trial examined the combination of 5-FU/leucovorin with nab-paclitaxel compared to gemcitabine plus nab-paclitaxel. The 4-month PFS as the primary endpoint of the trial was 56% for 5-FU/leucovorin plus nab-paclitaxel compared to 54% in the gemcitabine plus nab-paclitaxel group. The respective figures for mOS were 11.4 months vs. 9.2 months, respectively [28]. Thus, this protocol may be employed when gemcitabine cannot be used, e.g., in the case of gemcitabine intolerance.

Targeted Therapies

The combination of gemcitabine with targeted therapies has so far not shown a clinically relevant survival benefit for patients with mPDAC. An exemption is the combination of gemcitabine with erlotinib, an EGFR tyrosine kinase inhibitor. There was a significant improvement in overall survival in favor of the combination (HR 0.82, p = 0.038). However, in the overall population, the difference in mOS between the groups was just 10 days. According to unplanned subgroup analysis, the difference in mOS was only more pronounced in the subgroup of patients developing a skin rash ≥ grade 2 in response to erlotinib (mOS 10.5 months vs. 5.8 months). Thus, if employed, this combination should not be continued when there is no rash during the first 8 weeks of treatment [29, 30].

Second-Line and Further-Line Treatments

Upon progress under a first-line treatment, patients should receive a second-line treatment if their ECOG is ≤ 2. The majority of second-line treatments have been examined in patients receiving only gemcitabine as a first-line treatment (see Table 2).
Table 2
Metastatic pancreatic ductal adenocarcinoma: palliative second-line regimens
Regiment
Phase
n
mPFS
(months)
mOS
(months)
DCR
(%)
References
Second-line therapy (mostly after gemcitabine mono in first-line treatment)
 Nal-Iri/5-FU/LV (NAPOLI-1 trial)
III
236
3.1
6.1
n.a
[33]
 5-FU/LV
1.5
4.2
 OFF (CONKO-003 trial)
III
160
2.9
5.9
n.a
[35]
 5-FU/LV
2.0
3.3
 mFOLFOX6, (PANCREOX trial)
III
108
3.1
6.1
44.7
[37]
 5-FU/LV
2.9
9.9
55.3
 Gemcitabine/nab-paclitaxel (after FOLFIRINOX)
Cohort
57
5.1
8.8
58
[39]
mPFS median progression-free survival, mOS median overall survival, DCR disease control rate, OFF oxaliplatin, leucovorin, 5-FU, LV leucovorin, 5-FU 5-FU, nal-Iri nanoliposomal irinotecan, FOLFIRI 5-FU, leucovorin, irinotecan, FOLFOX 5-FU, leucovorin, oxaliplatin
The three-arm, phase III NAPOLI study examined the combination of nanoliposomal irinotecan (nal-Iri) plus 5-FU/FA, nal-Iri alone, and 5-FU/FA alone in gemcitabine-pretreated patients with mPDAC and a Karnofsky performance status of ≥ 70%. The combination of nal-iri/5-FU/FA significantly improved overall survival compared to 5-FU/FA alone (6.2 months vs. 4.2 months; HR 0.75; p = 0.039) [33]. Nal-Iri alone was not superior to 5-FU/FA. The most frequent ≥ grade 3 side effects of the combination were neutropenia (27%), diarrhea (13%), emesis (11%), and fatigue (14%). Health-related quality of life was comparable between the combination and 5-FU/FA [34].
Another option in the second-line setting is the OFF regimen consisting of 5-FU/LV and oxaliplatin. This regimen can be employed in patients with progress under gemcitabine, an ECOG of ≤ 2 and polyneuropathy ≤ 2. This regimen improved OS (5.9 vs. 3.3 months; HR 0.66; p = 0.010) and time to tumor progression (2.9 months vs. 2 months; HR 0.68; p = 0.019) compared to 5-FU/LV alone [35, 36]. The PANCREOX trial compared a modified FOLFOX6 (mFOLFOX6) protocol with 5-FU/LV. mFOLFOX6 has a higher-dose intensity and worse tolerability compared to OFF; 20% of patients in the mFOLFOX6 arm compared to 2% in the 5-FU/LV arm stopped the treatment due to side effects. The combination did not improve mPFS as the primary endpoint of the trial (mFOLFOX6: 3.1 months, 5-FU/LV: 2.9 months; p = 0.99). OS was even shorter in the mFOLFOX6 arm (6.1 months vs. 9.9 months; p = 0.02). This may be explained by the high rate of post-progression treatment in the 5-FU/LV arm (FU/LV: 25% vs. mFOLFOX6: 7%; p = 0.015) [37]. However, even with a high rate of post-progression therapy, an mOS of 9.9 months achieved with only 5-FU/LV in a second-line setting is difficult to explain when the optimal mOS with FOLFIRIOX in the first line is only 11 months.
Despite the fact that there are no data from prospective, randomized trials for the second-line treatment after gemcitabine plus nab-paclitaxel in the first line, 5-FU/FA plus nal-Iri or OFF is a therapeutic option in the second-line setting since its components have not been employed in the first line. However, oxaliplatin should only be employed when there is no ≥ 2 grade residual polyneuropathy from nab-paclitaxel.
The situation after FOLFIRINOX is more complex since the components of the NAPOLI-1 trial (nal-Iri, %-FU, LV) as well as the OFF (oxaliplatin, 5-FU, LV) protocol are also parts of the FOLFIRINOX protocol. There are no prospective, randomized trials evaluating a second-line treatment after first-line FOLFIRINOX. In the PRODIGE 4 Intergroup study, 47% of patients received second-line treatment, mainly gemcitabine (82.5%) or gemcitabine-based combinations (12.5%). The combination of gemcitabine plus nab-paclitaxel as second-line treatment after FOLFIRONOX has only been examined in retrospective analyses and small cohort studies [38]. The efficacy of gemcitabine plus nab-paclitaxel in the second-line setting may be high with an mOS of 8.8 months and an mPFS of 5.1 months in a small cohort study. However, the toxicity of the combination was also high, with a grade 3/4 toxicity of about 40% of the patients consisting of neutropenia (12.5%), neurotoxicity (12.5%), asthenia (9%), and thrombocytopenia (6.5%) [39]. Thus, if employed, this treatment should only be offered to patients with an ECOG of 0–1.
After the failure of second-line treatment, there is very few data suggesting the benefit of a third-line treatment and no large, randomized trials are available. In the NAPOLI trial, about 30% of patients had received more than one previous chemotherapy. The combination of 5-FU/FA plus nal-Iri was efficacious also in this setting, given the patients had not received irinotecan previously (no previous irinotecan: HR 0.62; previous irinotecan: HR 1.25). Thus, this combination can also be used in lines beyond the second line when irinotecan has not been used during the previous lines of treatment [33].

Therapeutic Options in Molecular Subgroups

Pancreatic cancer is characterized by high genomic heterogeneity. There are few subgroups that allow specific approaches. Recently, it could be shown that about 4–7% of an unselected Caucasian population exhibit germline mutation in the BRCA1 or -2 genes even without a clear family history [31, 40]. These tumors show a disturbed DNA homologous recombination leading to deficient repair of DNA double strand breaks. The tumors appear to be particularly sensitive to DNA crosslinking agents such as cisplatin or DNA repair inhibitors such as gemcitabine. This could be demonstrated in preclinical studies, but also in phase II trials examining the effect of gemcitabine plus cisplatin in mPDAC patients with a germline BRCA or PALB2 mutation [41].
The phase III POLO trial [31] examined maintenance treatment with the PARP inhibitor olaparib compared to placebo in patients with mPDAC and a germline mutation in BRCA1 or -2. Patients had to have at least stable disease under at least 16 weeks of a platinum-based first-line therapy; 81% of patients in the trial received FOLFIRINOX. In the overall population, mOS was about 19 months, stressing the beneficial role of a platinum-based treatment in patients with germ line BRCA1/2 mutations. Olaparib maintenance treatment significantly prolonged PFS, the primary endpoint of the study, compared to placebo (7.4 months vs. 3.8 months; HR 0.53; p = 0.004). However, there was no difference in overall survival between the two treatment arms (19 months vs. 19.2 months; HR 0.83, 95% CI 0.56–1.22; p = 0.3487) (see Table 1) [32]. Olaparib was well tolerated with anemia and fatigue as the most frequent ≥ grade 3 side effects.
Thus, patients with mPDAC should undergo testing for a germline BRCA1/2 mutation early after diagnosis since in these patients, a platinum-based treatment appears highly efficacious. At the moment, there is no direct comparison of whether cisplatin-based or oxaliplatin-based regimens are preferable in this situation[42]. Olaparib maintenance treatment is an interesting option for these specific patients. In case of proof of evidence of a BRCA-1/-2 germ line mutation, patients must be offered genetic counseling.

Immunotherapy

Immune checkpoint inhibitors like PD1/PD-L1 mAbs are particularly efficacious in tumors with a deficient mismatch repair system (dMMR) or high microsatellite instability (MSI-H) [43]. The frequency of dMMR pancreatic cancers is only about 1% [44, 45]. There are few data on the use of checkpoint inhibitors in MSI-H/dMMR mPDAC. In a cohort of 22 MSI-H patients with advanced PDAC, mPFS under treatment with pembrolizumab was 2.1 months and mOS was 4 months [46]. In patients responding to the treatment, the duration of response was long, with 13.4 months. Thus, unfortunately, only a subgroup of the patient with MSI-H /dMMR, but not the whole group, appears to benefit substantially from a checkpoint inhibitor treatment. So far, there is no evidence that checkpoint inhibitors have any benefit at all in PDACs without dMMR/MSI-H status.

Next Steps

Pancreatic cancer remains a difficult treatment area. However, identifying appropriate subgroups of pancreatic cancers may offer the chance to substantially improve outcomes. Such subgroups are tumors with somatic DNA damage repair deficiencies such as ATM [47, 48]. Also, the subgroup of KRAS wild-type tumors is interesting, exhibiting targetable fusions such as NRG1 fusions [49]. Another albeit very small subgroup PDACs with NTRK fusions that are also targetable. In addition, novel predictive screening approaches such as tumor–organoid pharmacotyping and treatment selection may also help to select a more efficacious treatment for an individual tumor [50]. Finally, the future potential availability of selective inhibitors of mutated KRAS could also mean substantial progress in PDAC treatment.

Declarations

Conflict of interest

Thomas J. Ettrich has received compensation from Servier and AstraZeneca for service on advisory boards, and has received travel support from Celgene. Thomas Seufferlein has received research funding from Celgene, and has received speaker's honoraria and compensation for service on advisory boards from Servier and Celgene.
Open AccessThis 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

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Jetzt bestellen und 100 € sparen!

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Jetzt bestellen und 100 € sparen!

Literatur
1.
Zurück zum Zitat Agalianos C, Gouvas N, Papaparaskeva K, Dervenis C. Positive para-aortic lymph nodes following pancreatectomy for pancreatic cancer. Systematic review and meta-analysis of impact on short term survival and association with clinicopathologic features. HPB. 2016;18(8):633–41.PubMedPubMedCentralCrossRef Agalianos C, Gouvas N, Papaparaskeva K, Dervenis C. Positive para-aortic lymph nodes following pancreatectomy for pancreatic cancer. Systematic review and meta-analysis of impact on short term survival and association with clinicopathologic features. HPB. 2016;18(8):633–41.PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat Cao F, Li J, Li A, Li F. Prognostic significance of positive peritoneal cytology in resectable pancreatic cancer: a systemic review and metaanalysis. Oncotarget. 2017;8(9):15004–13.PubMedPubMedCentralCrossRef Cao F, Li J, Li A, Li F. Prognostic significance of positive peritoneal cytology in resectable pancreatic cancer: a systemic review and metaanalysis. Oncotarget. 2017;8(9):15004–13.PubMedPubMedCentralCrossRef
3.
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.PubMedCrossRef 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.PubMedCrossRef
4.
Zurück zum Zitat Michalski CW, Erkan M, Huser N, Muller MW, Hartel M, Friess H, Kleeff J. Resection of primary pancreatic cancer and liver metastasis: a systematic review. Dig Surg. 2008;25(6):473–80.PubMedCrossRef Michalski CW, Erkan M, Huser N, Muller MW, Hartel M, Friess H, Kleeff J. Resection of primary pancreatic cancer and liver metastasis: a systematic review. Dig Surg. 2008;25(6):473–80.PubMedCrossRef
5.
Zurück zum Zitat National Comprehensive Cancer N: Pancreatic Adenocarcinoma, Version 1.2020. NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines). Fort Washington: NCCN; 2020. National Comprehensive Cancer N: Pancreatic Adenocarcinoma, Version 1.2020. NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines). Fort Washington: NCCN; 2020.
6.
Zurück zum Zitat Conroy T, Desseigne F, Ychou M, Bouche O, Guimbaud R, Becouarn Y, Adenis A, Raoul JL, Gourgou-Bourgade S, de la Fouchardiere C, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364(19):1817–25.PubMedCrossRef Conroy T, Desseigne F, Ychou M, Bouche O, Guimbaud R, Becouarn Y, Adenis A, Raoul JL, Gourgou-Bourgade S, de la Fouchardiere C, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364(19):1817–25.PubMedCrossRef
7.
Zurück zum Zitat Burris HA 3rd, Moore MJ, Andersen J, Green MR, Rothenberg ML, Modiano MR, Cripps MC, Portenoy RK, Storniolo AM, Tarassoff P, 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.PubMedCrossRef Burris HA 3rd, Moore MJ, Andersen J, Green MR, Rothenberg ML, Modiano MR, Cripps MC, Portenoy RK, Storniolo AM, Tarassoff P, 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.PubMedCrossRef
8.
Zurück zum Zitat National Institute for H, Care E: Pancreatic cancer in adults: diagnosis and management. NICE Guideline NG85. London: NICE; 2018. National Institute for H, Care E: Pancreatic cancer in adults: diagnosis and management. NICE Guideline NG85. London: NICE; 2018.
9.
Zurück zum Zitat Mallinson CN, Rake MO, Cocking JB, Fox CA, Cwynarski MT, Diffey BL, Jackson GA, Hanley J, Wass VJ. Chemotherapy in pancreatic cancer: results of a controlled, prospective, randomised, multicentre trial. Br Med J. 1980;281(6255):1589–91.PubMedPubMedCentralCrossRef Mallinson CN, Rake MO, Cocking JB, Fox CA, Cwynarski MT, Diffey BL, Jackson GA, Hanley J, Wass VJ. Chemotherapy in pancreatic cancer: results of a controlled, prospective, randomised, multicentre trial. Br Med J. 1980;281(6255):1589–91.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Palmer KR, Kerr M, Knowles G, Cull A, Carter DC, Leonard RC. Chemotherapy prolongs survival in inoperable pancreatic carcinoma. Br J Surg. 1994;81(6):882–5.PubMedCrossRef Palmer KR, Kerr M, Knowles G, Cull A, Carter DC, Leonard RC. Chemotherapy prolongs survival in inoperable pancreatic carcinoma. Br J Surg. 1994;81(6):882–5.PubMedCrossRef
11.
Zurück zum Zitat Glimelius B, Hoffman K, Sjoden PO, Jacobsson G, Sellstrom H, Enander LK, Linne T, Svensson C. Chemotherapy improves survival and quality of life in advanced pancreatic and biliary cancer. Ann Oncol. 1996;7(6):593–600.PubMedCrossRef Glimelius B, Hoffman K, Sjoden PO, Jacobsson G, Sellstrom H, Enander LK, Linne T, Svensson C. Chemotherapy improves survival and quality of life in advanced pancreatic and biliary cancer. Ann Oncol. 1996;7(6):593–600.PubMedCrossRef
12.
Zurück zum Zitat Gourgou-Bourgade S, Bascoul-Mollevi C, Desseigne F, Ychou M, Bouché O, Guimbaud R, Bécouar Y, Adenis A, Raoul JL, Boige V: Impact of FOLFIRINOX compared with gemcitabine on quality of life in patients with metastatic pancreatic cancer: results from the PRODIGE 4/ACCORD 11 randomized trial. J Clin Oncol. 2013;31(1):23.PubMedCrossRef Gourgou-Bourgade S, Bascoul-Mollevi C, Desseigne F, Ychou M, Bouché O, Guimbaud R, Bécouar Y, Adenis A, Raoul JL, Boige V: Impact of FOLFIRINOX compared with gemcitabine on quality of life in patients with metastatic pancreatic cancer: results from the PRODIGE 4/ACCORD 11 randomized trial. J Clin Oncol. 2013;31(1):23.PubMedCrossRef
13.
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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Macarulla T, Pazo-Cid R, Guillén-Ponce C, López R, Vera R, Reboredo M, Martin AM, Rivera F, Beveridge RD, La Casta A, et al. Phase I/II trial to evaluate the efficacy and safety of nanoparticle albumin-bound paclitaxel in combination with gemcitabine in patients with pancreatic cancer and an ECOG performance status of 2. J Clin Oncol. 2019;37(3):230–8.PubMedCrossRef Macarulla T, Pazo-Cid R, Guillén-Ponce C, López R, Vera R, Reboredo M, Martin AM, Rivera F, Beveridge RD, La Casta A, et al. Phase I/II trial to evaluate the efficacy and safety of nanoparticle albumin-bound paclitaxel in combination with gemcitabine in patients with pancreatic cancer and an ECOG performance status of 2. J Clin Oncol. 2019;37(3):230–8.PubMedCrossRef
15.
Zurück zum Zitat Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, Seay T, Tjulandin SA, Ma WW, Saleh MN et al: Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med 2013. Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, Seay T, Tjulandin SA, Ma WW, Saleh MN et al: Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med 2013.
16.
Zurück zum Zitat Goldstein D, El-Maraghi RH, Hammel P, Heinemann V, Kunzmann V, Sastre J, Scheithauer W, Siena S, Tabernero J, Teixeira L: Nab-paclitaxel plus gemcitabine for metastatic pancreatic cancer: long-term survival from a phase III trial. J Natl Cancer Inst 2015, 107(2). Goldstein D, El-Maraghi RH, Hammel P, Heinemann V, Kunzmann V, Sastre J, Scheithauer W, Siena S, Tabernero J, Teixeira L: Nab-paclitaxel plus gemcitabine for metastatic pancreatic cancer: long-term survival from a phase III trial. J Natl Cancer Inst 2015, 107(2).
17.
Zurück zum Zitat Blomstrand H, Scheibling U, Bratthäll C, Green H, Elander NO. Real world evidence on gemcitabine and nab-paclitaxel combination chemotherapy in advanced pancreatic cancer. BMC Cancer. 2019;19(1):40.PubMedPubMedCentralCrossRef Blomstrand H, Scheibling U, Bratthäll C, Green H, Elander NO. Real world evidence on gemcitabine and nab-paclitaxel combination chemotherapy in advanced pancreatic cancer. BMC Cancer. 2019;19(1):40.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Cutsem EV, Tempero MA, Sigal D, Oh D-Y, Fazio N, Macarulla T, Hitre E, Hammel P, Hendifar AE, Bates SE, et al. Randomized phase III trial of pegvorhyaluronidase alfa with nab-paclitaxel plus gemcitabine for patients with hyaluronan-high metastatic pancreatic adenocarcinoma. J Clin Oncol. 2020;38(27):3185–94.PubMedPubMedCentralCrossRef Cutsem EV, Tempero MA, Sigal D, Oh D-Y, Fazio N, Macarulla T, Hitre E, Hammel P, Hendifar AE, Bates SE, et al. Randomized phase III trial of pegvorhyaluronidase alfa with nab-paclitaxel plus gemcitabine for patients with hyaluronan-high metastatic pancreatic adenocarcinoma. J Clin Oncol. 2020;38(27):3185–94.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Thibodeau S, Voutsadakis IA. FOLFIRINOX chemotherapy in metastatic pancreatic cancer: a systematic review and meta-analysis of retrospective and phase II studies. J Clin Med. 2018;7(1):7.PubMedCentralCrossRef Thibodeau S, Voutsadakis IA. FOLFIRINOX chemotherapy in metastatic pancreatic cancer: a systematic review and meta-analysis of retrospective and phase II studies. J Clin Med. 2018;7(1):7.PubMedCentralCrossRef
20.
Zurück zum Zitat Hegewisch-Becker S, Aldaoud A, Wolf T, Krammer-Steiner B, Linde H, Scheiner-Sparna R, Hamm D, Janicke M, Marschner N, Group TPK. Results from the prospective German TPK clinical cohort study: treatment algorithms and survival of 1,174 patients with locally advanced, inoperable, or metastatic pancreatic ductal adenocarcinoma. Int J Cancer. 2019;144(5):981–90.CrossRef Hegewisch-Becker S, Aldaoud A, Wolf T, Krammer-Steiner B, Linde H, Scheiner-Sparna R, Hamm D, Janicke M, Marschner N, Group TPK. Results from the prospective German TPK clinical cohort study: treatment algorithms and survival of 1,174 patients with locally advanced, inoperable, or metastatic pancreatic ductal adenocarcinoma. Int J Cancer. 2019;144(5):981–90.CrossRef
21.
Zurück zum Zitat Tong H, Fan Z, Liu B, Lu T. The benefits of modified FOLFIRINOX for advanced pancreatic cancer and its induced adverse events: a systematic review and meta-analysis. Sci Rep. 2018;8(1):8666.PubMedPubMedCentralCrossRef Tong H, Fan Z, Liu B, Lu T. The benefits of modified FOLFIRINOX for advanced pancreatic cancer and its induced adverse events: a systematic review and meta-analysis. Sci Rep. 2018;8(1):8666.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Stein SM, James ES, Deng Y, Cong X, Kortmansky JS, Li J, Staugaard C, Indukala D, Boustani AM, Patel V, et al. Final analysis of a phase II study of modified FOLFIRINOX in locally advanced and metastatic pancreatic cancer. Br J Cancer. 2016;114(7):737–43.PubMedCrossRef Stein SM, James ES, Deng Y, Cong X, Kortmansky JS, Li J, Staugaard C, Indukala D, Boustani AM, Patel V, et al. Final analysis of a phase II study of modified FOLFIRINOX in locally advanced and metastatic pancreatic cancer. Br J Cancer. 2016;114(7):737–43.PubMedCrossRef
23.
Zurück zum Zitat Ramanathan RK, McDonough SL, Philip PA, Hingorani SR, Lacy J, Kortmansky JS, Thumar J, Chiorean EG, Shields AF, Behl D, et al. Phase IB/II randomized study of FOLFIRINOX plus pegylated recombinant human hyaluronidase versus FOLFIRINOX alone in patients with metastatic pancreatic adenocarcinoma: SWOG S1313. J Clin Oncol. 2019;37(13):1062–9.PubMedPubMedCentralCrossRef Ramanathan RK, McDonough SL, Philip PA, Hingorani SR, Lacy J, Kortmansky JS, Thumar J, Chiorean EG, Shields AF, Behl D, et al. Phase IB/II randomized study of FOLFIRINOX plus pegylated recombinant human hyaluronidase versus FOLFIRINOX alone in patients with metastatic pancreatic adenocarcinoma: SWOG S1313. J Clin Oncol. 2019;37(13):1062–9.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Maisey N, Chau I, Cunningham D, Norman A, Seymour M, Hickish T, Iveson T, O’Brien M, Tebbutt N, Harrington A, et al. Multicenter randomized phase III trial comparing protracted venous infusion (PVI) fluorouracil (5-FU) with PVI 5-FU plus mitomycin in inoperable pancreatic cancer. J Clin Oncol. 2002;20(14):3130–6.PubMedCrossRef Maisey N, Chau I, Cunningham D, Norman A, Seymour M, Hickish T, Iveson T, O’Brien M, Tebbutt N, Harrington A, et al. Multicenter randomized phase III trial comparing protracted venous infusion (PVI) fluorouracil (5-FU) with PVI 5-FU plus mitomycin in inoperable pancreatic cancer. J Clin Oncol. 2002;20(14):3130–6.PubMedCrossRef
25.
Zurück zum Zitat Ducreux M, Rougier P, Pignon JP, Douillard JY, Seitz JF, Bugat R, Bosset JF, Merouche Y, Raoul JL, Ychou M, et al. A randomised trial comparing 5-FU with 5-FU plus cisplatin in advanced pancreatic carcinoma. Ann Oncol. 2002;13(8):1185–91.PubMedCrossRef Ducreux M, Rougier P, Pignon JP, Douillard JY, Seitz JF, Bugat R, Bosset JF, Merouche Y, Raoul JL, Ychou M, et al. A randomised trial comparing 5-FU with 5-FU plus cisplatin in advanced pancreatic carcinoma. Ann Oncol. 2002;13(8):1185–91.PubMedCrossRef
26.
Zurück zum Zitat Dahan L, Bonnetain F, Ychou M, Mitry E, Gasmi M, Raoul JL, Cattan S, Phelip JM, Hammel P, Chauffert B, et al. Combination 5-fluorouracil, folinic acid and cisplatin (LV5FU2-CDDP) followed by gemcitabine or the reverse sequence in metastatic pancreatic cancer: final results of a randomised strategic phase III trial (FFCD 0301). Gut. 2010;59(11):1527–34.PubMedCrossRef Dahan L, Bonnetain F, Ychou M, Mitry E, Gasmi M, Raoul JL, Cattan S, Phelip JM, Hammel P, Chauffert B, et al. Combination 5-fluorouracil, folinic acid and cisplatin (LV5FU2-CDDP) followed by gemcitabine or the reverse sequence in metastatic pancreatic cancer: final results of a randomised strategic phase III trial (FFCD 0301). Gut. 2010;59(11):1527–34.PubMedCrossRef
27.
Zurück zum Zitat Trouilloud I, Dupont-Gossard A-C, Malka D, Artru P, Gauthier M, Lecomte T, Aparicio T, Thirot-Bidault A, Lobry C, Asnacios A, et al. Fixed-dose rate gemcitabine alone or alternating with FOLFIRI.3 (irinotecan, leucovorin and fluorouracil) in the first-line treatment of patients with metastatic pancreatic adenocarcinoma: an AGEO randomised phase II study (FIRGEM). Eur J Cancer. 2014;50(18):3116–24.PubMedCrossRef Trouilloud I, Dupont-Gossard A-C, Malka D, Artru P, Gauthier M, Lecomte T, Aparicio T, Thirot-Bidault A, Lobry C, Asnacios A, et al. Fixed-dose rate gemcitabine alone or alternating with FOLFIRI.3 (irinotecan, leucovorin and fluorouracil) in the first-line treatment of patients with metastatic pancreatic adenocarcinoma: an AGEO randomised phase II study (FIRGEM). Eur J Cancer. 2014;50(18):3116–24.PubMedCrossRef
28.
Zurück zum Zitat Bachet JB, Hammel P, Desramé J, Meurisse A, Chibaudel B, André T, Debourdeau P, Dauba J, Lecomte T, Seitz JF, et al. Nab-paclitaxel plus either gemcitabine or simplified leucovorin and fluorouracil as first-line therapy for metastatic pancreatic adenocarcinoma (AFUGEM GERCOR): a non-comparative, multicentre, open-label, randomised phase 2 trial. Lancet Gastroenterol Hepatol. 2017;2(5):337–46.PubMedCrossRef Bachet JB, Hammel P, Desramé J, Meurisse A, Chibaudel B, André T, Debourdeau P, Dauba J, Lecomte T, Seitz JF, et al. Nab-paclitaxel plus either gemcitabine or simplified leucovorin and fluorouracil as first-line therapy for metastatic pancreatic adenocarcinoma (AFUGEM GERCOR): a non-comparative, multicentre, open-label, randomised phase 2 trial. Lancet Gastroenterol Hepatol. 2017;2(5):337–46.PubMedCrossRef
29.
Zurück zum Zitat Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, Au HJ, Murawa P, Walde D, Wolff RA, et al. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol. 2007;25(15):1960–6.PubMedCrossRef Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, Au HJ, Murawa P, Walde D, Wolff RA, et al. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol. 2007;25(15):1960–6.PubMedCrossRef
30.
Zurück zum Zitat Heinemann V, Vehling-Kaiser U, Waldschmidt D, Kettner E, Marten A, Winkelmann C, Klein S, Kojouharoff G, Gauler TC, von Weikersthal LF, et al. Gemcitabine plus erlotinib followed by capecitabine versus capecitabine plus erlotinib followed by gemcitabine in advanced pancreatic cancer: final results of a randomised phase 3 trial of the ‘Arbeitsgemeinschaft Internistische Onkologie’ (AIO-PK0104). Gut. 2013;62(5):751–9.PubMedCrossRef Heinemann V, Vehling-Kaiser U, Waldschmidt D, Kettner E, Marten A, Winkelmann C, Klein S, Kojouharoff G, Gauler TC, von Weikersthal LF, et al. Gemcitabine plus erlotinib followed by capecitabine versus capecitabine plus erlotinib followed by gemcitabine in advanced pancreatic cancer: final results of a randomised phase 3 trial of the ‘Arbeitsgemeinschaft Internistische Onkologie’ (AIO-PK0104). Gut. 2013;62(5):751–9.PubMedCrossRef
31.
Zurück zum Zitat Golan T, Hammel P, Reni M, Van Cutsem E, Macarulla T, Hall MJ, Park JO, Hochhauser D, Arnold D, Oh DY et al: Maintenance olaparib for germline BRCA-mutated metastatic pancreatic cancer. N Engl J Med 2019. Golan T, Hammel P, Reni M, Van Cutsem E, Macarulla T, Hall MJ, Park JO, Hochhauser D, Arnold D, Oh DY et al: Maintenance olaparib for germline BRCA-mutated metastatic pancreatic cancer. N Engl J Med 2019.
32.
Zurück zum Zitat Golan T, Hammel P, Reni M, Cutsem EV, Macarulla T, Hall MJ, Park JO, Hochhauser D, Arnold D, Oh D-Y, et al. Overall survival from the phase 3 POLO trial: maintenance olaparib for germline BRCA-mutated metastatic pancreatic cancer. J Clin Oncol. 2021;39:378–378.CrossRef Golan T, Hammel P, Reni M, Cutsem EV, Macarulla T, Hall MJ, Park JO, Hochhauser D, Arnold D, Oh D-Y, et al. Overall survival from the phase 3 POLO trial: maintenance olaparib for germline BRCA-mutated metastatic pancreatic cancer. J Clin Oncol. 2021;39:378–378.CrossRef
33.
Zurück zum Zitat Wang-Gillam A, Chung-Pin L, Bodoky G, Dean A, Yan-Shen S, Jameson G, Macarulla T, Kyung-Hun L, Cunningham D, Blanc JF, et al. Nanoliposomal irinotecan with fluorouracil and folinic acid in metastatic pancreatic cancer after previous gemcitabine-based therapy (NAPOLI-1): a global, randomised, open-label, phase 3 trial. Lancet. 2016;387(10018):545–57.PubMedCrossRef Wang-Gillam A, Chung-Pin L, Bodoky G, Dean A, Yan-Shen S, Jameson G, Macarulla T, Kyung-Hun L, Cunningham D, Blanc JF, et al. Nanoliposomal irinotecan with fluorouracil and folinic acid in metastatic pancreatic cancer after previous gemcitabine-based therapy (NAPOLI-1): a global, randomised, open-label, phase 3 trial. Lancet. 2016;387(10018):545–57.PubMedCrossRef
34.
Zurück zum Zitat Hubner RA, Cubillo A, Blanc JF, Melisi D, Von Hoff DD, Wang-Gillam A, Chen LT, Becker C, Mamlouk K, Belanger B. Quality of life in metastatic pancreatic cancer patients receiving liposomal irinotecan plus 5-fluorouracil and leucovorin. Eur J Cancer. 2019;106:24–33.PubMedCrossRef Hubner RA, Cubillo A, Blanc JF, Melisi D, Von Hoff DD, Wang-Gillam A, Chen LT, Becker C, Mamlouk K, Belanger B. Quality of life in metastatic pancreatic cancer patients receiving liposomal irinotecan plus 5-fluorouracil and leucovorin. Eur J Cancer. 2019;106:24–33.PubMedCrossRef
35.
Zurück zum Zitat Oettle H, Riess H, Stieler JM, Heil G, Schwaner I, Seraphin J, Gorner M, Molle M, Greten TF, Lakner V, et al. Second-line oxaliplatin, folinic acid, and fluorouracil versus folinic acid and fluorouracil alone for gemcitabine-refractory pancreatic cancer: outcomes from the CONKO-003 trial. J Clin Oncol. 2014;32(23):2423–9.PubMedCrossRef Oettle H, Riess H, Stieler JM, Heil G, Schwaner I, Seraphin J, Gorner M, Molle M, Greten TF, Lakner V, et al. Second-line oxaliplatin, folinic acid, and fluorouracil versus folinic acid and fluorouracil alone for gemcitabine-refractory pancreatic cancer: outcomes from the CONKO-003 trial. J Clin Oncol. 2014;32(23):2423–9.PubMedCrossRef
36.
Zurück zum Zitat Pelzer U, Schwaner I, Stieler J, Adler M, Seraphin J, Dorken B, Riess H, Oettle H. Best supportive care (BSC) versus oxaliplatin, folinic acid and 5-fluorouracil (OFF) plus BSC in patients for second-line advanced pancreatic cancer: a phase III-study from the German CONKO-study group. Eur J Cancer. 2011;47(11):1676–81.PubMedCrossRef Pelzer U, Schwaner I, Stieler J, Adler M, Seraphin J, Dorken B, Riess H, Oettle H. Best supportive care (BSC) versus oxaliplatin, folinic acid and 5-fluorouracil (OFF) plus BSC in patients for second-line advanced pancreatic cancer: a phase III-study from the German CONKO-study group. Eur J Cancer. 2011;47(11):1676–81.PubMedCrossRef
37.
Zurück zum Zitat Gill S, Ko YJ, Cripps C, Beaudoin A, Dhesy-Thind S, Zulfiqar M, Zalewski P, Do T, Cano P, Lam WYH, et al. PANCREOX: a randomized phase III study of fluorouracil/leucovorin with or without oxaliplatin for second-line advanced pancreatic cancer in patients who have received gemcitabine-based chemotherapy. J Clin Oncol. 2016;34(32):3914–20.PubMedCrossRef Gill S, Ko YJ, Cripps C, Beaudoin A, Dhesy-Thind S, Zulfiqar M, Zalewski P, Do T, Cano P, Lam WYH, et al. PANCREOX: a randomized phase III study of fluorouracil/leucovorin with or without oxaliplatin for second-line advanced pancreatic cancer in patients who have received gemcitabine-based chemotherapy. J Clin Oncol. 2016;34(32):3914–20.PubMedCrossRef
38.
Zurück zum Zitat de Jesus VHF, Camandaroba MPG, Calsavara VF, Riechelmann RP. Systematic review and meta-analysis of gemcitabine-based chemotherapy after FOLFIRINOX in advanced pancreatic cancer. Ther Adv Med Oncol. 2020;12:1758835920905408.PubMedPubMedCentralCrossRef de Jesus VHF, Camandaroba MPG, Calsavara VF, Riechelmann RP. Systematic review and meta-analysis of gemcitabine-based chemotherapy after FOLFIRINOX in advanced pancreatic cancer. Ther Adv Med Oncol. 2020;12:1758835920905408.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Portal A, Pernot S, Tougeron D, Arbaud C, Bidault AT, de la Fouchardière C, Hammel P, Lecomte T, Dréanic J, Coriat R, et al. Nab-paclitaxel plus gemcitabine for metastatic pancreatic adenocarcinoma after Folfirinox failure: an AGEO prospective multicentre cohort. Br J Cancer. 2015;113(7):989–95.PubMedPubMedCentralCrossRef Portal A, Pernot S, Tougeron D, Arbaud C, Bidault AT, de la Fouchardière C, Hammel P, Lecomte T, Dréanic J, Coriat R, et al. Nab-paclitaxel plus gemcitabine for metastatic pancreatic adenocarcinoma after Folfirinox failure: an AGEO prospective multicentre cohort. Br J Cancer. 2015;113(7):989–95.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Holter S, Borgida A, Dodd A, Grant R, Semotiuk K, Hedley D, Dhani N, Narod S, Akbari M, Moore M, et al. Germline BRCA mutations in a large clinic-based cohort of patients with pancreatic adenocarcinoma. J Clin Oncol. 2015;33(28):3124–9.PubMedCrossRef Holter S, Borgida A, Dodd A, Grant R, Semotiuk K, Hedley D, Dhani N, Narod S, Akbari M, Moore M, et al. Germline BRCA mutations in a large clinic-based cohort of patients with pancreatic adenocarcinoma. J Clin Oncol. 2015;33(28):3124–9.PubMedCrossRef
41.
Zurück zum Zitat O’Reilly EM, Lee JW, Zalupski M, Capanu M, Park J, Golan T, Tahover E, Lowery MA, Chou JF, Sahai V, et al. Randomized, multicenter, phase II trial of gemcitabine and cisplatin with or without veliparib in patients with pancreas adenocarcinoma and a germline BRCA/PALB2 mutation. J Clin Oncol. 2020;38(13):1378–88.PubMedPubMedCentralCrossRef O’Reilly EM, Lee JW, Zalupski M, Capanu M, Park J, Golan T, Tahover E, Lowery MA, Chou JF, Sahai V, et al. Randomized, multicenter, phase II trial of gemcitabine and cisplatin with or without veliparib in patients with pancreas adenocarcinoma and a germline BRCA/PALB2 mutation. J Clin Oncol. 2020;38(13):1378–88.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Pokataev I, Fedyanin M, Polyanskaya E, Popova A, Agafonova J, Menshikova S, Tryakin A, Rumyantsev A, Tjulandin S. Efficacy of platinum-based chemotherapy and prognosis of patients with pancreatic cancer with homologous recombination deficiency: comparative analysis of published clinical studies. ESMO Open. 2020;5(1):e000578.PubMedPubMedCentralCrossRef Pokataev I, Fedyanin M, Polyanskaya E, Popova A, Agafonova J, Menshikova S, Tryakin A, Rumyantsev A, Tjulandin S. Efficacy of platinum-based chemotherapy and prognosis of patients with pancreatic cancer with homologous recombination deficiency: comparative analysis of published clinical studies. ESMO Open. 2020;5(1):e000578.PubMedPubMedCentralCrossRef
43.
44.
Zurück zum Zitat Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, Skora AD, Luber BS, Azad NS, Laheru D, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 2015;372(26):2509–20.PubMedPubMedCentralCrossRef Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, Skora AD, Luber BS, Azad NS, Laheru D, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 2015;372(26):2509–20.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Humphris JL, Patch AM, Nones K, Bailey PJ, Johns AL, McKay S, Chang DK, Miller DK, Pajic M, Kassahn KS, et al. Hypermutation in pancreatic cancer. 2017;152(1):68. Humphris JL, Patch AM, Nones K, Bailey PJ, Johns AL, McKay S, Chang DK, Miller DK, Pajic M, Kassahn KS, et al. Hypermutation in pancreatic cancer. 2017;152(1):68.
46.
Zurück zum Zitat Marabelle A, Le DT, Ascierto PA, Di Giacomo AM, De Jesus-Acosta A, Delord JP, Geva R, Gottfried M, Penel N, Hansen AR, et al. Efficacy of pembrolizumab in patients with noncolorectal high microsatellite instability/mismatch repair-deficient cancer: results from the phase II KEYNOTE-158 study. J Clin Oncol. 2020;38(1):1–10.PubMedCrossRef Marabelle A, Le DT, Ascierto PA, Di Giacomo AM, De Jesus-Acosta A, Delord JP, Geva R, Gottfried M, Penel N, Hansen AR, et al. Efficacy of pembrolizumab in patients with noncolorectal high microsatellite instability/mismatch repair-deficient cancer: results from the phase II KEYNOTE-158 study. J Clin Oncol. 2020;38(1):1–10.PubMedCrossRef
47.
Zurück zum Zitat Gout J, Perkhofer L, Morawe M, Arnold F, Ihle M, Biber S, Lange S, Roger E, Kraus JM, Stifter K, et al. Synergistic targeting and resistance to PARP inhibition in DNA damage repair-deficient pancreatic cancer. Gut. 2021;70(4):743–60.PubMedCrossRef Gout J, Perkhofer L, Morawe M, Arnold F, Ihle M, Biber S, Lange S, Roger E, Kraus JM, Stifter K, et al. Synergistic targeting and resistance to PARP inhibition in DNA damage repair-deficient pancreatic cancer. Gut. 2021;70(4):743–60.PubMedCrossRef
48.
Zurück zum Zitat Perkhofer L, Gout J, Roger E, Kude de Almeida F, Baptista Simões C, Wiesmüller L, Seufferlein T, Kleger A: DNA damage repair as a target in pancreatic cancer: state-of-the-art and future perspectives. Gut. 2021;70(3):606–17.PubMedCrossRef Perkhofer L, Gout J, Roger E, Kude de Almeida F, Baptista Simões C, Wiesmüller L, Seufferlein T, Kleger A: DNA damage repair as a target in pancreatic cancer: state-of-the-art and future perspectives. Gut. 2021;70(3):606–17.PubMedCrossRef
49.
Zurück zum Zitat Heining C, Horak P, Uhrig S, Codo PL, Klink B, Hutter B, Fröhlich M, Bonekamp D, Richter D, Steiger K, et al. NRG1 fusions in KRAS wild-type pancreatic cancer. Cancer Discov. 2018;8(9):1087–95.PubMedCrossRef Heining C, Horak P, Uhrig S, Codo PL, Klink B, Hutter B, Fröhlich M, Bonekamp D, Richter D, Steiger K, et al. NRG1 fusions in KRAS wild-type pancreatic cancer. Cancer Discov. 2018;8(9):1087–95.PubMedCrossRef
50.
Zurück zum Zitat Tiriac H, Belleau P, Engle DD, Plenker D, Deschênes A, Somerville TDD, Froeling FEM, Burkhart RA, Denroche RE, Jang GH, 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, Deschênes A, Somerville TDD, Froeling FEM, Burkhart RA, Denroche RE, Jang GH, et al. Organoid profiling identifies common responders to chemotherapy in pancreatic cancer. Cancer Discov. 2018;8(9):1112–29.PubMedPubMedCentralCrossRef
Metadaten
Titel
Systemic Therapy for Metastatic Pancreatic Cancer
verfasst von
Thomas J. Ettrich, M.D.
Thomas Seufferlein, M.D.
Publikationsdatum
01.11.2021
Verlag
Springer US
Erschienen in
Current Treatment Options in Oncology / Ausgabe 11/2021
Print ISSN: 1527-2729
Elektronische ISSN: 1534-6277
DOI
https://doi.org/10.1007/s11864-021-00895-4

Weitere Artikel der Ausgabe 11/2021

Current Treatment Options in Oncology 11/2021 Zur Ausgabe

Upper Gastrointestinal Cancers (JD Berlin, Section Editor)

Treatment Approach to Adenocarcinoma of the Ampulla of Vater

Neuro-oncology (GJ Lesser, Section Editor)

Targeted Therapy for BRAF Mutant Brain Tumors

Upper Gastrointestinal Cancers (JD Berlin, Section Editor)

Treatment Paradigms for Older Adults with Pancreatic Cancer: a Nuanced Approach

Upper Gastrointestinal Cancers (JD Berlin, Section Editor)

Immunotherapy in Gastroesophageal Cancers: Current Evidence and Ongoing Trials

Bei seelischem Stress sind Checkpoint-Hemmer weniger wirksam

03.06.2024 NSCLC Nachrichten

Wie stark Menschen mit fortgeschrittenem NSCLC von einer Therapie mit Immun-Checkpoint-Hemmern profitieren, hängt offenbar auch davon ab, wie sehr die Diagnose ihre psychische Verfassung erschüttert

Antikörper mobilisiert Neutrophile gegen Krebs

03.06.2024 Onkologische Immuntherapie Nachrichten

Ein bispezifischer Antikörper formiert gezielt eine Armee neutrophiler Granulozyten gegen Krebszellen. An den Antikörper gekoppeltes TNF-alpha soll die Zellen zudem tief in solide Tumoren hineinführen.

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.