Skip to main content
Erschienen in: Journal of Hematology & Oncology 1/2015

Open Access 01.12.2015 | Review

Novel approaches in the management of pancreatic ductal adenocarcinoma: potential promises for the future

verfasst von: Gaurav Goel, Weijing Sun

Erschienen in: Journal of Hematology & Oncology | Ausgabe 1/2015

Abstract

Despite a few breakthroughs in therapy for advanced disease in the recent years, pancreatic ductal adenocarcinoma continues to remain one of the most challenging human malignancies to treat. The overall prognosis for the majority of patients with pancreatic cancer is rather dismal, and therefore, more effective treatment options are being desperately sought. The practical goals of management are to improve the cure rates for patients with resectable disease, achieve a higher conversion rate of locally advanced tumor into potentially resectable disease, and finally, prolong the overall survival for those who develop metastatic disease. Our understanding of the complex genetic alterations, the implicated molecular pathways, and the role of desmoplastic stroma in pancreatic cancer tumorigenesis has increased several folds in the recent years. This has facilitated the development of novel therapeutic strategies against pancreatic cancer, some of which are currently under evaluation in ongoing preclinical and clinical studies. This review will summarize the existing treatment approaches for this devastating disease and also discuss the promising therapeutic approaches that are currently in different stages of clinical development.
Hinweise

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

GG drafted the manuscript and revised it critically for important intellectual content. WS revised the manuscript critically for important intellectual content. Both authors read and approved the final manuscript.

Introduction

Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer-related mortality in the US for both men and women [1]. In 2015 alone, it is estimated that 48,960 new cases of pancreatic cancer will be diagnosed in the US and 40,560 patients will die of this disease. As such, PDAC remains one of the most challenging malignancies with a dismal prognosis and limited therapeutic options. The 5-year survival rate for pancreatic cancer (all stages combined) is around 7%, which is the lowest among all different cancer sites. At the time of initial pancreatic cancer diagnosis, approximately 9% of patients present with localized disease, 28% have regional spread, and the remaining 53% of patients already have distant spread of their disease. There has been a very limited clinically meaningful improvement in survival rates for this disease during the past two decades. The poor prognosis of PDAC is largely attributed to delayed diagnosis due to nonspecific symptoms in the early stages of the disease, biological aggressiveness leading to rapid metastases, lack of effective screening methods, and resistance to radiation and chemotherapies.
It is now well established that PDAC is driven by alteration of multiple genes that regulate pathways and processes in the tumor cells and the neighboring microenvironment [2]. Despite our improved understanding of the molecular events underlying the multistep carcinogenesis of PDAC, the progress made towards improving the survival rates of these patients has been extremely slow. At present, multiple novel treatment strategies targeted against PDAC are under preclinical and clinical evaluation. This review will summarize the existing treatment approaches for this devastating disease and also discuss the promising therapeutic strategies that are currently in the different stages of clinical development.

Risk factors

The known risk factors that increase the likelihood of developing PDAC include cigarette smoking [3], alcohol abuse [4], high fat diet [5], and certain trace elements [6]. It is estimated that cigarette smoking doubles the risk of developing PDAC and accounts for approximately 20%–25% of the cases [3]. Chronic pancreatitis is also associated with an increased risk of PDAC, especially among smokers [7]. It has also been noted that the majority of patients with PDAC develop diabetes mellitus which is usually diagnosed in the preceding 1–2 years or concomitant with the new cancer diagnosis [7]. It is not entirely clear whether diabetes is a predisposing factor or a manifestation of PDAC itself. Obesity, which predisposes to insulin resistance, might be a common link between the two.
Approximately 5%–10% of patients with PDAC report a history of pancreatic cancer diagnosis in their family member [8]. The genetic syndromes such as familial breast cancer (BRCA2, BRCA1, and PALB2), the Peutz-Jeghers syndrome (LKB1/STK11), the familial atypical multiple mole melanoma (FAMMM) syndrome (p16/CDKN2A), hereditary pancreatitis (PRSS1), and the lynch syndrome (MLH1, MSH2, MSH6, PMS2) are also associated with an increased risk of developing PDAC [8,9]. Thus, patients with a family history of pancreatic cancer or these mutation carriers should undergo appropriate screening, as per the guidelines provided by the International Cancer of the Pancreas Screening Consortium [10].

Genetics and molecular pathogenesis

Pancreatic cancer most commonly originates in the exocrine cells of the pancreas [11]. Among the exocrine tumors, ‘ductal adenocarcinoma’ is the most frequently encountered pathological subtype and accounts for more than 90% of the cases. Initiation and development of PDAC involve a series of specific genetic alterations which promote growth and survival of aberrant precursors, initiation of a desmoplastic reaction in the stroma, and ultimately tissue invasion and metastases [12]. This oncogenic process begins with transformation of normal pancreatic duct epithelium into infiltrating cancer through a series of histologically defined precursors called pancreatic intraepithelial neoplasia (PanIN)-1, -2, and -3 [11]. These morphological changes occur in conjunction with several genetic alterations. Disease progression often involves development of distant metastases, which occurs late during the genetic evolution of pancreatic cancer [13]. Using genome sequencing, it has been determined that after the initiation of tumorigenesis, an average of 11.7 years is required for the birth of parental, non-metastatic founder pancreatic cancer clone, additional 6.8 years for the development of cancer cell subclones with metastatic potential, and an average of 2.7 years from then until the patient’s death [13].
It is now well established that pancreatic cancer cells contain one or more of the four primary genetic mutations that drive pancreatic cancer tumorigenesis [14]. These include KRAS, p16/CDKN2A, TP53, and SMAD4 mutations. KRAS plays a critical role in regulating important cellular functions including cell survival, cell differentiation, and proliferation [15]. Single point mutations in codon 12, 13, 59, or 61 of exon 2 and exon 3 of the KRAS oncogene lead to uncontrolled downstream signaling of RAF/MEK/ERK, leading to enhanced tumor cell proliferation and survival. It has been shown that these activating mutations in the KRAS are a necessary event for the initiation of pancreatic cancer and are therefore commonly found in the early precursor lesions (PanIN-1) [16,17]. With disease progression, the prevalence of oncogenic KRAS mutation increases and is present in over 90% of the tumors [17,18]. Inactivating mutation in the CDKN2A tumor suppressor gene results in the loss of p16 protein and thereby loss of regulation of the G1/S transition of the cell cycle. It is also thought to be a relatively early event in PDAC progression (PanIN-2 lesions) and is associated with larger tumors and early metastasis [17,19]. TP53 is a DNA checkpoint regulator in response to mutations from reactive oxygen species as well as telomere shortening. Abnormal TP53 gene allows cells to avoid DNA damage control checkpoints and subsequently apoptotic signals [20]. SMAD4 is a key component of the transforming growth factor-β (TGF-β) receptor signaling pathway and plays a role in activating transcription of cell cycle inhibitory factors. Inactivation of TP53 and SMAD4 occur at a later stage (PanIN-3) in pancreatic carcinogenesis [17].
A comprehensive genome analysis of 24 different human pancreatic cancers revealed an average of 63 genetic alterations per cancer, the majority of which were point mutations [2]. These mutations occur in several primary oncogenes and tumor suppressor genes and contribute to the genetic diversity of pancreatic cancer. This, in turn, leads to tumor heterogeneity, instability, and early metastasis. The genetic alterations associated with pancreatic cancer can be classified into a set of 12 core cellular signaling pathways: apoptosis, control of G1/S phase transition, sonic hedgehog (SHH) signaling, KRAS signaling, TGF-β signaling, Wnt/Notch signaling, DNA damage control, homophilic cell adhesion, integrin signaling, JNK signaling, invasion, and small GTPase signaling [2]. These pathways are responsible for some of the key cellular functions such as intracellular signaling, cell cycle regulation, metabolism, and DNA repair. Targeting these pathways has now become the main focus of drug development in pancreatic cancer.
A prominent histologic hallmark of PDAC is the presence of a desmoplastic reaction which consists of extracellular proliferation of leukocytes, fibroblasts, endothelial cells, neuronal cells, and collagen. This is mediated by paracrine signals from the pancreatic cancer cells which results in the formation of a dense stroma in the tumor microenvironment [21]. It is now established that the signals that promote this stromal reaction originate from the KRAS-mutant oncogene in the epithelium of pancreatic cancer cells. SHH signaling also acts in a paracrine fashion on the extracellular fibroblasts, resulting in their growth and differentiation [22]. The desmoplastic reaction not only acts as a mechanical barrier to the effective delivery of chemotherapeutic agents, it also provides an antiangiogenic and hypoxic microenvironment in which the pancreatic cancer cells like to grow and flourish.
Thus, it is now established beyond doubt that the wide range of genetic alterations and the stromal reaction play an important role in the initiation, progression, chemotherapeutic resistance, and recurrence of pancreatic cancer.

Clinical management

Localized disease

The recommended treatment for patients presenting with localized disease is surgery, since complete surgical resection with negative margins offers the only hope for cure in pancreatic cancer treatment [23]. Unfortunately, only 9% of the pancreatic cancer patients present with localized disease that is completely resectable at the time of initial diagnosis [1]. Depending upon the size and the location of the tumor, the operative procedure is either a cephalic pancreaticoduodenectomy (Whipple procedure), distal pancreatectomy, or total pancreatectomy [24]. The success of surgical resection depends on factors such as extent of lymph node involvement, tumor grade, tumor size, CA 19-9 levels, and the positivity of resection margins. Even following complete surgical resection, the 5-year survival rates are low at approximately 20%, and the overall prognosis remains discouraging [23]. Thus, postoperative treatment in the form of adjuvant chemotherapy [25] or chemoradiotherapy [26,27] is usually administered and is often gemcitabine- or 5-fluorouracil (5-FU)-based. The Charité Onkologie Clinical Studies in Gastrointestinal Cancers-001 (CONKO-001) trial randomized 354 patients with localized PDAC to receive either adjuvant gemcitabine or undergo observation after curative resection [28]. Gemcitabine arm was associated with a significant improvement in disease-free survival (DFS; 13.4 months vs. 6.9 months; P < 0.001). The median overall survival (OS) was however similar in the two arms (22.1 months vs. 20.2 months; P = 0.06) and was explained by the administration of gemcitabine to patients in the observation arm after disease progression. The European Study Group for Pancreatic Cancer (ESPAC)-3 trial compared gemcitabine vs. 5-FU in the adjuvant treatment of PDAC [25]. In this trial, 1,088 patients were randomized to receive either 5-FU/Leucovorin (LV) or gemcitabine. There was no difference in the median OS (23 months vs. 23.6 months; P = 0.39), progression-free survival (PFS; 14.1 months vs. 14.3 months; P = 0.53), and the global quality of life (QoL) scores between the treatment groups. In the Radiation Therapy Oncology Group (RTOG)-9704 trial, 451 patients with resected PDAC received either gemcitabine or 5-FU chemotherapy before and after 5-FU-based chemoradiation [29]. Among patients with pancreatic head tumors, a statistically non-significant improvement in median OS was seen in the gemcitabine containing arm (20.5 months vs. 16.9 months; P = 0.09). A phase III adjuvant trial (UNICANCER) comparing gemcitabine vs. modified FOLFIRINOX in surgically resected (R0 or R1) PDAC patients is currently ongoing (NCT01526135). Another phase III RTOG-0848 trial is comparing adjuvant gemcitabine with or without erlotinib in the first randomization and additional benefit of chemoradiation in the second randomization for patients with localized PDAC who have undergone R0 or R1 surgical resection (NCT01013649).
Another potential strategy for the treatment of localized pancreatic cancer is to administer chemotherapy in the neoadjuvant setting, and this approach has also been explored in multiple phase II clinical trials [30-34]. The rationale for this approach includes early treatment of micrometastatic disease, delivery of chemotherapy to an undisturbed tumor, and biological assessment of tumor aggressiveness [35]. An ongoing phase III study is comparing neoadjuvant gemcitabine/oxaliplatin combination vs. upfront surgery, to be followed by adjuvant gemcitabine in patients with resectable PDAC (NCT01314027).

Locally advanced pancreatic cancer

Locally advanced pancreatic cancer (LAPC) is the tumor that is confined locoregionally with some degree of involvement of the nearby major vascular structures but without any evidence of distant metastases. Approximately 30% of the patients are found to have locally advanced stage at the time of initial pancreatic cancer diagnosis [1]. Patients are usually classified as having either borderline-resectable or unresectable LAPC, depending upon the relationship of the tumor with the nearby vascular structures. Unfortunately, there are no standard guidelines for the management of LAPC. This is mainly due to the absence of a single standardized definition of LPAC, which limits a fair comparison of treatment strategies and results across the clinical trials.
The only potential way to achieve a cure in patients with LAPC is by maximizing upfront systemic and local therapy followed by a R0 surgical resection. For borderline-resectable LAPC patients that are deemed ineligible for upfront surgery, neoadjuvant treatment in the form of chemotherapy with or without radiation is usually performed [36-39]. Such neoadjuvant therapies have the potential to downstage borderline-resectable disease and make R0 resection feasible. For initially unresectable LAPC, neoadjuvant therapy should be offered as a bridge to potentially curative resection. Neoadjuvant therapy options include concurrent chemoradiation, chemotherapy alone, and chemotherapy followed by chemoradiation. A 1981 trial conducted by the Gastrointestinal Tumor Study Group (GITSG) established the superiority of combined 5-FU/radiation when compared with radiation therapy alone in unresectable LAPC [40]. Subsequently, gemcitabine-based chemoradiation was evaluated and established as an acceptable treatment option for LAPC, based on the results of the Eastern Cooperative Oncology Group (ECOG)-4201 trial [41] and the Taipei trial [42]. Chemotherapy alone is another management strategy in the neoadjuvant treatment of LAPC. Combination chemotherapy regimens consisting of gemcitabine backbone have failed to demonstrate survival advantage over single agent gemcitabine [43-45]. Non-gemcitabine-based regimens such as FOLFIRINOX and FOLFOX have also been explored in the neoadjuvant treatment of LAPC [46-50]. Yet another approach in the management of LAPC is chemotherapy followed by chemoradiotherapy prior to surgery. Such strategy has been looked at in retrospective series with encouraging results, but prospective phase II/III studies are needed before it can be incorporated into standard oncologic practice [50-52]. The four-arm phase III CONKO-7 trial will evaluate the efficacy of neoadjuvant chemotherapy plus chemoradiation vs. chemotherapy alone in an estimated 830 unresectable LAPC patients (NCT01827553). The chemotherapy options in this trial include either FOLFIRINOX or gemcitabine. A similar phase III trial in LAPC patients to evaluate neoadjuvant modified FOLFIRINOX followed by chemoradiation (with 5-FU as the radiosensitizer) is planned by the investigators from the Stanford University (NCT01926197).

Metastatic disease

Approximately 50%–60% of PDAC patients present with metastatic disease at the time of initial diagnosis [1], and the standard treatment option for these patients is chemotherapy. In 1997, gemcitabine monotherapy was established as the standard of care for metastatic disease. In this pivotal trial, single agent gemcitabine (1,000 mg/m2 weekly × 7 followed by 1 week of rest, then weekly × 3 every 4 weeks thereafter; n = 63) was compared with weekly bolus 5-FU (600 mg/m2; n = 63), and a modest survival benefit was demonstrated in the gemcitabine group (5.6 vs. 4.4 months; P = 0.0025) [53]. The primary efficacy measure was clinical benefit response (CBR), which was a composite of measurements of pain, Karnofsky Performance Status (KPS), and weight. The CBR was experienced by 23.8% of the gemcitabine-treated patients compared with 4.8% of 5-FU-treated patients (P = 0.0022). Despite the marginal improvement in 1-year survival, gemcitabine replaced 5-FU and was approved by the US Food and Drug Administration (FDA) as a standard treatment option for the treatment of metastatic pancreatic cancer largely based upon its efficacy in improving the disease-related symptoms [53]. Thereafter, gemcitabine continued as the chemotherapeutic standard of care in the management of patients with metastatic pancreatic cancer for a long time.
Erlotinib, an epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI), is the only biologic agent that has been approved for the treatment of advanced pancreatic cancer (APC). In the phase III PA.3 trial that randomized 569 patients with locally advanced and metastatic PDAC, the addition of erlotinib to gemcitabine resulted in a statistically significant improvement in median OS of approximately 10 days (6.24 months vs. 5.91 months; hazard ratio [HR] 0.82; P = 0.038) [54]. Despite the clinically insignificant benefit, the combination of gemcitabine plus erlotinib received the FDA approval in November 2005 due to a lack of more effective treatment options for this devastating disease at the time. However, due to the associated side effects and an exceedingly small clinical benefit, the use of this regimen in routine oncologic practice has remained virtually non-existent.
In recent years, the evidence has shifted from using single agent gemcitabine to combination regimens for front-line treatment of metastatic pancreatic cancer. FOLFIRINOX and gemcitabine plus nab-paclitaxel have now been established as the two standard combination chemotherapy options for metastatic disease. In the randomized phase III PRODIGE 4/ACCORD 11 trial that consisted of 342 metastatic PDAC patients, FOLFIRINOX (oxaliplatin 85 mg/m2, irinotecan 180 mg/m2, LV 400 mg/m2, and 5-FU 400 mg/m2 given as a bolus followed by 2,400 mg/m2 given as a 46-hour continuous infusion every 2 weeks) when compared with gemcitabine alone (1,000 mg/m2 weekly for 7 of 8 weeks and then weekly for 3 of 4 weeks) demonstrated a significantly better median OS (11.1 vs. 6.8 months; HR 0.57; 95% confidence interval [CI] 0.45 to 0.73; P < 0.001), PFS (6.4 months vs. 3.3 months; HR 0.47; 95% CI 0.37 to 0.59; P < 0.001), and objective response rate (ORR; 31.6% vs. 9.4%; P < 0.001) [55]. Based on the statistically significant and clinically meaningful improvement in the survival, FOLFIRINOX was approved for the first-line treatment of metastatic PDAC. This improvement in OS with FOLFIRINOX is, however, associated with significant toxicity (febrile neutropenia, thrombocytopenia, peripheral neuropathy, vomiting, and diarrhea), and therefore, this regimen is only indicated for patients with good performance status. To improve the tolerability of FOLFIRINOX, a modified regimen was recently proposed in which the 5-FU bolus is removed and growth factor prophylaxis is administered routinely [56].
An important contributor to the chemoresistance of PDAC is the presence of a dense stroma in the tumor microenvironment. Nab-paclitaxel is albumin-bound paclitaxel that has been developed to diminish the stromal tissue network. The ‘albumin’ in nab-paclitaxel interacts with secreted protein acidic and rich in cysteine (SPARC), a matrix glycoprotein that has a role in tumor invasion and facilitates the uptake of paclitaxel by the tumor cells [57]. In a phase I/II trial of 67 patients with metastatic PDAC, nab-paclitaxel plus gemcitabine combination was associated with an ORR of 48%, median OS of 12.2 months, and 1-year survival of 48% [58]. Subsequently, in a large phase III MPACT trial, 861 metastatic PDAC patients were randomized to receive nab-paclitaxel (125 mg/m2) followed by gemcitabine (1,000 mg/m2) on days 1, 8, and 15 every 4 weeks, vs. gemcitabine monotherapy (1,000 mg/m2) weekly for 7 of 8 weeks (cycle 1) and then on days 1, 8, and 15 every 4 weeks (cycle 2 onwards) [59]. The combination of nab-paclitaxel plus gemcitabine significantly improved the median OS from 6.7 months to 8.5 months (HR 0.72; 95% CI 0.62 to 0.83; P < 0.001), PFS from 3.7 months to 5.5 months (HR 0.69; 95% CI 0.58 to 0.82; P < 0.001) and RR from 7% to 23% (P < 0.001) when compared with gemcitabine alone. In September 2013, the FDA approved nab-paclitaxel plus gemcitabine as a first-line treatment option for metastatic pancreatic cancer based on the results of this study. Currently, the choice between the two approved standard first-line chemotherapy options for metastatic disease (FOLFIRINOX or gemcitabine/nab-paclitaxel) is usually guided by the patient’s age, performance status, preference of treatment frequency, and toxicity profiles of the two regimens.

Emerging novel therapeutic targets and treatment strategies

Currently, a multitude of innovative therapeutic approaches are being developed to target the known molecular pathways involved in pancreatic tumorigenesis. Table 1 provides a list of the selected clinical trials that are currently recruiting patients to evaluate the safety and efficacy of novel agents in PDAC.
Table 1
Summary of selected novel agents that are under evaluation in currently actively recruiting clinical trials
Primary modes of action
Study agent(s)
NCT identifier
Phase
Disease stage
MEK inhibitor + Bcl-2 inhibitor
Trametinib, navitoclax
NCT02079740
Ib/II
LA, metastatic
MEK inhibitor + ErbB inhibitor
PD-0325901, dacomitinib
NCT02039336
I/II
Metastatic
 
Trametinib, lapatinib
NCT02230553
I/II
Metastatic
EGFR TKI
Afatinib
NCT01728818
II
Metastatic
PI3K inhibitor
BKM120
NCT01571024
I
Metastatic
 
BYL719
NCT02155088
I
LA, metastatic
PI3K inhibitor + MEK inhibitor
BYL719, MEK162
NCT01449058
Ib/II
Metastatic
AKT inhibitor
MK2206
NCT01783171
I
LA, metastatic
PTEN inducer
AXP107-11
NCT01182246
Ib/II
LA, metastatic
Wnt signaling inhibitor
OMP-54 F28
NCT02050178
Ib
Metastatic
 
OMP-18R5
NCT02005315
Ib
Metastatic
 
PRI-724
NCT01764477
I
LA, metastatic
 
LGK974
NCT01351103
I
LA, metastatic
Glycogen synthase kinase-3 inhibitor
LY2090314
NCT01632306
I, II
Metastatic
Notch signaling inhibitor
MK0752
NCT01098344
I
LA, metastatic
 
PF-03084014
NCT02109445
Ib/II
Metastatic
 
OMP-59R5
NCT01647828
Ib/II
Metastatic
 
OMP-21 M18
NCT01189929
Ib
LA, metastatic
Anti-connective tissue growth factor mAb
FG-3019
NCT02210559
II
LA unresectable
Heparan sulfate mimetic
M402
NCT01621243
I/II
Metastatic
Hyaluronidase
PEGPH20
NCT01839487
II
Metastatic
  
NCT01959139
I/II
Metastatic
Hyaluronidase + anti-EGFR mAb
PEGPH20, cetuximab
NCT02241187
NP
Resectable
Oncolytic adenovirus encoding hyaluronidase
VCN-01
NCT02045589
I
LA, metastatic
Hedgehog inhibitor
IPI-926
NCT01383538
I
LA, metastatic
 
GDC-0449
NCT01088815
II
Metastatic
 
LDE-225
NCT01431794
I/II
LA
Hypoxia targeting agent
TH-302
NCT02047500
I
LA, metastatic
TGF-β receptor I inhibitor
LY2157299
NCT01373164
Ib/II
LA, metastatic
Hypomethylating agent
Azacitidine
NCT01845805
II
Resected
AMP-activated protein kinase (AMPK) activator
Metformin
NCT01954732
I
Localized
 
Metformin
NCT02005419
II
Localized
 
Metformin
NCT01666730
II
Metastatic
AMPK activator + mTOR inhibitor
Metformin, Rapamycin
NCT02048384
Ib/II
Metastatic
poly (ADP-ribose) polymerase (PARP) inhibitor
Veliparib
NCT01908478
I
LA
 
Veliparib
NCT01489865
I/II
Metastatic
 
Veliparib
NCT01585805
II
LA, metastatic
 
Rucaparib (AG-14699)
NCT02042378
II
LA, metastatic (BRCA mutant)
 
Olaparib (AZD2281)
NCT02184195
III
Metastatic (BRCA mutant)
Vascular targeting agent
ADH-1
NCT01825603
I
LA, metastatic
Antiangiogenic combination
Tl-118
NCT01509911
II
Metastatic
Arginine degrading enzyme
ADI-PEG 20
NCT02101580
Ib
LA, metastatic
Aurora A kinase inhibitor
Alisertib (MLN8237)
NCT01924260, NCT01677559
I
LA, metastatic
CDK inhibitor + AKT inhibitor
Dinaciclib, MK2206
NCT01783171
I
LA, metastatic
α-ketoglutarate dehydrogenase (KGDH) inhibitor
CPI-613
NCT01835041
I
Metastatic
 
CPI-613
NCT01839981
I
LA, metastatic
c-Met inhibitor
Cabozantinib (XL184)
NCT01663272
I
LA, metastatic
CRM-1 inhibitor
Selinexor (KPT-330)
NCT02178436
Ib/II
Metastatic
DNA minor groove binder
Lurbinectedin
NCT02210364
I
LA unresectable
Src inhibitor
Dasatinib
NCT01652976
II
Metastatic
Trk A, B, C inhibitors
PLX7486
NCT01804530
I
LA, metastatic
IDO inhibitor
Indoximod
NCT02077881
I/II
Metastatic
 
NLG919
NCT02048709
I
Refractory
Chemokine receptor 2 (CCR2) antagonist
PF-04136309
NCT01413022
I
LA
Anti-tissue factor mAb
MORAb-066
NCT01761240
I
LA, metastatic
Wee 1 inhibitor
MK1775
NCT02037230
I/II
LA unresectable
BET bromodomain inhibitor
OTX015
NCT02259114
Ib
LA, metastatic
SMAC mimetic
LCL161
NCT01934634
I
Metastatic
Cancer stemness inhibitor
BBI608
NCT02231723
Ib
Metastatic
Janus Kinase (JAK) inhibitor
Ruxolitinib
NCT01822756
I
Metastatic
 
Ruxolitinib
NCT02117479, NCT02119663
III
Metastatic
 
INCB039110
NCT01858883
Ib
Metastatic
 
Momelotinib
NCT02101021
II
Metastatic
Autophagy inhibitor
Hydroxychloroquine
NCT01506973
I/II
Metastatic
 
Hydroxychloroquine
NCT01978184
II
Resectable
 
Hydroxychloroquine
NCT01494155
II
Resectable
Cancer vaccine
GVAX
NCT01088789
II
Localized
 
Poly ICLC and dendritic cells
NCT01677962
0
LA unresectable
 
Autologous tumor-derived HSP gp96
NCT02133079
I/II
Resected
 
GVAX/CRS-207
NCT02004262
II
Metastatic
 
Algenpantucel-L
NCT01836432
III
LA
hTERT DNA cancer vaccine
INO-1400
NCT02327468
I
Non-metastatic
CTLA-4 inhibitor
Ipilimumab
NCT01473940
Ib
LA, metastatic
Anti-PD-1 mAb
CT-011
NCT01313416
II
Resected
Vaccine + CTLA-4 inhibitor
GVAX, ipilimumab
NCT01896869
II
Metastatic
Vaccine + anti-PD-1 mAb
GVAX/CRS-207, nivolumab
NCT02243371
II
Metastatic
CTLA-4 inhibitor + anti-PD-1 mAb
Ipilimumab, nivolumab
NCT01928394
I/II
LA, metastatic
Anti-CPAA mAb
NPC-1C
NCT01834235
I/II
LA, metastatic
 
NPC-1C
NCT01040000
II
LA, metastatic
Anti-MUC1 mAb
BTH1704
NCT02132403
I
LA, metastatic
Anti-CEA BiTE mAb
MEDI-565
NCT01284231
I
Refractory
Anti-CA-125 mAb
Oregovomab
NCT01959672
II
Non-metastatic
Pegylated recombinant human IL-10
AM0010
NCT02009449
I
Metastatic
IL-1 receptor antagonist
Anakinra
NCT02021422
I
Metastatic
RAS specific immunotherapy
TG01
NCT02261714
I/II
Resected
Radioimmunotherapy
90Y-clivatuzumab tetraxetan (IMMU-107)
NCT01956812
III
Metastatic
Activated T-cells
EGFRBi armed ATC infusions
NCT01420874
I
Metastatic
Dendritic cell/cytokine-induced killer cells
DC-CIK
NCT01781520
I/II
LA, metastatic
siRNA-transfected PBMC
APN401
NCT02166255
I
LA, metastatic
Autologous CAR T-cells
RNA mesothelin re-directed CAR T-cells
NCT01897415
I
Metastatic
 
Anti-mesothelin gene engineered lymphocytes
NCT01583686
I/II
Metastatic
Autologous natural killer T-cells
NKT cells
NCT01801852
I
Refractory
Activated dendritic cells
DCVax-Direct
NCT01882946
I/II
LA, metastatic
Autologous tumor infiltrating lymphocytes + interleukin
TIL, IL-2
NCT01174121
II
Metastatic
Antiguanylyl cyclase C antibody-drug conjugate (ADC)
MLN0264
NCT02202785
II
LA, metastatic
Micellar nanoparticle-encapsulated cisplatin
NC-6004
NCT02043288
III
LA or metastatic
Alkylating agent
Glufosfamide
NCT01954992
III
Metastatic
Source: http://​www.​clinicaltrials.​gov; Accessed on January 01, 2015. NCT National Clinical Trial, EGFR epidermal growth factor receptor, TKI tyrosine kinase inhibitor, PTEN Phosphatase and tensin homolog, TGF transforming growth factor receptor, mAb monoclonal antibody, CRM1 chromosome region maintenance 1, DNA deoxyribonucleic acid, BET bromodomain and extra-terminal, SMAC second mitochondrial-derived activator of caspases, hTERT telomerase reverse transcriptase, CTLA-4 cytotoxic T-lymphocyte-associated protein 4, PBMC peripheral blood mononuclear cell, siRNA small interfering RNA, LA locally advanced.

RAS/RAF/MEK/ERK signaling pathway

Despite being the most common mutation associated with PDAC, attempts to target KRAS by inhibiting its post-translational modification have been unsuccessful so far. Tipifarnib (R115777) is an inhibitor of farnesyltransferase (FTase) which is a dominant enzyme involved in post-translational modification of RAS [60]. So far, it has not demonstrated any significant antitumor activity both as a single agent and in combination with gemcitabine [44,61,62].
Attempts are being made to identify downstream targets (mitogen-activated protein kinase [MAPK], phosphatidylinositide 3-kinase [PI3K]) to block KRAS-dependent signaling pathways. Towards this goal, a number of MEK inhibitors are currently being evaluated in clinical trials [63]. Selumetenib (AZD6244) is a selective MEK inhibitor that was found to have similar efficacy as capecitabine in a phase II clinical trial that enrolled APC patients after failing first-line gemcitabine therapy [64]. It is also being tested in combination with erlotinib in APC patients resistant to gemcitabine (NCT01222689). Based on the encouraging results of a phase I clinical trial, trametinib (another MEK inhibitor) was combined with gemcitabine in a phase II clinical trial of metastatic pancreatic cancer patients but failed to demonstrate a clinical benefit [65]. Combinations of other novel MEK inhibitors (pimasertib [MSC1936369B], refametenib [BAY86-9766]) with gemcitabine are currently under evaluation in clinical trials.
It is now known that expression of RAS oncogene up-regulates basal autophagy, which is required for cancer cell survival in starvation and in tumorigenesis [66]. Autophagy is therefore believed to be a significant mechanism for pancreatic cancer cell survival. Hydroxychloroquine, an anti-malarial drug is being evaluated as an autophagy inhibitor for the treatment of these aggressive cancers.

Epidermal growth factor receptor pathway

ErbB-1 (EGFR) and ErbB-2 (HER2/neu) expression is found in 90% and 21% of pancreatic cancers, respectively [67,68]. Therapies targeted against EGFR (both TKIs and monoclonal antibody [mAb]) in pancreatic cancer have yielded overall disappointing results so far. As discussed previously, the phase III PA.3 trial evaluated gemcitabine in combination with erlotinib in the first-line treatment of APC and was associated with a clinically insignificant improvement in median OS when compared with gemcitabine alone [54]. In another phase II study of gemcitabine-refractory APC patients, a combination of erlotinib and capecitabine was associated with only 10% radiological response and a median OS of 6.5 months [69]. The combination of cetuximab and gemcitabine has also been evaluated in the treatment of APC patients [70,71]. The initial phase II study demonstrated stable disease (SD) in 63% and partial response (PR) in 12% of the EGFR-expressing APC patients that were treated with cetuximab plus gemcitabine combination [70]. In a subsequent phase III study (Southwest Oncology Group [SWOG]-directed intergroup trial S0205), this combination was not associated with any survival benefit when compared with the single agent gemcitabine (6.3 months vs. 5.9 months; HR 1.06; 95% CI 0.91 to 1.23; P = 0.23) [71]. A randomized phase II study of panitumumab, erlotinib, and gemcitabine combination suggested a trend towards OS benefit when compared with erlotinib plus gemcitabine [72]. However, this three-drug combination with dual inhibition of the EGFR pathway was associated with significant toxicities leading to early termination of the study. Anti-HER2 agent trastuzumab has been combined with gemcitabine in a phase II study that included metastatic pancreatic cancer patients with 2+ (88% patients) or 3+ (12% patients) HER2/neu overexpression by immunohistochemistry [73]. The response rate of this combination was very similar to gemcitabine alone.
One of the probable explanations for the lack of a meaningful benefit from anti-EGFR TKIs in pancreatic cancer could be the development of acquired resistance to these agents, which is a mechanism well studied in lung cancer [74]. Clinical trials evaluating newer EGFR TKIs such as afatinib (NCT01728818) and dacomitinib (NCT02039336) in pancreatic cancer are currently underway.

Anti-angiogenesis

Targeting vascular endothelial growth factor (VEGF) pathway has shown promising results in the treatment of many solid cancers. However, anti-VEGF therapies have been ineffective clinically in treating patients with PDAC. The phase III Cancer and Leukemia Group B (CALGB) 80303 trial randomized 602 patients with APC to receive gemcitabine with or without bevacizumab in the first-line setting [75]. The addition of bevacizumab to gemcitabine was associated with increased toxicity and without any improvement in survival (5.8 months vs. 5.9 months; P = 0.95). In another large, randomized phase III trial (AVITA), 607 metastatic pancreatic cancer patients were randomized to receive gemcitabine plus erlotinib with either bevacizumab or placebo [76]. The bevacizumab arm was associated with statistically significant PFS advantage (4.6 months vs. 3.6 months; HR 0.73; P = 0.0002) but a non-significant improvement in median OS (7.1 months vs. 6 months; HR 0.89; P = 0.2087). Axitinib is a selective oral inhibitor of VEGF receptor-1, -2, and -3 that has been combined with gemcitabine in a phase II clinical trial of APC patients and showed a statistically non-significant gain in OS [77]. A subsequent phase III study that randomized 632 APC patients to receive gemcitabine plus axitinib or placebo was terminated early due to the lack of survival benefit (8.5 months vs. 8.3 months; HR 1.014; P = 0.5436) at the time of planned interim analysis [78]. Ziv-Aflibercept is an anti-VEGF recombinant fusion protein that has also been combined with gemcitabine in a phase III trial for the treatment of metastatic pancreatic cancer patients [79]. However, this trial was terminated early as well due to the lack of efficacy at the time of planned interim analysis. Sorafenib and masitinib are oral multikinase inhibitors with antiangiogenic properties. In the phase III BAYPAN trial, addition of sorafenib to gemcitabine did not improve PFS in APC patients [80]. The phase III study of gemcitabine plus masitinib also did not result in improvement of OS in patients with unresectable pancreatic cancer [81].

Insulin-like growth factor-1 receptor

Insulin-like growth factor (IGF)-1 receptor is highly expressed in PDAC and participates in downstream signaling pathways that are involved in cancer cell survival and proliferation. Several mAbs against IGF-1 receptor (cixutumumab, ganitumab, dalotuzumab) are currently being tested in clinical trials. Ganitumab (AMG 479) was studied in combination with gemcitabine in a phase II trial of metastatic pancreatic cancer patients and showed a slight improvement in 6-month survival rate when compared with gemcitabine plus placebo (57% vs. 50%) [82]. However, the phase III GAMMA trial of ganitumab plus gemcitabine combination was terminated early due to lack of efficacy at the preplanned interim analysis. In another phase II trial that evaluated cixutumumab plus gemcitabine and erlotinib in metastatic pancreatic cancer patients, the three-drug combination did not improve the PFS or OS when compared with gemcitabine plus erlotinib [83].

PI3K/AKT/mTOR pathway

This is one of the major downstream effector pathways of KRAS gene that is being evaluated as a potential target for pancreatic cancer treatment [84]. Rigosertib is a small molecular inhibitor of PI3K that was combined with gemcitabine in a phase II/III clinical trial (ONTRAC trial). The study was terminated early due to lack of demonstration of benefit at the time of interim analysis. Buparlisib (BKM120) is another PI3K inhibitor being evaluated in combination with mFOLFOX6 regimen in a study of advanced stage solid tumors including pancreatic cancer (NCT01571024). MK2206 is an AKT inhibitor currently under clinical evaluation in patients with pancreatic cancer (NCT01783171, NCT01658943). Archexin (RX-0201) is another AKT inhibitor that was evaluated in combination with gemcitabine in a phase II study (NCT01028495). BEZ235 is a combined inhibitor of PI3K and mTOR. A phase I study evaluating the activity of BEZ235 plus a MEK inhibitor (MEK162) in advanced solid tumor patients (including pancreatic cancer) with KRAS, NRAS and/or BRAF mutations has recently completed (NCT01337765). Everolimus (RAD001) is a mTOR inhibitor that was associated with a PFS of 1.8 months and OS of 4.5 months in a phase II study consisting of 33 gemcitabine-refractory, metastatic pancreatic cancer patients [85]. It is also being evaluated as a part of combination regimens with other agents in ongoing clinical trials.

Wnt/β-catenin pathway

Wnt signals are transduced through the frizzled receptor and lipoprotein-related protein to the β-catenin signaling cascade. There is evidence to suggest that Wnt pathway plays a role in pancreatic cancer formation via involvement in pancreatic cancer stem cells (CSCs) [86,87]. Phase I trials using mAbs (OMP-54 F28, OMP-18R5) against frizzled receptors to inhibit Wnt signaling in PDAC are currently ongoing (NCT02050178, NCT02005315).

Notch signaling pathway

Notch signaling has been shown to be upregulated in many human cancers including PDACs [88,89]. It mediates pancreatic CSC function and contributes to chemotherapy resistance, tumor recurrence, and metastases. Gamma secretase is an enzyme that causes proteolytic cleavage and release of the intracellular domain of the Notch, leading to activation of the Notch signaling pathway. In preclinical models, inhibition of Notch pathways with a gamma-secretase inhibitor (GSI) in combination with gemcitabine showed enhanced antitumor activity [90]. A phase II study evaluating an oral GSI (RO4929097) in pretreated metastatic pancreatic cancer patients was recently completed (NCT01232829). MK0752 is another GSI being tested in combination with gemcitabine for first-line treatment of stage III and IV PDAC patients (NCT01098344). Tarextumab (anti-Notch2/3 mAb, OMP-59R5) and demcizumab (anti-DLL4 mAb, OMP-21 M18) also inhibit Notch signaling and are being evaluated in clinical trials. ALPINE trial is studying the combination of tarextumab with nab-paclitaxel and gemcitabine (NCT01647828).

Targeting desmoplastic tumor microenvironment

The desmoplastic stroma in the tumor microenvironment is now regarded as a key component of pancreatic cancer biology which not only acts as a physical barrier to effective drug delivery inside the tumor but also facilitates tumor growth and promotes metastases. Strategies aimed at targeting the stromal compartment may enhance the delivery of chemotherapeutic agents to the tumor cells leading to improved efficacy. The most promising targets include the SHH signaling pathway, hyaluronic acid, and SPARC.
SHH pathway is an important signaling system that can activate the characteristic desmoplastic reaction present in the microenvironment of pancreatic tumors [22]. Sustained activation of this pathway enhances tumor growth during pancreatic oncogenesis [91]. Several clinical trials have been initiated to investigate the activity of SHH inhibitors in patients with PDAC. Vismodegib (GDC-0449) is a SHH inhibitor under clinical evaluation in combination with gemcitabine and nab-paclitaxel (NCT01088815). Saridegib (IPI-926) is another agent targeting the SHH pathway that was combined with gemcitabine in a phase II study of APC, but the trial was prematurely terminated since the combination was associated with a shorter survival than gemcitabine alone (NCT01130142). A study combining the SHH inhibitor sonidegib (LDE225) with FOLFIRINOX in untreated APC patients is ongoing (NCT01485744).
Hyaluronan is a glycosaminoglycan present in the extracellular matrix of PDAC, and high levels within the tumor are usually associated with a poor prognosis. Pegylated human recombinant PH20 hyaluronidase (PEGPH20) degrades hyaluronan and has been shown to decrease the hyaluronic acid content in a genetically-engineered PDAC mouse model, allowing for re-expansion of the PDAC blood vessels and enhanced intratumoral delivery of chemotherapeutic agents which leads to decreased tumor growth [92]. Clinically, the combination of PEGPH20 plus gemcitabine has shown promising activity in a phase Ib study of metastatic pancreatic cancer patients [93], and a phase II study of this combination is ongoing (NCT01453153). Another phase Ib/II study of PEGPH20 plus modified FOLFIRINOX combination in metastatic pancreatic cancer patients is currently under clinical evaluation (NCT01959139).
SPARC (osteonectin) is an extracellular matrix protein that plays a role in collagen turnover in the dense stroma. It is associated with invasion and metastasis in PDAC, and elevated levels are associated with poor prognosis. Nab-paclitaxel is albumin-bound paclitaxel that increases tumor accumulation of paclitaxel through binding of albumin to the stroma rich in overexpression of SPARC. As described previously, the efficacy of nab-paclitaxel plus gemcitabine combination in the first-line treatment of metastatic pancreatic cancer was demonstrated in the phase III MPACT trial which ultimately led to its FDA approval [59].
Some of the other novel strategies aimed at targeting the desmoplastic stroma within the pancreatic tumor include the use of matrix metalloproteinase (MMP) inhibitors, heparin derivatives, and hypoxia targeting agents. MMP inhibitors have been tried for the treatment of PDAC without much success to date. Marimastat (BB-2516) is a broad spectrum MMP inhibitor that was combined with gemcitabine in the treatment of APC but did not show any demonstrable clinical benefit [94]. Tanomastat (BAY12-9566) is another biphenyl MMP inhibitor with antiangiogenic and antimetastatic properties that was compared with gemcitabine for the treatment of APC patients and was found to be inferior to gemcitabine [95]. Heparin-derivative agents such as 2-0, 3-0 desulfated heparin (ODSH) and necuparanib (M402) are currently being studied in combination with gemcitabine and nab-paclitaxel for treatment of patients with metastatic pancreatic cancer (NCT01461915, NCT01621243). It is now well established the pancreatic tumor microenvironment is characterized by hypoxia. Consequently, hypoxia-targeting agents are being developed to evaluate this novel therapeutic strategy. TH-302 is a hypoxia-activated prodrug that is activated into a potent DNA-alkylating agent, bromo-isophosphoramide mustard selectively under hypoxic conditions. A recent phase II study of TH-302 plus gemcitabine showed a significant improvement in primary end point of PFS when compared with gemcitabine alone (5.6 months vs. 3.6 months; HR 0.61; 95% CI 0.43 to 0.87; P = 0.005) [96]. A phase III trial of this combination is currently in progress (MAESTRO study; NCT01746979).

TGF-β signaling pathway

TGF-β participates in stimulating stromal reaction, invasion, metastases, and angiogenesis in PDAC [97]. Examples of novel agents that target TGF-β signaling include trabedersen (AP-12009) and galunisertib (LY2157299). Trabedersen is a specific inhibitor of TGF-β2 that has demonstrated good safety and encouraging survival results in the phase I/II clinical study [98]. Galunisertib is being evaluated in combination with gemcitabine for the treatment of patients with APC in an ongoing phase Ib/II clinical trial (NCT01373164).

Epigenetic modification

Epigenetic changes such as histone deacetylation (HDAC) and DNA methylation (cytosine methylation within CG dinucleotides) can result in inactivation of the tumor suppressor genes leading to tumor growth and progression. Vorinostat is a HDAC inhibitor being tested in a phase I/II study of LAPC patients in combination with capecitabine and radiotherapy (NCT00948688). 5-Azacytidine is a cytosine analog that inhibits DNA methyltransferase, and a phase I study of its combination with gemcitabine in APC patients was recently terminated (NCT01167816).

Adenosine monophosphate-activated protein kinase pathway

The oral anti-diabetic drug metformin is an activator of adenosine monophosphate-activated protein kinase (AMPK) and disrupts the crosstalk between insulin receptor and G protein-coupled receptors (GPCR) signaling in pancreatic cancer cells, via inhibition of mTOR and suppression of its downstream effectors [99]. In xenograft mice models, metformin has been shown to inhibit pancreatic cancer growth [99]. Clinically, the available data surrounding the benefit of metformin in pancreatic cancer is conflicting and is mostly derived from retrospective studies. In a hospital-based case-control study, metformin was shown to decrease the risk of developing pancreatic cancer among diabetics [100]. In another retrospective analysis, metformin use was associated with an improvement in survival for the PDAC patients with diabetes [101]. In contrast, a more recent study from the UK failed to demonstrate a survival benefit from metformin use in PDAC patients [102]. There are multiple ongoing phase I and phase II trials that are evaluating the efficacy of metformin in PDAC. A phase I study of metformin plus erlotinib and gemcitabine in patients with APC has recently completed accrual (NCT01210911).

Synthetic lethality

The DNA double-strand breaks are repaired by a process of homologous recombination that is mediated via BRCA1 and BRCA2 proteins. Mutations in BRCA render this repair mechanism dysfunctional and are known to occur in both sporadic and familial cases of pancreatic cancer. Poly (ADP-ribose) polymerase (PARP) is another critical enzyme that mediates repair of DNA single-strand breaks. PARP pathway assumes the major role for DNA repair when BRCA mutation occurs. Consequently, inhibition of the PARP pathway results in ‘synthetic lethality’ via inhibition of DNA repair in BRCA-deficient tumor cells. A phase II study of veliparib alone or in combination with gemcitabine plus cisplatin for locally advanced and metastatic, BRCA 1–2, and PALB2-mutated pancreatic cancer patients is ongoing (NCT01585805). Other PARP inhibitors that are being evaluated in pancreatic cancer clinical trials include rucaparib (AG-14699; NCT02042378) and olaparib (AZD2281; NCT00515866).

Immunotherapy-based approaches

Despite significant efforts, no immunotherapeutic strategy against pancreatic cancer has demonstrated clinical benefit in a randomized phase III trial till date. This has been attributed to the immunologically quiescent microenvironment of pancreatic cancer. Currently, several approaches aimed at stimulating the host immune system against the pancreatic cancer tumor cells are under evaluation.
GI-4000, a form of RAS-specific immunotherapy, is heat-killed recombinant Saccharomyces cerevisiae yeast that expresses mutant RAS peptides [103]. A phase II trial of GI-4000 plus adjuvant gemcitabine is ongoing (NCT00300950). Reovirus is a tumor-targeted replication-competent virus with specificity for RAS-activated cells [104]. It is being combined with chemotherapy for the treatment of APC patients in two phase II clinical trials (NCT00998322, NCT01280058).
Developing vaccines against tumor antigens is another potential immunotherapeutic strategy to treat pancreatic cancer. Several antigens have been explored as potential targets for vaccine-based treatment in pancreatic cancer including carcinoembryonic antigen (CEA) [105], MUC1 [106,107], and heat shock proteins (HSP) [108]. Algenpantucel-L immunotherapy is a whole-cell allogeneic pancreatic cancer vaccine composed of two irradiated human pancreatic cell lines that have been genetically modified to overexpress murine alpha(1,3)-galactosyltransferase, resulting in expression of alpha-galactosyl (Alpha-Gal) epitopes on membrane glycoproteins and glycolipids. Since human cells do not express these epitopes, an immediate hyperacute rejection response ensues leading to the development of strong T-cell mediated antitumor immunity. This immunotherapeutic agent has demonstrated encouraging activity when combined with radiation and 5-FU plus gemcitabine in a phase II adjuvant trial of resected PDAC patients [109]. The phase III adjuvant trial that compares gemcitabine with or without algenpantucel-L, followed by chemoradiation has also completed recently (NCT01072981). Another phase III neoadjuvant trial is evaluating FOLFIRINOX with or without algenpantucel-L, followed by chemoradiation in borderline-resectable and unresectable LAPC patients (NCT01836432). GV1001 is a telomerase peptide vaccine shown to prolong survival when combined with granulocyte-macrophage colony-stimulating factor (GM-CSF) in a phase I/II study of unresectable LAPC patients [110]. However, the phase III study comparing GV1001 and gemcitabine in sequential combination, vs. gemcitabine monotherapy in advanced unresectable pancreatic cancer was terminated early due to lack of survival benefit in the GV1001 arm (NCT00358566). G17D (Gastrimmune) is an antigastrin-17 immunogen that was evaluated in a randomized, multicenter, placebo-controlled study of APC patients and showed a non-significant improvement in OS when compared to placebo [111]. Another potential therapeutic strategy that has been explored is combining two vaccines (GVAX plus CRS-207) with the hope to achieve an enhanced efficacy. GVAX is composed of pancreatic cancer cells that have been genetically modified to secrete GM-CSF and can induce T-cell responses. CRS-207 is a live-attenuated Listeria-based vaccine that can induce listeriolysin O and mesothelin-specific T-cell responses. The combination of GVAX plus CRS-207 is being evaluated in a phase IIb clinical trial (ECLIPSE) that consists of previously treated metastatic pancreatic cancer patients (NCT02004262).
Prostate stem cell antigen (PSCA) is a glycosylphosphatidylinositol-linked cell surface antigen expressed in pancreatic cancers. AGS-1C4D4 is a fully human IgG1 mAb against PSCA that has been combined with gemcitabine for the treatment of metastatic pancreatic cancer patients and has shown encouraging results [112].
It is now established that CD40 activation can reverse immune suppression and drive antitumor T-cell responses. Utilizing this concept, agonist CD40 antibody has been used in combination with gemcitabine in a phase I study to shrink PDAC by stimulating tumor macrophages against pancreatic cancer stroma [113].
Immunoinhibitory checkpoint pathways (cytotoxic T lymphocyte-associated protein-4 [CTLA-4]/B7, programmed cell death-1 [PD-1]/programmed cell death ligand-1 [PD-L1]) are emerging as interesting immunotherapeutic targets for the treatment of cancer. Single agent ipilimumab was evaluated in a phase II trial of APC and failed to demonstrate an appreciable antitumor activity [114]. The combination of ipilimumab with gemcitabine is currently under phase I evaluation (NCT01473940).
Radioimmunotherapy with 90Y-clivatuzumab tetraxetan (radioimmunoconjugate comprised of the humanized mAb HuPAM4 that is radiolabeled with yttrium-90) is another potential therapeutic strategy that is being evaluated in clinical trials. The combination of 90Y-clivatuzumab tetraxetan with low-dose gemcitabine demonstrated a median OS of 7.7 months in a phase I study of untreated APC patients [115]. The phase III study (PANCRIT-1) of this combination in pretreated metastatic pancreatic cancer patients is ongoing (NCT01956812).
An additional therapeutic strategy is adoptive cell transfer (ACT) approach which utilizes introduction of engineered T-cells with chimeric antigen receptors (CARs) to specifically recognize a tumor antigen of interest. This personalized immunotherapy approach is still under preclinical stages of development in the field of pancreatic cancer [116].

Novel cytotoxic agents

PEP02 (MM-398) is a novel nanoparticle liposomal formulation of irinotecan. In a phase II study of gemcitabine-refractory metastatic PDAC patients, treatment with single agent PEP02 was associated with a median PFS and OS of 9 weeks and 21.6 weeks, respectively [117]. A phase III trial (NAPOLI 1) is evaluating the combination of PEP02 with 5-FU in metastatic pancreatic cancer patients who have failed prior gemcitabine-based therapy (NCT01494506).
S-1 is a fourth-generation oral fluoropyrimidine that contains tegafur (FT, a prodrug of 5-FU), 5-chloro-2,4-dihydropyrimidine (CHDP), and potassium oxonate (Oxo). It has been evaluated in the treatment of both resectable and advanced pancreatic cancer with encouraging results. In a phase II Japanese study (PC-01), 116 patients with unresectable APC were randomized to receive gemcitabine plus S-1 vs. gemcitabine alone [118]. There was significant improvement in the ORR (28.3% vs. 6.8%; P = 0.005) and median OS (13.7 months vs. 8.0 months; P = 0.035) in the S-1 arm. Japan Adjuvant Study Group of Pancreatic Cancer (JASPAC-01) is a phase III non-inferiority trial that compared S-1 with gemcitabine as adjuvant chemotherapy for patients with curatively resected pancreatic cancer [119]. The interim analysis showed that S-1 was non-inferior to gemcitabine (OS at 2 years was 70% vs. 53%; HR 0.56; 95% CI 0.42 to 0.74; P < 0.0001 for non-inferiority) [120]. Based on the results of this study, the authors proposed that S-1 should be considered as a new standard treatment for patient with resected pancreatic cancer.

Identification of biomarkers

Development of more efficacious approaches for pancreatic cancer treatment would require identification of biomarkers that can predict the response and toxicity to various therapeutic agents. Research efforts geared towards this objective are underway.
The human equilibrative nucleoside transporter-1 (hENT1) plays an important role in the uptake of gemcitabine in cells and has been evaluated as a potential predictive biomarker of gemcitabine response. In a study that evaluated the expression pattern of genes involved in gemcitabine activity in 102 pancreatic tumor specimens, it was found that low hENT-1 expression levels were associated with a poorer prognosis [121]. However, in the pivotal phase II Low hENT1 and Adenocarcinoma of the Pancreas (LEAP) study, the hENT1 status was shown to have no clinical utility for predicting gemcitabine sensitivity [122]. Some additional potential biomarkers that have been evaluated in pancreatic cancer treatment include deoxycytidine kinase (dCK), ribonucleoside reductase-M1 (RRM1), and -M2 (RRM2) [123,124], KRAS status, SPARC staining [58], IGF-1R expression, and rs9582036 single nucleotide polymorphism (SNP) in the VEGF receptor-1 region [125]. Recently, pharmacogenomic profiling of circulating tumor and invasive cells (CTICs) isolated from patients with PDAC was evaluated as a predictor of tumor response, progression, and resistance [126].

Future directions and conclusion

The therapeutic advances in the field of pancreatic cancer have been painstakingly slow. Although we have seen a few breakthroughs in therapy for advanced disease in the recent years, the overall progress made in the field of pancreatic cancer has been relatively small in comparison to the some other tumor types. It should be noted that the majority of pancreatic cancer clinical trials over the past 5 years have failed to demonstrate any significant clinical benefit. A substantial fraction of these studies evaluated drug combinations using gemcitabine as the chemotherapy backbone. This should make us think that maybe it is time to move away from gemcitabine-based combinations and focus attention on developing innovative strategies to attack the pancreatic cancer oncogenesis. Selecting drug combinations with novel agents that target not only the primary tumor but also the surrounding stroma might be one such approach. Since our ability to safely combine drugs will be enhanced if the drug selection is based on biomarkers, we would also need prospective studies to validate potential biomarkers in well-defined patient populations in order to maximize the clinical efficacy while minimizing the toxicity of the therapeutic agents.
It is now well established that pancreatic cancer is a heterogeneous and a genetically diverse disease that results from successive accumulation of mutations over a long period of time, and these mutations affect multiple molecular pathways involved in pancreatic tumorigenesis. The presence of desmoplastic reaction in the tumor microenvironment and its role in pancreatic cancer initiation, invasion, and metastases is also being recognized. Consequently, multiple potential therapeutic approaches against pancreatic cancer are being developed and evaluated in several ongoing preclinical and clinical trials. We are hopeful that at least some of these novel strategies will demonstrate clinically meaningful benefit in future phase III studies and add to our armamentarium for treating this lethal malignancy.
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​4.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

GG drafted the manuscript and revised it critically for important intellectual content. WS revised the manuscript critically for important intellectual content. Both authors read and approved the final manuscript.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

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

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

e.Med Innere Medizin

Kombi-Abonnement

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

Literatur
1.
Zurück zum Zitat Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin. 2015;65:5–29.PubMed Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin. 2015;65:5–29.PubMed
2.
Zurück zum Zitat Jones S, Zhang X, Parsons DW, Lin JC, Leary RJ, Angenendt P, et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science. 2008;321:1801–6.PubMedCentralPubMed Jones S, Zhang X, Parsons DW, Lin JC, Leary RJ, Angenendt P, et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science. 2008;321:1801–6.PubMedCentralPubMed
3.
Zurück zum Zitat Blackford A, Parmigiani G, Kensler TW, Wolfgang C, Jones S, Zhang X, et al. Genetic mutations associated with cigarette smoking in pancreatic cancer. Cancer Res. 2009;69:3681–8.PubMedCentralPubMed Blackford A, Parmigiani G, Kensler TW, Wolfgang C, Jones S, Zhang X, et al. Genetic mutations associated with cigarette smoking in pancreatic cancer. Cancer Res. 2009;69:3681–8.PubMedCentralPubMed
4.
Zurück zum Zitat Genkinger JM, Spiegelman D, Anderson KE, Bergkvist L, Bernstein L, van den Brandt PA, et al. Alcohol intake and pancreatic cancer risk: a pooled analysis of fourteen cohort studies. Cancer Epidemiol Biomarkers Prev. 2009;18:765–76.PubMedCentralPubMed Genkinger JM, Spiegelman D, Anderson KE, Bergkvist L, Bernstein L, van den Brandt PA, et al. Alcohol intake and pancreatic cancer risk: a pooled analysis of fourteen cohort studies. Cancer Epidemiol Biomarkers Prev. 2009;18:765–76.PubMedCentralPubMed
5.
Zurück zum Zitat Philip B, Roland CL, Daniluk J, Liu Y, Chatterjee D, Gomez SB, et al. A high-fat diet activates oncogenic Kras and COX2 to induce development of pancreatic ductal adenocarcinoma in mice. Gastroenterology. 2013;145:1449–58.PubMed Philip B, Roland CL, Daniluk J, Liu Y, Chatterjee D, Gomez SB, et al. A high-fat diet activates oncogenic Kras and COX2 to induce development of pancreatic ductal adenocarcinoma in mice. Gastroenterology. 2013;145:1449–58.PubMed
6.
Zurück zum Zitat Amaral AF, Porta M, Silverman DT, Milne RL, Kogevinas M, Rothman N, et al. Pancreatic cancer risk and levels of trace elements. Gut. 2012;61:1583–8.PubMedCentralPubMed Amaral AF, Porta M, Silverman DT, Milne RL, Kogevinas M, Rothman N, et al. Pancreatic cancer risk and levels of trace elements. Gut. 2012;61:1583–8.PubMedCentralPubMed
7.
8.
Zurück zum Zitat Shi C, Hruban RH, Klein AP. Familial pancreatic cancer. Arch Pathol Lab Med. 2009;133:365–74.PubMed Shi C, Hruban RH, Klein AP. Familial pancreatic cancer. Arch Pathol Lab Med. 2009;133:365–74.PubMed
9.
Zurück zum Zitat Hruban RH, Canto MI, Goggins M, Schulick R, Klein AP. Update on familial pancreatic cancer. Adv Surg. 2010;44:293–311.PubMedCentralPubMed Hruban RH, Canto MI, Goggins M, Schulick R, Klein AP. Update on familial pancreatic cancer. Adv Surg. 2010;44:293–311.PubMedCentralPubMed
10.
Zurück zum Zitat Canto MI, Harinck F, Hruban RH, Offerhaus GJ, Poley JW, Kamel I, et al. International Cancer of the Pancreas Screening (CAPS) Consortium summit on the management of patients with increased risk for familial pancreatic cancer. Gut. 2013;62:339–47.PubMedCentralPubMed Canto MI, Harinck F, Hruban RH, Offerhaus GJ, Poley JW, Kamel I, et al. International Cancer of the Pancreas Screening (CAPS) Consortium summit on the management of patients with increased risk for familial pancreatic cancer. Gut. 2013;62:339–47.PubMedCentralPubMed
11.
Zurück zum Zitat Hruban RH, Goggins M, Parsons J, Kern SE. Progression model for pancreatic cancer. Clin Cancer Res. 2000;6:2969–72.PubMed Hruban RH, Goggins M, Parsons J, Kern SE. Progression model for pancreatic cancer. Clin Cancer Res. 2000;6:2969–72.PubMed
12.
13.
Zurück zum Zitat Yachida S, Jones S, Bozic I, Antal T, Leary R, Fu B, et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature. 2010;467:1114–7.PubMedCentralPubMed Yachida S, Jones S, Bozic I, Antal T, Leary R, Fu B, et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature. 2010;467:1114–7.PubMedCentralPubMed
15.
Zurück zum Zitat Caldas C, Kern SE. K-ras mutation and pancreatic adenocarcinoma. Int J Pancreatol. 1995;18:1–6.PubMed Caldas C, Kern SE. K-ras mutation and pancreatic adenocarcinoma. Int J Pancreatol. 1995;18:1–6.PubMed
16.
Zurück zum Zitat di Magliano MP, Logsdon CD. Roles for KRAS in pancreatic tumor development and progression. Gastroenterology. 2013;144:1220–9.PubMedCentralPubMed di Magliano MP, Logsdon CD. Roles for KRAS in pancreatic tumor development and progression. Gastroenterology. 2013;144:1220–9.PubMedCentralPubMed
17.
Zurück zum Zitat Yachida S, Iacobuzio-Donahue CA. Evolution and dynamics of pancreatic cancer progression. Oncogene. 2013;32:5253–60.PubMed Yachida S, Iacobuzio-Donahue CA. Evolution and dynamics of pancreatic cancer progression. Oncogene. 2013;32:5253–60.PubMed
18.
Zurück zum Zitat Agbunag C, Bar-Sagi D. Oncogenic K-ras drives cell cycle progression and phenotypic conversion of primary pancreatic duct epithelial cells. Cancer Res. 2004;64:5659–63.PubMed Agbunag C, Bar-Sagi D. Oncogenic K-ras drives cell cycle progression and phenotypic conversion of primary pancreatic duct epithelial cells. Cancer Res. 2004;64:5659–63.PubMed
19.
Zurück zum Zitat Sasaki S, Yamamoto H, Kaneto H, Ozeki I, Adachi Y, Takagi H, et al. Differential roles of alterations of p53, p16, and SMAD4 expression in the progression of intraductal papillary-mucinous tumors of the pancreas. Oncol Rep. 2003;10:21–5.PubMed Sasaki S, Yamamoto H, Kaneto H, Ozeki I, Adachi Y, Takagi H, et al. Differential roles of alterations of p53, p16, and SMAD4 expression in the progression of intraductal papillary-mucinous tumors of the pancreas. Oncol Rep. 2003;10:21–5.PubMed
20.
Zurück zum Zitat Yachida S, Iacobuzio-Donahue CA. The pathology and genetics of metastatic pancreatic cancer. Arch Pathol Lab Med. 2009;133:413–22.PubMed Yachida S, Iacobuzio-Donahue CA. The pathology and genetics of metastatic pancreatic cancer. Arch Pathol Lab Med. 2009;133:413–22.PubMed
21.
Zurück zum Zitat Neesse A, Michl P, Frese KK, Feig C, Cook N, Jacobetz MA, et al. Stromal biology and therapy in pancreatic cancer. Gut. 2011;60:861–8.PubMed Neesse A, Michl P, Frese KK, Feig C, Cook N, Jacobetz MA, et al. Stromal biology and therapy in pancreatic cancer. Gut. 2011;60:861–8.PubMed
22.
Zurück zum Zitat Tian H, Callahan CA, DuPree KJ, Darbonne WC, Ahn CP, Scales SJ, et al. Hedgehog signaling is restricted to the stromal compartment during pancreatic carcinogenesis. Proc Natl Acad Sci U S A. 2009;106:4254–9.PubMedCentralPubMed Tian H, Callahan CA, DuPree KJ, Darbonne WC, Ahn CP, Scales SJ, et al. Hedgehog signaling is restricted to the stromal compartment during pancreatic carcinogenesis. Proc Natl Acad Sci U S A. 2009;106:4254–9.PubMedCentralPubMed
23.
Zurück zum Zitat Wagner M, Redaelli C, Lietz M, Seiler CA, Friess H, Buchler MW. Curative resection is the single most important factor determining outcome in patients with pancreatic adenocarcinoma. Br J Surg. 2004;91:586–94.PubMed Wagner M, Redaelli C, Lietz M, Seiler CA, Friess H, Buchler MW. Curative resection is the single most important factor determining outcome in patients with pancreatic adenocarcinoma. Br J Surg. 2004;91:586–94.PubMed
24.
Zurück zum Zitat Hidalgo M. Pancreatic cancer. N Engl J Med. 2010;362:1605–17.PubMed Hidalgo M. Pancreatic cancer. N Engl J Med. 2010;362:1605–17.PubMed
25.
Zurück zum Zitat Neoptolemos JP, Stocken DD, Bassi C, Ghaneh P, Cunningham D, Goldstein D, et al. Adjuvant chemotherapy with fluorouracil plus folinic acid vs gemcitabine following pancreatic cancer resection: a randomized controlled trial. JAMA. 2010;304:1073–81.PubMed Neoptolemos JP, Stocken DD, Bassi C, Ghaneh P, Cunningham D, Goldstein D, et al. Adjuvant chemotherapy with fluorouracil plus folinic acid vs gemcitabine following pancreatic cancer resection: a randomized controlled trial. JAMA. 2010;304:1073–81.PubMed
26.
Zurück zum Zitat Neoptolemos JP, Stocken DD, Friess H, Bassi C, Dunn JA, Hickey H, et al. A randomized trial of chemoradiotherapy and chemotherapy after resection of pancreatic cancer. N Engl J Med. 2004;350:1200–10.PubMed Neoptolemos JP, Stocken DD, Friess H, Bassi C, Dunn JA, Hickey H, et al. A randomized trial of chemoradiotherapy and chemotherapy after resection of pancreatic cancer. N Engl J Med. 2004;350:1200–10.PubMed
27.
Zurück zum Zitat Regine WF, Winter KA, Abrams R, Safran H, Hoffman JP, Konski A, et al. Fluorouracil-based chemoradiation with either gemcitabine or fluorouracil chemotherapy after resection of pancreatic adenocarcinoma: 5-year analysis of the U.S. Intergroup/RTOG 9704 phase III trial. Ann Surg Oncol. 2011;18:1319–26.PubMedCentralPubMed Regine WF, Winter KA, Abrams R, Safran H, Hoffman JP, Konski A, et al. Fluorouracil-based chemoradiation with either gemcitabine or fluorouracil chemotherapy after resection of pancreatic adenocarcinoma: 5-year analysis of the U.S. Intergroup/RTOG 9704 phase III trial. Ann Surg Oncol. 2011;18:1319–26.PubMedCentralPubMed
28.
Zurück zum Zitat Oettle H, Post S, Neuhaus P, Gellert K, Langrehr J, Ridwelski K, et al. Adjuvant chemotherapy with gemcitabine vs observation in patients undergoing curative-intent resection of pancreatic cancer: a randomized controlled trial. JAMA. 2007;297:267–77.PubMed Oettle H, Post S, Neuhaus P, Gellert K, Langrehr J, Ridwelski K, et al. Adjuvant chemotherapy with gemcitabine vs observation in patients undergoing curative-intent resection of pancreatic cancer: a randomized controlled trial. JAMA. 2007;297:267–77.PubMed
29.
Zurück zum Zitat Regine WF, Winter KA, Abrams RA, Safran H, Hoffman JP, Konski A, et al. Fluorouracil vs gemcitabine chemotherapy before and after fluorouracil-based chemoradiation following resection of pancreatic adenocarcinoma: a randomized controlled trial. JAMA. 2008;299:1019–26.PubMed Regine WF, Winter KA, Abrams RA, Safran H, Hoffman JP, Konski A, et al. Fluorouracil vs gemcitabine chemotherapy before and after fluorouracil-based chemoradiation following resection of pancreatic adenocarcinoma: a randomized controlled trial. JAMA. 2008;299:1019–26.PubMed
30.
Zurück zum Zitat Evans DB, Rich TA, Byrd DR, Cleary KR, Connelly JH, Levin B, et al. Preoperative chemoradiation and pancreaticoduodenectomy for adenocarcinoma of the pancreas. Arch Surg. 1992;127:1335–9.PubMed Evans DB, Rich TA, Byrd DR, Cleary KR, Connelly JH, Levin B, et al. Preoperative chemoradiation and pancreaticoduodenectomy for adenocarcinoma of the pancreas. Arch Surg. 1992;127:1335–9.PubMed
31.
Zurück zum Zitat Pisters PW, Abbruzzese JL, Janjan NA, Cleary KR, Charnsangavej C, Goswitz MS, et al. Rapid-fractionation preoperative chemoradiation, pancreaticoduodenectomy, and intraoperative radiation therapy for resectable pancreatic adenocarcinoma. J Clin Oncol. 1998;16:3843–50.PubMed Pisters PW, Abbruzzese JL, Janjan NA, Cleary KR, Charnsangavej C, Goswitz MS, et al. Rapid-fractionation preoperative chemoradiation, pancreaticoduodenectomy, and intraoperative radiation therapy for resectable pancreatic adenocarcinoma. J Clin Oncol. 1998;16:3843–50.PubMed
32.
Zurück zum Zitat Pisters PW, Wolff RA, Janjan NA, Cleary KR, Charnsangavej C, Crane CN, et al. Preoperative paclitaxel and concurrent rapid-fractionation radiation for resectable pancreatic adenocarcinoma: toxicities, histologic response rates, and event-free outcome. J Clin Oncol. 2002;20:2537–44.PubMed Pisters PW, Wolff RA, Janjan NA, Cleary KR, Charnsangavej C, Crane CN, et al. Preoperative paclitaxel and concurrent rapid-fractionation radiation for resectable pancreatic adenocarcinoma: toxicities, histologic response rates, and event-free outcome. J Clin Oncol. 2002;20:2537–44.PubMed
33.
Zurück zum Zitat Evans DB, Varadhachary GR, Crane CH, Sun CC, Lee JE, Pisters PW, et al. Preoperative gemcitabine-based chemoradiation for patients with resectable adenocarcinoma of the pancreatic head. J Clin Oncol. 2008;26:3496–502.PubMed Evans DB, Varadhachary GR, Crane CH, Sun CC, Lee JE, Pisters PW, et al. Preoperative gemcitabine-based chemoradiation for patients with resectable adenocarcinoma of the pancreatic head. J Clin Oncol. 2008;26:3496–502.PubMed
34.
Zurück zum Zitat Varadhachary GR, Wolff RA, Crane CH, Sun CC, Lee JE, Pisters PW, et al. Preoperative gemcitabine and cisplatin followed by gemcitabine-based chemoradiation for resectable adenocarcinoma of the pancreatic head. J Clin Oncol. 2008;26:3487–95.PubMed Varadhachary GR, Wolff RA, Crane CH, Sun CC, Lee JE, Pisters PW, et al. Preoperative gemcitabine and cisplatin followed by gemcitabine-based chemoradiation for resectable adenocarcinoma of the pancreatic head. J Clin Oncol. 2008;26:3487–95.PubMed
35.
Zurück zum Zitat Paulson AS, Tran Cao HS, Tempero MA, Lowy AM. Therapeutic advances in pancreatic cancer. Gastroenterology. 2013;144:1316–26.PubMed Paulson AS, Tran Cao HS, Tempero MA, Lowy AM. Therapeutic advances in pancreatic cancer. Gastroenterology. 2013;144:1316–26.PubMed
36.
Zurück zum Zitat Mehta VK, Fisher G, Ford JA, Poen JC, Vierra MA, Oberhelman H, et al. Preoperative chemoradiation for marginally resectable adenocarcinoma of the pancreas. J Gastrointest Surg. 2001;5:27–35.PubMed Mehta VK, Fisher G, Ford JA, Poen JC, Vierra MA, Oberhelman H, et al. Preoperative chemoradiation for marginally resectable adenocarcinoma of the pancreas. J Gastrointest Surg. 2001;5:27–35.PubMed
37.
Zurück zum Zitat Small Jr W, Berlin J, Freedman GM, Lawrence T, Talamonti MS, Mulcahy MF, et al. Full-dose gemcitabine with concurrent radiation therapy in patients with nonmetastatic pancreatic cancer: a multicenter phase II trial. J Clin Oncol. 2008;26:942–7.PubMed Small Jr W, Berlin J, Freedman GM, Lawrence T, Talamonti MS, Mulcahy MF, et al. Full-dose gemcitabine with concurrent radiation therapy in patients with nonmetastatic pancreatic cancer: a multicenter phase II trial. J Clin Oncol. 2008;26:942–7.PubMed
38.
Zurück zum Zitat Stokes JB, Nolan NJ, Stelow EB, Walters DM, Weiss GR, de Lange EE, et al. Preoperative capecitabine and concurrent radiation for borderline resectable pancreatic cancer. Ann Surg Oncol. 2011;18:619–27.PubMed Stokes JB, Nolan NJ, Stelow EB, Walters DM, Weiss GR, de Lange EE, et al. Preoperative capecitabine and concurrent radiation for borderline resectable pancreatic cancer. Ann Surg Oncol. 2011;18:619–27.PubMed
39.
Zurück zum Zitat Patel M, Hoffe S, Malafa M, Hodul P, Klapman J, Centeno B, et al. Neoadjuvant GTX chemotherapy and IMRT-based chemoradiation for borderline resectable pancreatic cancer. J Surg Oncol. 2011;104:155–61.PubMedCentralPubMed Patel M, Hoffe S, Malafa M, Hodul P, Klapman J, Centeno B, et al. Neoadjuvant GTX chemotherapy and IMRT-based chemoradiation for borderline resectable pancreatic cancer. J Surg Oncol. 2011;104:155–61.PubMedCentralPubMed
40.
Zurück zum Zitat Moertel CG, Frytak S, Hahn RG, O’Connell MJ, Reitemeier RJ, Rubin J, et al. Therapy of locally unresectable pancreatic carcinoma: a randomized comparison of high dose (6000 rads) radiation alone, moderate dose radiation (4000 rads + 5-fluorouracil), and high dose radiation + 5-fluorouracil: The Gastrointestinal Tumor Study Group. Cancer. 1981;48:1705–10.PubMed Moertel CG, Frytak S, Hahn RG, O’Connell MJ, Reitemeier RJ, Rubin J, et al. Therapy of locally unresectable pancreatic carcinoma: a randomized comparison of high dose (6000 rads) radiation alone, moderate dose radiation (4000 rads + 5-fluorouracil), and high dose radiation + 5-fluorouracil: The Gastrointestinal Tumor Study Group. Cancer. 1981;48:1705–10.PubMed
41.
Zurück zum Zitat Loehrer Sr PJ, Feng Y, Cardenes H, Wagner L, Brell JM, Cella D, et al. Gemcitabine alone versus gemcitabine plus radiotherapy in patients with locally advanced pancreatic cancer: an Eastern Cooperative Oncology Group trial. J Clin Oncol. 2011;29:4105–12.PubMedCentralPubMed Loehrer Sr PJ, Feng Y, Cardenes H, Wagner L, Brell JM, Cella D, et al. Gemcitabine alone versus gemcitabine plus radiotherapy in patients with locally advanced pancreatic cancer: an Eastern Cooperative Oncology Group trial. J Clin Oncol. 2011;29:4105–12.PubMedCentralPubMed
42.
Zurück zum Zitat Li CP, Chao Y, Chi KH, Chan WK, Teng HC, Lee RC, et al. Concurrent chemoradiotherapy treatment of locally advanced pancreatic cancer: gemcitabine versus 5-fluorouracil, a randomized controlled study. Int J Radiat Oncol, Biol, Phys. 2003;57:98–104. Li CP, Chao Y, Chi KH, Chan WK, Teng HC, Lee RC, et al. Concurrent chemoradiotherapy treatment of locally advanced pancreatic cancer: gemcitabine versus 5-fluorouracil, a randomized controlled study. Int J Radiat Oncol, Biol, Phys. 2003;57:98–104.
43.
Zurück zum Zitat Rocha Lima CM, Green MR, Rotche R, Miller Jr WH, Jeffrey GM, Cisar LA, et al. 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;22:3776–83.PubMed Rocha Lima CM, Green MR, Rotche R, Miller Jr WH, Jeffrey GM, Cisar LA, et al. 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;22:3776–83.PubMed
44.
Zurück zum Zitat Van Cutsem E, van de Velde H, Karasek P, Oettle H, Vervenne WL, Szawlowski A, et al. Phase III trial of gemcitabine plus tipifarnib compared with gemcitabine plus placebo in advanced pancreatic cancer. J Clin Oncol. 2004;22:1430–8.PubMed Van Cutsem E, van de Velde H, Karasek P, Oettle H, Vervenne WL, Szawlowski A, et al. Phase III trial of gemcitabine plus tipifarnib compared with gemcitabine plus placebo in advanced pancreatic cancer. J Clin Oncol. 2004;22:1430–8.PubMed
45.
Zurück zum Zitat Louvet C, Labianca R, Hammel P, Lledo G, Zampino MG, Andre T, et al. 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;23:3509–16.PubMed Louvet C, Labianca R, Hammel P, Lledo G, Zampino MG, Andre T, et al. 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;23:3509–16.PubMed
46.
Zurück zum Zitat Ghosn M, Farhat F, Kattan J, Younes F, Moukadem W, Nasr F, et al. FOLFOX-6 combination as the first-line treatment of locally advanced and/or metastatic pancreatic cancer. Am J Clin Oncol. 2007;30:15–20.PubMed Ghosn M, Farhat F, Kattan J, Younes F, Moukadem W, Nasr F, et al. FOLFOX-6 combination as the first-line treatment of locally advanced and/or metastatic pancreatic cancer. Am J Clin Oncol. 2007;30:15–20.PubMed
47.
Zurück zum Zitat Boone BA, Steve J, Krasinskas AM, Zureikat AH, Lembersky BC, Gibson MK, et al. Outcomes with FOLFIRINOX for borderline resectable and locally unresectable pancreatic cancer. J Surg Oncol. 2013;108:236–41.PubMed Boone BA, Steve J, Krasinskas AM, Zureikat AH, Lembersky BC, Gibson MK, et al. Outcomes with FOLFIRINOX for borderline resectable and locally unresectable pancreatic cancer. J Surg Oncol. 2013;108:236–41.PubMed
48.
Zurück zum Zitat Faris JE, Blaszkowsky LS, McDermott S, Guimaraes AR, Szymonifka J, Huynh MA, et al. FOLFIRINOX in locally advanced pancreatic cancer: the Massachusetts General Hospital Cancer Center experience. Oncologist. 2013;18:543–8.PubMedCentralPubMed Faris JE, Blaszkowsky LS, McDermott S, Guimaraes AR, Szymonifka J, Huynh MA, et al. FOLFIRINOX in locally advanced pancreatic cancer: the Massachusetts General Hospital Cancer Center experience. Oncologist. 2013;18:543–8.PubMedCentralPubMed
49.
Zurück zum Zitat Gunturu KS, Yao X, Cong X, Thumar JR, Hochster HS, Stein SM, et al. FOLFIRINOX for locally advanced and metastatic pancreatic cancer: single institution retrospective review of efficacy and toxicity. Med Oncol. 2013;30:361.PubMed Gunturu KS, Yao X, Cong X, Thumar JR, Hochster HS, Stein SM, et al. FOLFIRINOX for locally advanced and metastatic pancreatic cancer: single institution retrospective review of efficacy and toxicity. Med Oncol. 2013;30:361.PubMed
50.
Zurück zum Zitat Hosein PJ, Macintyre J, Kawamura C, Maldonado JC, Ernani V, Loaiza-Bonilla A, et al. A retrospective study of neoadjuvant FOLFIRINOX in unresectable or borderline-resectable locally advanced pancreatic adenocarcinoma. BMC Cancer. 2012;12:199.PubMedCentralPubMed Hosein PJ, Macintyre J, Kawamura C, Maldonado JC, Ernani V, Loaiza-Bonilla A, et al. A retrospective study of neoadjuvant FOLFIRINOX in unresectable or borderline-resectable locally advanced pancreatic adenocarcinoma. BMC Cancer. 2012;12:199.PubMedCentralPubMed
51.
Zurück zum Zitat Huguet F, Andre T, Hammel P, Artru P, Balosso J, Selle F, et al. Impact of chemoradiotherapy after disease control with chemotherapy in locally advanced pancreatic adenocarcinoma in GERCOR phase II and III studies. J Clin Oncol. 2007;25:326–31.PubMed Huguet F, Andre T, Hammel P, Artru P, Balosso J, Selle F, et al. Impact of chemoradiotherapy after disease control with chemotherapy in locally advanced pancreatic adenocarcinoma in GERCOR phase II and III studies. J Clin Oncol. 2007;25:326–31.PubMed
52.
Zurück zum Zitat Krishnan S, Rana V, Janjan NA, Varadhachary GR, Abbruzzese JL, Das P, et al. Induction chemotherapy selects patients with locally advanced, unresectable pancreatic cancer for optimal benefit from consolidative chemoradiation therapy. Cancer. 2007;110:47–55.PubMed Krishnan S, Rana V, Janjan NA, Varadhachary GR, Abbruzzese JL, Das P, et al. Induction chemotherapy selects patients with locally advanced, unresectable pancreatic cancer for optimal benefit from consolidative chemoradiation therapy. Cancer. 2007;110:47–55.PubMed
53.
Zurück zum Zitat Burris 3rd HA, Moore MJ, Andersen J, Green MR, Rothenberg ML, Modiano MR, et al. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. J Clin Oncol. 1997;15:2403–13.PubMed Burris 3rd HA, Moore MJ, Andersen J, Green MR, Rothenberg ML, Modiano MR, et al. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. J Clin Oncol. 1997;15:2403–13.PubMed
54.
Zurück zum Zitat Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, et al. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol. 2007;25:1960–6.PubMed Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, et al. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol. 2007;25:1960–6.PubMed
55.
Zurück zum Zitat Conroy T, Desseigne F, Ychou M, Bouche O, Guimbaud R, Becouarn Y, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364:1817–25.PubMed Conroy T, Desseigne F, Ychou M, Bouche O, Guimbaud R, Becouarn Y, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364:1817–25.PubMed
56.
Zurück zum Zitat Mahaseth H, Kauh JS, Brutcher E, Hawk NN, Kim S, Chen Z. Safety and efficacy of modified FOLFIRINOX in pancreatic cancer: a retrospective experience. In: ASCO Meeting. 2012. Mahaseth H, Kauh JS, Brutcher E, Hawk NN, Kim S, Chen Z. Safety and efficacy of modified FOLFIRINOX in pancreatic cancer: a retrospective experience. In: ASCO Meeting. 2012.
57.
Zurück zum Zitat Edmonds C, Cengel KA. Tumor-stroma interactions in pancreatic cancer: Will this SPARC prove a raging fire? Cancer Biol Ther. 2008;7:1816–7.PubMed Edmonds C, Cengel KA. Tumor-stroma interactions in pancreatic cancer: Will this SPARC prove a raging fire? Cancer Biol Ther. 2008;7:1816–7.PubMed
58.
Zurück zum Zitat Von Hoff DD, Ramanathan RK, Borad MJ, Laheru DA, Smith LS, Wood TE, et al. Gemcitabine plus nab-paclitaxel is an active regimen in patients with advanced pancreatic cancer: a phase I/II trial. J Clin Oncol. 2011;29:4548–54. Von Hoff DD, Ramanathan RK, Borad MJ, Laheru DA, Smith LS, Wood TE, et al. Gemcitabine plus nab-paclitaxel is an active regimen in patients with advanced pancreatic cancer: a phase I/II trial. J Clin Oncol. 2011;29:4548–54.
59.
Zurück zum Zitat Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369:1691–703. Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369:1691–703.
60.
Zurück zum Zitat Rowinsky EK, Windle JJ, Von Hoff DD. Ras protein farnesyltransferase: a strategic target for anticancer therapeutic development. J Clin Oncol. 1999;17:3631–52.PubMed Rowinsky EK, Windle JJ, Von Hoff DD. Ras protein farnesyltransferase: a strategic target for anticancer therapeutic development. J Clin Oncol. 1999;17:3631–52.PubMed
61.
Zurück zum Zitat Cohen SJ, Ho L, Ranganathan S, Abbruzzese JL, Alpaugh RK, Beard M, et al. Phase II and pharmacodynamic study of the farnesyltransferase inhibitor R115777 as initial therapy in patients with metastatic pancreatic adenocarcinoma. J Clin Oncol. 2003;21:1301–6.PubMed Cohen SJ, Ho L, Ranganathan S, Abbruzzese JL, Alpaugh RK, Beard M, et al. Phase II and pharmacodynamic study of the farnesyltransferase inhibitor R115777 as initial therapy in patients with metastatic pancreatic adenocarcinoma. J Clin Oncol. 2003;21:1301–6.PubMed
62.
Zurück zum Zitat Macdonald JS, McCoy S, Whitehead RP, Iqbal S, Wade 3rd JL, Giguere JK, et al. A phase II study of farnesyl transferase inhibitor R115777 in pancreatic cancer: a Southwest oncology group (SWOG 9924) study. Invest New Drugs. 2005;23:485–7.PubMed Macdonald JS, McCoy S, Whitehead RP, Iqbal S, Wade 3rd JL, Giguere JK, et al. A phase II study of farnesyl transferase inhibitor R115777 in pancreatic cancer: a Southwest oncology group (SWOG 9924) study. Invest New Drugs. 2005;23:485–7.PubMed
63.
Zurück zum Zitat Akinleye A, Furqan M, Mukhi N, Ravella P, Liu D. MEK and the inhibitors: from bench to bedside. J Hematol Oncol. 2013;6:27.PubMedCentralPubMed Akinleye A, Furqan M, Mukhi N, Ravella P, Liu D. MEK and the inhibitors: from bench to bedside. J Hematol Oncol. 2013;6:27.PubMedCentralPubMed
64.
Zurück zum Zitat Bodoky G, Timcheva C, Spigel DR, La Stella PJ, Ciuleanu TE, Pover G, et al. A phase II open-label randomized study to assess the efficacy and safety of selumetinib (AZD6244 [ARRY-142886]) versus capecitabine in patients with advanced or metastatic pancreatic cancer who have failed first-line gemcitabine therapy. Invest New Drugs. 2012;30:1216–23.PubMed Bodoky G, Timcheva C, Spigel DR, La Stella PJ, Ciuleanu TE, Pover G, et al. A phase II open-label randomized study to assess the efficacy and safety of selumetinib (AZD6244 [ARRY-142886]) versus capecitabine in patients with advanced or metastatic pancreatic cancer who have failed first-line gemcitabine therapy. Invest New Drugs. 2012;30:1216–23.PubMed
65.
Zurück zum Zitat Infante JR, Somer BG, Park JO, Li CP, Scheulen ME, Kasubhai SM, et al. A randomised, double-blind, placebo-controlled trial of trametinib, an oral MEK inhibitor, in combination with gemcitabine for patients with untreated metastatic adenocarcinoma of the pancreas. Eur J Cancer. 2014;50:2072–81.PubMed Infante JR, Somer BG, Park JO, Li CP, Scheulen ME, Kasubhai SM, et al. A randomised, double-blind, placebo-controlled trial of trametinib, an oral MEK inhibitor, in combination with gemcitabine for patients with untreated metastatic adenocarcinoma of the pancreas. Eur J Cancer. 2014;50:2072–81.PubMed
66.
Zurück zum Zitat Guo JY, Chen HY, Mathew R, Fan J, Strohecker AM, Karsli-Uzunbas G, et al. Activated Ras requires autophagy to maintain oxidative metabolism and tumorigenesis. Genes Dev. 2011;25:460–70.PubMedCentralPubMed Guo JY, Chen HY, Mathew R, Fan J, Strohecker AM, Karsli-Uzunbas G, et al. Activated Ras requires autophagy to maintain oxidative metabolism and tumorigenesis. Genes Dev. 2011;25:460–70.PubMedCentralPubMed
67.
Zurück zum Zitat Lemoine NR, Hughes CM, Barton CM, Poulsom R, Jeffery RE, Kloppel G, et al. The epidermal growth factor receptor in human pancreatic cancer. J Pathol. 1992;166:7–12.PubMed Lemoine NR, Hughes CM, Barton CM, Poulsom R, Jeffery RE, Kloppel G, et al. The epidermal growth factor receptor in human pancreatic cancer. J Pathol. 1992;166:7–12.PubMed
68.
Zurück zum Zitat Safran H, Steinhoff M, Mangray S, Rathore R, King TC, Chai L, et al. Overexpression of the HER-2/neu oncogene in pancreatic adenocarcinoma. Am J Clin Oncol. 2001;24:496–9.PubMed Safran H, Steinhoff M, Mangray S, Rathore R, King TC, Chai L, et al. Overexpression of the HER-2/neu oncogene in pancreatic adenocarcinoma. Am J Clin Oncol. 2001;24:496–9.PubMed
69.
Zurück zum Zitat Kulke MH, Blaszkowsky LS, Ryan DP, Clark JW, Meyerhardt JA, Zhu AX, et al. Capecitabine plus erlotinib in gemcitabine-refractory advanced pancreatic cancer. J Clin Oncol. 2007;25:4787–92.PubMed Kulke MH, Blaszkowsky LS, Ryan DP, Clark JW, Meyerhardt JA, Zhu AX, et al. Capecitabine plus erlotinib in gemcitabine-refractory advanced pancreatic cancer. J Clin Oncol. 2007;25:4787–92.PubMed
70.
Zurück zum Zitat Xiong HQ, Rosenberg A, LoBuglio A, Schmidt W, Wolff RA, Deutsch J, et al. Cetuximab, a monoclonal antibody targeting the epidermal growth factor receptor, in combination with gemcitabine for advanced pancreatic cancer: a multicenter phase II trial. J Clin Oncol. 2004;22:2610–6.PubMed Xiong HQ, Rosenberg A, LoBuglio A, Schmidt W, Wolff RA, Deutsch J, et al. Cetuximab, a monoclonal antibody targeting the epidermal growth factor receptor, in combination with gemcitabine for advanced pancreatic cancer: a multicenter phase II trial. J Clin Oncol. 2004;22:2610–6.PubMed
71.
Zurück zum Zitat Philip PA, Benedetti J, Corless CL, Wong R, O’Reilly EM, Flynn PJ, et al. 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;28:3605–10.PubMedCentralPubMed Philip PA, Benedetti J, Corless CL, Wong R, O’Reilly EM, Flynn PJ, et al. 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;28:3605–10.PubMedCentralPubMed
72.
Zurück zum Zitat Kim GP, Foster NR, Salim M, Flynn PJ, Moore DF, Zon R, et al. Randomized phase II trial of panitumumab, erlotinib, and gemcitabine (PGE) versus erlotinib-gemcitabine (GE) in patients with untreated, metastatic pancreatic adenocarcinoma. J Clin Oncol. 2011;29(suppl):abstr 4030. Kim GP, Foster NR, Salim M, Flynn PJ, Moore DF, Zon R, et al. Randomized phase II trial of panitumumab, erlotinib, and gemcitabine (PGE) versus erlotinib-gemcitabine (GE) in patients with untreated, metastatic pancreatic adenocarcinoma. J Clin Oncol. 2011;29(suppl):abstr 4030.
73.
Zurück zum Zitat Safran H, Iannitti D, Ramanathan R, Schwartz JD, Steinhoff M, Nauman C, et al. Herceptin and gemcitabine for metastatic pancreatic cancers that overexpress HER-2/neu. Cancer Invest. 2004;22:706–12.PubMed Safran H, Iannitti D, Ramanathan R, Schwartz JD, Steinhoff M, Nauman C, et al. Herceptin and gemcitabine for metastatic pancreatic cancers that overexpress HER-2/neu. Cancer Invest. 2004;22:706–12.PubMed
74.
75.
Zurück zum Zitat Kindler HL, Niedzwiecki D, Hollis D, Sutherland S, Schrag D, Hurwitz H, et al. Gemcitabine plus bevacizumab compared with gemcitabine plus placebo in patients with advanced pancreatic cancer: phase III trial of the Cancer and Leukemia Group B (CALGB 80303). J Clin Oncol. 2010;28:3617–22.PubMedCentralPubMed Kindler HL, Niedzwiecki D, Hollis D, Sutherland S, Schrag D, Hurwitz H, et al. Gemcitabine plus bevacizumab compared with gemcitabine plus placebo in patients with advanced pancreatic cancer: phase III trial of the Cancer and Leukemia Group B (CALGB 80303). J Clin Oncol. 2010;28:3617–22.PubMedCentralPubMed
76.
Zurück zum Zitat Van Cutsem E, Vervenne WL, Bennouna J, Humblet Y, Gill S, Van Laethem JL, et al. Phase III trial of bevacizumab in combination with gemcitabine and erlotinib in patients with metastatic pancreatic cancer. J Clin Oncol. 2009;27:2231–7.PubMed Van Cutsem E, Vervenne WL, Bennouna J, Humblet Y, Gill S, Van Laethem JL, et al. Phase III trial of bevacizumab in combination with gemcitabine and erlotinib in patients with metastatic pancreatic cancer. J Clin Oncol. 2009;27:2231–7.PubMed
77.
Zurück zum Zitat Spano JP, Chodkiewicz C, Maurel J, Wong R, Wasan H, Barone C, et al. Efficacy of gemcitabine plus axitinib compared with gemcitabine alone in patients with advanced pancreatic cancer: an open-label randomised phase II study. Lancet. 2008;371:2101–8.PubMed Spano JP, Chodkiewicz C, Maurel J, Wong R, Wasan H, Barone C, et al. Efficacy of gemcitabine plus axitinib compared with gemcitabine alone in patients with advanced pancreatic cancer: an open-label randomised phase II study. Lancet. 2008;371:2101–8.PubMed
78.
Zurück zum Zitat Kindler HL, Ioka T, Richel DJ, Bennouna J, Letourneau R, Okusaka T, et al. Axitinib plus gemcitabine versus placebo plus gemcitabine in patients with advanced pancreatic adenocarcinoma: a double-blind randomised phase 3 study. Lancet Oncol. 2011;12:256–62.PubMed Kindler HL, Ioka T, Richel DJ, Bennouna J, Letourneau R, Okusaka T, et al. Axitinib plus gemcitabine versus placebo plus gemcitabine in patients with advanced pancreatic adenocarcinoma: a double-blind randomised phase 3 study. Lancet Oncol. 2011;12:256–62.PubMed
79.
Zurück zum Zitat Rougier P, Riess H, Manges R, Karasek P, Humblet Y, Barone C, et al. Randomised, placebo-controlled, double-blind, parallel-group phase III study evaluating aflibercept in patients receiving first-line treatment with gemcitabine for metastatic pancreatic cancer. Eur J Cancer. 2013;49:2633–42.PubMed Rougier P, Riess H, Manges R, Karasek P, Humblet Y, Barone C, et al. Randomised, placebo-controlled, double-blind, parallel-group phase III study evaluating aflibercept in patients receiving first-line treatment with gemcitabine for metastatic pancreatic cancer. Eur J Cancer. 2013;49:2633–42.PubMed
80.
Zurück zum Zitat Goncalves A, Gilabert M, Francois E, Dahan L, Perrier H, Lamy R, et al. BAYPAN study: a double-blind phase III randomized trial comparing gemcitabine plus sorafenib and gemcitabine plus placebo in patients with advanced pancreatic cancer. Ann Oncol. 2012;23:2799–805.PubMed Goncalves A, Gilabert M, Francois E, Dahan L, Perrier H, Lamy R, et al. BAYPAN study: a double-blind phase III randomized trial comparing gemcitabine plus sorafenib and gemcitabine plus placebo in patients with advanced pancreatic cancer. Ann Oncol. 2012;23:2799–805.PubMed
81.
Zurück zum Zitat Deplanque G, Demarchi M, Hebbar M, Flynn PJ, Melichar B, Atkins J, et al. Masitinib in nonresectable pancreatic cancer: results of a phase III randomized placebo-controlled trial. J Clin Oncol. 2013;31(suppl):abstr 158. Deplanque G, Demarchi M, Hebbar M, Flynn PJ, Melichar B, Atkins J, et al. Masitinib in nonresectable pancreatic cancer: results of a phase III randomized placebo-controlled trial. J Clin Oncol. 2013;31(suppl):abstr 158.
82.
Zurück zum Zitat Kindler HL, Richards DA, Garbo LE, Garon EB, Stephenson Jr JJ, Rocha-Lima CM, et al. A randomized, placebo-controlled phase 2 study of ganitumab (AMG 479) or conatumumab (AMG 655) in combination with gemcitabine in patients with metastatic pancreatic cancer. Ann Oncol. 2012;23:2834–42.PubMed Kindler HL, Richards DA, Garbo LE, Garon EB, Stephenson Jr JJ, Rocha-Lima CM, et al. A randomized, placebo-controlled phase 2 study of ganitumab (AMG 479) or conatumumab (AMG 655) in combination with gemcitabine in patients with metastatic pancreatic cancer. Ann Oncol. 2012;23:2834–42.PubMed
83.
Zurück zum Zitat Philip PA, Goldman BH, Ramanathan RK, Lenz H, Lowy AM, Philip PA, et al. Phase I randomized phase II trial of gemcitabine, erlotinib, and cixutumumab versus gemcitabine plus erlotinib as first-line treatment in patients with metastatic pancreatic cancer (SWOG-0727). J Clin Oncol. 2012;30 suppl 4:abstr 198. Philip PA, Goldman BH, Ramanathan RK, Lenz H, Lowy AM, Philip PA, et al. Phase I randomized phase II trial of gemcitabine, erlotinib, and cixutumumab versus gemcitabine plus erlotinib as first-line treatment in patients with metastatic pancreatic cancer (SWOG-0727). J Clin Oncol. 2012;30 suppl 4:abstr 198.
84.
Zurück zum Zitat Akinleye A, Avvaru P, Furqan M, Song Y, Liu D. Phosphatidylinositol 3-kinase (PI3K) inhibitors as cancer therapeutics. J Hematol Oncol. 2013;6:88.PubMedCentralPubMed Akinleye A, Avvaru P, Furqan M, Song Y, Liu D. Phosphatidylinositol 3-kinase (PI3K) inhibitors as cancer therapeutics. J Hematol Oncol. 2013;6:88.PubMedCentralPubMed
85.
Zurück zum Zitat Wolpin BM, Hezel AF, Abrams T, Blaszkowsky LS, Meyerhardt JA, Chan JA, et al. Oral mTOR inhibitor everolimus in patients with gemcitabine-refractory metastatic pancreatic cancer. J Clin Oncol. 2009;27:193–8.PubMedCentralPubMed Wolpin BM, Hezel AF, Abrams T, Blaszkowsky LS, Meyerhardt JA, Chan JA, et al. Oral mTOR inhibitor everolimus in patients with gemcitabine-refractory metastatic pancreatic cancer. J Clin Oncol. 2009;27:193–8.PubMedCentralPubMed
86.
Zurück zum Zitat Cui J, Jiang W, Wang S, Wang L, Xie K. Role of Wnt/β-catenin signaling in drug resistance of pancreatic cancer. Curr Pharm Des. 2012;18:2464–71.PubMed Cui J, Jiang W, Wang S, Wang L, Xie K. Role of Wnt/β-catenin signaling in drug resistance of pancreatic cancer. Curr Pharm Des. 2012;18:2464–71.PubMed
87.
Zurück zum Zitat Farhana L, Dawson MI, Das JK, Murshed F, Xia Z, Hadden TJ, et al. Adamantyl retinoid-related molecules induce apoptosis in pancreatic cancer cells by inhibiting IGF-1R and Wnt/β-catenin pathways. J Oncol. 2012;2012:796729.PubMedCentralPubMed Farhana L, Dawson MI, Das JK, Murshed F, Xia Z, Hadden TJ, et al. Adamantyl retinoid-related molecules induce apoptosis in pancreatic cancer cells by inhibiting IGF-1R and Wnt/β-catenin pathways. J Oncol. 2012;2012:796729.PubMedCentralPubMed
88.
Zurück zum Zitat Sjolund J, Manetopoulos C, Stockhausen MT, Axelson H. The Notch pathway in cancer: differentiation gone awry. Eur J C ancer. 2005;41:2620–9. Sjolund J, Manetopoulos C, Stockhausen MT, Axelson H. The Notch pathway in cancer: differentiation gone awry. Eur J C ancer. 2005;41:2620–9.
89.
Zurück zum Zitat Yuan X, Wu H, Han N, Xu H, Chu Q, Yu S, et al. Notch signaling and EMT in non-small cell lung cancer: biological significance and therapeutic application. J Hematol Oncol. 2014;7:87.PubMedCentralPubMed Yuan X, Wu H, Han N, Xu H, Chu Q, Yu S, et al. Notch signaling and EMT in non-small cell lung cancer: biological significance and therapeutic application. J Hematol Oncol. 2014;7:87.PubMedCentralPubMed
90.
Zurück zum Zitat Mizuma M, Rasheed ZA, Yabuuchi S, Omura N, Campbell NR, de Wilde RF, et al. The gamma secretase inhibitor MRK-003 attenuates pancreatic cancer growth in preclinical models. Mol Cancer Ther. 2012;11:1999–2009.PubMedCentralPubMed Mizuma M, Rasheed ZA, Yabuuchi S, Omura N, Campbell NR, de Wilde RF, et al. The gamma secretase inhibitor MRK-003 attenuates pancreatic cancer growth in preclinical models. Mol Cancer Ther. 2012;11:1999–2009.PubMedCentralPubMed
91.
Zurück zum Zitat Morton JP, Mongeau ME, Klimstra DS, Morris JP, Lee YC, Kawaguchi Y, et al. Sonic hedgehog acts at multiple stages during pancreatic tumorigenesis. Proc Natl Acad Sci U S A. 2007;104:5103–8.PubMedCentralPubMed Morton JP, Mongeau ME, Klimstra DS, Morris JP, Lee YC, Kawaguchi Y, et al. Sonic hedgehog acts at multiple stages during pancreatic tumorigenesis. Proc Natl Acad Sci U S A. 2007;104:5103–8.PubMedCentralPubMed
92.
Zurück zum Zitat Jacobetz MA, Chan DS, Neesse A, Bapiro TE, Cook N, Frese KK, et al. Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer. Gut. 2013;62:112–20.PubMedCentralPubMed Jacobetz MA, Chan DS, Neesse A, Bapiro TE, Cook N, Frese KK, et al. Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer. Gut. 2013;62:112–20.PubMedCentralPubMed
93.
Zurück zum Zitat Hingorani SR, Harris WP, Beck JT, Berdov BA, Wagner SA, Pshevlotsky EM, et al. A phase Ib study of gemcitabine plus PEGPH20 (pegylated recombinant human hyaluronidase) in patients with stage IV previously untreated pancreatic cancer. J Clin Oncol. 2013;31(suppl):abstr 4010. Hingorani SR, Harris WP, Beck JT, Berdov BA, Wagner SA, Pshevlotsky EM, et al. A phase Ib study of gemcitabine plus PEGPH20 (pegylated recombinant human hyaluronidase) in patients with stage IV previously untreated pancreatic cancer. J Clin Oncol. 2013;31(suppl):abstr 4010.
94.
Zurück zum Zitat Bramhall SR, Schulz J, Nemunaitis J, Brown PD, Baillet M, Buckels JA. A double-blind placebo-controlled, randomised study comparing gemcitabine and marimastat with gemcitabine and placebo as first line therapy in patients with advanced pancreatic cancer. Br J Cancer. 2002;87:161–7.PubMedCentralPubMed Bramhall SR, Schulz J, Nemunaitis J, Brown PD, Baillet M, Buckels JA. A double-blind placebo-controlled, randomised study comparing gemcitabine and marimastat with gemcitabine and placebo as first line therapy in patients with advanced pancreatic cancer. Br J Cancer. 2002;87:161–7.PubMedCentralPubMed
95.
Zurück zum Zitat Moore MJ, Hamm J, Dancey J, Eisenberg PD, Dagenais M, Fields A, et al. Comparison of gemcitabine versus the matrix metalloproteinase inhibitor BAY 12–9566 in patients with advanced or metastatic adenocarcinoma of the pancreas: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol. 2003;21:3296–302.PubMed Moore MJ, Hamm J, Dancey J, Eisenberg PD, Dagenais M, Fields A, et al. Comparison of gemcitabine versus the matrix metalloproteinase inhibitor BAY 12–9566 in patients with advanced or metastatic adenocarcinoma of the pancreas: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol. 2003;21:3296–302.PubMed
96.
Zurück zum Zitat Borad MJ, Reddy SG, Bahary N, Uronis HE, Sigal D, Cohn AL, Schelman WR, Stephenson J, Jr., Chiorean EG, Rosen PJ, et al.: Randomized phase II trial of gemcitabine plus TH-302 versus gemcitabine in patients with advanced pancreatic cancer. J Clin Oncol 2014. Borad MJ, Reddy SG, Bahary N, Uronis HE, Sigal D, Cohn AL, Schelman WR, Stephenson J, Jr., Chiorean EG, Rosen PJ, et al.: Randomized phase II trial of gemcitabine plus TH-302 versus gemcitabine in patients with advanced pancreatic cancer. J Clin Oncol 2014.
97.
Zurück zum Zitat Fuxe J, Karlsson MC. TGF-β-induced epithelial-mesenchymal transition: a link between cancer and inflammation. Semin Cancer Biol. 2012;22:455–61.PubMed Fuxe J, Karlsson MC. TGF-β-induced epithelial-mesenchymal transition: a link between cancer and inflammation. Semin Cancer Biol. 2012;22:455–61.PubMed
98.
Zurück zum Zitat Oettle H, Seufferlein T, Luger T, Schmid RM, Wichert GV, Endlicher E, et al. Final results of a phase I/II study in patients with pancreatic cancer, malignant melanoma, and colorectal carcinoma with trabedersen. J Clin Oncol. 2012;30(suppl):abstr 4034. Oettle H, Seufferlein T, Luger T, Schmid RM, Wichert GV, Endlicher E, et al. Final results of a phase I/II study in patients with pancreatic cancer, malignant melanoma, and colorectal carcinoma with trabedersen. J Clin Oncol. 2012;30(suppl):abstr 4034.
99.
Zurück zum Zitat Kisfalvi K, Eibl G, Sinnett-Smith J, Rozengurt E. Metformin disrupts crosstalk between G protein-coupled receptor and insulin receptor signaling systems and inhibits pancreatic cancer growth. Cancer Res. 2009;69:6539–45.PubMedCentralPubMed Kisfalvi K, Eibl G, Sinnett-Smith J, Rozengurt E. Metformin disrupts crosstalk between G protein-coupled receptor and insulin receptor signaling systems and inhibits pancreatic cancer growth. Cancer Res. 2009;69:6539–45.PubMedCentralPubMed
100.
Zurück zum Zitat Li D, Yeung SC, Hassan MM, Konopleva M, Abbruzzese JL. Antidiabetic therapies affect risk of pancreatic cancer. Gastroenterology. 2009;137:482–8.PubMedCentralPubMed Li D, Yeung SC, Hassan MM, Konopleva M, Abbruzzese JL. Antidiabetic therapies affect risk of pancreatic cancer. Gastroenterology. 2009;137:482–8.PubMedCentralPubMed
101.
Zurück zum Zitat Sadeghi N, Abbruzzese JL, Yeung SC, Hassan M, Li D. Metformin use is associated with better survival of diabetic patients with pancreatic cancer. Clin Cancer Res. 2012;18:2905–12.PubMedCentralPubMed Sadeghi N, Abbruzzese JL, Yeung SC, Hassan M, Li D. Metformin use is associated with better survival of diabetic patients with pancreatic cancer. Clin Cancer Res. 2012;18:2905–12.PubMedCentralPubMed
102.
Zurück zum Zitat Hwang AL, Haynes K, Hwang WT, Yang YX. Metformin and survival in pancreatic cancer: a retrospective cohort study. Pancreas. 2013;42:1054–9.PubMedCentralPubMed Hwang AL, Haynes K, Hwang WT, Yang YX. Metformin and survival in pancreatic cancer: a retrospective cohort study. Pancreas. 2013;42:1054–9.PubMedCentralPubMed
103.
Zurück zum Zitat Lu Y, Bellgrau D, Dwyer-Nield LD, Malkinson AM, Duke RC, Rodell TC, et al. Mutation-selective tumor remission with Ras-targeted, whole yeast-based immunotherapy. Cancer Res. 2004;64:5084–8.PubMed Lu Y, Bellgrau D, Dwyer-Nield LD, Malkinson AM, Duke RC, Rodell TC, et al. Mutation-selective tumor remission with Ras-targeted, whole yeast-based immunotherapy. Cancer Res. 2004;64:5084–8.PubMed
104.
Zurück zum Zitat Strong JE, Coffey MC, Tang D, Sabinin P, Lee PW. The molecular basis of viral oncolysis: usurpation of the Ras signaling pathway by reovirus. EMBO J. 1998;17:3351–62.PubMedCentralPubMed Strong JE, Coffey MC, Tang D, Sabinin P, Lee PW. The molecular basis of viral oncolysis: usurpation of the Ras signaling pathway by reovirus. EMBO J. 1998;17:3351–62.PubMedCentralPubMed
105.
Zurück zum Zitat Morse MA, Nair SK, Boczkowski D, Tyler D, Hurwitz HI, Proia A, et al. The feasibility and safety of immunotherapy with dendritic cells loaded with CEA mRNA following neoadjuvant chemoradiotherapy and resection of pancreatic cancer. Int J Gastrointest Cancer. 2002;32:1–6.PubMed Morse MA, Nair SK, Boczkowski D, Tyler D, Hurwitz HI, Proia A, et al. The feasibility and safety of immunotherapy with dendritic cells loaded with CEA mRNA following neoadjuvant chemoradiotherapy and resection of pancreatic cancer. Int J Gastrointest Cancer. 2002;32:1–6.PubMed
106.
Zurück zum Zitat Ramanathan RK, Lee KM, McKolanis J, Hitbold E, Schraut W, Moser AJ, et al. Phase I study of a MUC1 vaccine composed of different doses of MUC1 peptide with SB-AS2 adjuvant in resected and locally advanced pancreatic cancer. Cancer Immunol Immunother. 2005;54:254–64.PubMed Ramanathan RK, Lee KM, McKolanis J, Hitbold E, Schraut W, Moser AJ, et al. Phase I study of a MUC1 vaccine composed of different doses of MUC1 peptide with SB-AS2 adjuvant in resected and locally advanced pancreatic cancer. Cancer Immunol Immunother. 2005;54:254–64.PubMed
107.
Zurück zum Zitat Rong Y, Qin X, Jin D, Lou W, Wu L, Wang D, et al. A phase I pilot trial of MUC1-peptide-pulsed dendritic cells in the treatment of advanced pancreatic cancer. Clin Exp Med. 2012;12:173–80.PubMed Rong Y, Qin X, Jin D, Lou W, Wu L, Wang D, et al. A phase I pilot trial of MUC1-peptide-pulsed dendritic cells in the treatment of advanced pancreatic cancer. Clin Exp Med. 2012;12:173–80.PubMed
108.
Zurück zum Zitat Maki RG, Livingston PO, Lewis JJ, Janetzki S, Klimstra D, Desantis D, et al. A phase I pilot study of autologous heat shock protein vaccine HSPPC-96 in patients with resected pancreatic adenocarcinoma. Dig Dis Sci. 2007;52:1964–72.PubMed Maki RG, Livingston PO, Lewis JJ, Janetzki S, Klimstra D, Desantis D, et al. A phase I pilot study of autologous heat shock protein vaccine HSPPC-96 in patients with resected pancreatic adenocarcinoma. Dig Dis Sci. 2007;52:1964–72.PubMed
109.
Zurück zum Zitat Hardacre JM, Mulcahy M, Small W, Talamonti M, Obel J, Krishnamurthi S, et al. Addition of algenpantucel-L immunotherapy to standard adjuvant therapy for pancreatic cancer: a phase 2 study. J Gastrointest Surg. 2013;17:94–100. discussion p 100–101.PubMed Hardacre JM, Mulcahy M, Small W, Talamonti M, Obel J, Krishnamurthi S, et al. Addition of algenpantucel-L immunotherapy to standard adjuvant therapy for pancreatic cancer: a phase 2 study. J Gastrointest Surg. 2013;17:94–100. discussion p 100–101.PubMed
110.
Zurück zum Zitat Bernhardt SL, Gjertsen MK, Trachsel S, Moller M, Eriksen JA, Meo M, et al. Telomerase peptide vaccination of patients with non-resectable pancreatic cancer: a dose escalating phase I/II study. Br J Cancer. 2006;95:1474–82.PubMedCentralPubMed Bernhardt SL, Gjertsen MK, Trachsel S, Moller M, Eriksen JA, Meo M, et al. Telomerase peptide vaccination of patients with non-resectable pancreatic cancer: a dose escalating phase I/II study. Br J Cancer. 2006;95:1474–82.PubMedCentralPubMed
111.
Zurück zum Zitat Gilliam AD, Broome P, Topuzov EG, Garin AM, Pulay I, Humphreys J, et al. An international multicenter randomized controlled trial of G17DT in patients with pancreatic cancer. Pancreas. 2012;41:374–9.PubMed Gilliam AD, Broome P, Topuzov EG, Garin AM, Pulay I, Humphreys J, et al. An international multicenter randomized controlled trial of G17DT in patients with pancreatic cancer. Pancreas. 2012;41:374–9.PubMed
112.
Zurück zum Zitat Wolpin BM, O’Reilly EM, Ko YJ, Blaszkowsky LS, Rarick M, Rocha-Lima CM, et al. Global, multicenter, randomized, phase II trial of gemcitabine and gemcitabine plus AGS-1C4D4 in patients with previously untreated, metastatic pancreatic cancer. Ann Oncol. 2013;24:1792–801.PubMedCentralPubMed Wolpin BM, O’Reilly EM, Ko YJ, Blaszkowsky LS, Rarick M, Rocha-Lima CM, et al. Global, multicenter, randomized, phase II trial of gemcitabine and gemcitabine plus AGS-1C4D4 in patients with previously untreated, metastatic pancreatic cancer. Ann Oncol. 2013;24:1792–801.PubMedCentralPubMed
113.
Zurück zum Zitat Beatty GL, Chiorean EG, Fishman MP, Saboury B, Teitelbaum UR, Sun W, et al. CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science. 2011;331:1612–6.PubMedCentralPubMed Beatty GL, Chiorean EG, Fishman MP, Saboury B, Teitelbaum UR, Sun W, et al. CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science. 2011;331:1612–6.PubMedCentralPubMed
114.
Zurück zum Zitat Royal RE, Levy C, Turner K, Mathur A, Hughes M, Kammula US, et al. Phase 2 trial of single agent ipilimumab (anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma. J Immunother. 2010;33:828–33.PubMed Royal RE, Levy C, Turner K, Mathur A, Hughes M, Kammula US, et al. Phase 2 trial of single agent ipilimumab (anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma. J Immunother. 2010;33:828–33.PubMed
115.
Zurück zum Zitat Ocean AJ, Pennington KL, Guarino MJ, Sheikh A, Bekaii-Saab T, Serafini AN, et al. Fractionated radioimmunotherapy with (90) Y-clivatuzumab tetraxetan and low-dose gemcitabine is active in advanced pancreatic cancer: A phase 1 trial. Cancer. 2012;118:5497–506.PubMedCentralPubMed Ocean AJ, Pennington KL, Guarino MJ, Sheikh A, Bekaii-Saab T, Serafini AN, et al. Fractionated radioimmunotherapy with (90) Y-clivatuzumab tetraxetan and low-dose gemcitabine is active in advanced pancreatic cancer: A phase 1 trial. Cancer. 2012;118:5497–506.PubMedCentralPubMed
116.
Zurück zum Zitat Chmielewski M, Hahn O, Rappl G, Nowak M, Schmidt-Wolf IH, Hombach AA, et al. T cells that target carcinoembryonic antigen eradicate orthotopic pancreatic carcinomas without inducing autoimmune colitis in mice. Gastroenterology. 2012;143:1095–107.PubMed Chmielewski M, Hahn O, Rappl G, Nowak M, Schmidt-Wolf IH, Hombach AA, et al. T cells that target carcinoembryonic antigen eradicate orthotopic pancreatic carcinomas without inducing autoimmune colitis in mice. Gastroenterology. 2012;143:1095–107.PubMed
117.
Zurück zum Zitat Ko AH, Tempero MA, Shan Y, Su W, Lin Y, Dito E, et al. A multinational phase II study of PEP02 (liposome irinotecan) for patients with gemcitabine-refractory metastatic pancreatic cancer. J Clin Oncol. 2011;29(suppl):abstr 4069. Ko AH, Tempero MA, Shan Y, Su W, Lin Y, Dito E, et al. A multinational phase II study of PEP02 (liposome irinotecan) for patients with gemcitabine-refractory metastatic pancreatic cancer. J Clin Oncol. 2011;29(suppl):abstr 4069.
118.
Zurück zum Zitat Ozaka M, Matsumura Y, Ishii H, Omuro Y, Itoi T, Mouri H, et al. Randomized phase II study of gemcitabine and S-1 combination versus gemcitabine alone in the treatment of unresectable advanced pancreatic cancer (Japan Clinical Cancer Research Organization PC-01 study). Cancer Chemother Pharmacol. 2012;69:1197–204.PubMed Ozaka M, Matsumura Y, Ishii H, Omuro Y, Itoi T, Mouri H, et al. Randomized phase II study of gemcitabine and S-1 combination versus gemcitabine alone in the treatment of unresectable advanced pancreatic cancer (Japan Clinical Cancer Research Organization PC-01 study). Cancer Chemother Pharmacol. 2012;69:1197–204.PubMed
119.
Zurück zum Zitat Maeda A, Boku N, Fukutomi A, Kondo S, Kinoshita T, Nagino M, et al. Randomized phase III trial of adjuvant chemotherapy with gemcitabine versus S-1 in patients with resected pancreatic cancer: Japan Adjuvant Study Group of Pancreatic Cancer (JASPAC-01). Jpn J Clin Oncol. 2008;38:227–9.PubMed Maeda A, Boku N, Fukutomi A, Kondo S, Kinoshita T, Nagino M, et al. Randomized phase III trial of adjuvant chemotherapy with gemcitabine versus S-1 in patients with resected pancreatic cancer: Japan Adjuvant Study Group of Pancreatic Cancer (JASPAC-01). Jpn J Clin Oncol. 2008;38:227–9.PubMed
120.
Zurück zum Zitat Fukutomi A, Uesaka K, Boku N, Kanemoto H, Konishi M, Matsumoto I, et al. JASPAC 01: Randomized phase III trial of adjuvant chemotherapy with gemcitabine versus S-1 for patients with resected pancreatic cancer. J Clin Oncol. 2013;31(suppl):abstr 4008. Fukutomi A, Uesaka K, Boku N, Kanemoto H, Konishi M, Matsumoto I, et al. JASPAC 01: Randomized phase III trial of adjuvant chemotherapy with gemcitabine versus S-1 for patients with resected pancreatic cancer. J Clin Oncol. 2013;31(suppl):abstr 4008.
121.
Zurück zum Zitat Giovannetti E, Del Tacca M, Mey V, Funel N, Nannizzi S, Ricci S, et al. Transcription analysis of human equilibrative nucleoside transporter-1 predicts survival in pancreas cancer patients treated with gemcitabine. Cancer Res. 2006;66:3928–35.PubMed Giovannetti E, Del Tacca M, Mey V, Funel N, Nannizzi S, Ricci S, et al. Transcription analysis of human equilibrative nucleoside transporter-1 predicts survival in pancreas cancer patients treated with gemcitabine. Cancer Res. 2006;66:3928–35.PubMed
122.
Zurück zum Zitat Poplin E, Wasan H, Rolfe L, Raponi M, Ikdahl T, Bondarenko I, et al. Randomized, multicenter, phase II study of CO-101 versus gemcitabine in patients with metastatic pancreatic ductal adenocarcinoma: including a prospective evaluation of the role of hENT1 in gemcitabine or CO-101 sensitivity. J Clin Oncol. 2013;31:4453–61.PubMed Poplin E, Wasan H, Rolfe L, Raponi M, Ikdahl T, Bondarenko I, et al. Randomized, multicenter, phase II study of CO-101 versus gemcitabine in patients with metastatic pancreatic ductal adenocarcinoma: including a prospective evaluation of the role of hENT1 in gemcitabine or CO-101 sensitivity. J Clin Oncol. 2013;31:4453–61.PubMed
123.
Zurück zum Zitat Xie H, Jiang W, Jiang J, Wang Y, Kim R, Liu X, et al. Predictive and prognostic roles of ribonucleotide reductase M1 in resectable pancreatic adenocarcinoma. Cancer. 2013;119:173–81.PubMed Xie H, Jiang W, Jiang J, Wang Y, Kim R, Liu X, et al. Predictive and prognostic roles of ribonucleotide reductase M1 in resectable pancreatic adenocarcinoma. Cancer. 2013;119:173–81.PubMed
124.
Zurück zum Zitat Nakano Y, Tanno S, Koizumi K, Nishikawa T, Nakamura K, Minoguchi M, et al. Gemcitabine chemoresistance and molecular markers associated with gemcitabine transport and metabolism in human pancreatic cancer cells. Br J Cancer. 2007;96:457–63.PubMedCentralPubMed Nakano Y, Tanno S, Koizumi K, Nishikawa T, Nakamura K, Minoguchi M, et al. Gemcitabine chemoresistance and molecular markers associated with gemcitabine transport and metabolism in human pancreatic cancer cells. Br J Cancer. 2007;96:457–63.PubMedCentralPubMed
125.
Zurück zum Zitat Lambrechts D, Claes B, Delmar P, Reumers J, Mazzone M, Yesilyurt BT, et al. VEGF pathway genetic variants as biomarkers of treatment outcome with bevacizumab: an analysis of data from the AViTA and AVOREN randomised trials. Lancet Oncol. 2012;13:724–33.PubMed Lambrechts D, Claes B, Delmar P, Reumers J, Mazzone M, Yesilyurt BT, et al. VEGF pathway genetic variants as biomarkers of treatment outcome with bevacizumab: an analysis of data from the AViTA and AVOREN randomised trials. Lancet Oncol. 2012;13:724–33.PubMed
126.
Zurück zum Zitat Yu KH, Ricigliano M, Hidalgo M, Abou-Alfa GK, Lowery MA, Saltz LB, et al. Pharmacogenomic modeling of circulating tumor and invasive cells for prediction of chemotherapy response and resistance in pancreatic cancer. Clin Cancer Res. 2014;20:5281–9.PubMed Yu KH, Ricigliano M, Hidalgo M, Abou-Alfa GK, Lowery MA, Saltz LB, et al. Pharmacogenomic modeling of circulating tumor and invasive cells for prediction of chemotherapy response and resistance in pancreatic cancer. Clin Cancer Res. 2014;20:5281–9.PubMed
Metadaten
Titel
Novel approaches in the management of pancreatic ductal adenocarcinoma: potential promises for the future
verfasst von
Gaurav Goel
Weijing Sun
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
Journal of Hematology & Oncology / Ausgabe 1/2015
Elektronische ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-015-0141-5

Weitere Artikel der Ausgabe 1/2015

Journal of Hematology & Oncology 1/2015 Zur Ausgabe

Update Onkologie

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