Minireviews Open Access
Copyright ©The Author(s) 2017. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Aug 28, 2017; 23(32): 5875-5886
Published online Aug 28, 2017. doi: 10.3748/wjg.v23.i32.5875
Nano albumin bound-paclitaxel in pancreatic cancer: Current evidences and future directions
Guido Giordano, Marianna Velocci, Tania Di Raimo, Mario Rosario D’Andrea, Medical Oncology Unit, San Filippo Neri Hospital, 00135 Roma, Italy
Guido Giordano, CRO Aviano National Cancer Institute, 33081 Aviano, Italy
Massimo Pancione, Department of Sciences and Technologies, University of Sannio, 82100 Benevento, Italy
Massimo Pancione, Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, Complutense University, 28040 Madrid, Spain
Nunzio Olivieri, Department of Biology, University of Naples, Federico II, Via Mezzocannone, 80134 Napoli, Italy
Pietro Parcesepe, Department of Pathology and Diagnostics, University of Verona Strada, 37134 Verona, Italy
Luigi Coppola, Anatomic Pathology Unit, San Filippo Neri, 00135 Roma, Italy
Giuseppe Toffoli, Experimental and Clinical Pharmacology Unit, CRO-National Cancer Institute Via F, 33081 Aviano (Pordenone), Italy
Author contributions: Giordano G and Olivieri N performed the literature research; Giordano G and Olivieri N wrote the paper; Giordano G, Olivieri N, Parcesepe P and Di Raimo T assessed figure and tables; Pancione M, Parcesepe P, Velocci M, Di Raimo T, Coppola L, Toffoli G and D’Andrea MR reviewed the paper; Giordano G supervised the project.
Conflict-of-interest statement: Giordano G discloses: advisor role for Celgene, Sanofi, Novartis; consultant for Celgene, expenses reimbursement from Celgene. All other authors disclose no conflict of interest. This article does not contain any studies with human or animal subjects performed by the any of the authors.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (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/
Correspondence to: Guido Giordano, MD, Medical Oncology Unit, San Filippo Neri Hospital, ASL Roma 1, Via Giovanni Martinotti 20, 00135 Roma, Italy. giordano.guido81@gmail.com
Telephone: +39-6-33062437 Fax: +39-6-33062445
Received: March 29, 2017
Peer-review started: April 1, 2017
First decision: June 5, 2017
Revised: July 3, 2017
Accepted: August 1, 2017
Article in press: August 2, 2017
Published online: August 28, 2017

Abstract

Pancreatic cancer (PDAC) is an aggressive and chemoresistant disease, representing the fourth cause of cancer related deaths in western countries. Majority of patients have unresectable, locally advanced or metastatic disease at time of diagnosis and the 5-year survival rate in these conditions is extremely low. For more than a decade gemcitabine has been the cornerstone of metastatic PDAC treatment, although survival benefit was very poor. PDAC cells are surrounded by an intense desmoplastic reaction that may create a barrier to the drugs penetration within the tumor. Recently PDAC stroma has been addressed as a potential therapeutic target. Nano albumin bound (Nab)-paclitaxel is an innovative molecule depleting tumor stroma, through interaction between albumin and secreted protein acidic and rich in cysteine. Addition of nab-paclitaxel to gemcitabine has showed activity and efficacy in metastatic PDAC first-line treatment improving survival and overall response rate vs gemcitabine alone in the MPACT phase III study. This combination represents one of the standards of care in advanced PDAC therapy and is suitable to a broader spectrum of patients compared to other schedules. Nab-paclitaxel is under investigation as a backbone of chemotherapy in novel combinations with target agents or immunotherapy in locally advanced or metastatic PDAC. In this article, we provide an updated and critical overview about the role of nab-paclitaxel in PDAC treatment based on the latest advances in preclinical and clinical research. Furthermore, we focus on the use of nab-paclitaxel within the context of metastatic PDAC treatment landscape and we discuss about future implications in the light of current clinical ongoing trials.

Key Words: Nano albumin bound-paclitaxel, Pancreatic cancer, Metastatic disease, Gemcitabine, Folfirinox

Core tip: In this article, we provide an updated and critical overview about the role of nano albumin bound (Nab)-paclitaxel in pancreatic cancer (PDAC) treatment based on the latest advances in preclinical and clinical research. Furthermore, we focus on the use of Nab-paclitaxel within the context of metastatic PDAC treatment landscape and we discuss about future implications in the light of current clinical ongoing trials.



INTRODUCTION

Pancreatic cancer (PDAC) is a lethal disease with an incidence rate nearly equal to its mortality and it is the fourth cause of cancer-related death worldwide[1,2]. Five-year survival rate is about 5% and it increases to 20% in patients receiving radical surgery, chemotherapy and radiation[3-5]. The majority of patients have locally advanced or metastatic disease at the time of diagnosis and chemotherapy represents the only curative chance, even if prognosis remains very poor[6]. For more than a decade, gemcitabine has been the cornerstone of treatment in metastatic setting, despite of a small advantage in terms of survival[7]. Recently, PDAC stroma has been addressed as a possible target for novel molecules[8,9]. PDAC is characterized by an intense desmoplastic reaction that may explain its high chemoresistance. Tumor stroma contributes to poor vascularization and high intratumoral pressure resulting in reduced drugs penetration within cancer cells[10,11]. Among stromal components, secreted protein acidic and rich in cysteine (SPARC) has been investigated as a potential therapeutic target because of its involvement in PDAC cells proliferation, migration, metastasization and escape mechanisms[12-15]. Nano albumin bound (Nab)-paclitaxel is an innovative molecule obtained by the combination of paclitaxel with nano-particles of albumin[16]. SPARC binds albumin and it has been postulated that “nab-technology” may enhance selective delivery and uptake of paclitaxel in cancer cells[17]. Following the results of the MPACT phase III study, nab-paclitaxel in combination with gemcitabine has become a standard of care in metastatic PDAC first-line therapy[18]. Accordingly, this doublet represents also a valid backbone for the new developing schedules. In this paper, we will focus on molecular structure, mechanism of action, preclinical data, clinical studies and future perspectives regarding nab-paclitaxel in PDAC treatment.

NAB-PACLITAXEL: MOLECULAR STRUCTURE AND MECHANISM OF ACTION

Nab-paclitaxel (ABI 007 or Abraxane®; Celgene) is a 130-nm, albumin-bound, formulation of paclitaxel without any solvents or ethanol[19]. The Cremophor® EL free formulation reduces the incidence of infusion adverse reactions and makes premedication with steroids not necessary[19]. This agent is prepared by homogenization of human serum albumin at 3%-4% concentration with paclitaxel[20]. Nab-paclitaxel particles have a reduced diameter that enhances intracellular paclitaxel delivery and thus higher antitumor activity[21]. This molecule has a greater distribution volume, higher concentration and a faster clearance than conventional paclitaxel[22]. After administration of equal doses of nab-paclitaxel and radiolabeled paclitaxel-cremophor to MX-1 xenograft athymic mice, nab-paclitaxel had a more rapid tumor uptake. This resulted in overall increase in the area under the curve and a more effective intratumoral accumulation of nab-paclitaxel compared to paclitaxel-cremophor[23]. Preclinical studies have shown that albumin facilitates transport of paclitaxel across endothelial cells through the gp60 albumin receptor/caveolin-1 pathway (Figure 1). Notably, endothelial transcytosis of nab-paclitaxel was inhibited by methyl β-cyclodextrin, a gp60/caveolin-1 transport inhibitor. In vitro, a significant 9.9 times increase of nab-paclitaxel binding to the endothelial cells of human umbilical vein compared to conventional paclitaxel was recorded. Accordingly, a 4.2 times increased endothelial transcytosis of paclitaxel linked to nano-particles of albumin was demonstrated[22,24]. Furthermore, passive transport through permeable peritumoral vessels could have a role in paclitaxel delivery to cancer cells[25]. Peculiar and tumor-selective mechanism of action may be partially elucidated by interactions between SPARC and albumin (Figure 1). SPARC is an albumin-binding glycoprotein, also known as osteonectin, overexpressed in different types of tumor such as breast, lung, PDAC and melanoma[26]. SPARC is expressed both in PDAC stroma and tumor cells, representing a potential target for nab-paclitaxel mechanism of action[26]. Nevertheless, preclinical data on engineered mouse models (KPC models) demonstrated that SPARC had no role in nab-paclitaxel internalization in tumor cells[27]. Consistently, tumor delivery of nab-paclitaxel was not related to SPARC expression in patient derived xenograft (PDX) PDAC murine models[28].

Figure 1
Figure 1 Nab-paclitaxel mechanism of action. A: After intravenous administration nab-paclitaxel is carried through bloodstream; B: Nab-paclitaxel binds gp60 albumin receptor on the endothelial cell; C: Endothelial transcytosis of nab-paclitaxel through the gp60 albumin receptor/caveolin - 1 pathway; D: Interaction between SPARC and nab-paclitaxel both on the pancreatic cancer cells and stromal fibroblasts. SPARC: Secreted protein acidic and rich in cysteine.
DEVELOPMENT OF NAB-PACLITAXEL IN PDAC PRECLINICAL MODELS

In the context of a phase I-II study by Von Hoff et al[29], PDAC PDX murine models were randomly assigned to control, gemcitabine 100 mg/kg intraperitoneally on days 1 and 5 weekly for 4 wk, nab-paclitaxel 30 mg/kg per day intravenously for 5 consecutive days, and gemcitabine plus nab-paclitaxel in the preceding regimens for 4 wk. This study aimed to evaluate: (1) tumor progression; (2) changes in the pancreatic stroma; and (3) intratumoral drug penetration. Tumor regression was observed in 18%, 36% and 64% of xenografts treated with gemcitabine, nab-paclitaxel alone and both in combination, respectively. A depleted desmoplastic stroma, combined with dilated blood vessels, was found in each of nab-paclitaxel treatment group. Nab-paclitaxel plus gemcitabine resulted in 2.8 fold increased intra-tumor concentration of gemcitabine, compared to gemcitabine alone[29]. Another study investigated nab-paclitaxel, gemcitabine, docetaxel or control in PDAC cell-lines AsPC-1, BxPC-3, MIA PaCa-2 and Panc-1a. Addition of docetaxel or nab-paclitaxel at IC25, reduced gemcitabine IC50. Tumor growth inhibition after gemcitabine, docetaxel or nab-paclitaxel was 67%, 31% and 72%, respectively. Tumor stromal density, measured through reduction in α-smooth muscle actin, S100A4 and collagen 1 expression, was decreased by nab-paclitaxel better than docetaxel. In the same study, xenograft model was used to evaluate the efficacy of gemcitabine, docetaxel or nab-paclitaxel. Nab-paclitaxel followed by gemcitabine resulted in a longer median survival vs control or gemcitabine alone[30]. Notably, combination of nab-paclitaxel and gemcitabine improved survival, reduced incidence of metastasis and increased intratumoral gemcitabine levels in KPC mouse models. Those effects were explained by the decrease of cytidine-deaminase, a gemcitabine-metabolizing enzyme[31]. Engineered mice models of PDAC treated with nab-paclitaxel and gemcitabine, showed a distorted collagen with low cellular content in a preclinical-clinical study. Conversely, cancer-associated fibroblasts were increased in gemcitabine-treated mice. In the clinical section of this study, surgical specimen from patients with resectable PDAC, receiving preoperative treatment with two cycles of nab-paclitaxel and gemcitabine were studied to determine the collagen content and cancer associated fibroblasts density. Tumor samples of patients treated with nab-paclitaxel plus gemcitabine, showed a less abundant fibrillar collagen matrix around the tumor. Consistently, cancer associated fibroblasts were decreased in number, without effects on their activity[32]. Recently, a preclinical study definitively demonstrated that nab-paclitaxel was superior to conventional paclitaxel in blocking primary tumor progression, depletion of tumor stroma and resulted in a longer survival in PDAC murine models[33].

NAB-PACLITAXEL AND GEMCITABINE: A STANDARD OF CARE IN METASTATIC PDAC TREATMENT

Combination of nab-paclitaxel and gemcitabine in metastatic PDAC patients was investigated in an open label phase I-II study conducted in four United States centers. In the phase I part of this trial, primary endpoint was to identify the maximum tolerated dose (MTD) and dose-limiting toxicities (DLTs) of gemcitabine 1000 mg/m2 followed by nab-paclitaxel 100, 125 or 150 mg/m2 administered intravenously on day 1, 8 and 15 of a 28 d cycle. DLTs were sepsis and neutropenia and the MTD was fixed at level 2 (nab-paclitaxel 125 mg/m2). Phase II part of this study continued accrual at MTD to evaluate activity and safety of this combination. A cohort of 67 patients was enrolled and 44 received treatment at MTD. Median overall survival (OS) and progression free survival (PFS) in patients treated at MTD were 12.2 and 7.9 mo, respectively. Overall response rate (ORR) was 48% and overall disease control rate (DCR) was 68%. Most common grade ≥ 3 toxicities were represented by neutropenia (67%), leukopenia (44%), thrombocytopenia (23%), fatigue (21%) and neuropathy (15%). SPARC levels analysis was performed by immunohistochemistry in 36 tumor samples and patients were classified into high-SPARC and low-SPARC. High-SPARC was related to longer OS than low-SPARC (17.8 mo vs 8.1 mo, P = 0.0431) and it was significant predictor for OS at multivariate Cox regression model (P = 0.041)[29]. Phase I-II studies have been performed also in Asian patients. In Japanese population, no differences were observed in grade 3 or higher toxicities compared to the American phase I-II study, using the same schedule[34]. A Chinese phase I-II trial evaluated a different schedule of nab-paclitaxel plus gemcitabine. MTD was fixed at nab-paclitaxel 120 mg/m2 in combination with gemcitabine 1000 mg/m2 on day 1 and 8 every 3 wk[35].

MPACT phase III trial

The MPACT trial was an international, multicenter, open label, randomized phase III study involving 11 countries worldwide. Eligible patients were randomly assigned 1:1 to receive either nab-paclitaxel 125 mg/m2 plus gemcitabine 1000 mg/m2 on days 1, 8, 15 every 4 wk or gemcitabine alone weekly for 7 of 8 wk during first cycle and days 1, 8, 15 every 4 wk in subsequent cycles. Treatment was administered until disease progression or unacceptable toxicity. Eight-hundred sixty-one patients (431 in the experimental arm) were enrolled and stratified according Karnofsky Performance Status (KPS), presence of liver metastases and geographic region. Primary end-point was OS; secondary objectives were PFS and ORR. Age, gender, race, region and KPS were comparable in both study arms. No significant differences in pancreatic tumor site, location of metastases, number of metastatic sites, median CA19.9 levels, previous surgical or medical treatment and biliary stent implantation were observed between nab-paclitaxel plus gemcitabine and single agent gemcitabine groups. In the experimental arm, median duration of treatment was 3.9 mo and median dose intensity was 81% for nab-paclitaxel and 75% for gemcitabine. Addition of nab-paclitaxel to gemcitabine increased significantly median OS over gemcitabine alone (8.5 mo, 95%CI: 7.89-9.53 vs 6.7 mo, 95%CI: 6.01-7.23) with a 28% reduced risk of death (HR = 0.72, 95%CI: 0.62-0.82, P < 0.001). One and two-year survival rates were significantly higher in nab-paclitaxel and gemcitabine group compared to gemcitabine alone (35% vs 22%, P < 0.001 and 9% vs 4%, P = 0.02, respectively). Median PFS was prolonged by combination schedule (5.5 mo, 95%CI: 4.5-5.9 vs 3.7 mo, 95%CI: 3.6-4.0; HR = 0.69, 95%CI: 0.58-0.82, P < 0.001) with a significantly higher 12 mo- PFS rate than single agent gemcitabine (16% vs 9%). ORR was increased by addition of nab-paclitaxel to gemcitabine vs gemcitabine alone as assessed both by independent and investigator review (23% vs 7%, P < 0.001 and 29% vs 8%, P < 0.001, respectively). Notably, DCR, as evaluated by independent review, was higher in combination than single agent arm (48% vs 33%). Nab-paclitaxel and gemcitabine combination benefit was consistent across the different subgroups in terms of OS and PFS. Patients with more advanced disease, poorer PS, presence of liver metastases, more than 3 metastatic sites and CA19.9 higher than 59 times the upper limit of the normal range had the highest reduction in risk of death. Most common grade 3-4 hematologic events in the experimental arm were neutropenia (38%), leukopenia (31%), thrombocytopenia (13%) and most common non-haematological events were fatigue (17%) and peripheral neuropathy (17%)[18]. An updated analysis of MPACT trial with a longer median follow-up (13.9 mo) confirmed the superior OS in nab-paclitaxel plus gemcitabine vs single agent gemcitabine group (8.7 mo vs 6.6 mo, respectively) and identified 10%, 4% and 3% of long survivors in the combination arm at 24, 36 and 42 mo, respectively[36].

MPACT study: post hoc analyses

Nab-paclitaxel plus gemcitabine remained an independent predictor of improved OS and PFS also after correction for prognostic factors. In fact, MPACT investigators evaluated the effect of treatments according stratification factors and known prognostic factors (age, sex, KPS, peritoneal carcinomatosis, primary tumor location, pulmonary metastases, liver metastases, Whipple procedure, biliary stent implantation, stage at diagnosis, CA19.9 level). Combination regimen was more effective than gemcitabine in a larger spectrum of patients[37]. In addition, an elevated neutrophil to lymphocytes ratio (NLR ≥ 5) at baseline resulted as an independent prognostic factor related to a worse OS in an updated multivariate analysis[36]. Nab-paclitaxel plus gemcitabine combination showed activity both on primary and metastatic lesions in terms of tumor shrinkage benefit in a further analysis[38]. In MPACT trial, baseline CA19.9 levels were not a significant predictor of survival; conversely, CA19.9 decrease was related to better outcomes in both arms. An exploratory analysis confirmed the CA19.9 decrease at 8 wk as an early marker of treatment efficacy in terms of OS and ORR[39]. A pre-specified analysis evaluated the role of metabolic response detected by PET scan and compared metabolic response rate with treatment efficacy. Metabolic response rates at 8 wk as well as best metabolic response were higher in nab-paclitaxel plus gemcitabine arm. Metabolic response appeared as predictor of longer OS in both study arms[40]. Nab-paclitaxel plus gemcitabine was related to 54% of any grade neuropathy. Majority of patients experiencing grade 3 neuropathy, improved to grade 1 or less within one-month. Patients who developed peripheral neuropathy received a longer treatment and obtained a longer OS. Survival benefit increased according to the grade of peripheral neuropathy[41]. An exploratory analysis was performed in order to gain insight the role of SPARC expression as prognostic or predictive factor of survival. Neither baseline plasma SPARC levels, nor changes from baseline were predictors of OS. Stromal and tumor levels of SPARC, measured by immunohistochemistry had no correlation with OS[42]. Nab-paclitaxel plus gemcitabine increased OS, PFS and ORR, compared to gemcitabine alone in patients treated until disease progression. Accordingly, more than 50% of patients treated until progression received a second line therapy[43]. In the MPACT study 41% of patients had a nab-paclitaxel dose reduction, 71% had a dose delay, mostly after two cycles of treatment. In nab-paclitaxel plus gemcitabine arm, those modifications led to a longer treatment exposure and OS respect of patients who did not receive reductions or delays[44]. Forty-percent of patients treated in nab-paclitaxel and gemcitabine group received a second line treatment, mostly (74%) with fluoropyrimidines in combination regimens. Second line therapies were related to a significant benefit in OS in both study arms compared to patients without second line therapy. Notably, OS was significantly increased in patients treated with first line nab-paclitaxel plus gemcitabine vs gemcitabine alone (12.8 mo vs 9.0 mo, HR = 0.76, P = 0.015)[45].

NOVEL INVESTIGATIONS WITH NAB-PACLITAXEL IN PDAC

Hypoxia represents a common feature of several malignancies and it correlates with a worse prognosis and resistance to chemo- and radio-therapy[46,47]. TH-302 is a bromo-isophosphoramide mustard that works under conditions of hypoxia. The triplet containing nab-paclitaxel, gemcitabine and TH-302 exhibited superior efficacy in reducing stromal density and cell proliferation compared to TH-302 alone or nab-paclitaxel plus gemcitabine, in human PDAC xenograft models[48]. Combination of nab-paclitaxel with the oral fluoropyrimidine S-1 demonstrated to inhibit proliferation in PDAC cell lines and to decrease relative tumor volume in murine xenograft models[49,50]. An important pathway involved in PDAC stem cells maintenance is represented by Notch signaling[51]. Recently, the oncolytic adenovirus AdNuPARmE1A has showed to interfere with Notch pathway. In PDAC PDX mouse models, administration of nab-paclitaxel and gemcitabine combined with AdNuPARmE1A resulted in a higher therapeutic response than nab-paclitaxel plus gemcitabine combination and AdNuPARmE1A alone[52]. Tumor microenvironment plays a crucial role in PDAC aggressiveness and drug-resistance[53,54]. In this context, pancreatic stellate cells have been addressed as responsible for fibrosis and they have similar characteristics to the monocyte-macrophage lineage. Pamidronate and zoledronic acid are nitrogen-containing bisphosphonates that have showed in vitro anti-proliferative and pro-apoptotic activity on macrophages. In vivo, antitumor effect of nab-paclitaxel is enhanced by the combination with bisphosphonates in PDAC PDX mouse models[55]. Depending on predominant signals within the tumor microenvironment, macrophages can adopt a variety of functional states. In particular, macrophages with M2 phenotype acquire immuno-suppressive and tumor-promoting features[56]. In vitro and in vivo studies showed that nab-paclitaxel is internalized by macrophages via macro-pinocytosis, leading to a polarization towards a potentially tumoricidal M1 phenotype[57]. Insulin-like growth factor (IGF) signaling proteins are overexpressed in PDAC, predicting poor prognosis and higher aggressiveness. A small IGF-1 receptor and Insulin receptor reversible inhibitor (BMS-754807) reduced PDAC relative volume when combined with nab-paclitaxel in PDX mouse models[58].

Phase I-II studies

Several clinical trials in locally advanced or metastatic PDAC treatment included nab-paclitaxel in new therapeutic schedules. A phase I study has investigated the combination of FOLFOX plus nab-paclitaxel in a cohort of 35 patients, aiming to establish the nab-paclitaxel MTD. A dose of 150 mg/m2 every 2 wk was identified as MTD and a 20% reduction in oxaliplatin dose was recommended in patients who developed grade 2 neuropathy. This schedule showed a promising activity with 60% response rate and median survival of 15 mo[59]. The four-drug schedule combining cisplatin, nab-paclitaxel, capecitabine and gemcitabine (PAXG) has been tested in unresectable or borderline resectable PDAC patients. The PAXG regimen was investigated in a phase I trial on 24 patients with the objective to define the recommended phase 2 dose (RP2D) of nab-paclitaxel. A dose of 150 mg/m2 every 2 wk resulted as RP2D and major grade 3-4 toxicities were represented by neutropenia (31%), anemia (12%), fatigue (19%), hand-foot syndrome (12%). Disease control was obtained in all 21 patients and 67% of them showed a partial response. Six- and twelve-months PFS rates were 96% and 50% respectively[60]. This novel combination was evaluated in a randomized phase II study in which PAXG was compared to standard nab-paclitaxel plus gemcitabine. Fifty-four patients with unresectable or borderline resectable PDAC were randomized and 26 received PAXG. Primary endpoint of the study was resectability rate and each regimen would have been considered active if at least 4 patients per arm underwent to surgical resection. Both schedules resulted to be active and 5 patients in each arm underwent to surgery[61]. Combination of nab-paclitaxel and gemcitabine has been tested as a therapeutic backbone with target agents. A phase Ib trial evaluated the addition of erlotinib to nab-paclitaxel and gemcitabine in previously untreated advanced PDAC patients. The triplet was not feasible at standard dose of each single agent and it was necessary to reduce both nab-paclitaxel and erlotinib doses to 75 mg/m2 and 75 mg/d, respectively. Due to the small number of patients treated at dose level 3, no valid information about activity outcomes were obtained[62]. Recently, combination of nab-paclitaxel with both oral and intravenous fluoropyrimidines has been investigated. A single arm phase II trial evaluated the combination of capecitabine 825 mg/m2 orally bis in die on days 1-15 and nab-paclitaxel 125 mg/m2 intravenously on days 1 and 8 every 3 wk as a first line treatment in 30 PDAC patients. In patients without relevant adverse reaction after first cycle, the nab-paclitaxel dose was escalated to 100 mg/m2 on days 1, 8, 15 every 4 wk. ORR and DCR were 41.4% and 76%, respectively. Major grade 3 toxicities were peripheral neuropathy (23%), neutropenia (17%), hand-foot syndrome (13%), and phototoxic skin reaction (10%)[63]. A randomized phase II study compared the combination of nab-paclitaxel 125 mg/m2, leucovorin 400 mg/m2, 5-fluorouracil 400 mg/m2 bolus, followed by 2400 mg/m2 given as 46 h continuous infusion on day 1 and 15 every 4 wk (NABFU schedule) vs standard nab-paclitaxel plus gemcitabine. One hundred-fourteen patients were included with 2:1 randomization in favor of experimental arm. PFS at 4 mo was the primary endpoint. No differences were observed between the experimental arm and the standard treatment (55% vs 54%). NABFU regimen resulted in a higher median OS, as well as higher OS rates at 12 and 18 mo vs nab-paclitaxel plus gemcitabine (11.4 mo vs 9.2 mo, 48% vs 41% and 34% vs 31% respectively). Safety profile was similar in both study arms, except for grade 3 mucositis and diarrhea that were higher in the NABFU treatment group, reflecting the fluorouracil-related toxicities[64,65].

PUTTING NAB-PACLITAXEL IN PDAC LANDSCAPE: PAST, PRESENT…

Despite of promising preclinical and “early” clinical activity, paclitaxel and docetaxel have never showed superiority compared to gemcitabine in PDAC therapy[66]. Conversely, nano-technology improved bioavailability and intratumoral delivery of paclitaxel as well as avoided effects due to cremophor and therefore premedication with steroids. The albumin-bound formulation, acts on tumor microenvironment and depletes stroma through interaction between SPARC and albumin. This mechanism of action favors gemcitabine increased intratumoral concentration, making nab-paclitaxel an ideal partner for this drug. Nab-paclitaxel plus gemcitabine was the first combination to increase survival and response rates in metastatic PDAC treatment compared to gemcitabine alone. This schedule represents one of the standards of care in advanced PDAC first line treatment together with FOLFIRINOX and single agent gemcitabine[7,67,68]. Among these schedules, the most appropriate therapeutic choice should be guided by patient-related features (age, performance status, comorbidities, preferences), disease characteristics (tumor burden, metastatic sites, biliary stent implantation, bilirubin levels), also taking into consideration previous therapies and cost-effectiveness. In this context, FOLFIRINOX, according to the ACCORD/PRODIGE phase II-III study, could be an option in young, healthy and good performance status patients. Single agent gemcitabine could be used in unfit patients, with poor performance status and few comorbidities where a doublet or a triplet would not be feasible. Nab-paclitaxel and gemcitabine combination appears suitable for a broader spectrum of patients. In fact, MPACT trial was performed on a larger and more heterogeneous population than FOLFIRINOX and gemcitabine pivotal trials (Table 1). This study demonstrated that nab-paclitaxel plus gemcitabine improved survival in metastatic PDAC patients regardless to age, gender, geographic region, KPS, primary tumor location, presence of liver metastases, number of metastatic sites and baseline CA19.9 PDAC incidence is higher in elderly patients although this population is scarcely included in clinical trials[69]. Patients aged ≥ 75 years were not enrolled in the ACCORD/PRODIGE study; conversely they represented 10% of the MPACT population. Nab-paclitaxel plus gemcitabine showed efficacy and improved survival regardless age at a cut-off of 65 years; nevertheless data about “truly elderly” patients, aged over 75 years were available only on a small proportion of subjects. Real life experiences, with the limitation of retrospective series, suggest that the combination schedule was feasible, active, effective and well tolerated in elderly patients aged more than 70 and 75[70,71]. No data are available about the use of FOLFIRINOX in patients with poor performance status, in fact only subjects with ECOG PS 0-1 were included in the pivotal trial. Addition of nab-paclitaxel to gemcitabine did not improve survival in the KPS 70 (corresponding to ECOG PS of 2) compared to single agent gemcitabine, although these results were limited only to the 8% of MPACT study population. In order to make the best therapeutic choice in this population, physicians should differentiate between ECOG PS 2 due to “tumor burden” or due to comorbidities. In fact, as stated by European Society for Medical Oncology Guidelines for PDAC treatment, nab-paclitaxel plus gemcitabine could be considered an option in patients with ECOG PS 2 due to “heavy tumor load” for best chance of response[72]. Feasibility and flexibility of nab-paclitaxel plus gemcitabine combination allow to treat patients until disease progression. Data from MPACT study post-hoc analysis confirm that the higher exposure to both drugs correlates to better outcomes. Metastatic PDAC subjects treated until disease progression as well as receiving dose delays or modifications within the study, had a longer OS than patients who did not. Introduction of new, active schedules in PDAC first line treatment let to higher proportion of patients to benefit also from further lines of therapy. In fact, in ACCORD/PRODIGE and MPACT study population, 46% and 40% of patients received a second line chemotherapy in the FOLFIRINOX and nab-paclitaxel plus gemcitabine arm, respectively. Real life data show that this number is even higher in nab-paclitaxel and gemcitabine treated subjects, reaching more than 50%[73]. Second-line options that have demonstrated efficacy are oxaliplatin plus fluoro-folate (OFF schedule) and nano-liposomal irinotecan plus fluoro-folate according to CONKO-003 and NAPOLI-1 phase III trials results, respectively[74-76]. The choice of nab-paclitaxel as a first-line regimen allows the use of a non-cross-resistant second-line chemotherapy both with oxaliplatin and nano-liposomal irinotecan, whereas FOLFIRINOX does not. Finally, the optimal first-line therapeutic decision should take into account treatment-related costs. Recent data show that patient who initiated first line therapy with nab-paclitaxel plus gemcitabine has similar treatment duration but lower all-cause costs than FOLFIRINOX[77].

Table 1 Patients characteristics in metastatic pancreatic cancer pivotal trials.
GemcitabineFOLFIRINOXNab-paclitaxel + gemcitabine
Trial (yr)Gemcitabine vs 5-fluorouracil (1997)FOLFIRINOX vs Gemcitabine (2011)Nab-paclitaxel + Gemcitabine vs Gemcitabine (2013)
Phase1II-IIIIII
CountriesUnited states-Canada-United KingdomFranceWorldwide
Patients126342861
Median age61.56162
Elderly ( ≥ 75 yr)YesYes
ECOG PS 0-1YesYesYes
ECOG PS 2YesYes
Biliary StentYesYesYes
Multiple metastatic sitesYesYesYes
CONCLUSION

Innovative technology, peculiar mechanism of action and clinical activity of nab-paclitaxel have made this molecule an intriguing weapon for further studies in PDAC treatment. Several phase I, II and III trials are actually recruiting patients in PDAC therapy in order to investigate nab-paclitaxel based chemotherapy plus immunotherapy or target agents in the metastatic setting (Table 2). Furthermore, recent data from a randomized phase II trial suggest that sequential administration of nab-paclitaxel on day 1, 8, 15 and gemcitabine day 2, 9, 16 every 4 wk as first-line therapy in metastatic PDAC, may be more effective than a concomitant schedule and this approach warrants further studies[78]. Notably, combination of nab-paclitaxel and gemcitabine is under investigation in adjuvant setting (Table 3). APACT is a phase III, multicenter, open label, randomized study of nab-paclitaxel plus gemcitabine vs gemcitabine alone in subjects with surgically resected PDAC. This trial planned to enroll 846 patients evaluating the disease free survival as primary endpoint and study results are expected in the next future. Several studies are ongoing in the locally advanced, borderline resectable or unresectable PDAC (Table 3). In particular, LAPACT is a phase II, international, multicenter, open label, single-arm study in subjects with unresectable, locally advanced PDAC who have not received prior treatment. Nab-paclitaxel and gemcitabine was administered for a maximum of six cycles as induction therapy. Only in patients without disease progression or unacceptable toxicity, the investigator could choose among different options: (1) continuing chemotherapy with the same schedule; (2) chemo-radiation; and (3) surgical resection. Primary end-point was the time to treatment failure (time from the first day of treatment and disease progression or death). An interim analysis evaluated preliminary safety and efficacy data on 47 of 110 planned patients. Induction phase was completed in 60% of patients and 40% discontinued treatment, due to adverse events in the majority of cases. DCR was 80% and 13% of patients underwent to surgical resection. Major grade 3-4 toxicities were neutropenia (45%), anemia (13%) and fatigue (11%)[79]. Clinical trials in neo-adjuvant treatment or in locally advanced disease are evaluating integrated strategies containing nab-paclitaxel with both intensity modulated, image guided and stereotactic radiotherapy (Table 3). Actually there are no predictive biomarkers that may guide therapeutic choice in PDAC treatment and may help clinicians to choose the best regimen for each patient. An intense research is ongoing in order to investigate the potential role of micro-RNA as predictive of sensitivity and resistance to the drugs included in the FOLFIRINOX and nab-paclitaxel plus gemcitabine schedules[80].

Table 2 Principal clinical trials with nab-paclitaxel in metastatic pancreatic cancer.
Study IDSettingStudy drugsPhaseStatus
NCT02993731MetastaticNab-paclitaxel + Gemcitabine +/-NapabucasinIIIRecruiting
NCT02101021MetastaticNab-paclitaxel + Gemcitabine +/- MomelotinibIIIActive not recruiting
NCT02715804MetastaticNab-paclitaxel + Gemcitabine +/-PEGPH20IIIRecruiting
NCT02436668MetastaticNab-paclitaxel + Gemcitabine +/- IbrutinibII-IIIRecruiting
NCT02827201MetastaticSequential Nab-paclitaxel + Gemcitabine /FOLFIRIIIActive not recruiting
NCT02767557MetastaticNab-paclitaxel + Gemcitabine +/-tocilizumabIIRecruiting
NCT03086369MetastaticNab-paclitaxel + Gemcitabine +/- OlaratumabIINot yet recruiting
NCT02879318MetastaticNab-paclitaxel + Gemcitabine +/- Durvalumab + TremelimumabIIRecruiting
NCT02399137MetastaticNab-paclitaxel + Gemcitabine +IIRecruiting
MM-141
NCT02340117MetastaticNab-paclitaxel + Gemcitabine +IIRecruiting
SGT-53
NCT02124317MetastaticNab-paclitaxel + S-1IIActive not recruiting
NCT03076216MetastaticNab-paclitaxel + Gemcitabine + ONCOsilIINot yet recruiting
NCT02905578MetastaticNab-paclitaxel + Gemcitabine +IINot yet recruiting
High-dose ascorbate
NCT02732938MetastaticNab-paclitaxel + Gemcitabine + PF-04136309IIRecruiting
NCT02551991MetastaticNal-Iri or Gemcitabine +IIRecruiting
Nab-paclitaxel
NCT03009058MetastaticNab-paclitaxel + Gemcitabine +I-IINot yet recruiting
IMM 101
NCT02559674MetastaticNab-paclitaxel + Gemcitabine +I-IIRecruiting
ALT-803
NCT02705196MetastaticNab-paclitaxel + Gemcitabine + LOAd703 oncolytic virusI-IIRecruiting
NCT01834235MetastaticNab-paclitaxel + Gemcitabine +I-IIActive not recruiting
NPC-1C
NCT01730222MetastaticNab-paclitaxel + Gemcitabine + Capecitabine + CisplatinI-IIRecruiting
NCT02620800MetastaticNab-paclitaxel + 5-fluorouracil + Leucovorin + Bevacizumab + OxaliplatinI-IIRecruiting
NCT01506973MetastaticNab-paclitaxel + Gemcitabine + HydroxychloroquineI-IIRecruiting
NCT02504333MetastaticNab-paclitaxel + Gemcitabine followed by FOLFOXI-IIRecruiting
NCT02050178MetastaticNab-paclitaxel + Gemcitabine + OMP-54F28IActive not recruiting
NCT02309177MetastaticNab-paclitaxel + Nivolumab +/- GemcitabineIRecruiting
NCT02514031MetastaticNab-paclitaxel + Gemcitabine +IRecruiting
ARQ-761
NCT01934634MetastaticNab-paclitaxel + Gemcitabine + LCL161IActive not recruiting
NCT02101580MetastaticNab-paclitaxel + Gemcitabine +IActive not recruiting
ADI-PEG 20
NCT02138383MetastaticNab-paclitaxel + Gemcitabine + EnzalutamideIActive not recruiting
NCT02975141MetastaticNab-paclitaxel + Gemcitabine + AfatinibIRecruiting
NCT02227940MetastaticNab-paclitaxel + Gemcitabine + CeritinibIRecruiting
NCT02231723MetastaticNab-paclitaxel + Gemcitabine +IRecruiting
BBI608
NCT02501902MetastaticNab-paclitaxel + PalbociclibIRecruiting
Table 3 Principal clinical trials with nab-paclitaxel in pancreatic cancer “early settings”.
Study IDSettingStudy drugsPhaseStatus
NCT01964430AdjuvantNab-paclitaxel + Gemcitabine vs GemcitabineIIIRecruiting
NCT02506842AdjuvantNab-paclitaxel + Gemcitabine vs OFFIIIRecruiting
NCT02023021AdjuvantNab-paclitaxel + GemcitabineIIRecruiting
NCT02125136NeoadjuvantNab-paclitaxel + Gemcitabine vs FOLFIFINOXIIRecruiting
NCT02243007NeoadjuvantNab-paclitaxel + Gemcitabine vs FOLFIFINOXIIRecruiting
NCT02717091Neoadjuvant/borderline resectableNab-paclitaxel + Gemcitabine vs FOLFIFINOXIIRecruiting
NCT02481635NEO/adjuvantNab-paclitaxel + Gemcitabine + radiotherapyI-IIRecruiting
NCT01431794NeoadjuvantNab-paclitaxel + Gemcitabine + LDE-225I-IIActive not recruiting
NCT02588443NEO/adjuvantNab-paclitaxel + Gemcitabine + RO7009789IRecruiting
NCT02272738NeoadjuvantNab-paclitaxel + Gemcitabine + radiotherapyIRecruiting
NCT02506803NeoadjuvantNab-paclitaxel + GemcitabineIRecruiting
NCT02427841ResectableNab-paclitaxel + Gemcitabine followed by radiotherapyIIRecruiting
NCT02550327ResectableNab-paclitaxel + Gemcitabine + Cisplatin + AnakinraIRecruiting
NCT02210559Locally advancedNab-paclitaxel + Gemcitabine + FG-3019I-IIRecruiting
NCT02394535Locally advancedNab-paclitaxel + Capecitabine + radiotherapyIRecruiting
NCT02124369Borderline unresectableNab-paclitaxel + GemcitabineIIRecruiting
NCT02043730Unresectable locally advacedNab-paclitaxel + GemcitabineIIRecruiting
NCT02207465Unresectable locally advancedNab-paclitaxel + Gemcitabine + radiotherapyIRecruiting
NCT02272738Unresectable locally advancedNab-paclitaxel + GemcitabineIRecruiting
NCT01978184PreoperativeNab-paclitaxel + Gemcitabine + HydroxychloroquineIIRecruiting
NCT02427841PreoperativeNab-paclitaxel + Gemcitabine + radiotherapyIIRecruiting
NCT02723331PreoperativeNab-paclitaxel + Gemcitabine followed by SBRTIIRecruiting
NCT02930902PreoperativeNab-paclitaxel + Gemcitabine + Pembrolizumab + ParicalcitolIRecruiting

Introduction of nab-paclitaxel in Oncologist’s portfolio of drugs has represented a very important step forward in PDAC treatment. Nab-paclitaxel plus gemcitabine is one of the standards of care in advanced PDAC therapy and this combination is suitable for a wide spectrum of patients with different features and clinical presentations. This scenario could change rapidly as the result of nab-paclitaxel anticipation in “early settings” and further studies could be required in order to investigate a “re-challenge” in the metastatic disease.

ACKNOWLEDGMENTS

All authors thank Celgene Corporation for kind permission to adapt and modify Figure 1 about nab-paclitaxel mechanism of action from their educational slides kit.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Gastroenterology and Hepatology

Country of origin: Italy

Peer-review report classification

Grade A (Excellent): 0

Grade B (Very good): B, B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P- Reviewer: Giovannetti E, Lee KT S- Editor: Gong ZM L- Editor: A E- Editor: Zhang FF

References
1.  Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014;74:2913-2921.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3888]  [Cited by in F6Publishing: 4585]  [Article Influence: 458.5]  [Reference Citation Analysis (0)]
2.  Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin. 2015;65:5-29.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9172]  [Cited by in F6Publishing: 9815]  [Article Influence: 1090.6]  [Reference Citation Analysis (0)]
3.  SEER Stat Fact Sheets: Pancreas Cancer. 2015. Accessed on 2016-04-15.  Available from: http://seer.cancer.gov/statfacts/html/pancreas.html.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Brunner TB, Seufferlein T. Pancreatic cancer chemoradiotherapy. Best Pract Res Clin Gastroenterol. 2016;30:617-628.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 9]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
5.  Kim SM, Eads JR. Adjuvant and Neoadjuvant Therapy for Resectable Pancreatic and Periampullary Cancer. Surg Clin North Am. 2016;96:1287-1300.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
6.  Ilic M, Ilic I. Epidemiology of pancreatic cancer. World J Gastroenterol. 2016;22:9694-9705.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 877]  [Cited by in F6Publishing: 835]  [Article Influence: 104.4]  [Reference Citation Analysis (21)]
7.  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: 4042]  [Article Influence: 149.7]  [Reference Citation Analysis (0)]
8.  Garber K. Stromal depletion goes on trial in pancreatic cancer. J Natl Cancer Inst. 2010;102:448-450.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 55]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
9.  Hidalgo M. Pancreatic cancer. N Engl J Med. 2010;362:1605-1617.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2078]  [Cited by in F6Publishing: 2117]  [Article Influence: 151.2]  [Reference Citation Analysis (2)]
10.  Heinemann V, Reni M, Ychou M, Richel DJ, Macarulla T, Ducreux M. Tumour-stroma interactions in pancreatic ductal adenocarcinoma: rationale and current evidence for new therapeutic strategies. Cancer Treat Rev. 2014;40:118-128.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 90]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
11.  Li X, Ma Q, Xu Q, Duan W, Lei J, Wu E. Targeting the cancer-stroma interaction: a potential approach for pancreatic cancer treatment. Curr Pharm Des. 2012;18:2404-2415.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 51]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
12.  Arnold SA, Brekken RA. SPARC: a matricellular regulator of tumorigenesis. J Cell Commun Signal. 2009;3:255-273.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in F6Publishing: 128]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
13.  Podhajcer OL, Benedetti LG, Girotti MR, Prada F, Salvatierra E, Llera AS. The role of the matricellular protein SPARC in the dynamic interaction between the tumor and the host. Cancer Metastasis Rev. 2008;27:691-705.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 126]  [Cited by in F6Publishing: 129]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
14.  Infante JR, Matsubayashi H, Sato N, Tonascia J, Klein AP, Riall TA, Yeo C, Iacobuzio-Donahue C, Goggins M. Peritumoral fibroblast SPARC expression and patient outcome with resectable pancreatic adenocarcinoma. J Clin Oncol. 2007;25:319-325.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 281]  [Cited by in F6Publishing: 305]  [Article Influence: 17.9]  [Reference Citation Analysis (0)]
15.  Neuzillet C, Tijeras-Raballand A, Cros J, Faivre S, Hammel P, Raymond E. Stromal expression of SPARC in pancreatic adenocarcinoma. Cancer Metastasis Rev. 2013;32:585-602.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 94]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
16.  Borazanci E, Von Hoff DD. Nab-paclitaxel and gemcitabine for the treatment of patients with metastatic pancreatic cancer. Expert Rev Gastroenterol Hepatol. 2014;8:739-747.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 27]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
17.  Desai N. Nab Technology: A Drug Delivery Platform Utilizing Endothelial gp60 Receptor-based Transport and Tumor-derived SPARC for Targeting. Drug Deliv Rep. 2008;37-41.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, Seay T, Tjulandin SA, Ma WW, Saleh MN. 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: 4330]  [Article Influence: 393.6]  [Reference Citation Analysis (0)]
19.  Gradishar WJ. Albumin-bound paclitaxel: a next-generation taxane. Expert Opin Pharmacother. 2006;7:1041-1053.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 448]  [Cited by in F6Publishing: 435]  [Article Influence: 25.6]  [Reference Citation Analysis (0)]
20.  European Commission  Available from: https://ec.europa.eu/health/documents/communityregister/2015/.../anx_132551_it.pd.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Desai N. Challenges in development of nanoparticle-based therapeutics. AAPS J. 2012;14:282-295.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 487]  [Cited by in F6Publishing: 467]  [Article Influence: 38.9]  [Reference Citation Analysis (0)]
22.  Desai N, Trieu V, Yao Z, Louie L, Ci S, Yang A, Tao C, De T, Beals B, Dykes D. Increased antitumor activity, intratumor paclitaxel concentrations, and endothelial cell transport of cremophor-free, albumin-bound paclitaxel, ABI-007, compared with cremophor-based paclitaxel. Clin Cancer Res. 2006;12:1317-1324.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 807]  [Cited by in F6Publishing: 811]  [Article Influence: 45.1]  [Reference Citation Analysis (0)]
23.  Desai N, Yao Z, Trieu V. Evidence of a novel transporter mechanism for a Cremophorfree, protein-engineered paclitaxel (ABI-007) and enhanced in vivo antitumor activity in an MX-1 human breast tumor xenograft model. 25th Annual San Antonio Breast Conference Symposium, San Antonio, TX; 2002: 11-14. .  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Yardley DA. nab-Paclitaxel mechanisms of action and delivery. J Control Release. 2013;170:365-372.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 232]  [Cited by in F6Publishing: 297]  [Article Influence: 27.0]  [Reference Citation Analysis (0)]
25.  Chen N, Brachmann C, Liu X, Pierce DW, Dey J, Kerwin WS, Li Y, Zhou S, Hou S, Carleton M. Albumin-bound nanoparticle (nab) paclitaxel exhibits enhanced paclitaxel tissue distribution and tumor penetration. Cancer Chemother Pharmacol. 2015;76:699-712.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 72]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
26.  Seidel D, Cremer P. [Prevention of cardiovascular diseases by intervention in lipid metabolism]. Z Kardiol. 1992;81 Suppl 4:191-197.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Neesse A, Frese KK, Chan DS, Bapiro TE, Howat WJ, Richards FM, Ellenrieder V, Jodrell DI, Tuveson DA. SPARC independent drug delivery and antitumour effects of nab-paclitaxel in genetically engineered mice. Gut. 2014;63:974-983.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 144]  [Article Influence: 14.4]  [Reference Citation Analysis (0)]
28.  Kim H, Samuel S, Lopez-Casas P, Grizzle W, Hidalgo M, Kovar J, Oelschlager D, Zinn K, Warram J, Buchsbaum D. SPARC-Independent Delivery of Nab-Paclitaxel without Depleting Tumor Stroma in Patient-Derived Pancreatic Cancer Xenografts. Mol Cancer Ther. 2016;15:680-688.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 37]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
29.  Von Hoff DD, Ramanathan RK, Borad MJ, Laheru DA, Smith LS, Wood TE, Korn RL, Desai N, Trieu V, Iglesias JL. Gemcitabine plus nab-paclitaxel is an active regimen in patients with advanced pancreatic cancer: a phase I/II trial. J Clin Oncol. 2011;29:4548-4554.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 755]  [Cited by in F6Publishing: 835]  [Article Influence: 64.2]  [Reference Citation Analysis (2)]
30.  Hoge CW, Sethabutr O, Bodhidatta L, Echeverria P, Robertson DC, Morris JG Jr. Use of a synthetic oligonucleotide probe to detect strains of non-serovar O1 Vibrio cholerae carrying the gene for heat-stable enterotoxin (NAG-ST). J Clin Microbiol. 1990;28:1473-1476.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 99]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
31.  Frese KK, Neesse A, Cook N, Bapiro TE, Lolkema MP, Jodrell DI, Tuveson DA. nab-Paclitaxel potentiates gemcitabine activity by reducing cytidine deaminase levels in a mouse model of pancreatic cancer. Cancer Discov. 2012;2:260-269.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 312]  [Cited by in F6Publishing: 328]  [Article Influence: 27.3]  [Reference Citation Analysis (0)]
32.  Alvarez R, Musteanu M, Garcia-Garcia E, Lopez-Casas PP, Megias D, Guerra C, Muñoz M, Quijano Y, Cubillo A, Rodriguez-Pascual J. Stromal disrupting effects of nab-paclitaxel in pancreatic cancer. Br J Cancer. 2013;109:926-933.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 195]  [Cited by in F6Publishing: 252]  [Article Influence: 22.9]  [Reference Citation Analysis (0)]
33.  Rajeshkumar NV, Yabuuchi S, Pai SG, Tong Z, Hou S, Bateman S, Pierce DW, Heise C, Von Hoff DD, Maitra A. Superior therapeutic efficacy of nab-paclitaxel over cremophor-based paclitaxel in locally advanced and metastatic models of human pancreatic cancer. Br J Cancer. 2016;115:442-453.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 30]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
34.  Ueno H, Ikeda M, Ueno M, Mizuno N, Ioka T, Omuro Y, Nakajima TE, Furuse J. Phase I/II study of nab-paclitaxel plus gemcitabine for chemotherapy-naive Japanese patients with metastatic pancreatic cancer. Cancer Chemother Pharmacol. 2016;77:595-603.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 104]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
35.  Zhang DS, Wang DS, Wang ZQ, Wang FH, Luo HY, Qiu MZ, Wang F, Li YH, Xu RH. Phase I/II study of albumin-bound nab-paclitaxel plus gemcitabine administered to Chinese patients with advanced pancreatic cancer. Cancer Chemother Pharmacol. 2013;71:1065-1072.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 26]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
36.  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:pii: dju413.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 356]  [Cited by in F6Publishing: 418]  [Article Influence: 46.4]  [Reference Citation Analysis (0)]
37.  Tabernero J, Chiorean EG, Infante JR, Hingorani SR, Ganju V, Weekes C, Scheithauer W, Ramanathan RK, Goldstein D, Penenberg DN. Prognostic factors of survival in a randomized phase III trial (MPACT) of weekly nab-paclitaxel plus gemcitabine versus gemcitabine alone in patients with metastatic pancreatic cancer. Oncologist. 2015;20:143-150.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 104]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
38.  Kunzmann V, Ramanathan RK, Goldstein D, Liu H, Ferrara S, Lu B, Renschler MF, Von Hoff DD. Tumor Reduction in Primary and Metastatic Pancreatic Cancer Lesions With nab-Paclitaxel and Gemcitabine: An Exploratory Analysis From a Phase 3 Study. Pancreas. 2017;46:203-208.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
39.  Chiorean EG, Von Hoff DD, Reni M, Arena FP, Infante JR, Bathini VG, Wood TE, Mainwaring PN, Muldoon RT, Clingan PR. CA19-9 decrease at 8 weeks as a predictor of overall survival in a randomized phase III trial (MPACT) of weekly nab-paclitaxel plus gemcitabine versus gemcitabine alone in patients with metastatic pancreatic cancer. Ann Oncol. 2016;27:654-660.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 73]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
40.  Ramanathan RK, Goldstein D, Korn RL, Arena F, Moore M, Siena S, Teixeira L, Tabernero J, Van Laethem JL, Liu H. Positron emission tomography response evaluation from a randomized phase III trial of weekly nab-paclitaxel plus gemcitabine versus gemcitabine alone for patients with metastatic adenocarcinoma of the pancreas. Ann Oncol. 2016;27:648-653.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 33]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
41.  Goldstein D, Von Hoff DD, Moore M, Greeno E, Tortora G, Ramanathan RK, Macarulla T, Liu H, Pilot R, Ferrara S. Development of peripheral neuropathy and its association with survival during treatment with nab-paclitaxel plus gemcitabine for patients with metastatic adenocarcinoma of the pancreas: A subset analysis from a randomised phase III trial (MPACT). Eur J Cancer. 2016;52:85-91.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 31]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
42.  Hidalgo M, Plaza C, Musteanu M, Illei P, Brachmann CB, Heise C, Pierce D, Lopez-Casas PP, Menendez C, Tabernero J. SPARC Expression Did Not Predict Efficacy of nab-Paclitaxel plus Gemcitabine or Gemcitabine Alone for Metastatic Pancreatic Cancer in an Exploratory Analysis of the Phase III MPACT Trial. Clin Cancer Res. 2015;21:4811-4818.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 106]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
43.  Yamabe S, Katayama K, Mochizuki M. The effect of RU486 and progesterone on luteal function during pregnancy. Nihon Naibunpi Gakkai Zasshi. 1989;65:497-511.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Scheithauer W, Ramanathan RK, Moore M, Macarulla T, Goldstein D, Hammel P, Kunzmann V, Liu H, McGovern D, Romano A. Dose modification and efficacy of nab-paclitaxel plus gemcitabine vs. gemcitabine for patients with metastatic pancreatic cancer: phase III MPACT trial. J Gastrointest Oncol. 2016;7:469-478.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 44]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
45.  Chiorean EG, Von Hoff DD, Tabernero J, El-Maraghi R, Ma WW, Reni M, Harris M, Whorf R, Liu H, Li JS. Second-line therapy after nab-paclitaxel plus gemcitabine or after gemcitabine for patients with metastatic pancreatic cancer. Br J Cancer. 2016;115:188-194.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 58]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
46.  Vaupel P, Mayer A. Hypoxia in cancer: significance and impact on clinical outcome. Cancer Metastasis Rev. 2007;26:225-239.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1540]  [Cited by in F6Publishing: 1598]  [Article Influence: 94.0]  [Reference Citation Analysis (0)]
47.  Subarsky P, Hill RP. The hypoxic tumour microenvironment and metastatic progression. Clin Exp Metastasis. 2003;20:237-250.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Sun JD, Liu Q, Ahluwalia D, Li W, Meng F, Wang Y, Bhupathi D, Ruprell AS, Hart CP. Efficacy and safety of the hypoxia-activated prodrug TH-302 in combination with gemcitabine and nab-paclitaxel in human tumor xenograft models of pancreatic cancer. Cancer Biol Ther. 2015;16:438-449.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 45]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
49.  Suenaga M, Yamada S, Fujii T, Tanaka C, Kanda M, Nakayama G, Sugimoto H, Koike M, Fujiwara M, Kodera Y. S-1 plus nab-paclitaxel is a promising regimen for pancreatic cancer in a preclinical model. J Surg Oncol. 2016;113:413-419.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 13]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
50.  Li JA, Xu XF, Han X, Fang Y, Shi CY, Jin DY, Lou WH. Nab-Paclitaxel Plus S-1 Shows Increased Antitumor Activity in Patient-Derived Pancreatic Cancer Xenograft Mouse Models. Pancreas. 2016;45:425-433.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
51.  Miyamoto Y, Maitra A, Ghosh B, Zechner U, Argani P, Iacobuzio-Donahue CA, Sriuranpong V, Iso T, Meszoely IM, Wolfe MS. Notch mediates TGF alpha-induced changes in epithelial differentiation during pancreatic tumorigenesis. Cancer Cell. 2003;3:565-576.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Mato-Berciano A, Raimondi G, Maliandi MV, Alemany R, Montoliu L, Fillat C. A NOTCH-sensitive uPAR-regulated oncolytic adenovirus effectively suppresses pancreatic tumor growth and triggers synergistic anticancer effects with gemcitabine and nab-paclitaxel. Oncotarget. 2017;8:22700-22715.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 14]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
53.  Delitto D, Black BS, Sorenson HL, Knowlton AE, Thomas RM, Sarosi GA, Moldawer LL, Behrns KE, Liu C, George TJ. The inflammatory milieu within the pancreatic cancer microenvironment correlates with clinicopathologic parameters, chemoresistance and survival. BMC Cancer. 2015;15:783.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 35]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
54.  Dauer P, Nomura A, Saluja A, Banerjee S. Microenvironment in determining chemo-resistance in pancreatic cancer: Neighborhood matters. Pancreatology. 2017;17:7-12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 97]  [Article Influence: 13.9]  [Reference Citation Analysis (0)]
55.  Gonzalez-Villasana V, Rodriguez-Aguayo C, Arumugam T, Cruz-Monserrate Z, Fuentes-Mattei E, Deng D, Hwang RF, Wang H, Ivan C, Garza RJ. Bisphosphonates inhibit stellate cell activity and enhance antitumor effects of nanoparticle albumin-bound paclitaxel in pancreatic ductal adenocarcinoma. Mol Cancer Ther. 2014;13:2583-2594.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 18]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
56.  Kurahara H, Shinchi H, Mataki Y, Maemura K, Noma H, Kubo F, Sakoda M, Ueno S, Natsugoe S, Takao S. Significance of M2-polarized tumor-associated macrophage in pancreatic cancer. J Surg Res. 2011;167:e211-e219.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 380]  [Cited by in F6Publishing: 426]  [Article Influence: 28.4]  [Reference Citation Analysis (0)]
57.  Cullis J, Siolas D, Avanzi A, Barui S, Maitra A, Bar-Sagi D. Macropinocytosis of Nab-paclitaxel Drives Macrophage Activation in Pancreatic Cancer. Cancer Immunol Res. 2017;5:182-190.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 103]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
58.  Awasthi N, Scire E, Monahan S, Grojean M, Zhang E, Schwarz MA, Schwarz RE. Augmentation of response to nab-paclitaxel by inhibition of insulin-like growth factor (IGF) signaling in preclinical pancreatic cancer models. Oncotarget. 2016;7:46988-47001.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
59.  Safran H, Charpentier KP, Perez K, Mantripragada K, Miner T, DiPetrillo T, Kuritzky B, Apor E, Bishop K, Luppe D. FOLFOX+Nab-Paclitaxel (FOLFOX-A) for Advanced Pancreatic Cancer: A Brown University Oncology Research Group Phase I Study. Am J Clin Oncol. 2016;39:619-622.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 10]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
60.  Reni M, Balzano G, Zanon S, Passoni P, Nicoletti R, Arcidiacono PG, Pepe G, Doglioni C, Fugazza C, Ceraulo D. Phase 1B trial of Nab-paclitaxel plus gemcitabine, capecitabine, and cisplatin (PAXG regimen) in patients with unresectable or borderline resectable pancreatic adenocarcinoma. Br J Cancer. 2016;115:290-296.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 29]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
61.  Reni M, Zanon S, Balzano G, Passoni P, Costantino A, Pircher C, Chiaravalli M, Nicoletti R, Arcidiacono PG, Pepe G. Randomized phase 2 trial of nab-paclitaxel plus gemcitabine, ± capecitabine, cisplatin (PAXG regimen) in unresectable or borderline resectable pancreatic cancer. Ann Oncol. 2016;27:681P.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
62.  Cohen SJ, O’Neil BH, Berlin J, Ames P, McKinley M, Horan J, Catalano PM, Davies A, Weekes CD, Leichman L. A phase 1b study of erlotinib in combination with gemcitabine and nab-paclitaxel in patients with previously untreated advanced pancreatic cancer: an Academic Oncology GI Cancer Consortium study. Cancer Chemother Pharmacol. 2016;77:693-701.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 8]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
63.  Scheithauer W, Kornek G, Prager G, Stranzl N, Laengle F, Schindl M, Friedl J, Klech J, Roethlin S, Zielinski C. Phase II trial of capecitabine plus nab-paclitaxel in patients with metastatic pancreatic adenocarcinoma. J Gastrointest Oncol. 2016;7:234-238.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 6]  [Reference Citation Analysis (0)]
64.  Bachet JB, Chibaudel B, Bonnetain F, Validire P, Hammel P, André T, Louvet C; GERCOR group. A randomized phase II study of weekly nab-paclitaxel plus gemcitabine or simplified LV5FU2 as first-line therapy in patients with metastatic pancreatic cancer: the AFUGEM GERCOR trial. BMC Cancer. 2015;15:653.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 7]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
65.  Dubreuil O, Bachet JB, Hammel P, Desrame J, Meurisse A, Andre T, Debourdeau P, Dauba J, Lecomte T, Seitz JF. Nab-paclitaxel plus gemcitabine or plus simplified LV5FU2 as first-line therapy in patients with metastatic pancreatic adenocarcinoma: A GERCOR randomized phase II study (AFUGEM). J Clin Oncol. 2017;35:350.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Chiorean EG, Von Hoff DD. Taxanes: impact on pancreatic cancer. Anticancer Drugs. 2014;25:584-592.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 16]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
67.  Conroy T, Desseigne F, Ychou M, Bouché O, Guimbaud R, Bécouarn Y, Adenis A, Raoul JL, Gourgou-Bourgade S, de la Fouchardière C. 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: 5099]  [Article Influence: 392.2]  [Reference Citation Analysis (1)]
68.  Vaccaro V, Sperduti I, Vari S, Bria E, Melisi D, Garufi C, Nuzzo C, Scarpa A, Tortora G, Cognetti F. Metastatic pancreatic cancer: Is there a light at the end of the tunnel? World J Gastroenterol. 2015;21:4788-4801.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 51]  [Cited by in F6Publishing: 53]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
69.  Li D, Capanu M, Yu KH, Lowery MA, Kelsen DP, O’Reilly EM. Treatment, Outcomes, and Clinical Trial Participation in Elderly Patients With Metastatic Pancreas Adenocarcinoma. Clin Colorectal Cancer. 2015;14:269-276.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 21]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
70.  Giordano G, Vaccaro V, Lucchini E, Musettini G, Bertocchi P, Bergamo F, Giommoni E, Santoni M, Russano M, Campidoglio S. Nab-paclitaxel (Nab-P) and gemcitabine (G) as first-line chemotherapy (CT) in advanced pancreatic cancer (APDAC) elderly patients (pts): A “real-life” study. J Clin Oncol. 2015;33:424.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
71.  Giordano G, De Vita F, Melisi D, Vaccaro V, Zaniboni A, Zagonel V, vasile E, Passardi A, Leo L, Ventriglia J. Analysis of activity, efficacy and safety of first line Nab Paclitaxel (Nab-P) and Gemcitabine (G) in advanced pancreatic cancer (APDAC) frail and elderly patients (pts). Eur J Cancer. 2015;51 Suppl 3:S445.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
72.  Ducreux M, Cuhna AS, Caramella C, Hollebecque A, Burtin P, Goéré D, Seufferlein T, Haustermans K, Van Laethem JL, Conroy T. Cancer of the pancreas: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2015;26 Suppl 5:v56-v68.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 905]  [Cited by in F6Publishing: 844]  [Article Influence: 93.8]  [Reference Citation Analysis (0)]
73.  Giordano G, Febbraro A, Milella M, Vaccaro V, Melisi D, Luisa Foltran L, Zagonel V, Zaniboni A, Bertocchi P, Bergamo F. Impact of second-line treatment (2L T) in advanced pancreatic cancer (APDAC) patients (pts) receiving first line Nab-Paclitaxel (nab-P) + Gemcitabine (G): an Italian multicentre real life experience. J Clin Oncol. 2016;34:4124.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Pelzer U, Schwaner I, Stieler J, Adler M, Seraphin J, Dörken 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;47:1676-1681.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 239]  [Cited by in F6Publishing: 237]  [Article Influence: 18.2]  [Reference Citation Analysis (0)]
75.  Oettle H, Riess H, Stieler JM, Heil G, Schwaner I, Seraphin J, Görner M, Mölle M, Greten TF, Lakner V. 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)]
76.  Wang-Gillam A, Li CP, Bodoky G, Dean A, Shan YS, Jameson G, Macarulla T, Lee KH, Cunningham D, Blanc JF. 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)]
77.  McBride A, Bonafede M, Cai Q, Princic N, Tran O, Pelletier C, Patel M. Health care costs and treatment patterns among metastatic pancreatic cancer (MPC) patients (pts) initiating first-line (1L) on nab-paclitaxel/gemcitabine (nab-P+G) or FOLFIRINOX (FFX). J Clin Oncol. 2017;35:415.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Corrie P, Qian W, Basu B, Ian Jodrell D, Falk S, Iwuji C, Wasan H, Palmer DH, Scott-Brown M, Wadsley J. Babaoglan B, Dalchau KM, Gopinathan A, Chhabra A, Machin A, Neesse A, Tuveson DA, Valle JW, SIEGE Trial Investigators. Health care costs and treatment patterns among metastatic pancreatic cancer (MPC) patients (pts) initiating first-line (1L) on nab-paclitaxel/gemcitabine (nab-P+G) or FOLFIRINOX (FFX). J Clin Oncol. 2017;35:415.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Lacy J, Portales F, Hammel P, Pazo Cid RA, Manzano Mozo JL, Jae-hoon Kim E, Dowden SD, Borg C, Sastre J, Bathini VG. Interim results of a multicenter phase II trial of nab-paclitaxel (nab-P) plus gemcitabine (G) for patients (Pts) with locally advanced pancreatic cancer (LAPC). J Clin Oncol. 2017;35:358.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Caparello C, Meijer LL, Garajova I, Falcone A, Le Large TY, Funel N, Kazemier G, Peters GJ, Vasile E, Giovannetti E. FOLFIRINOX and translational studies: Towards personalized therapy in pancreatic cancer. World J Gastroenterol. 2016;22:6987-7005.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 54]  [Cited by in F6Publishing: 50]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]