Editorial Open Access
Copyright ©The Author(s) 2015. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Oncol. Oct 15, 2015; 7(10): 172-177
Published online Oct 15, 2015. doi: 10.4251/wjgo.v7.i10.172
Targeted therapies for pancreatic adenocarcinoma: Where do we stand, how far can we go?
Dimitra Grapsa, Konstantinos Syrigos, Oncology Unit, 3rd Department of Medicine, “Sotiria” General Hospital, Athens University School of Medicine, 11527 Athens, Greece
Muhammad Wasif Saif, Tufts Cancer Center, Tufts University School of Medicine, Boston, MA 02111, United States
Author contributions: Grapsa D drafted the manuscript; Saif MW and Syrigos K revised the manuscript for intellectual content.
Conflict-of-interest statement: The authors declare that they have no relevant conflicts of interest.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Konstantinos Syrigos, MD, PhD, Professor, Head, Oncology Unit, 3rd Department of Medicine, “Sotiria” General Hospital, Athens University School of Medicine, Mesogion 152, 11527 Athens, Greece. knsyrigos@usa.net
Telephone: +30-210-7475034 Fax: +30-210-7781035
Received: May 26, 2015
Peer-review started: May 28, 2015
First decision: June 18, 2015
Revised: July 10, 2015
Accepted: August 30, 2015
Article in press: August 31, 2015
Published online: October 15, 2015

Abstract

Pancreatic adenocarcinoma (usually referred to as pancreatic cancer) is a highly lethal and aggressive malignancy with a disease-related mortality almost equaling its incidence, and one of the most challenging cancers to treat. The notorious resistance of pancreatic cancer not only to conventional cytotoxic therapies but also to almost all targeted agents developed to date, continues to puzzle the oncological community and represents one of the biggest hurdles to reducing the death toll from this ominous disease. This editorial highlights the most important recent advances in preclinical and clinical research, with regards to targeted therapeutics for pancreatic cancer, outlines current challenges and provides an overview of potential future perspectives in this rapidly evolving field.

Key Words: Clinical, Cytotoxic chemotherapy, Pancreatic cancer, Preclinical, Targeted agents

Core tip: Expansion of our knowledge regarding the molecular basis of pancreatic cancer has facilitated the development of a significant number of innovative targeted therapies for this lethal disease. Almost all these agents have, nevertheless, failed to produce statistically significant survival benefits when tested in clinical trial settings; therefore, successful clinical translation of preclinical advancements in pancreatic cancer research has yet to be materialized. Future treatment options might include multi-targeted and individualized molecular therapies, ideally guided by patient-specific genomic data, in combination with conventional cytotoxic or other regimens.



INTRODUCTION

Despite recent advances in our understanding of the molecular mechanisms involved in the development and progression of pancreatic adenocarcinoma and an abundance of preclinical data suggesting the potential value of several targeted agents in treatment of this lethal disease, pancreatic cancer statistics remain grim and nearly the same as they were almost 30 years ago[1-3]. Pancreatic adenocarcinoma - usually referred to as “pancreatic cancer” - currently ranks as the fourth most frequent cause of cancer-related death among males and the fifth among females in the Western world, and is sadly expected to rise to the second leading position within the next decade[3,4]. Median survival is 4 to 6 mo following diagnosis while long term (5-year) survival rates do not exceed 4%-5%, for all stages combined[5]. The only treatment option with a curative potential is surgery, but less than 20% of patients are eligible for this approach, while the survival rates are poor (25%-30%) even among those with localized node-negative disease undergoing complete surgical resection and adjuvant chemotherapy[6].

This dismal clinical record inevitably leads to the following questions: Why have we failed thus far to reduce the death toll from this lethal disease? And, most importantly, what can we do to widen the range of available treatment options and improve their clinical effectiveness?

PRECLINICAL AND CLINICAL DATA: DISCREPANCY PREVAILS

In the preclinical arena of pancreatic cancer research the picture is much rosier; a significant and rather rapidly expanding number of different targeted agents have shown considerable efficacy in controlling growth of human pancreatic cancer cells, both in vitro and in vivo, and prolonging survival of pancreatic cancer models, as summarized in recent reviews on this topic[5-11]. This rather extensive armamentarium includes, among others, inhibitors of epidermal growth factor receptor (EGFR)[12,13], human epidermal growth factor receptor 2 (HER2)[14,15], vascular endothelial growth factor (VEGF) and VEGF receptors[16], insulin-like growth factor receptor[17-19], KRAS and its downstream effectors (mainly mitogen-activated protein kinase)[20,21], the developmental Wnt, Hedgehog and Notch signaling pathways[22-24], as well as reagents targeting the tumor extracellular matrix/stromal microenvironment or molecules overexpressed in the surface of pancreatic cancer cells (i.e., mesothelin, carcinoembryonic antigen, epithelial cell adhesion molecule, MUC1)[25-29]. Dual-agent and multi-kinase molecular targeting represent additional exciting therapeutic possibilities and are gaining increasing research attention and popularity[30-34]. Alternative approaches, such as targeting the cellular process of autophagy - which plays a key role in the development and progression of malignancy or combined targeting of oncogene-driven signaling pathways and critical energy sources (such as mitochondrial respiration) of the subpopulation of dormant tumor cells surviving oncogene ablation, have also been studied as potential treatment options in pancreatic cancer, but are still in their infancy[7,35,36]. Interestingly, in accordance with increasing data suggesting potential preventive and therapeutic effects of aspirin and non-steroidal inflammatory drugs in gastrointestinal cancers, particularly colorectal cancer[37,38], aspirin is being explored as a targeted therapeutic agent for pancreatic cancer as well[39,40]. As shown in recent preclinical studies, aspirin, either alone or in combination with the antidiabetic drug metformin, may inhibit pancreatic cancer cell growth, counteract desmoplasia and cancer stem cell features and enhance the therapeutic efficacy of cytotoxic agents-such as gemcitabine- in pancreatic cancer by sensitizing pancreatic cancer cells to chemotherapy-mediated cytotoxicity[41-43].

Modified cytotoxic agents, mainly including nab-paclitaxel (paclitaxel conjugated with albumin nanoparticles) or other nanovector-based anticancer drugs, such as cationic liposome encapsulated paclitaxel (EndoTAGTM-1) or liposomal doxorubicin, cisplatin and irinotecan, have been recently developed using sophisticated nanotechnology and tested in preclinical studies of pancreatic cancer, with some encouraging results[7,44-49]. These selective drug formulations offer the advantage of improved drug delivery to the tumor tissue and selective targeting via binding to tumor-associated receptors or macromolecules, thus positively modulating the pharmacokinetics and therapeutic index of cytotoxic chemotherapy[44]. Nab-paclitaxel, in particular, can bind to SPARC (secreted protein acid and rich in cysteine), an extracellular matrix protein which is frequently overexpressed in pancreatic adenocarcinomas[10,50,51], and, presumably, result in depletion of desmoplastic tumor stroma and an increase in vascularization, thus enhancing transvascular transport and delivery of cytotoxic agents to tumor cells[52].

The overwhelming majority of the abovementioned targeted therapies have, nevertheless, failed to demonstrate any statistically significant efficacy in clinical trials of pancreatic cancer patients; the EGFR and VEGF monoclonal antibodies cetuximab and bevacizumab, respectively, and the multikinase inhibitor sorafenib are representative examples of once-promising targeted agents who failed to produce a statistically significant improvement of survival when used in combination with gemcitabine vs gemcitabine alone in phase III randomized trials[53-55]. Hence, successful translation of our otherwise encouraging preclinical achievements into tangible clinical benefit remains an elusive goal. Two notable exceptions, though, leave some room for optimism. Erlotinib, an EGFR tyrosine kinase inhibitor which was United States Food and Drug Administration (FDA)-approved in 2007 for the treatment of advanced pancreatic cancer, is the first targeted agent which succeeded in producing a significant-albeit modest-survival benefit when administered as an adjunct to gemcitabine, especially among patients experiencing erlotinib-induced skin rash[7,56]; still, given the marginal effect of erlotinib on survival and its unclear therapeutic value in localized, resectable disease this drug has yet to be widely adopted as standard of care in routine clinical practice[8,10]. Based on the results of the recent phase III Metastatic Pancreatic Adenocarcinoma Clinical Trial[57] of nab-paclitaxel and gemcitabine combination vs gemcitabine alone in 861 patients with metastatic pancreatic cancer, showing a statistically significant survival benefit (as regards overall, progression-free and 1-year survival) in the combinatorial arm, nab-paclitaxel was also approved by the FDA in 2013 to be administered in combination with gemcitabine as first-line therapy for metastatic pancreatic cancer.

CONCLUSION

Considering all available evidence, as summarized above, we should first acknowledge that, although some revolutionary progress has indeed been achieved on the theoretical front, preclinical enthusiasm has been severely tempered by clinical disappointment. The reasons behind this discrepancy remain largely unknown and can only be speculated upon at this point. Resistance of pancreatic cancer to anticancer drugs, including both standard cytotoxic and novel targeted agents, is often attributed to the abundant, dense, fibroinflammatory stroma surrounding pancreatic tumor tissue, which is believed to function as a barrier to efficient delivery of drug formulations to their target tumor cells by restricting blood supply and limiting diffusion of large molecules[10,58,59]. The high genetic heterogeneity and complexity of pancreatic cancer may also explain why targeting a specific mutation in a tumor containing 63 genetic alterations on average -as shown by previous genomic studies[22,60] - or “randomly combining drugs in the hope of achieving a better outcome in an unselected patient population”[10], may be doomed to fail.

Hopefully, the results of ongoing clinical trials on current and emerging targeted therapeutics, including, among others, the anti-EGFR and anti-HER2/neu monoclonal antibodies nimotuzumab (NCT02395016) and trastuzumab (NCT01204372), respectively, the hedgehog inhibitors vismodegib (NCT01195415) and LDE225 (NCT01485744) and agents targeting the Notch pathway, such as the gamma-secretase inhibitor MK-0752 (NCT01098344), may help bridge the gap between preclinical and clinical outcomes. The increasing advances in structural and functional genomics are also expected to further elucidate the key molecular events underlying pancreatic tumorigenesis and identify additional targets for novel agents. Based on data derived from global genomic analyses of pancreatic tumors, previous authors have suggested that agents broadly targeting downstream mediators of critical physiologic functions (such as neo-angiogenesis or cell cycle alterations) may be preferable to agents targeting specific mutated genes[60]. Most importantly, personalized genomic medicine, utilizing patient-specific genomic data for guidance of treatment selection in each individual patient, may not only significantly enhance the clinical efficacy of molecular targeted therapy but also reduce the burden of unnecessary - and potentially harmful-drugs.

As previously commented by Kleger et al[7], in a recent review article critically discussing current and future targeted therapies for pancreatic cancer, “smart drugs need smart applications”. Indeed, most experts concur that the latter applications should include multi-targeted and, ideally, individualized molecular therapies, in combination with conventional cytotoxic agents or other regimens (such as immunotherapy)[61], guided by reliable biomarkers of treatment response. Increased toxicity resulting from these combinatorial approaches as well as their cost-effectiveness and socioeconomic implications should, nevertheless, be carefully considered and may represent major limiting factors for their widespread use. In a disease as aggressive and lethal as pancreatic cancer, maintaining the highest possible quality of life for as long as possible is the most important target, and expectations should always be based on realistic goals.

Footnotes

P- Reviewer: Georgoulias V, Ogino S S- Editor: Tian YL L- Editor: A E- Editor: Wu HL

References
1.  Tanaka S. Molecular Pathogenesis and Targeted Therapy of Pancreatic Cancer. Ann Surg Oncol. 2015;Mar 7; Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 30]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
2.  Vincent A, Herman J, Schulick R, Hruban RH, Goggins M. Pancreatic cancer. Lancet. 2011;378:607-620.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1720]  [Cited by in F6Publishing: 1893]  [Article Influence: 145.6]  [Reference Citation Analysis (3)]
3.  Krejs GJ. Pancreatic cancer: epidemiology and risk factors. Dig Dis. 2010;28:355-358.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 80]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
4.  Cardin DB, Berlin JD. Pancreas cancer on the rise: are we up to the challenge? J Natl Cancer Inst. 2013;105:1675-1676.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 11]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
5.  Saif MW. Pancreatic neoplasm in 2011: an update. JOP. 2011;12:316-321.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Teague A, Lim KH, Wang-Gillam A. Advanced pancreatic adenocarcinoma: a review of current treatment strategies and developing therapies. Ther Adv Med Oncol. 2015;7:68-84.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 109]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
7.  Kleger A, Perkhofer L, Seufferlein T. Smarter drugs emerging in pancreatic cancer therapy. Ann Oncol. 2014;25:1260-1270.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 62]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
8.  Antoniou G, Kountourakis P, Papadimitriou K, Vassiliou V, Papamichael D. Adjuvant therapy for resectable pancreatic adenocarcinoma: review of the current treatment approaches and future directions. Cancer Treat Rev. 2014;40:78-85.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 29]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
9.  Huang ZQ, Buchsbaum DJ. Monoclonal antibodies in the treatment of pancreatic cancer. Immunotherapy. 2009;1:223-229.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 30]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
10.  Oettle H. Progress in the knowledge and treatment of advanced pancreatic cancer: from benchside to bedside. Cancer Treat Rev. 2014;40:1039-1047.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 74]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
11.  Ozmen F, Şahin TT, Ozmen MM. Current adjuvant therapeutic approaches for pancreatic cancer. Adv Ther. 2015;32:42-56.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 6]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
12.  Huang ZQ, Buchsbaum DJ, Raisch KP, Bonner JA, Bland KI, Vickers SM. Differential responses by pancreatic carcinoma cell lines to prolonged exposure to Erbitux (IMC-C225) anti-EGFR antibody. J Surg Res. 2003;111:274-283.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 32]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
13.  Morgan MA, Parsels LA, Kollar LE, Normolle DP, Maybaum J, Lawrence TS. The combination of epidermal growth factor receptor inhibitors with gemcitabine and radiation in pancreatic cancer. Clin Cancer Res. 2008;14:5142-5149.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 105]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
14.  Saeki H, Yanoma S, Takemiya S, Sugimasa Y, Akaike M, Yukawa N, Rino Y, Imada T. Antitumor activity of a combination of trastuzumab (Herceptin) and oral fluoropyrimidine S-1 on human epidermal growth factor receptor 2-overexpressing pancreatic cancer. Oncol Rep. 2007;18:433-439.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Kimura K, Sawada T, Komatsu M, Inoue M, Muguruma K, Nishihara T, Yamashita Y, Yamada N, Ohira M, Hirakawa K. Antitumor effect of trastuzumab for pancreatic cancer with high HER-2 expression and enhancement of effect by combined therapy with gemcitabine. Clin Cancer Res. 2006;12:4925-4932.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 77]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
16.  Wilhelm SM, Carter C, Tang L, Wilkie D, McNabola A, Rong H, Chen C, Zhang X, Vincent P, McHugh M. BAY 43-9006 exhibits broad spectrum oral antitumor activity and targets the RAF/MEK/ERK pathway and receptor tyrosine kinases involved in tumor progression and angiogenesis. Cancer Res. 2004;64:7099-7109.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2966]  [Cited by in F6Publishing: 3039]  [Article Influence: 152.0]  [Reference Citation Analysis (0)]
17.  Neid M, Datta K, Stephan S, Khanna I, Pal S, Shaw L, White M, Mukhopadhyay D. Role of insulin receptor substrates and protein kinase C-zeta in vascular permeability factor/vascular endothelial growth factor expression in pancreatic cancer cells. J Biol Chem. 2004;279:3941-3948.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 45]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
18.  Liu W, Bloom DA, Cance WG, Kurenova EV, Golubovskaya VM, Hochwald SN. FAK and IGF-IR interact to provide survival signals in human pancreatic adenocarcinoma cells. Carcinogenesis. 2008;29:1096-1107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 83]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
19.  Rowinsky EK, Youssoufian H, Tonra JR, Solomon P, Burtrum D, Ludwig DL. IMC-A12, a human IgG1 monoclonal antibody to the insulin-like growth factor I receptor. Clin Cancer Res. 2007;13:5549s-5555s.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 126]  [Cited by in F6Publishing: 141]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
20.  End DW, Smets G, Todd AV, Applegate TL, Fuery CJ, Angibaud P, Venet M, Sanz G, Poignet H, Skrzat S. Characterization of the antitumor effects of the selective farnesyl protein transferase inhibitor R115777 in vivo and in vitro. Cancer Res. 2001;61:131-137.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Zimmermann G, Papke B, Ismail S, Vartak N, Chandra A, Hoffmann M, Hahn SA, Triola G, Wittinghofer A, Bastiaens PI. Small molecule inhibition of the KRAS-PDEδ interaction impairs oncogenic KRAS signalling. Nature. 2013;497:638-642.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 460]  [Cited by in F6Publishing: 461]  [Article Influence: 41.9]  [Reference Citation Analysis (0)]
22.  Witkiewicz AK, McMillan EA, Balaji U, Baek G, Lin WC, Mansour J, Mollaee M, Wagner KU, Koduru P, Yopp A. Whole-exome sequencing of pancreatic cancer defines genetic diversity and therapeutic targets. Nat Commun. 2015;6:6744.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 704]  [Cited by in F6Publishing: 763]  [Article Influence: 84.8]  [Reference Citation Analysis (0)]
23.  Olive KP, Jacobetz MA, Davidson CJ, Gopinathan A, McIntyre D, Honess D, Madhu B, Goldgraben MA, Caldwell ME, Allard D. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science. 2009;324:1457-1461.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2272]  [Cited by in F6Publishing: 2409]  [Article Influence: 160.6]  [Reference Citation Analysis (0)]
24.  Yen WC, Fischer MM, Axelrod F, Bond C, Cain J, Cancilla B, Henner WR, Meisner R, Sato A, Shah J. Targeting Notch signaling with a Notch2/Notch3 antagonist (tarextumab) inhibits tumor growth and decreases tumor-initiating cell frequency. Clin Cancer Res. 2015;21:2084-2095.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 396]  [Article Influence: 30.5]  [Reference Citation Analysis (0)]
25.  Golfier S, Kopitz C, Kahnert A, Heisler I, Schatz CA, Stelte-Ludwig B, Mayer-Bartschmid A, Unterschemmann K, Bruder S, Linden L. Anetumab ravtansine: a novel mesothelin-targeting antibody-drug conjugate cures tumors with heterogeneous target expression favored by bystander effect. Mol Cancer Ther. 2014;13:1537-1548.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 146]  [Reference Citation Analysis (0)]
26.  Showalter SL, Huang YH, Witkiewicz A, Costantino CL, Yeo CJ, Green JJ, Langer R, Anderson DG, Sawicki JA, Brody JR. Nanoparticulate delivery of diphtheria toxin DNA effectively kills Mesothelin expressing pancreatic cancer cells. Cancer Biol Ther. 2008;7:1584-1590.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 43]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
27.  Maawy AA, Hiroshima Y, Zhang Y, Heim R, Makings L, Garcia-Guzman M, Luiken GA, Kobayashi H, Hoffman RM, Bouvet M. Near infra-red photoimmunotherapy with anti-CEA-IR700 results in extensive tumor lysis and a significant decrease in tumor burden in orthotopic mouse models of pancreatic cancer. PLoS One. 2015;10:e0121989.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 47]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
28.  Lund K, Bostad M, Skarpen E, Braunagel M, Kiprijanov S, Krauss S, Duncan A, Høgset A, Selbo PK. The novel EpCAM-targeting monoclonal antibody 3-17I linked to saporin is highly cytotoxic after photochemical internalization in breast, pancreas and colon cancer cell lines. MAbs. 2014;6:1038-1050.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 32]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
29.  Tholey RM, Lal S, Jimbo M, Burkhart RA, Blanco FF, Cozzitorto JA, Eisenberg JD, Jiang W, Iacobuzio-Donahue CA, Witkiewicz AK. MUC1 Promoter-Driven DTA as a Targeted Therapeutic Strategy against Pancreatic Cancer. Mol Cancer Res. 2015;13:439-448.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
30.  Larbouret C, Robert B, Bascoul-Mollevi C, Penault-Llorca F, Ho-Pun-Cheung A, Morisseau S, Navarro-Teulon I, Mach JP, Pèlegrin A, Azria D. Combined cetuximab and trastuzumab are superior to gemcitabine in the treatment of human pancreatic carcinoma xenografts. Ann Oncol. 2010;21:98-103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 41]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
31.  Larbouret C, Gaborit N, Chardès T, Coelho M, Campigna E, Bascoul-Mollevi C, Mach JP, Azria D, Robert B, Pèlegrin A. In pancreatic carcinoma, dual EGFR/HER2 targeting with cetuximab/trastuzumab is more effective than treatment with trastuzumab/erlotinib or lapatinib alone: implication of receptors’ down-regulation and dimers’ disruption. Neoplasia. 2012;14:121-130.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 58]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
32.  Bianco C, Giovannetti E, Ciardiello F, Mey V, Nannizzi S, Tortora G, Troiani T, Pasqualetti F, Eckhardt G, de Liguoro M. Synergistic antitumor activity of ZD6474, an inhibitor of vascular endothelial growth factor receptor and epidermal growth factor receptor signaling, with gemcitabine and ionizing radiation against pancreatic cancer. Clin Cancer Res. 2006;12:7099-7107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 40]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
33.  Pan Y, Zheng M, Zhong L, Yang J, Zhou S, Qin Y, Xiang R, Chen Y, Yang SY. A preclinical evaluation of SKLB261, a multikinase inhibitor of EGFR/Src/VEGFR2, as a therapeutic agent against pancreatic cancer. Mol Cancer Ther. 2015;14:407-418.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
34.  Ulivi P, Arienti C, Zoli W, Scarsella M, Carloni S, Fabbri F, Tesei A, Chiadini E, Orlandi A, Passeri D. In vitro and in vivo antitumor efficacy of docetaxel and sorafenib combination in human pancreatic cancer cells. Curr Cancer Drug Targets. 2010;10:600-610.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 11]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
35.  Donadelli M, Dando I, Zaniboni T, Costanzo C, Dalla Pozza E, Scupoli MT, Scarpa A, Zappavigna S, Marra M, Abbruzzese A. Gemcitabine/cannabinoid combination triggers autophagy in pancreatic cancer cells through a ROS-mediated mechanism. Cell Death Dis. 2011;2:e152.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 150]  [Cited by in F6Publishing: 167]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
36.  Viale A, Pettazzoni P, Lyssiotis CA, Ying H, Sánchez N, Marchesini M, Carugo A, Green T, Seth S, Giuliani V. Oncogene ablation-resistant pancreatic cancer cells depend on mitochondrial function. Nature. 2014;514:628-632.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 887]  [Cited by in F6Publishing: 885]  [Article Influence: 88.5]  [Reference Citation Analysis (0)]
37.  Liao X, Lochhead P, Nishihara R, Morikawa T, Kuchiba A, Yamauchi M, Imamura Y, Qian ZR, Baba Y, Shima K. Aspirin use, tumor PIK3CA mutation, and colorectal-cancer survival. N Engl J Med. 2012;367:1596-1606.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 651]  [Cited by in F6Publishing: 626]  [Article Influence: 52.2]  [Reference Citation Analysis (0)]
38.  Domingo E, Church DN, Sieber O, Ramamoorthy R, Yanagisawa Y, Johnstone E, Davidson B, Kerr DJ, Tomlinson IP, Midgley R. Evaluation of PIK3CA mutation as a predictor of benefit from nonsteroidal anti-inflammatory drug therapy in colorectal cancer. J Clin Oncol. 2013;31:4297-4305.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 162]  [Cited by in F6Publishing: 156]  [Article Influence: 14.2]  [Reference Citation Analysis (0)]
39.  Shen X, Han L, Ma Z, Chen C, Duan W, Yu S, Li P, Zhang L, Li W, Xu Q. Aspirin: a potential therapeutic approach in pancreatic cancer. Curr Med Chem. 2013;20:4153-4162.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 9]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
40.  Yue W, Yang CS, DiPaola RS, Tan XL. Repurposing of metformin and aspirin by targeting AMPK-mTOR and inflammation for pancreatic cancer prevention and treatment. Cancer Prev Res (Phila). 2014;7:388-397.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 108]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
41.  Zhang Y, Liu L, Fan P, Bauer N, Gladkich J, Ryschich E, Bazhin AV, Giese NA, Strobel O, Hackert T. Aspirin counteracts cancer stem cell features, desmoplasia and gemcitabine resistance in pancreatic cancer. Oncotarget. 2015;6:9999-10015.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Ou YQ, Zhu Wb, Li Y, Qiu PX, Huang YJ, Xie J, He SM, Zheng XK, Leng TD, Xu D. Aspirin inhibits proliferation of gemcitabine-resistant human pancreatic cancer cells and augments gemcitabine-induced cytotoxicity. Acta Pharmacol Sin. 2010;31:73-80.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 21]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
43.  Yue W, Zheng X, Lin Y, Yang CS, Xu Q, Carpizo D, Huang H, DiPaola RS, Tan XL. Metformin combined with aspirin significantly inhibit pancreatic cancer cell growth in vitro and in vivo by suppressing anti-apoptotic proteins Mcl-1 and Bcl-2. Oncotarget. 2015;6:21208-21224.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Tsai CS, Park JW, Chen LT. Nanovector-based therapies in advanced pancreatic cancer. J Gastrointest Oncol. 2011;2:185-194.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 10]  [Reference Citation Analysis (0)]
45.  Neesse A, Frese KK, Chan DS, Bapiro TE, Howat WJ, Richards FM, Ellenrieder V, Jodrell DI, Tuveson DA. SPARC independent drug delivery and antitumour effects of nab-paclitaxel in genetically engineered mice. Gut. 2014;63:974-983.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 144]  [Article Influence: 14.4]  [Reference Citation Analysis (0)]
46.  Eichhorn ME, Ischenko I, Luedemann S, Strieth S, Papyan A, Werner A, Bohnenkamp H, Guenzi E, Preissler G, Michaelis U. Vascular targeting by EndoTAG-1 enhances therapeutic efficacy of conventional chemotherapy in lung and pancreatic cancer. Int J Cancer. 2010;126:1235-1245.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 60]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
47.  Mamidi RN, Weng S, Stellar S, Wang C, Yu N, Huang T, Tonelli AP, Kelley MF, Angiuoli A, Fung MC. Pharmacokinetics, efficacy and toxicity of different pegylated liposomal doxorubicin formulations in preclinical models: is a conventional bioequivalence approach sufficient to ensure therapeutic equivalence of pegylated liposomal doxorubicin products? Cancer Chemother Pharmacol. 2010;66:1173-1184.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 35]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
48.  Yoshida M, Takimoto R, Murase K, Sato Y, Hirakawa M, Tamura F, Sato T, Iyama S, Osuga T, Miyanishi K. Targeting anticancer drug delivery to pancreatic cancer cells using a fucose-bound nanoparticle approach. PLoS One. 2012;7:e39545.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 39]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
49.  Pal A, Khan S, Wang YF, Kamath N, Sarkar AK, Ahmad A, Sheikh S, Ali S, Carbonaro D, Zhang A. Preclinical safety, pharmacokinetics and antitumor efficacy profile of liposome-entrapped SN-38 formulation. Anticancer Res. 2005;25:331-341.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Neuzillet C, Tijeras-Raballand A, Cros J, Faivre S, Hammel P, Raymond E. Stromal expression of SPARC in pancreatic adenocarcinoma. Cancer Metastasis Rev. 2013;32:585-602.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 94]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
51.  Sinn M, Sinn BV, Striefler JK, Lindner JL, Stieler JM, Lohneis P, Bischoff S, Bläker H, Pelzer U, Bahra M. SPARC expression in resected pancreatic cancer patients treated with gemcitabine: results from the CONKO-001 study. Ann Oncol. 2014;25:1025-1032.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 65]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
52.  Al-Batran SE, Geissler M, Seufferlein T, Oettle H. Nab-paclitaxel for metastatic pancreatic cancer: clinical outcomes and potential mechanisms of action. Oncol Res Treat. 2014;37:128-134.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 24]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
53.  Philip PA, Benedetti J, Corless CL, Wong R, O’Reilly EM, Flynn PJ, Rowland KM, Atkins JN, Mirtsching BC, Rivkin SE. Phase III study comparing gemcitabine plus cetuximab versus gemcitabine in patients with advanced pancreatic adenocarcinoma: Southwest Oncology Group-directed intergroup trial S0205. J Clin Oncol. 2010;28:3605-3610.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 446]  [Cited by in F6Publishing: 478]  [Article Influence: 34.1]  [Reference Citation Analysis (0)]
54.  Van Cutsem E, Vervenne WL, Bennouna J, Humblet Y, Gill S, Van Laethem JL, Verslype C, Scheithauer W, Shang A, Cosaert J. Phase III trial of bevacizumab in combination with gemcitabine and erlotinib in patients with metastatic pancreatic cancer. J Clin Oncol. 2009;27:2231-2237.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 469]  [Cited by in F6Publishing: 478]  [Article Influence: 31.9]  [Reference Citation Analysis (0)]
55.  Kindler HL, Wroblewski K, Wallace JA, Hall MJ, Locker G, Nattam S, Agamah E, Stadler WM, Vokes EE. Gemcitabine plus sorafenib in patients with advanced pancreatic cancer: a phase II trial of the University of Chicago Phase II Consortium. Invest New Drugs. 2012;30:382-386.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 62]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
56.  Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, Au HJ, Murawa P, Walde D, Wolff RA. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol. 2007;25:1960-1966.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Tabernero J, Chiorean EG, Infante JR, Hingorani SR, Ganju V, Weekes C, Scheithauer W, Ramanathan RK, Goldstein D, Penenberg DN. Prognostic factors of survival in a randomized phase III trial (MPACT) of weekly nab-paclitaxel plus gemcitabine versus gemcitabine alone in patients with metastatic pancreatic cancer. Oncologist. 2015;20:143-150.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 104]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
58.  Hidalgo M. Pancreatic cancer. N Engl J Med. 2010;362:1605-1617.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2078]  [Cited by in F6Publishing: 2117]  [Article Influence: 151.2]  [Reference Citation Analysis (2)]
59.  Trédan O, Galmarini CM, Patel K, Tannock IF. Drug resistance and the solid tumor microenvironment. J Natl Cancer Inst. 2007;99:1441-1454.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Jones S, Zhang X, Parsons DW, Lin JC, Leary RJ, Angenendt P, Mankoo P, Carter H, Kamiyama H, Jimeno A. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science. 2008;321:1801-1806.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3082]  [Cited by in F6Publishing: 2881]  [Article Influence: 180.1]  [Reference Citation Analysis (0)]
61.  Springett GM. Novel pancreatic cancer vaccines could unleash the army within. Cancer Control. 2014;21:242-246.  [PubMed]  [DOI]  [Cited in This Article: ]