Topic Highlight Open Access
Copyright ©2014 Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Jul 28, 2014; 20(28): 9354-9360
Published online Jul 28, 2014. doi: 10.3748/wjg.v20.i28.9354
Pancreatic cancer: Advances in treatment
Somala Mohammed, George Van Buren II, William E Fisher, The Elkins Pancreas Center, Michael E DeBakey Department of Surgery, and Dan L Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, United States
Author contributions: Mohammed S, Van Buren II G, Fisher WE contributed to concept and design, drafting of article, critical revision and gave final approval of this paper.
Correspondence to: William E Fisher, MD, Director, Professor, The Elkins Pancreas Center, Michael E DeBakey Department of Surgery, and Dan L Duncan Cancer Center, Baylor College of Medicine, 6620 Main St, Suite 1450, Houston, TX 77030, United States. wfisher@bcm.edu
Telephone: +1-832-3551490 Fax: +1-713-6102489
Received: October 21, 2013
Revised: January 20, 2014
Accepted: February 17, 2014
Published online: July 28, 2014
Processing time: 269 Days and 23.8 Hours

Abstract

Pancreatic cancer is a leading cause of cancer mortality and the incidence of this disease is expected to continue increasing. While patients with pancreatic cancer have traditionally faced a dismal prognosis, over the past several years various advances in diagnosis and treatment have begun to positively impact this disease. Identification of effective combinations of existing chemotherapeutic agents, such as the FOLFIRINOX and the gemcitabine + nab-paclitaxel regimen, has improved survival for selected patients although concerns regarding their toxicity profiles remain. A better understanding of pancreatic carcinogenesis has identified several pre-malignant precursor lesions, such as pancreatic intraepithelial neoplasias, intraductal papillary mucinous neoplasms, and cystic neoplasms. Imaging technology has also evolved dramatically so as to allow early detection of these lesions and thereby facilitate earlier management. Surgery remains a cornerstone of treatment for patients with resectable pancreatic tumors, and advances in surgical technique have allowed patients to undergo resection with decreasing perioperative morbidity and mortality. Surgery has also become feasible in selected patients with borderline resectable tumors as a result of neoadjuvant therapy. Furthermore, pancreatectomy involving vascular reconstruction and pancreatectomy with minimally invasive techniques have demonstrated safety without significantly compromising oncologic outcomes. Lastly, a deeper understanding of molecular aberrations contributing to the development of pancreatic cancer shows promise for future development of more targeted and safe therapeutic agents.

Key Words: Pancreatic cancer, Pancreaticoduodenectomy, Treatment advances, Pancreatic oncology, Chemotherapy, FOLFIRINOX, Pancreatic resection

Core tip: Pancreatic cancer is a leading cause of cancer mortality. However, recent advances have improved our ability to treat patients with this highly lethal disease. This review article discusses some of the salient advances in the field, such as improvements in chemotherapeutic regimens, imaging technology, surgical technique, and our understanding of the pathogenesis of pancreatic cancer.



INTRODUCTION

Pancreatic cancer is the tenth most commonly diagnosed cancer and the fourth leading cause of cancer mortality in the United States; the overall 5-year survival is only 5%[1]. Even patients who undergo complete resection, chemotherapy, and radiation have a 5-year survival of only 20%[2], underscoring the need for novel therapies. In the year 2012, 43000 cases of pancreatic cancer were diagnosed and, as the general population continues to age, this incidence is expected to increase[3].

This review article discusses recent advances made in the treatment of pancreatic cancer, such as new chemotherapeutic regimens that have improved survival, the recognition of potentially pre-malignant lesions, the emergence of improved imaging modalities allowing early detection of pancreatic masses, the growing practice of minimally invasive and robotic pancreatic surgery, and an improved understanding of the molecular changes contributing to pancreatic cancer development.

ADVANCES IN CHEMOTHERAPY REGIMENS

Few effective chemotherapeutic options exist for metastatic pancreatic cancer. Since the 1990s, gemcitabine has been considered the standard agent of choice, and, although multiple different agents have been evaluated in combination with gemcitabine or alone, few have demonstrated positive impact on survival in patients with advanced disease[4-9]. More recently, higher response rates have been observed with the FOLFIRINOX (5-fluorouracil, leucovorin, irinotecan, oxaliplatin) regimen and with the gemcitabine + nab-Paclitaxel regimen than with gemcitabine alone. In the Actions Concertées dans les Cancer Colo-Rectaux et Digestifs (ACCORD) 11 trial, 342 patients with metastatic pancreatic cancer were randomly assigned to receive either FOLFIRINOX or single-agent gemcitabine as first-line treatment for pancreatic cancer[10]. Endpoints included overall survival, progression-free survival, tumor response (RECIST criteria), safety, and quality of life. On interim analysis, a significantly improved median overall survival was observed in the FOLFIRINOX arm (11.1 mo vs 6.8 mo, HR = 0.57, P < 0.001) compared with the gemcitabine arm[10]. However, there were significantly more grade 3-4 toxicities, such as cytopenias, diarrhea, and neutropenic fever, in the treatment group (all P < 0.01). Subsequent studies have confirmed the efficacy of the FOLFIRINOX regimen[11,12], but have questioned its applicability to patients of older age, with poor performance status, and with co-morbid conditions[13]. FOLFIRINOX also remains controversial with respect to its tolerability; studies report manageable side effects as well as significant toxicity resulting in treatment discontinuation[11,14]. In small studies, components of the FOLFIRINOX regimen have been dose-attenuated, raising the concern that physician modification of the regimen may affect patient outcomes[15].

Favorable outcomes are also beginning to be observed with the use of this regimen in the neoadjuvant setting for patients with borderline resectable or locally unresectable disease. In a recent study of 21 patients with either unresectable or borderline resectable pancreatic cancer who received neoadjuvant FOLFIRINOX, a 33% R0 resection rate was achieved (55% borderline resectable, 10% locally unresectable) and 24% of patients demonstrated a significant pathologic response[16]. Despite concerns of toxicity and tolerability, in the carefully selected patient with good performance status and early or advanced disease, FOLFIRINOX demonstrates potential for improved oncologic outcomes.

Nab-paclitaxel (trade name, Abraxane) is a nanoparticle albumin-bound (nab) paclitaxel that was initially developed to avoid hypersensitivity reactions resulting from solvents used to dissolve the agent[17]. It was approved by the FDA in 2004 for use in metastatic breast cancer and metastatic non-small cell lung cancer[18,19]. In the phase III Metastatic Pancreatic Adenocarcinoma Clinical Trial (MPACT), the addition of nab-paclitaxel to gemcitabine demonstrated improved median overall survival (8.5 mo vs 6.7 mo, HR = 0.72, P < 0.001), improved 1-year survival (35% vs 22%), improved 2-year survival (9% vs 4%), and improved objective response rate (23% vs 7%) when compared with gemcitabine alone[20]. Although this response is not as dramatic as those observed with FOLFIRINOX, this regimen was well-tolerated and demonstrated a safer toxicity profile. It has emerged as an option for patients who cannot tolerate FOLFIRNOX because of poor performance status.

The role of nab-paclitaxel was investigated in pancreatic cancer after molecular profiling done on pancreatic tumors demonstrated high levels of the albumin-binding protein SPARC (secreted protein acidic and rich in cysteine)[21]. Nab-paclitaxel has demonstrated anti-tumor activity in cancers of the breast and lung, particularly in tissues that express high levels of SPARC[22]. It is believed that among patients with pancreatic cancer, tumors with high SPARC expression serve as albumin-binding sites that sequester nab-paclitaxel and concentrate drug levels intratumorally[23]. Another mechanism proposed involves an albumin receptor (gp60) on endothelial cells that transports paclitaxel into the tumoral interstitial space[24].

Gemcitabine plus erlotinib is another multi-drug regimen that has shown improved progression-free survival and overall survival[25,26]. However, due to their greater potential for improved outcomes, FOLFIRINOX and gemcitabine + nab-paclitaxel are the preferred treatment options for patients with acceptable performance status.

EARLY IDENTIFICATION AND TREATMENT OF PREMALIGNANT LESIONS

Early detection and management of adenomatous polyps, in situ lesions, and other premalignant or potentially malignant entities of the colon and breast have resulted in less mortality due to these cancers. It is now believed that pancreatic ductal adenocarcinoma also arises from a series of similar progressive genetic mutations and specific precursor lesions, such as pancreatic intraepithelial neoplasia (PanIN), intraductal papillary mucinous neoplasms (IPMN), and mucinous cystic neoplasms.

PanINs are by far the most common of these precursor lesions[27]. Autopsy studies have shown that panINs increase with age, are more common in the head of the pancreas, and are seen at much higher rates in pancreata with tumors than those with pancreatitis[28-30]. They involve the same molecular events seen in the development of adenocarcinoma of other organs, such as activation of K-ras mutants, overexpression of p53, and loss of p16 and SMAD4[31]. Although no specific sequence has been elucidated, certain mutations (K-ras, p16) occur before others (TP53, SMAD4), and higher grades of panIN indicate higher levels of mutations[32].

IPMN belong to a heterogenous group of cystic lesions and are also considered precursor lesions for the development of invasive carcinoma. Main-duct IPMNs connect to the main duct of Wirsung while side-branch IPMNs originate from smaller branches off the main duct. Main duct and branch duct IPMNs were associated with malignancy in 70% and 25% of cases, respectively. Other groups have produced similar findings[33]. There is a strong consensus for resection of main-duct IPMNs due to their higher risk for associated malignancy.

Mucinous cystic neoplasms comprise around 25% of all resected cystic neoplasms[34]. They are characterized by dense stroma surrounding a tumor with mucin-producing epithelial cells, which are susceptible to various degrees of atypia. In a study of mucinous cystic neoplasms by the Massachusetts General Hospital, the risk of malignancy among 163 cases was 17.5%, and all malignant tumors had either nodules or were greater than 4 cm in size[35]. Patients are typically managed by surgical resection. If non-operative management is pursued, lifelong surveillance is essential.

With the widespread use of radiographic imaging and improvement in its resolution, there has been an increase in the incidence of cystic lesions, which are now found in approximately 1% of all abdominal computed tomographic scans obtained[36]. Given the variable potential for malignancy, groups have developed criteria to characterize these lesions and risk-stratify patients. The diagnostic algorithm often includes endoscopic ultrasound (EUS) with fine needle aspiration of cyst fluid to assess cytology, the presence of mucin, tumor markers carcinoembryonic antigen, and DNA for loss of heterozygosity and K-ras mutations.

High resolution endoscopic ultrasound (EUS) is an imaging modality that is able to detect focal lesions as small as 2-3 mm in size[37]. Studies have shown that EUS is superior or at least equal to computed tomography (CT) or magnetic resonance imaging in its sensitivity for detecting lesions, determining tumor size and extent, and assessing lymph node involvement and vascular invasion[38,39]. Conventional CT scans also provide detailed high-resolution views of pancreatic tumors in relation to the superior mesenteric artery, celiac axis, superior mesenteric vein, and portal vein, and this imaging modality remains the preferred choice for initial evaluation of most patients suspected to have pancreatic cancer.

Early detection of pre-malignant and potentially malignant lesions represents a significant advance in the treatment of pancreatic cancer. Since invasive pancreatic cancer is rarely cured, resection of these premalignant lesions is believed to be warranted. However, further refinements in our understanding of premalignant lesions and more accurate risk-stratification of patients is necessary so that patients with a low risk of malignancy can avoid an operation.

ADVANCES IN SURGICAL PRACTICE

Surgery plays a critical role in the management of pancreatic cancer, and many advances in surgical practice patterns as well as surgical technique have resulted in reduced perioperative morbidity and mortality. Centralization of pancreaticoduodenectomy, for example, to higher-volume centers with higher-volume surgeons, has contributed to a reduction in postoperative mortality, such that the risk of mortality at high volume centers is currently as low as 3%[40].

Historically, pancreatic tumors were considered either resectable or unresectable. In 2003, the National Comprehensive Cancer Network introduced the “borderline resectable” classification for pancreatic cancer, which refers to tumors that are involved with nearby structures so as to be neither clearly resectable nor clearly unresectable[41]. Aggressive management of this group of patients with neoadjuvant chemotherapy has made surgery feasible and may have improved survival in selected patients. The safety of vascular reconstruction in conjunction with pancreaticoduodenectomy has also been demonstrated in a systematic review of the literature[42].

Aside from more complex open surgeries, pancreatic cancer is also being increasingly approached laparoscopically. Early studies show that minimally invasive approaches can be performed safely and facilitate shorter hospital stay, earlier return to preoperative activity level, and reduced postoperative recuperation allowing for less delay in time to adjuvant chemotherapy or radiation[43-45]. With evolving technology and experience, laparoscopic distal pancreatectomy has become a standard approach for benign and malignant lesions of the pancreatic body and tail. In a multicenter study comparing open and laparoscopic distal pancreatectomy for patients with pancreatic ductal adenocarcinoma, Kooby et al[46] showed that there were no significant differences in positive margin rates, number of nodes examined, number of patients with at least one positive node, or overall survival, and that there was shorter hospital stay (7.4 d vs 9.4 d, P = 0.06) in the laparoscopic distal pancreatectomy group.

Laparoscopy has been extended to pancreaticoduodenectomy as well, and several case series have demonstrated feasibility, safety, and efficacy of this approach as compared to open surgery[47]. The robotic platform is also being increasingly adopted in pancreatic surgery. This approach overcomes limitations of laparoscopy, such as two-dimensional visualization, lack of dexterity, and poor ergonomics. In a series of 30 patients undergoing robot-assisted major pancreatectomy and reconstruction, Zureikat et al[48] reported an overall pancreatic fistula rate of 27% and a 90-d Clavien grade III-IV complication rate of 23%. They concluded that robot-assisted surgery can be performed safely with postoperative complication rates comparable to those of open pancreatectomy[48]. Further experience and larger, controlled studies are needed to clearly define potential benefits and elucidate long-term oncologic outcomes of minimally invasive pancreaticoduodenectomy.

ADVANCED UNDERSTANDING OF GENETIC AND MOLECULAR FACTORS

Many genetic alterations, including germ line and somatic mutations, contribute to the development of pancreatic cancer. Recent studies indicate that pancreatic cancer cells carry an average of 63 genetic mutations per cancer, and these mutations can be grouped into twelve core signaling pathways[49]. Over 90% of pancreatic cancers possess mutations in the k-ras oncogene, which is mutated in 20%-30% of all human malignancies[50]. Mutations within this oncogene are most often located on exon 1 of codon 12 and sometimes on codons 61 and 13[50,51]. Mutated k-ras upregulates several pathways, such as the PI3K-AKT pathway, which is involved in a series of important cellular functions, including survival and proliferation[52]. Other oncogenes involved in pancreatic carcinogenesis include those involved with the Notch signaling pathway[53] and the sonic hedgehog pathway[54].

The most widely recognized tumor suppressor gene (TSG) implicated in pancreatic cancer development is p53, which is found to be mutated in over 75% of specimens[55]. Other TSGs of importance include DPC4 (Deleted in Pancreatic Cancer, locus 4), LKB1 (liver kinase B1), p16, MAPK (mitogen activated protein kinase), and BRCA 2. These various discoveries contribute to the development of more targeted therapies and may also provide prognostic information. Over 50% of pancreatic adenocarcinomas have been demonstrated to have an inactivating mutation in SMAD4[56]. Tascilar et al[56] measured SMAD4 protein expression in 249 pancreatic adenocarcinomas and found that patients with this mutation had significantly longer survival than those without it (19.2 mo vs 14.7 mo), even after adjusting for other factors such as tumor size, margins, lymph node status, pathological stage, blood loss, and use of adjuvant chemoradiotherapy.

Whole exome sequencing and copy number analysis of a prospective cohort of 142 patients with pancreatic cancer recently defined 16 significantly mutated genes, ranging from those which were previously known to contribute to pancreatic cancer pathogenesis (KRAS, TP53, CDKN2A, SMAD4, MLL3, TGFBR2, ARID1A, SF3B1) to newly discovered genes involved in chromatin modification (EPC1, ARID2), DNA damage repair (ATM), and other mechanisms (ZIM2, MAP2K4, NALCN, SLC16A4, MAGEA6)[57]. Larger studies are needed to determine whether these mutations are more prevalent among specific demographic groups or whether they affect oncologic outcomes.

CONCLUSION

Pancreatic cancer remains a highly lethal disease. By the time patients are diagnosed, the disease may often be advanced, precluding patients from surgery. Recent advances in chemotherapeutic regimens have not only improved our ability to treat patients with metastatic disease, but have also shown favorable outcomes in the neoadjuvant setting. Advances in imaging technology and a better understanding of the pathogenesis of pancreatic cancer are allowing earlier diagnosis and early aggressive management of potentially pre-malignant entities. Emergence of high volume centers, the incorporation of imaging technology, and the availability of specialty services, such as interventional radiology, have reduced perioperative morbidity and mortality associated with pancreaticoduodenectomy. Furthermore, advances in surgical technology are allowing these procedures to be performed in less invasive fashion and are demonstrating safety and feasibility. Despite these advances, there remains room for improvement. Today’s pancreatic oncologists must focus on further understanding the genetic and molecular factors contributing to oncogenesis and on the development of more targeted and less toxic systemic therapies.

ACKNOWLEDGMENTS

The authors would like to thank Ana María Rodríguez, PhD, a member of the Baylor College of Medicine Michael E. DeBakey Department of Surgery Research Core Team, for her editorial assistance during the preparation of this manuscript.

Footnotes

P- Reviewer: Behrns KE, Mezhir JJ, Matsumoto I, Ogura T S- Editor: Song XX L- Editor: A E- Editor: Zhang DN

References
1.  Siegel R, Naishadham D, Jemal A. Cancer statistics, 2013. CA Cancer J Clin. 2013;63:11-30.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9215]  [Cited by in F6Publishing: 9751]  [Article Influence: 886.5]  [Reference Citation Analysis (3)]
2.  Jemal A, Siegel R, Xu J, Ward E. Cancer statistics, 2010. CA Cancer J Clin. 2010;60:277-300.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10002]  [Cited by in F6Publishing: 10364]  [Article Influence: 740.3]  [Reference Citation Analysis (0)]
3.  American Cancer Society. Cancer Facts & Figures.  Available from: http://www.cancer.org/acs/groups/content/@epidemiologysurveilance/documents/document/acspc-026238.pdf.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Burris HA, Moore MJ, Andersen J, Green MR, Rothenberg ML, Modiano MR, Cripps MC, Portenoy RK, Storniolo AM, Tarassoff P. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. J Clin Oncol. 1997;15:2403-2413.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Hertel LW, Boder GB, Kroin JS, Rinzel SM, Poore GA, Todd GC, Grindey GB. Evaluation of the antitumor activity of gemcitabine (2’,2’-difluoro-2’-deoxycytidine). Cancer Res. 1990;50:4417-4422.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Cullinan SA, Moertel CG, Fleming TR, Rubin JR, Krook JE, Everson LK, Windschitl HE, Twito DI, Marschke RF, Foley JF. A comparison of three chemotherapeutic regimens in the treatment of advanced pancreatic and gastric carcinoma. Fluorouracil vs fluorouracil and doxorubicin vs fluorouracil, doxorubicin, and mitomycin. JAMA. 1985;253:2061-2067.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 233]  [Cited by in F6Publishing: 210]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
7.  DeCaprio JA, Mayer RJ, Gonin R, Arbuck SG. Fluorouracil and high-dose leucovorin in previously untreated patients with advanced adenocarcinoma of the pancreas: results of a phase II trial. J Clin Oncol. 1991;9:2128-2133.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Hansen R, Quebbeman E, Ritch P, Chitambar C, Anderson T. Continuous 5-fluorouracil (5FU) infusion in carcinoma of the pancreas: a phase II study. Am J Med Sci. 1988;295:91-93.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 61]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
9.  Conroy T, Gavoille C, Adenis A. Metastatic pancreatic cancer: old drugs, new paradigms. Curr Opin Oncol. 2011;23:390-395.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 29]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
10.  Conroy T, Desseigne F, Ychou M, Bouché O, Guimbaud R, Bécouarn Y, Adenis A, Raoul JL, Gourgou-Bourgade S, de la Fouchardière C. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364:1817-1825.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4838]  [Cited by in F6Publishing: 5227]  [Article Influence: 402.1]  [Reference Citation Analysis (1)]
11.  Faris JE, Blaszkowsky LS, McDermott S, Guimaraes AR, Szymonifka J, Huynh MA, Ferrone CR, Wargo JA, Allen JN, Dias LE. FOLFIRINOX in locally advanced pancreatic cancer: the Massachusetts General Hospital Cancer Center experience. Oncologist. 2013;18:543-548.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 204]  [Cited by in F6Publishing: 216]  [Article Influence: 19.6]  [Reference Citation Analysis (0)]
12.  Gunturu KS, Yao X, Cong X, Thumar JR, Hochster HS, Stein SM, Lacy J. FOLFIRINOX for locally advanced and metastatic pancreatic cancer: single institution retrospective review of efficacy and toxicity. Med Oncol. 2013;30:361.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 93]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
13.  Gill S, Kennecke HF, Renouf DJ, Cheung WY, Lim HJ. Defining elibibility of FOLFIRINOX for first-line metastatic pancreatic adenocarcinoma in the province of British Columbia: a population-based retrospective study. ASCO Meet Abstr. 2012;30:14588.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Peddi PF, Lubner S, McWilliams R, Tan BR, Picus J, Sorscher SM, Suresh R, Lockhart AC, Wang J, Menias C. Multi-institutional experience with FOLFIRINOX in pancreatic adenocarcinoma. JOP. 2012;13:497-501.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 55]  [Reference Citation Analysis (0)]
15.  Valsecchi ME, Díaz-Cantón E, de la Vega M, Littman SJ. Recent treatment advances and novel therapies in pancreas cancer: a review. J Gastrointest Cancer. 2014;45:190-201.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Boone BA, Steve J, Krasinskas AM, Zureikat AH, Lembersky BC, Gibson MK, Stoller RG, Zeh HJ, Bahary N. Outcomes with FOLFIRINOX for borderline resectable and locally unresectable pancreatic cancer. J Surg Oncol. 2013;108:236-241.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 140]  [Cited by in F6Publishing: 156]  [Article Influence: 14.2]  [Reference Citation Analysis (0)]
17.  Peddi PF, Cho M, Wang J, Gao F, Wang-Gillam A. Nab-paclitaxel monotherapy in refractory pancreatic adenocarcinoma. J Gastrointest Oncol. 2013;4:370-373.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 4]  [Reference Citation Analysis (0)]
18.  Gradishar WJ, Tjulandin S, Davidson N, Shaw H, Desai N, Bhar P, Hawkins M, O’Shaughnessy J. Phase III trial of nanoparticle albumin-bound paclitaxel compared with polyethylated castor oil-based paclitaxel in women with breast cancer. J Clin Oncol. 2005;23:7794-7803.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1422]  [Cited by in F6Publishing: 1425]  [Article Influence: 75.0]  [Reference Citation Analysis (0)]
19.  Socinski MA, Bondarenko I, Karaseva NA, Makhson AM, Vynnychenko I, Okamoto I, Hon JK, Hirsh V, Bhar P, Zhang H. Weekly nab-paclitaxel in combination with carboplatin versus solvent-based paclitaxel plus carboplatin as first-line therapy in patients with advanced non-small-cell lung cancer: final results of a phase III trial. J Clin Oncol. 2012;30:2055-2062.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 488]  [Cited by in F6Publishing: 563]  [Article Influence: 46.9]  [Reference Citation Analysis (0)]
20.  Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, Seay T, Tjulandin SA, Ma WW, Saleh MN. Randomized phase III study of weekly nab-paclitaxel plus gemcitabine vs. gemcitabine alone in patients with metastatic adenocarcinooma of the pancreas (MPACT). ASCO Meet Abstr. 2013;31:LBA148.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  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)]
22.  Nab-Paclitaxel for treatment of solid tumors: beyond breast cancer Commun Oncol. 2008;5:8-15.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Desai NP, Trieu V, Hwang LY, Wu R, Soon-Shiong P, Gradishar WJ. Improved effectiveness of nanoparticle albumin-bound (nab) paclitaxel versus polysorbate-based docetaxel in multiple xenografts as a function of HER2 and SPARC status. Anticancer Drugs. 2008;19:899-909.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 163]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
24.  Cucinotto I, Fiorillo L, Gualtieri S, Arbitrio M, Ciliberto D, Staropoli N, Grimaldi A, Luce A, Tassone P, Caraglia M. Nanoparticle albumin bound Paclitaxel in the treatment of human cancer: nanodelivery reaches prime-time? J Drug Deliv. 2013;2013:905091.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 50]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
25.  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: ]  [Cited by in Crossref: 2835]  [Cited by in F6Publishing: 2688]  [Article Influence: 158.1]  [Reference Citation Analysis (0)]
26.  Yang ZY, Yuan JQ, Di MY, Zheng DY, Chen JZ, Ding H, Wu XY, Huang YF, Mao C, Tang JL. Gemcitabine plus erlotinib for advanced pancreatic cancer: a systematic review with meta-analysis. PLoS One. 2013;8:e57528.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 48]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
27.  Hruban RH, Maitra A, Goggins M. Update on pancreatic intraepithelial neoplasia. Int J Clin Exp Pathol. 2008;1:306-316.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Andea A, Sarkar F, Adsay VN. Clinicopathological correlates of pancreatic intraepithelial neoplasia: a comparative analysis of 82 cases with and 152 cases without pancreatic ductal adenocarcinoma. Mod Pathol. 2003;16:996-1006.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 204]  [Cited by in F6Publishing: 188]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
29.  Brat DJ, Lillemoe KD, Yeo CJ, Warfield PB, Hruban RH. Progression of pancreatic intraductal neoplasias to infiltrating adenocarcinoma of the pancreas. Am J Surg Pathol. 1998;22:163-169.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 216]  [Cited by in F6Publishing: 209]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
30.  Zamboni G, Hirabayashi K, Castelli P, Lennon AM. Precancerous lesions of the pancreas. Best Pract Res Clin Gastroenterol. 2013;27:299-322.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 46]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
31.  Sasaki S, Yamamoto H, Kaneto H, Ozeki I, Adachi Y, Takagi H, Matsumoto T, Itoh H, Nagakawa T, Miyakawa H. Differential roles of alterations of p53, p16, and SMAD4 expression in the progression of intraductal papillary-mucinous tumors of the pancreas. Oncol Rep. 2003;10:21-25.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Löhr M, Klöppel G, Maisonneuve P, Lowenfels AB, Lüttges J. Frequency of K-ras mutations in pancreatic intraductal neoplasias associated with pancreatic ductal adenocarcinoma and chronic pancreatitis: a meta-analysis. Neoplasia. 2005;7:17-23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 224]  [Cited by in F6Publishing: 229]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
33.  Tanaka M, Chari S, Adsay V, Fernandez-del Castillo C, Falconi M, Shimizu M, Yamaguchi K, Yamao K, Matsuno S. International consensus guidelines for management of intraductal papillary mucinous neoplasms and mucinous cystic neoplasms of the pancreas. Pancreatology. 2006;6:17-32.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1539]  [Cited by in F6Publishing: 1401]  [Article Influence: 77.8]  [Reference Citation Analysis (0)]
34.  Valsangkar NP, Morales-Oyarvide V, Thayer SP, Ferrone CR, Wargo JA, Warshaw AL, Fernández-del Castillo C. 851 resected cystic tumors of the pancreas: a 33-year experience at the Massachusetts General Hospital. Surgery. 2012;152:S4-12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 257]  [Cited by in F6Publishing: 260]  [Article Influence: 21.7]  [Reference Citation Analysis (0)]
35.  Crippa S, Salvia R, Warshaw AL, Domínguez I, Bassi C, Falconi M, Thayer SP, Zamboni G, Lauwers GY, Mino-Kenudson M. Mucinous cystic neoplasm of the pancreas is not an aggressive entity: lessons from 163 resected patients. Ann Surg. 2008;247:571-579.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 340]  [Cited by in F6Publishing: 260]  [Article Influence: 16.3]  [Reference Citation Analysis (0)]
36.  Spinelli KS, Fromwiller TE, Daniel RA, Kiely JM, Nakeeb A, Komorowski RA, Wilson SD, Pitt HA. Cystic pancreatic neoplasms: observe or operate. Ann Surg. 2004;239:651-67; discussion 651-67;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 405]  [Cited by in F6Publishing: 438]  [Article Influence: 21.9]  [Reference Citation Analysis (0)]
37.  Helmstaedter L, Riemann JF. Pancreatic cancer--EUS and early diagnosis. Langenbecks Arch Surg. 2008;393:923-927.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 45]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
38.  Legmann P, Vignaux O, Dousset B, Baraza AJ, Palazzo L, Dumontier I, Coste J, Louvel A, Roseau G, Couturier D. Pancreatic tumors: comparison of dual-phase helical CT and endoscopic sonography. AJR Am J Roentgenol. 1998;170:1315-1322.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 300]  [Cited by in F6Publishing: 315]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
39.  Rösch T, Braig C, Gain T, Feuerbach S, Siewert JR, Schusdziarra V, Classen M. Staging of pancreatic and ampullary carcinoma by endoscopic ultrasonography. Comparison with conventional sonography, computed tomography, and angiography. Gastroenterology. 1992;102:188-199.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Schmidt CM, Turrini O, Parikh P, House MG, Zyromski NJ, Nakeeb A, Howard TJ, Pitt HA, Lillemoe KD. Effect of hospital volume, surgeon experience, and surgeon volume on patient outcomes after pancreaticoduodenectomy: a single-institution experience. Arch Surg. 2010;145:634-640.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 312]  [Cited by in F6Publishing: 334]  [Article Influence: 23.9]  [Reference Citation Analysis (0)]
41.  National Comprehensive Cancer Network. Clinical Practice Guidelines in Oncology: Pancreatic Adenocarcinoma. Version 1.2014. Accessed 31 Dec 2013.  Available from: http://www.nccn.org/professionals/physician_gls/recently_updated.asp.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Ramacciato G, Mercantini P, Petrucciani N, Giaccaglia V, Nigri G, Ravaioli M, Cescon M, Cucchetti A, Del Gaudio M. Does portal-superior mesenteric vein invasion still indicate irresectability for pancreatic carcinoma? Ann Surg Oncol. 2009;16:817-825.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 103]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
43.  Kooby DA, Gillespie T, Bentrem D, Nakeeb A, Schmidt MC, Merchant NB, Parikh AA, Martin RC, Scoggins CR, Ahmad S. Left-sided pancreatectomy: a multicenter comparison of laparoscopic and open approaches. Ann Surg. 2008;248:438-446.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 292]  [Cited by in F6Publishing: 320]  [Article Influence: 20.0]  [Reference Citation Analysis (0)]
44.  Eom BW, Jang JY, Lee SE, Han HS, Yoon YS, Kim SW. Clinical outcomes compared between laparoscopic and open distal pancreatectomy. Surg Endosc. 2008;22:1334-1338.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 116]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
45.  Kim SC, Park KT, Hwang JW, Shin HC, Lee SS, Seo DW, Lee SK, Kim MH, Han DJ. Comparative analysis of clinical outcomes for laparoscopic distal pancreatic resection and open distal pancreatic resection at a single institution. Surg Endosc. 2008;22:2261-2268.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 137]  [Cited by in F6Publishing: 123]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
46.  Kooby DA, Hawkins WG, Schmidt CM, Weber SM, Bentrem DJ, Gillespie TW, Sellers JB, Merchant NB, Scoggins CR, Martin RC. A multicenter analysis of distal pancreatectomy for adenocarcinoma: is laparoscopic resection appropriate? J Am Coll Surg. 2010;210:779-85, 786-7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 229]  [Cited by in F6Publishing: 234]  [Article Influence: 16.7]  [Reference Citation Analysis (0)]
47.  Kendrick ML, Cusati D. Total laparoscopic pancreaticoduodenectomy: feasibility and outcome in an early experience. Arch Surg. 2010;145:19-23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 251]  [Cited by in F6Publishing: 282]  [Article Influence: 20.1]  [Reference Citation Analysis (0)]
48.  Zureikat AH, Nguyen KT, Bartlett DL, Zeh HJ, Moser AJ. Robotic-assisted major pancreatic resection and reconstruction. Arch Surg. 2011;146:256-261.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 91]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
49.  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: 2922]  [Article Influence: 182.6]  [Reference Citation Analysis (0)]
50.  Caldas C, Kern SE. K-ras mutation and pancreatic adenocarcinoma. Int J Pancreatol. 1995;18:1-6.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Slebos RJ, Hoppin JA, Tolbert PE, Holly EA, Brock JW, Zhang RH, Bracci PM, Foley J, Stockton P, McGregor LM. K-ras and p53 in pancreatic cancer: association with medical history, histopathology, and environmental exposures in a population-based study. Cancer Epidemiol Biomarkers Prev. 2000;9:1223-1232.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Fang Y, Yao Q, Chen Z, Xiang J, William FE, Gibbs RA, Chen C. Genetic and molecular alterations in pancreatic cancer: implications for personalized medicine. Med Sci Monit. 2013;19:916-926.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 33]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
53.  Sjölund J, Manetopoulos C, Stockhausen MT, Axelson H. The Notch pathway in cancer: differentiation gone awry. Eur J Cancer. 2005;41:2620-2629.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 93]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
54.  Feldmann G, Dhara S, Fendrich V, Bedja D, Beaty R, Mullendore M, Karikari C, Alvarez H, Iacobuzio-Donahue C, Jimeno A. Blockade of hedgehog signaling inhibits pancreatic cancer invasion and metastases: a new paradigm for combination therapy in solid cancers. Cancer Res. 2007;67:2187-2196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 515]  [Cited by in F6Publishing: 516]  [Article Influence: 30.4]  [Reference Citation Analysis (0)]
55.  Scarpa A, Capelli P, Mukai K, Zamboni G, Oda T, Iacono C, Hirohashi S. Pancreatic adenocarcinomas frequently show p53 gene mutations. Am J Pathol. 1993;142:1534-1543.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Tascilar M, Skinner HG, Rosty C, Sohn T, Wilentz RE, Offerhaus GJ, Adsay V, Abrams RA, Cameron JL, Kern SE. The SMAD4 protein and prognosis of pancreatic ductal adenocarcinoma. Clin Cancer Res. 2001;7:4115-4121.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Biankin AV, Waddell N, Kassahn KS, Gingras MC, Muthuswamy LB, Johns AL, Miller DK, Wilson PJ, Patch AM, Wu J. Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature. 2012;491:399-405.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1442]  [Cited by in F6Publishing: 1546]  [Article Influence: 128.8]  [Reference Citation Analysis (0)]