Minireviews Open Access
Copyright ©The Author(s) 2021. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Clin Oncol. Sep 24, 2021; 12(9): 787-799
Published online Sep 24, 2021. doi: 10.5306/wjco.v12.i9.787
Real-world evidence on first- and second-line palliative chemotherapy in advanced pancreatic cancer
Hakon Blomstrand, Nils Oskar Elander, Department of Biomedical and Clinical Sciences, Linköping University, Linköping 58185, Sweden
Atul Batra, Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi 110029, India
Winson Y Cheung, Department of Oncology, University of Calgary, Calgary T2N 4N1, Canada
ORCID number: Hakon Blomstrand (0000-0002-9845-1410); Atul Batra (0000-0002-1934-8408); Winson Y Cheung (0000-0002-3679-4290); Nils Oskar Elander (0000-0001-9345-0278).
Author contributions: Blomstrand H, Cheung WY and Elander NO planned the study; Blomstrand H and Batra A performed the literature search; Blomstrand H, Batra A, Cheung WY and Elander NO assessed and interpreted the search results, and all were major contributors to the manuscript; all authors read and approved the final manuscript.
Conflict-of-interest statement: The authors declare that they do not have any conflicting interests.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Nils Oskar Elander, MD, PhD, Doctor, Department of Biomedical and Clinical Sciences, Linköping University, Linköping 58185, Sweden. nils.elander@liu.se
Received: February 28, 2021
Peer-review started: February 28, 2021
First decision: April 27, 2021
Revised: May 9, 2021
Accepted: August 12, 2021
Article in press: August 12, 2021
Published online: September 24, 2021

Abstract

In spite of recent diagnostic and therapeutic advances, the prognosis of pancreatic ductal adenocarcinoma (PDAC) remains very poor. As most patients are not amenable to curative intent treatments, optimized palliative management is highly needed. One key question is to what extent promising results produced by randomized controlled trials (RCTs) correspond to clinically meaningful outcomes in patients treated outside the strict frames of a clinical trial. To answer such questions, real-world evidence is necessary. The present paper reviews and discusses the current literature on first- and second-line palliative chemotherapy in PDAC. Notably, a growing number of studies report that the outcomes of the two predominant first-line multidrug regimens, i.e. gemcitabine plus nab-paclitaxel (GnP) and folfirinox (FFX), is similar in RCTs and real-life populations. Outcomes of second-line therapy following failure of first-line regimens are still dismal, and considerable uncertainty of the optimal management remains. Additional RCTs and real-world evidence studies focusing on the optimal treatment sequence, such as FFX followed by GnP or vice versa, are urgently needed. Finally, the review highlights the need for prognostic and predictive biomarkers to inform clinical decision making and enable personalized management in advanced PDAC.

Key Words: Pancreatic cancer, Palliative therapy, Cancer chemotherapy, Gemcitabine, Paclitaxel, nano albumin-bound, Folfirinox

Core Tip: This review summarizes and interprets published real-world evidence of the effectiveness and safety of treatment strategies in advanced pancreatic cancer. The real-world outcomes of first-line chemotherapy regimens such as folfirinox and gemcitabine/ nab-paclitaxel are thoroughly reviewed. The results of randomized controlled trials (RCTs) exploring the regimens seem to be largely generalizable in a real-world context. On second-line options, i.e. salvage chemotherapy following failure of first-line therapy, significant uncertainties remain. Additional RCTs and real-world evidence studies addressing current and novel regimens, and the optimal sequence of these, are needed.



INTRODUCTION

Over the past decades, mortality has decreased for many types of cancer. One exception is pancreatic cancer (pancreatic ductal adenocarcinoma, PDAC), which is soon expected to overtake breast cancer as the second most common cause of cancer-related death[1]. The majority of PDAC patients still present with either locally advanced or metastasized disease, and hence are considered beyond curative potential. For those individuals, as well as for those resected patients who suffer from relapses, palliative systemic therapy and/or radiotherapy are the only treatment options available.

Historically, palliative treatment of PDAC was limited mostly to regimens based on 5-fluorouracil (5-FU), usually with modest results at best. In that setting, 5-FU treatment was more or less experimental, but evidence from a randomized trial in 1996 showed that palliative chemotherapy in PDAC improved median overall survival (mOS) as well as quality of life compared with the best supportive care only[2]. The year after, gemcitabine replaced 5-FU as the gold standard in this clinical scenario based on the results of another randomized trial with prolonged mOS in favor of gemcitabine[3].

For a period of almost 15 years thereafter, many attempts to further improve the treatment in the setting of palliative PDAC were made by adding various cytotoxic drugs and monoclonal antibodies to gemcitabine, often resulting in increased toxicity without any significant survival benefit for patients[4-10]. A first breakthrough came in 2011, when a randomized controlled trial (RCT) showed a significant and clinically meaningful survival benefit over gemcitabine with the triplet combination chemotherapy known as Folfirinox (FFX, 11.1 mo vs 6.8 mo for gemcitabine monotherapy)[11]. The survival advantage occurred at the expense of considerably increased hematological and non-hematological toxicity in the intervention group. Another transformative RCT introduced the combination of gemcitabine and nano-albumin-bound paclitaxel (GnP). The regimen produced a smaller effect on overall survival (8.5 mo vs 6.8 mo for gemcitabine monotherapy). Nonetheless, it also resulted in increased toxicity, especially myelosuppression and chemotherapy-induced peripheral neuropathy[12]. The finding of more than tripled objective response ratio for the intervention group compared with gemcitabine (roughly 30% vs 10%) in both trials indicated a substantial antitumoral effect with the use of those combination regimens. Conversely, the treatment response duration of first-line therapy was usually short, and the RCT-population typically included highly selected patients with lower comorbidity and frailty compared with real-life patients. Whether survival and toxicity data from trials are generalizable to patients treated in routine clinical practice is unclear.

Regarding second-line treatment in PDAC, evidence is scarce. Empirical chemotherapy has been used in highly selected patients, and is usually reserved for very fit or young patients who responded to first-line treatment. Most often, gemcitabine and 5-FU have been used either as monotherapy or in combination with either oxaliplatin or irinotecan. In one of the few RCTs conducted, Oettle et al[13] compared the combination of folinic acid and fluorouracil (FF) in a 42-day cycle with FF in combination with oxaliplatin (OFF). The latter regimen gave significantly longer median progression-free survival (mPFS) and mOS, even though the absolute increase in months was rather small (mPFS 2.0 mo vs 2.9 mo, and mOS 3.3 mo vs 5.9 mo for FF and OFF, respectively). The occurrence of low-grade neuropathy was more than five times higher (38% of patients) in the OFF group. In contrast, the PANCREOX RCT, which compared the commonly used regimens of 5-FU/leucovorin infusion (5-FU/LV) and modified FOLFOX6 (mFOLFOX6) did not show any advantage with the addition of oxaliplatin[14]. There was no significant difference in the primary mPFS endpoint (3.1 mo vs 2.9 mo for mFOLFOX6 and FU/LV). The mOS favored 5-FU/LV (9.9 mo vs 6.1 mo for mFOLFOX6). Furthermore, substantial toxicity was observed in the mFOLFOX6-arm, with grade 3-4 adverse events affecting a majority (63%) of participants[14]. A more recent RCT[15] explored the role of 5-FU/LV and liposomal irinotecan in the second-line setting. The combination showed a small survival benefit over 5-FU/LV alone (6.1 vs 4.2 mo). However, the 5-FU/LV and liposomal irinotecan combination has not gained widespread traction in countries such as Canada and Sweden because regulatory authorities and health technology assessment bodies have considered the treatment to be not economically justifiable[16-18].

As the results of RCTs may be difficult to interpret and properly implement as standard healthcare, it is essential to complement the basis of knowledge with real-world evidence. The aim of this review was to summarize and assess available studies reporting real-world evidence in support of first- and second-line palliative chemotherapy in advanced PDAC. In first-line therapy, the focus was restricted to the two most established multidrug regimens, i.e. FFX and GnP. For second-line therapy, where the evidence on the optimal regimen is weak, no restriction in terms of regimen was applied.

LITERATURE SEARCH

PubMed was searched on December 19, 2020 for studies with titles containing the phrases “pancreatic cancer” and “real world”. All results were assessed for potential relevance. Only studies of human pancreatic cancer in the palliative setting and written in English were selected for possible inclusion in this review. Additional requirements for inclusion were information related to chemotherapy (FFX and/or GnP in the first-line setting, or any regimen in the second-line setting); survival [(overall survival (OS) data were required, progression-free survival (PFS) data were optional, and surrogate markers for OS were not accepted); real-world study population, and study type (retrospective or prospective cohort trials). RCTs, published study protocols, case studies, and meeting abstracts were not included. Studies reporting data on several treatment regimens were included as long as either FFX or GnP was among them, and specific survival data and treatment intention for the regimens were clearly distinguishable and compatible with the criteria mentioned above. Included studies are presented in a structured way with key data in tables sorted by topic and year of publication.

RESULTS

The PubMed query on first-line therapy returned 87 publications. Following careful review with regard to the inclusion criteria and scope of this review, 14 articles were selected, four with data on GnP (Table 1), one reporting FFX data (Table 2), eight that compared FFX and GnP (Table 3), and one covering several first-line treatments. The PubMed search of second-line setting returned 17 articles of which 15 were potentially relevant. The articles were subclassified according to which first-line treatment (FFX or GnP) had been administered (Tables 4 and 5). In addition to the above mentioned articles, several papers that did not focus on a specific first- or second-line regimen, and/or described the treatment pattern in general terms, were identified and will be discussed in the relevant section of this review.

Table 1 Real-world studies of gemcitabine/nab-paclitaxel in the first-line setting[19-23].
Ref.LocationStudy design Stage M1 nRegimenmOS in moSubgroup analysismPFS in mo Remarks
Prager et al[19], 2021 AustriaProspective cohort100%317GnP10.6/10.2Age < 70/> 705.6/5.5No difference in frequent toxicities
Blomstrand et al[20,21], 2019/2020SwedenRetrospective cohort71%75GnP10.9Alb <3 7, age < 65 with shorter survival5.2Less hematotoxicity than MPACT
Ostwal et al[22], 2018IndiaRetrospective cohort83%78GnP11.65.6Grade III-IV toxicity 35%
Quinton et al[23], 2018United KingdomRetrospective cohort100%748.4-Hematotoxicity similar to MPACT
Table 2 Real-world studies of Folfirinox in the first-line setting[24].
Ref.LocationStudy design nM1RegimenmOS in momPFS in mo Remarks
Cavanna et al[24], 2019ItalyRetrospective cohort5074%FFX/mFFX10.15.6mFFX sign less toxicity
Table 3 Real-world studies comparing Folfirinox and Gemcitabine/Nab-paclitaxel in the first-line setting[25-32].
Ref.LocationStudy design nM1 RegimenmOS in mo, P valuePrognostic factorsmPFS in mo, P valueRemarks
Franco et al[25], 2020 SpainRetrospective cohort11950%FFX 59; GnP 60FFX 12.7; GnP 10.2; P = 0.912Ca19-9, NLR-Toxicity data not reported
Wang et al[26], 2019CanadaRetrospective cohort22558%FFX 92; GnP 87; Gem 46FFX 14.1; GnP 10.5; Gem 4.2-FFX 8.4; GnP 8.5; Gem 3.7Sign more hematotoxicity in FFX
Pusceddu et al[27], 2019-Review3813NAFFX 1690; GnP 21231.15 longer for FFX. P = 0.03--GnP more neurotoxicity and anemia. FFX more neutropenia
Chiorean et al[28], 2019-Review> 6915NAFFX > 3556; GnP > 3359FFX 15.9; GnP 14.4-FFX 11.7; GnP 8.5FFX more neutropenia, GnP more neuropathy
Papneja et al[29], 2019CanadaRetrospective cohort11977%FFX 86; GnP 33FFX 9.0; GnP 9.0S-Alb, male sex, 2nd line therapyFFX 6.0; GnP 4.0 Grade 1-2 thromboembolism, mucositis and neuropathy sign more in FFX. Among grade 3-4 toxicity only fatigue sign more in GnP group
Kordes et al[32], 2019SwedenRetrospective cohort595-FFX 31; GnP 66; Gem 185FFX 9.9; GnP 9.8; Gem 6.6--No sign differences in toxicity comparing FFX vs GnP
Cartwright et al[30], 2018United StatesRetrospective cohort486100%FFX 159; GnP 255; Gem 72FFX 11.4; GnP 9.8; Gem 4.4--No sign differences in toxicity comparing FFX vs GnP
Kim et al[31], 2018United StatesRetrospective cohort654100%FFX 317; GnP 337FFX 13.8; GnP 12.1; P = 0.96Age-Less toxicity in GnP group
Table 4 Real-world studies of second-line therapy following failure of Folfirinox[36-44].
Ref.nM12L regimenmPFS in momOS in moRemarksAE
Portal et al[36], 201557100%GnP5.18.8Prospective cohort38% grade 3-4 toxicity
Mita et al[37], 20193080%GnP3.87.6Phase II70% grade 3-4 toxicity
Tsang et al[38], 201915967%GnP 78; Gem 81-5.8; 4.6Population-based, three Canadian provinces-
Zhang et al[39], 20186073%; 75%; 73%GnP 30; Gem 8; BSC 223.6; 2.55.7; 3.8Single centerMore grade 3-4 fatigue in Gem
Nguyen et al[40], 20173077%GnP3.712.4Single centerGrade 3-4 thrombocytopenia (33%), anemia (23%), nausea (17%)
Bertocchi et al[41], 201523100%GnP3.05.0Single center-
Zhang et al[42], 20152882%GnP3.0 5.7Single centerGrade 3-4, anemia (25%), thrombocytopenia (25%), neutropenia (18%)
Caparello et al[44], 201671-GnP2.56.2Single center-
Rissy et al[43], 201712100%GnP4.9-Single-centerNo grade 3-4 toxicity reported
Table 5 Real-world studies of second-line treatment with Folfirinox following failure of gemcitabine/nab-paclitaxel or single-agent gemcitabine[46-51].
Ref.nM11L regimen2L regimenmPFS in momOS in moRemarks
Sawada et al[46], 2020104100%GnPModified; FFX3.97.0Bolus 5-FU omitted. 55% grade 3-4 toxicity
Matsumoto et al[47], 20202383%GnPFFX 12; mFFX 115.3; 4.36.9; 12.8No sign difference in toxicity between FFX/mFFX
Assaf et al[50], 201127100%GemFFX3.08.556% grade 3-4 neutropenia
Kobayashi et al[48], 201718100%GemFFX2.89.8Phase I/II. 83% grade 3-4 toxicity
Kim et al[51], 20183982%GemAttenuated; FFX3.88.5Oxaliplatin: 65 mg/m2. 41% grade 3-4 neutropenia
Chung et al[49], 20184879%GemReduced irinotecan and oxaliplatin; FFX5.89.0Phase IIIrinotecan: 120 mg/m2; Oxaliplatin: 60 mg/m2; 65% grade 3-4 neutropenia
First-line GnP combination chemotherapy

Studies evaluating the effect of GnP in the real-world setting are listed in chronological order in Table 1. One study prospectively evaluated the efficacy of the regimen in younger (< 70 years) vs older (> 70 years) patients and found no significant between-group differences of either mOS (10.6 mo and 10.2 mo) or adverse events[19]. Real-world survival outcomes were superior to those observed in the phase III MPACT trial[12]. The authors suggested that the difference could be explained by a larger fraction of patients proceeding to second-line treatment, 47.4%-56.2% in the real-world studies compared with 38% in the MPACT study[12]. It is also noteworthy that the proportion of patients with performance status (PS) 0 or 1 or corresponding Karnofsky score was somewhat higher than in the MPACT trial. Another study retrospectively evaluated the benefit of GnP in advanced PDAC and found an mOS of 10.9 mo in the entire cohort, and an mOS of 17.1 mo in the locally advanced group[20]. Hematological toxicity was less frequent than in the MPACT study. In the same cohort, multivariate analysis found that low albumin (< 36 g/L) and age (< 65) were significant predictors of worse survival[21].

An additional study found comparable survival outcomes with the use of non-cremophore-based paclitaxel and gemcitabine, with an mOS of 11.6 mo and an mPFS of 5.6 mo[22]. In this retrospective cohort, the majority of patients had metastatic disease (83%) and PS 1 (80%). Grade III-IV toxicity was reported in 36% of patients, with hematological toxicity as the most frequent type of adverse event. In another retrospective cohort analysis where all patients had metastatic disease, mOS was 8.4 mo[23]. Most patients were in PS 1 (66%) at the time of treatment initiation. Similar frequencies of hematological toxicity were seen, with grade III-IV neutropenia being the most frequently reported adverse event (35% of patients).

First-line FFX combination chemotherapy

Studies evaluating the effectiveness of FFX in the real-world setting are listed in chronological order in Table 2. One study evaluated FFX treatment in a retrospective cohort and reported an mPFS of 5.6 mo and an mOS of 10.1 mo[24]. The first 18 consecutive patients received full-dose FFX and the following 32 cases received dose-reduced modified FFX (mFFX), resulting in significantly lower toxicity with fewer hematological and non-hematological side-effects.

First-line FFX vs GnP

Studies comparing the real-world effectiveness of FFX and GnP are listed in chronological order in Table 3. One retrospective cohort study of first-line treatment of patients with metastatic PDAC reported an mOS of 12.7 mo with FFX and 10.2 mo with GnP[25]. Tumor marker serum CA-19-9 and neutrophil-lymphocyte-ratio (NLR) were associated with survival. Authors intended to analyze patients aged above 70 years separately but this group was too small. Hematological toxicity was evenly distributed between the two treatments. Of interest, neuropathy was only reported in two patients receiving FFX. A study that compared the real-world effectiveness of FFX, GnP and gemcitabine reported OS durations of 14.1, 10.5 and 4.2 mo for the three treatments, respectively[26]. FFX treated patients were significantly younger and had better PS, and OS was significantly longer in both FFX- and GnP-treated patients compared with gemcitabine. The majority of patients had metastatic disease (68%). For the subgroups with localized disease, median OS had not been reached at the time of publication. The occurrence of neutropenia, febrile neutropenia, and neuropathy was significantly more frequent in FFX treated patients. In a review article, slightly longer survival (an additional 1.2 mo) was noted in favor of FFX over GnP. Despite the numerical difference, the overall adjusted risk of death was similar regardless of the regimen administered[27]. Neurotoxicity and anemia were seen more frequently in GnP-treated patients; neutropenia was more often associated with FFX treatment. In another review, a similar, non-significant, survival benefit was seen for FFX, with a reported OS of 15.9 mo vs 14.4 mo for GnP[28]. PFS was 11.7 mo with FFX and 8.5 mo for GnP. Toxicity data were not consistently reported in the studies, but neutropenia was more often associated with FFX than with GnP. The opposite was observed for neuropathy. In a retrospective study that largely focused on metastatic PDAC patients (77%), equivalent survival for FFX and GnP was reported (OS 9.0 mo for both regimens, P = 0.88). However, PFS was slightly longer with FFX, although the difference was not statistically significant (6.0 mo for FFX vs 4.0 mo for GnP, P = 0.38)[29]. There were no significant differences in the frequencies of severe toxicity between the two regimens. Another retrospective study reported OS of 11.4, 9.8 and 4.4 mo for FFX, GnP and gemcitabine monotherapy, respectively. Again, the differences were not significant[30]. Patients receiving GnP were significantly older and had worse PS. Toxicities were evenly distributed between the treatment groups. No significant prognostic factors were found in multivariate analysis, except for PS 2+, which was associated with worse survival. In another Celgene-funded real-world retrospective cohort study, there was a slight, non-significant, trend that favored FFX over GnP, with an OS of 13.8 mo compared with 12.1 mo[31]. All patients had metastatic disease. Common side-effects such as nausea, vomiting, diarrhea and mucositis were less frequent in the GnP group. A Swedish retrospective study comparing palliative first-line treatment in a PDAC patient cohort that included 31 FFX, 66 GnP, and 185 gemcitabine patients reported OS of 9.9, 9.8 and 6.6 mo, respectively[32]. Patient characteristics, including age and PS, varied substantially among the three groups. No significant differences in grade 3 or higher toxicities were reported between FFX and GnP.

Second-line real-world studies

Second-line treatment in PDAC: Despite advancements in the first-line treatment of advanced PDAC, most patients progress and succumb to the disease. To date, three phase III randomized clinical trials have been reported in the second-line treatment space[13-15] and are thoroughly described above under the background heading. These three trials compared 5-FU alone vs 5-FU/oxaliplatin doublets[13,14] or 5-FU vs nal-irinothecan vs 5-FU/nal-irinothecan doublet[15], and were all conducted after the patients progressed on gemcitabine-based chemotherapy as first-line treatment for advanced PDAC. However, the contemporary first-line standard treatment includes FFX or GnP combinations for patients with good PS[11,12]. There are no randomized clinical trial data for second-line treatment specifically after failure on FFX and GnP. Second-line treatment of advanced pancreatic cancer is largely driven by the chemotherapy regimen administered in the first-line setting. In a large real-world study that examined the outcome of 167 patients with advanced PDAC using several treatment regimens, the mOS from start of second-line therapy (OS2) was 5.2 mo, and plasma albumin, serum CA-19-9, and performance status were identified as key prognostic factors[33].

Second-line treatment after first-line FFX: In the real world, such patients are usually treated with GnP combination or gemcitabine monotherapy. The initial supportive evidence for use of GnP after first-line use of FFX in advanced pancreatic cancer was published in the form of case reports[34,35]. Subsequently, a prospective multicenter cohort study of 57 patients treated with GnP after FFX failure reported an mPFS of 5.1 mo and an OS2 of 8.8 mo[36]. It is noteworthy that just over half of the patients who received FFX for advanced pancreatic cancer in the frontline setting were eligible to receive salvage therapy with GnP in this cohort study. The objective response rate was 17.5%, while the disease control rate was 58.0%. From the start of first-line chemotherapy, the median OS was 18.0 mo. Grade 3-4 toxicities were observed in 40.0% of patients, of which neutropenia and neuropathy were the two most common. Recently, a phase II study of 30 patients reported in this setting described an mPFS of 3.8 mo and an OS2 of 7.6 mo[37]. The corresponding figures from the start of first-line chemotherapy were 9.3 and 14.2 mo, respectively. The overall response rate was 13.3% and the disease control rate was 46.7%. Grade 3-4 toxicities were reported in 70.0% patients, the most common being neutropenia and neuropathy. Furthermore, several real-world studies have been reported to support the use of GnP as second-line treatment. A large population-based Canadian study compared the real-world data of 368 patients with advanced PDAC treated with first-line FFX across two provinces with differential access to second-line treatment[38]. Of these, 159 patients (43.2%) received second-line treatment that was equally allocated as GnP (49.1%) and single-agent gemcitabine (50.9%). In a secondary analysis, the mOS counted from the initiation of second-line chemotherapy (OS2) was slightly longer for GnP compared with (5.8 mo vs 4.6 mo, P = 0.01).

Another Canadian study included 60 patients with advanced PDAC who received FFX as the first-line treatment[39]. Of these, 30 patients (50.0%) were treated with GnP, 8 (13.3%) with gemcitabine alone, and 22 patients (37.7%) received optimal supportive care. The mPFS (3.6 mo vs 2.5 mo, P = 0.03), and OS2 (5.7 mo vs 3.8 mo, P = 0.03) were longer in patients who received GnP compared with gemcitabine (Table 4). Other real-world studies have reported similar PFS and OS2 with the use of GnP after failure of FFX[40-44]. Furthermore, a recently published systematic review that included 16 studies reported a higher overall response rate (14.4% vs 8.4%, P = 0.038), disease control rate (53.5% vs 30.2%, P < 0.001), PFS (3.6 mo vs 2.5 mo, P = 0.030), and OS2 (5.7 mo vs 3.8 mo, P = 0.030) with GnP than with gemcitabine monotherapy[45]. Similar grade 3/4 event rates were reported in the prespecified analysis (22.9% vs 34.6%, P = 0.415). Overall, GnP appears to be a reasonable second-line treatment after FFX and patients considered unfit for GnP may benefit from gemcitabine monotherapy, while those with a poor performance status should be offered the best supportive care.

Second-line treatment after first-line GnP: In the absence of a head-to-head comparison of FFX and GnP in advanced PDAC, a substantial proportion of patients are treated with GnP in the first-line setting. Several chemotherapy regimens using a combination of fluoropyrimidines with irinotecan and/or oxaliplatin have been used in the real-world as salvage, second-line therapy of such patients. It is intuitive to consider FFX in this setting. A recent retrospective analysis of 104 patients treated with modified FFX (i.e. intravenous oxaliplatin 85 mg/m2, intravenous irinotecan 150 mg/m2, and continuous infusion of 5-fluorouracil 2400 mg/m2 for 46 h without bolus infusion) in that setting reported an objective response rate of 10.6% and a disease control rate of 56.7%[46]. The median PFS and OS2 were 3.9 mo and 7.0 mo, respectively. Grade 3-4 adverse events were reported in 54.8% patients and included hematological toxicities and peripheral sensory neuropathy. A smaller study of 23 patients who received standard FFX (n = 12) and modified FFX (n = 11) reported a median PFS of 5.3 mo and an OS of 6.9 mo in patients who received standard dosages. The corresponding numbers for those receiving modified FFX were 4.3 and 12.8 mo, respectively[47]. The observed differences in survival between the FFX and mFFX groups were not statistically significant.

Other real-world studies have reported the effectiveness of either the standard or modified FFX regimen after failure of single-agent gemcitabine as first-line therapy[48-51]. The studies, which adopted several modifications of the original FFX regimen, reported a PFS of 2.8-5.8 mo and OS2 of 8.5-9.8 mo (Table 2). Overall, limited data from real-world studies supports the use of modified FFX after failure of GnP. However, it is an intensive chemotherapy regimen and a high rate of grade 3-4 adverse events has been reported in above-mentioned studies, primarily hematological events and peripheral neuropathy. Patient selection remains paramount for electing to use such a regimen.

A real-world study of 52 patients with gemcitabine-refractory advanced PDAC reported that nano-liposomal irinotecan with FF was associated with a median PFS of 3.8 mo and OS2 of 6.8 mo[52]. The figures closely mirror the outcome reported from the phase III NAPOLI-1 study[15]. Capecitabine combined with oxaliplatin has also been used in this setting,, and several studies have reported a PFS of around 3 mo and OS2 of approximately 6 mo[53-55]. The median PFS and OS with single-agent capecitabine in 41 patients who failed first-line therapy were reported to be 1.5 mo and 4.3 mo, respectively[56].

Therefore, in patients considered unfit for FFX as second-line treatment, a doublet chemotherapy with fluoropyrimidine and oxaliplatin or nano-liposomal irinotecan is reasonable, while monotherapy with capecitabine may be considered for those with borderline performance status. There are no clinical trials that have compared the efficacy of oxaliplatin with irinotecan in this setting. However, a meta-analysis reported that the combination of a fluoropyrimidine plus irinotecan significantly improved both PFS and OS2, while the oxaliplatin combination modestly improved PFS but not OS2[57]. The modest benefit with these regimens should be balanced with the associated adverse events, and best supportive care should be considered a viable option for patients with poor general condition.

Targeted therapy and immunotherapy

As survival is still short, even when the most effective modern combinations of cytotoxic drugs are administered to patients with good performance status, it is tempting to look for alternatives such as targeted therapies or immune checkpoint inhibitors for the treatment of advanced PDAC. While the major breakthrough is yet to come, some recent findings may have the potential to become game-changing treatments of at least some types of PDAC in the future.

Approximately one in every five patients with advanced PDAC harbors a germline or somatic mutation in the DNA damage repair pathway[58]. There are limited data to suggest that Poly (ADP-ribose) polymerase (PARP) inhibitors may be effective in such patients. For example, a retrospective analysis of patients with previously treated PDAC (median prior therapies = 2) harboring a mutation in the DNA damage repair pathway reported an objective response rate of 23%, PFS of 7.6 mo and OS of 16.5 mo with olaparib[59]. Another report of 30 patients with BRCA1/2 mutations and no available standard treatment options reported disease control rate of 31% and an objective response rate of 4% with olaparib[60]. The role of immunotherapy in advanced PDAC is still evolving. However, a low prevalence (< 2%) of deficient mismatch repair suggests a limited role of immune check point inhibitors in this setting, at least with the currently available drugs[61,62].

CONCLUSION

Pancreatic cancer not amenable to surgical resection remains one of the most difficult challenges for medical oncologists around the globe. Despite improved diagnostic imaging tools, most cases are detected at a stage where cure or long-term survival are not achievable. Nevertheless, there is reason for cautious optimism. Large RCTs over the last decade have introduced first-line FFX and GnP regimens as the current standard of care, which has significantly changed the treatment landscape. Although extrapolation of the outcomes observed in highly selected RCT populations should be done with great care, combined evidence from real-world studies across different countries and health care systems indicates that the regimens are effective and reasonably safe in the real-world setting. In several of the real-world evidence publications, FFX was associated with a slightly better median OS than GnP, but selection bias was probable. Thus, it is possible that the differences observed might be the result of less fit patients being prescribed GnP rather than FFX. A sufficiently large head-to-head RCT comparing first-line FFX and GnP would potentially resolve these issues, but such a study is unlikely to occur.

In terms of second-line therapies, there are still considerable gaps in our knowledge. The few available RCTs provide only limited guidance, and it is difficult to translate their results into real-life practice. Notably, none of the published RCTs addresses whether the sequence of FFX followed by GnP or GnP followed by FFX is the most feasible or beneficial approach. Still, those sequences are often advocated by expert guidelines, and several real-world experience studies support that strategy. The extrapolation of RCTs into the real world is, at least in theory, even more complex in the second-line setting because patients at that point in their disease trajectory are likely to be frailer than patients eligible for first-line therapy.

The accumulating real-world evidence presented in this review points to some key conclusions. Several multidrug regimens show promising potency and acceptable toxicity in the first-line scenario, and to a somewhat lesser extent, the second-line setting. Outcomes reported in RCTs seem to be relatively consistent when the respective regimens are administered in real-life patients. Larger and/or pooled real-world studies are needed to further explore prognostic and predictive parameters such as serum albumin, serum CA-19-9, NLR and other novel biomarkers. Regarding second-line chemotherapy, the RCTs and real-world studies published to date are not fully aligned, and the key question regarding the optimal sequence of regimens remains uncertain. While most patients in this situation have very short expected survival, the identification of reliable clinical and biochemical biomarkers could be very helpful to inform treatment decision making.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Oncology

Country/Territory of origin: Sweden

Peer-review report’s scientific quality classification

Grade A (Excellent): A

Grade B (Very good): 0

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Sugimoto M S-Editor: Gong ZM L-Editor: Filipodia P-Editor: Xing YX

References
1.  Ferlay J, Partensky C, Bray F. More deaths from pancreatic cancer than breast cancer in the EU by 2017. Acta Oncol. 2016;55:1158-1160.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 291]  [Cited by in F6Publishing: 261]  [Article Influence: 32.6]  [Reference Citation Analysis (0)]
2.  Glimelius B, Hoffman K, Sjödén PO, Jacobsson G, Sellström H, Enander LK, Linné T, Svensson C. Chemotherapy improves survival and quality of life in advanced pancreatic and biliary cancer. Ann Oncol. 1996;7:593-600.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 569]  [Cited by in F6Publishing: 602]  [Article Influence: 21.5]  [Reference Citation Analysis (0)]
3.  Burris HA 3rd, Moore MJ, Andersen J, Green MR, Rothenberg ML, Modiano MR, Cripps MC, Portenoy RK, Storniolo AM, Tarassoff P, Nelson R, Dorr FA, Stephens CD, Von Hoff DD. 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;15:2403-2413.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4270]  [Cited by in F6Publishing: 4043]  [Article Influence: 149.7]  [Reference Citation Analysis (0)]
4.  Heinemann V, Quietzsch D, Gieseler F, Gonnermann M, Schönekäs H, Rost A, Neuhaus H, Haag C, Clemens M, Heinrich B, Vehling-Kaiser U, Fuchs M, Fleckenstein D, Gesierich W, Uthgenannt D, Einsele H, Holstege A, Hinke A, Schalhorn A, Wilkowski R. Randomized phase III trial of gemcitabine plus cisplatin compared with gemcitabine alone in advanced pancreatic cancer. J Clin Oncol. 2006;24:3946-3952.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 464]  [Cited by in F6Publishing: 482]  [Article Influence: 26.8]  [Reference Citation Analysis (0)]
5.  Herrmann R, Bodoky G, Ruhstaller T, Glimelius B, Bajetta E, Schüller J, Saletti P, Bauer J, Figer A, Pestalozzi B, Köhne CH, Mingrone W, Stemmer SM, Tàmas K, Kornek GV, Koeberle D, Cina S, Bernhard J, Dietrich D, Scheithauer W; Swiss Group for Clinical Cancer Research;  Central European Cooperative Oncology Group. Gemcitabine plus capecitabine compared with gemcitabine alone in advanced pancreatic cancer: a randomized, multicenter, phase III trial of the Swiss Group for Clinical Cancer Research and the Central European Cooperative Oncology Group. J Clin Oncol. 2007;25:2212-2217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 418]  [Cited by in F6Publishing: 422]  [Article Influence: 24.8]  [Reference Citation Analysis (0)]
6.  Kindler HL, Wroblewski K, Wallace JA, Hall MJ, Locker G, Nattam S, Agamah E, Stadler WM, Vokes EE. Gemcitabine plus sorafenib in patients with advanced pancreatic cancer: a phase II trial of the University of Chicago Phase II Consortium. Invest New Drugs. 2012;30:382-386.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 62]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
7.  Louvet C, Labianca R, Hammel P, Lledo G, Zampino MG, André T, Zaniboni A, Ducreux M, Aitini E, Taïeb J, Faroux R, Lepere C, de Gramont A; GERCOR;  GISCAD. Gemcitabine in combination with oxaliplatin compared with gemcitabine alone in locally advanced or metastatic pancreatic cancer: results of a GERCOR and GISCAD phase III trial. J Clin Oncol. 2005;23:3509-3516.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 681]  [Cited by in F6Publishing: 704]  [Article Influence: 37.1]  [Reference Citation Analysis (1)]
8.  Rocha Lima CM, Green MR, Rotche R, Miller WH Jr, Jeffrey GM, Cisar LA, Morganti A, Orlando N, Gruia G, Miller LL. Irinotecan plus gemcitabine results in no survival advantage compared with gemcitabine monotherapy in patients with locally advanced or metastatic pancreatic cancer despite increased tumor response rate. J Clin Oncol. 2004;22:3776-3783.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 407]  [Cited by in F6Publishing: 418]  [Article Influence: 20.9]  [Reference Citation Analysis (0)]
9.  Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, Au HJ, Murawa P, Walde D, Wolff RA, Campos D, Lim R, Ding K, Clark G, Voskoglou-Nomikos T, Ptasynski M, Parulekar W; National Cancer Institute of Canada Clinical Trials Group. 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;25:1960-1966.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2755]  [Cited by in F6Publishing: 2667]  [Article Influence: 156.9]  [Reference Citation Analysis (0)]
10.  Philip PA, Benedetti J, Corless CL, Wong R, O'Reilly EM, Flynn PJ, Rowland KM, Atkins JN, Mirtsching BC, Rivkin SE, Khorana AA, Goldman B, Fenoglio-Preiser CM, Abbruzzese JL, Blanke CD. Phase III study comparing gemcitabine plus cetuximab versus gemcitabine in patients with advanced pancreatic adenocarcinoma: Southwest Oncology Group-directed intergroup trial S0205. J Clin Oncol. 2010;28:3605-3610.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 446]  [Cited by in F6Publishing: 478]  [Article Influence: 34.1]  [Reference Citation Analysis (0)]
11.  Conroy T, Desseigne F, Ychou M, Bouché O, Guimbaud R, Bécouarn Y, Adenis A, Raoul JL, Gourgou-Bourgade S, de la Fouchardière C, Bennouna J, Bachet JB, Khemissa-Akouz F, Péré-Vergé D, Delbaldo C, Assenat E, Chauffert B, Michel P, Montoto-Grillot C, Ducreux M; Groupe Tumeurs Digestives of Unicancer;  PRODIGE Intergroup. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364:1817-1825.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4838]  [Cited by in F6Publishing: 5100]  [Article Influence: 392.3]  [Reference Citation Analysis (1)]
12.  Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, Seay T, Tjulandin SA, Ma WW, Saleh MN, Harris M, Reni M, Dowden S, Laheru D, Bahary N, Ramanathan RK, Tabernero J, Hidalgo M, Goldstein D, Van Cutsem E, Wei X, Iglesias J, Renschler MF. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369:1691-1703.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4035]  [Cited by in F6Publishing: 4332]  [Article Influence: 393.8]  [Reference Citation Analysis (0)]
13.  Oettle H, Riess H, Stieler JM, Heil G, Schwaner I, Seraphin J, Görner M, Mölle M, Greten TF, Lakner V, Bischoff S, Sinn M, Dörken B, Pelzer U. 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;32:2423-2429.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 304]  [Cited by in F6Publishing: 305]  [Article Influence: 30.5]  [Reference Citation Analysis (0)]
14.  Gill S, Ko YJ, Cripps C, Beaudoin A, Dhesy-Thind S, Zulfiqar M, Zalewski P, Do T, Cano P, Lam WYH, Dowden S, Grassin H, Stewart J, Moore M. 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;34:3914-3920.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 165]  [Article Influence: 20.6]  [Reference Citation Analysis (0)]
15.  Wang-Gillam A, Li CP, Bodoky G, Dean A, Shan YS, Jameson G, Macarulla T, Lee KH, Cunningham D, Blanc JF, Hubner RA, Chiu CF, Schwartsmann G, Siveke JT, Braiteh F, Moyo V, Belanger B, Dhindsa N, Bayever E, Von Hoff DD, Chen LT; NAPOLI-1 Study Group. 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;387:545-557.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 691]  [Cited by in F6Publishing: 703]  [Article Influence: 87.9]  [Reference Citation Analysis (0)]
16.  United States Food and Drug Administration  FDA approves ONIVYDE (irinotecan liposome injection) for advanced pancreatic cancer. 2015 [cited 9 February 2021]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/Label/2015/207793 Lbl.pdf.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Pan-Canadian Oncology Drug Review  Irinotecan liposome (Onivyde) for metastatic pancreatic cancer. 2017 [cited 9 February 2021]. Available from: https://cadth.ca/sites/default/files/pcodr/pcodr_irinotecan_liposome_onivyde_mpc_in_cgr.pdf.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  NT-rådet  Onivyde (irinotekan) vid bukspottkörtelcancer. 2018 [cited 9 February 2021]. Available from: https://janusinfo.se/download/18.840e7ca163033c061f1e082/1535626615047/Irinotekan-(Onivyde)-180302.pdf.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Prager GW, Oehler L, Gerger A, Mlineritsch B, Andel J, Petzer A, Wilthoner K, Sliwa T, Pichler P, Winder T, Heibl S, Gruenberger B, Laengle F, Hubmann E, Korger M, Pecherstorfer M, Djanani A, Neumann HJ, Philipp-Abbrederis K, Wöll E, Trondl R, Arnold-Schrauf C, Eisterer W. Comparison of nab-paclitaxel plus gemcitabine in elderly versus younger patients with metastatic pancreatic cancer: Analysis of a multicentre, prospective, non-interventional study. Eur J Cancer. 2021;143:101-112.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 12]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
20.  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;19:40.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 39]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
21.  Blomstrand H, Green H, Fredrikson M, Gränsmark E, Björnsson B, Elander NO. Clinical characteristics and blood/serum bound prognostic biomarkers in advanced pancreatic cancer treated with gemcitabine and nab-paclitaxel. BMC Cancer. 2020;20:950.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 7]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
22.  Ostwal V, Sahu A, Zanwar S, Nayak L, Shrikhande SV, Shetty N, Gupta S, Ramaswamy A. Experience with non-cremophor-based paclitaxel-gemcitabine regimen in advanced pancreatic cancer: Results from a single tertiary cancer centre. Indian J Med Res. 2018;148:284-290.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
23.  Quinton AE, Gwynne SH, Yim KL. Nab-paclitaxel in combination with gemcitabine for the treatment of metastatic pancreas cancer: the South Wales experience. Med Oncol. 2018;35:115.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
24.  Cavanna L, Stroppa EM, Citterio C, Mordenti P, Di Nunzio C, Peveri S, Orlandi E, Vecchia S. Modified FOLFIRINOX for unresectable locally advanced/metastatic pancreatic cancer. A real-world comparison of an attenuated with a full dose in a single center experience. Onco Targets Ther. 2019;12:3077-3085.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
25.  Franco F, Camara JC, Martín-Valadés JI, López-Alfonso A, Marrupe D, Gutiérrez-Abad D, Martínez-Amores B, León A, Juez I, Pérez M, Royuela A, Ruiz-Casado A. Clinical outcomes of FOLFIRINOX and gemcitabine-nab paclitaxel for metastatic pancreatic cancer in the real world setting. Clin Transl Oncol. 2021;23:812-819.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 7]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
26.  Wang Y, Camateros P, Cheung WY. A Real-World Comparison of FOLFIRINOX, Gemcitabine Plus nab-Paclitaxel, and Gemcitabine in Advanced Pancreatic Cancers. J Gastrointest Cancer. 2019;50:62-68.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 40]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
27.  Pusceddu S, Ghidini M, Torchio M, Corti F, Tomasello G, Niger M, Prinzi N, Nichetti F, Coinu A, Di Bartolomeo M, Cabiddu M, Passalacqua R, de Braud F, Petrelli F. Comparative Effectiveness of Gemcitabine plus Nab-Paclitaxel and FOLFIRINOX in the First-Line Setting of Metastatic Pancreatic Cancer: A Systematic Review and Meta-Analysis. Cancers (Basel). 2019;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 65]  [Article Influence: 13.0]  [Reference Citation Analysis (1)]
28.  Chiorean EG, Cheung WY, Giordano G, Kim G, Al-Batran SE. Real-world comparative effectiveness of nab-paclitaxel plus gemcitabine versus FOLFIRINOX in advanced pancreatic cancer: a systematic review. Ther Adv Med Oncol. 2019;11:1758835919850367.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 38]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
29.  Papneja N, Zaidi A, Chalchal H, Moser M, Tan K, Olson C, Haider K, Shaw J, Ahmed S. Comparisons of Outcomes of Real-World Patients With Advanced Pancreatic Cancer Treated With FOLFIRINOX Versus Gemcitabine and Nab-Paclitaxel: A Population-Based Cohort Study. Pancreas. 2019;48:920-926.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 20]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
30.  Cartwright TH, Parisi M, Espirito JL, Wilson TW, Pelletier C, Patel M, Babiker HM. Clinical Outcomes with First-Line Chemotherapy in a Large Retrospective Study of Patients with Metastatic Pancreatic Cancer Treated in a US Community Oncology Setting. Drugs Real World Outcomes. 2018;5:149-159.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 29]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
31.  Kim S, Signorovitch JE, Yang H, Patterson-Lomba O, Xiang CQ, Ung B, Parisi M, Marshall JL. Comparative Effectiveness of nab-Paclitaxel Plus Gemcitabine vs FOLFIRINOX in Metastatic Pancreatic Cancer: A Retrospective Nationwide Chart Review in the United States. Adv Ther. 2018;35:1564-1577.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 45]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
32.  Kordes M, Yu J, Malgerud O, Gustafsson Liljefors M, Löhr J-. Survival Benefits of Chemotherapy for Patients with Advanced Pancreatic Cancer in A Clinical Real-World Cohort. Cancers (Basel). 2019;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 16]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
33.  Gränsmark E, Bågenholm Bylin N, Blomstrand H, Fredrikson M, Åvall-Lundqvist E, Elander NO. Real World Evidence on Second-Line Palliative Chemotherapy in Advanced Pancreatic Cancer. Front Oncol. 2020;10:1176.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
34.  Berger AK, Weber TF, Jäger D, Springfeld C. Successful treatment with nab-paclitaxel and gemcitabine after FOLFIRINOX failure in a patient with metastasized pancreatic adenocarcinoma. Onkologie. 2013;36:763-765.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 5]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
35.  Portal A, Pernot S, Siauve N, Landi B, Lepère C, Colussi O, Rougier P, Zaanan A, Verrière B, Taieb J. Sustained response with gemcitabine plus Nab-paclitaxel after folfirinox failure in metastatic pancreatic cancer: report of an effective new strategy. Clin Res Hepatol Gastroenterol. 2014;38:e23-e26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 18]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
36.  Portal A, Pernot S, Tougeron D, Arbaud C, Bidault AT, de la Fouchardière C, Hammel P, Lecomte T, Dréanic J, Coriat R, Bachet JB, Dubreuil O, Marthey L, Dahan L, Tchoundjeu B, Locher C, Lepère C, Bonnetain F, Taieb J. Nab-paclitaxel plus gemcitabine for metastatic pancreatic adenocarcinoma after Folfirinox failure: an AGEO prospective multicentre cohort. Br J Cancer. 2015;113:989-995.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 124]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
37.  Mita N, Iwashita T, Uemura S, Yoshida K, Iwasa Y, Ando N, Iwata K, Okuno M, Mukai T, Shimizu M. Second-Line Gemcitabine Plus Nab-Paclitaxel for Patients with Unresectable Advanced Pancreatic Cancer after First-Line FOLFIRINOX Failure. J Clin Med. 2019;8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 25]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
38.  Tsang ES, Spratlin J, Cheung WY, Kim CA, Kong S, Xu Y, Gill S. Real-world Outcomes Among Patients Treated With Gemcitabine-based Therapy Post-FOLFIRINOX Failure in Advanced Pancreatic Cancer. Am J Clin Oncol. 2019;42:903-908.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
39.  Zhang H, Kellett C, Lambert P, Kim CA. Efficacy and Tolerability of Second-line Nab-paclitaxel and Gemcitabine After Failure of First-line FOLFIRINOX for Advanced Pancreas Cancer: A Single-institution Experience. Clin Colorectal Cancer. 2018;17:e451-e456.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
40.  Nguyen KT, Kalyan A, Beasley HS, Singhi AD, Sun W, Zeh HJ, Normolle D, Bahary N. Gemcitabine/nab-paclitaxel as second-line therapy following FOLFIRINOX in metastatic/advanced pancreatic cancer-retrospective analysis of response. J Gastrointest Oncol. 2017;8:556-565.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 21]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
41.  Bertocchi P, Abeni C, Meriggi F, Rota L, Rizzi A, Di Biasi B, Aroldi F, Ogliosi C, Savelli G, Rosso E, Zaniboni A. Gemcitabine Plus Nab-Paclitaxel as Second-Line and Beyond Treatment for Metastatic Pancreatic Cancer: a Single Institution Retrospective Analysis. Rev Recent Clin Trials. 2015;10:142-145.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 21]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
42.  Zhang Y, Hochster H, Stein S, Lacy J. Gemcitabine plus nab-paclitaxel for advanced pancreatic cancer after first-line FOLFIRINOX: single institution retrospective review of efficacy and toxicity. Exp Hematol Oncol. 2015;4:29.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 44]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
43.  El Rassy E, Assi T, El Karak F, Ghosn M, Kattan J. Could the combination of Nab-paclitaxel plus gemcitabine salvage metastatic pancreatic adenocarcinoma after folfirinox failure? Clin Res Hepatol Gastroenterol. 2017;41:e26-e28.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 8]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
44.  Caparello C, Vivaldi C, Fornaro L, Musettini G, Pasquini G, Catanese S, Masi G, Lencioni M, Falcone A, Vasile E. Second-line therapy for advanced pancreatic cancer: evaluation of prognostic factors and review of current literature. Future Oncol. 2016;12:901-908.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 14]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
45.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
46.  Sawada M, Kasuga A, Mie T, Furukawa T, Taniguchi T, Fukuda K, Yamada Y, Takeda T, Kanata R, Matsuyama M, Sasaki T, Ozaka M, Sasahira N. Modified FOLFIRINOX as a second-line therapy following gemcitabine plus nab-paclitaxel therapy in metastatic pancreatic cancer. BMC Cancer. 2020;20:449.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 35]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
47.  Matsumoto T, Kurioka Y, Okazaki U, Matsuo Y, Kimura S, Miura K, Tsuduki T, Takagi S, Takatani M, Morishita H. FOLFIRINOX for Advanced Pancreatic Cancer Patients After Nab-Paclitaxel Plus Gemcitabine Failure. Pancreas. 2020;49:574-578.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 9]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
48.  Kobayashi N, Shimamura T, Tokuhisa M, Goto A, Endo I, Ichikawa Y. Effect of FOLFIRINOX as second-line chemotherapy for metastatic pancreatic cancer after gemcitabine-based chemotherapy failure. Medicine (Baltimore). 2017;96:e6769.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 18]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
49.  Chung MJ, Kang H, Kim HG, Hyun JJ, Lee JK, Lee KH, Noh MH, Kang DH, Lee SH, Bang S; Pancreatobiliary Cancer Study Group of Korean Society of Gastrointestinal Cancer. Multicenter phase II trial of modified FOLFIRINOX in gemcitabine-refractory pancreatic cancer. World J Gastrointest Oncol. 2018;10:505-515.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 23]  [Cited by in F6Publishing: 24]  [Article Influence: 4.0]  [Reference Citation Analysis (1)]
50.  Assaf E, Verlinde-Carvalho M, Delbaldo C, Grenier J, Sellam Z, Pouessel D, Bouaita L, Baumgaertner I, Sobhani I, Tayar C, Paul M, Culine S. 5-fluorouracil/leucovorin combined with irinotecan and oxaliplatin (FOLFIRINOX) as second-line chemotherapy in patients with metastatic pancreatic adenocarcinoma. Oncology. 2011;80:301-306.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 79]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
51.  Kim JH, Lee SC, Oh SY, Song SY, Lee N, Nam EM, Lee S, Hwang IG, Lee HR, Lee KT, Bae SB, Kim HJ, Jang JS, Lim DH, Lee HW, Kang SY, Kang JH. Attenuated FOLFIRINOX in the salvage treatment of gemcitabine-refractory advanced pancreatic cancer: a phase II study. Cancer Commun (Lond). 2018;38:32.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 20]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
52.  Kieler M, Unseld M, Bianconi D, Scheithauer W, Prager GW. A real-world analysis of second-line treatment options in pancreatic cancer: liposomal-irinotecan plus 5-fluorouracil and folinic acid. Ther Adv Med Oncol. 2019;11:1758835919853196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
53.  Bullock A, Stuart K, Jacobus S, Abrams T, Wadlow R, Goldstein M, Miksad R. Capecitabine and oxaliplatin as first and second line treatment for locally advanced and metastatic pancreatic ductal adenocarcinoma. J Gastrointest Oncol. 2017;8:945-952.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
54.  Chung KH, Ryu JK, Son JH, Lee JW, Jang DK, Lee SH, Kim YT. Efficacy of Capecitabine Plus Oxaliplatin Combination Chemotherapy for Advanced Pancreatic Cancer after Failure of First-Line Gemcitabine-Based Therapy. Gut Liver. 2017;11:298-305.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
55.  Bayoglu IV, Varol U, Yildiz I, Muslu U, Alacacioglu A, Kucukzeybek Y, Akyol M, Demir L, Dirican A, Cokmert S, Yildiz Y, Karabulut B, Uslu R, Tarhan MO. Second-line capecitabine and oxaliplatin combination for gemcitabine-resistant advanced pancreatic cancer. Asian Pac J Cancer Prev. 2014;15:7119-7123.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
56.  Park SJ, Kim H, Shin K, Lee MA, Hong TH. Oral chemotherapy for second-line treatment in patients with gemcitabine-refractory advanced pancreatic cancer. World J Gastrointest Oncol. 2019;11:1021-1030.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 10]  [Cited by in F6Publishing: 8]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
57.  Sonbol MB, Firwana B, Wang Z, Almader-Douglas D, Borad MJ, Makhoul I, Ramanathan RK, Ahn DH, Bekaii-Saab T. Second-line treatment in patients with pancreatic ductal adenocarcinoma: A meta-analysis. Cancer. 2017;123:4680-4686.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 25]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
58.  Park W, Chen J, Chou JF, Varghese AM, Yu KH, Wong W, Capanu M, Balachandran V, McIntyre CA, El Dika I, Khalil DN, Harding JJ, Ghalehsari N, McKinnell Z, Chalasani SB, Makarov V, Selenica P, Pei X, Lecomte N, Kelsen DP, Abou-Alfa GK, Robson ME, Zhang L, Berger MF, Schultz N, Chan TA, Powell SN, Reis-Filho JS, Iacobuzio-Donahue CA, Riaz N, O'Reilly EM. Genomic Methods Identify Homologous Recombination Deficiency in Pancreas Adenocarcinoma and Optimize Treatment Selection. Clin Cancer Res. 2020;26:3239-3247.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 128]  [Cited by in F6Publishing: 114]  [Article Influence: 28.5]  [Reference Citation Analysis (0)]
59.  Borazanci E, Korn R, Liang WS, Guarnieri C, Haag S, Snyder C, Hendrickson K, Caldwell L, Von Hoff D, Jameson G. An Analysis of Patients with DNA Repair Pathway Mutations Treated with a PARP Inhibitor. Oncologist. 2020;25:e60-e67.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 7]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
60.  Ahn ER, Garrett-Mayer E, Halabi S, Mangat PK, Calfa CJ, Alva AS, Suhag VS, Hamid O, Dotan E, Yang ESH, Alese OB, Yost KJ, Marr AS, Palmer MC, Thompson FL, Rygiel AL, Anderson ST, Islam S, Schilsky RL. Olaparib (O) in patients (pts) with pancreatic cancer with BRCA1/2 inactivating mutations: Results from the Targeted Agent and Profiling Utilization Registry (TAPUR) study. J Clin Oncol. 2020;38:4637-4637.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
61.  Middha S, Zhang L, Nafa K, Jayakumaran G, Wong D, Kim HR, Sadowska J, Berger MF, Delair DF, Shia J, Stadler Z, Klimstra DS, Ladanyi M, Zehir A, Hechtman JF. Reliable Pan-Cancer Microsatellite Instability Assessment by Using Targeted Next-Generation Sequencing Data. JCO Precis Oncol. 2017;2017.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 164]  [Article Influence: 23.4]  [Reference Citation Analysis (0)]
62.  Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, Lu S, Kemberling H, Wilt C, Luber BS, Wong F, Azad NS, Rucki AA, Laheru D, Donehower R, Zaheer A, Fisher GA, Crocenzi TS, Lee JJ, Greten TF, Duffy AG, Ciombor KK, Eyring AD, Lam BH, Joe A, Kang SP, Holdhoff M, Danilova L, Cope L, Meyer C, Zhou S, Goldberg RM, Armstrong DK, Bever KM, Fader AN, Taube J, Housseau F, Spetzler D, Xiao N, Pardoll DM, Papadopoulos N, Kinzler KW, Eshleman JR, Vogelstein B, Anders RA, Diaz LA Jr. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357:409-413.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3799]  [Cited by in F6Publishing: 4326]  [Article Influence: 618.0]  [Reference Citation Analysis (0)]