Minireviews Open Access
Copyright ©The Author(s) 2017. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Oncol. Dec 15, 2017; 9(12): 457-465
Published online Dec 15, 2017. doi: 10.4251/wjgo.v9.i12.457
Neoadjuvant therapy for resectable pancreatic cancer
Sheikh Hasibur Rahman, Robin Urquhart, Department of Surgery, Dalhousie University, Halifax B3H 2Y9, Nova Scotia, Canada
Michele Molinari, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, United States
ORCID number: Sheikh Hasibur Rahman (0000-0001-7877-2703); Robin Urquhart (0000-0001-8864-5716); Michele Molinari (0000-0001-8864-5719).
Author contributions: Rahman SH participated in the concept, design and search of the scientific literature, extracted the data and wrote the manuscript including the tables; Urquhart R participated in the concept and design and participated in revising the manuscript; Molinari M formulated the research question, designed the literature search, supervised and co-authored the manuscript with Rahman SH.
Conflict-of-interest statement: The authors certify that they have no affiliations with or involvement in any organization or entity with any financial interest (such as honoraria; educational grants; participation in speakers’ bureaus; membership, employment, consultancies, stock ownership, or other equity interest; and expert testimony or patent-licensing arrangements), or non-financial interest (such as personal or professional relationships, affiliations, knowledge or beliefs) in the subject matter or materials discussed in this manuscript.
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: Michele Molinari, MD, MSc, Senior Scientist, Surgeon, Associate Professor, Department of Surgery, University of Pittsburgh Medical Center, 3459 Fifth Avenue, N758, Pittsburgh, PA 15213, United States. molinarim@upmc.edu
Telephone: +1-412-6475734 Fax: +1-412-6475736
Received: June 25, 2017
Peer-review started: June 27, 2017
First decision: August 7, 2017
Revised: August 24, 2017
Accepted: September 15, 2017
Article in press: September 15, 2017
Published online: December 15, 2017

Abstract

The use of neoadjuvant therapies has played a major role for borderline resectable and locally advanced pancreatic cancers (PCs). For this group of patients, preoperative chemotherapy or chemoradiation has increased the likelihood of surgery with negative resection margins and overall survival. On the other hand, for patients with resectable PC, the main rationale for neoadjuvant therapy is that the overall survival with current strategies is unsatisfactory. There is a consensus that we need new treatments to improve the overall survival and quality of life of patients with PC. However, without strong scientific evidence supporting the theoretical advantages of neoadjuvant therapies, these potential benefits might turn out not to be worth the risk of tumors progression while waiting for surgery. The focus of this paper is to provide the readers an overview of the most recent evidence on this subject.

Key Words: Pancreatic adenocarcinoma, Neoadjuvant chemotherapy, Neoadjuvant chemoradiation therapy, Meta-analysis, Decision analysis, Borderline resectable, Locally advanced, Randomized controlled trial, Phase I trial, Phase II trial, Phase III trial

Core tip: The use of neoadjuvant therapy for patients with resectable pancreatic cancer (PC) has been used by an increasing number of cancer centers around the world. The main rationale of using neoadjuvant therapies in resectable PC is the hope that patients’ likelihood of long-term overall survival will benefit from the chemo or chemoradiation therapy administered when their overall conditions allow them to tolerate the treatment. At this time, there is no phase III trial to support the use of neoadjuvant therapies in resectable PC. Without strong scientific evidence supporting the theoretical advantages of neoadjuvant therapies, these potential benefits might turn out not to be worth the risk of tumors progression while waiting for surgery.



INTRODUCTION

The most common form of pancreatic cancers (PCs) originates from the ductal cells of the exocrine gland[1,2]. In the United States, it represents the fourth leading cause of cancer-related deaths with 44000 new cases per year[2,3]. The prognosis of patients with PC remains poor with only 5%-10% of patients alive after five years[4]. Their outcome is significantly improved if they undergo surgery; however, even in this case, 5-year survival is only 25%-40%[1,4]. PC is a difficult tumor to cure as it behaves as a systemic disease even in its early stages. Although surgery remains the only potential cure, it is still inadequate for most of the patients who will develop recurrent disease within five years. The use of multimodality therapy (surgery, chemotherapy and radiation therapy) provides the best chance for long-term survival[5], but the ideal sequence and duration of these treatments remain unknown due to the lack of scientific evidence.

Despite these limitations, there is a consensus that, because of the poor outcomes observed with old treatment modalities, new strategies are necessary[6]. Among them, the use of neoadjuvant chemotherapy has gained traction and, in recent years, an increasing number of oncologists and surgeons are recommending it[7,8].

For borderline resectable and locally advanced PC, there is evidence that neoadjuvant therapy increases the probability of negative resection margins and the number of patients who can undergo surgery[8,9]. On the other hand, for resectable PC, neoadjuvant chemotherapy or chemoradiation remains debatable because of the conflicting data on its effectiveness, and because there is no phase III trial to support their use[10-12]. The focus of this publication is to provide an overview of the most recent evidence on this topic, appraise the potential benefits and disadvantages of neoadjuvant vs surgery first approach, and finally, to review the ongoing phase III trials that might address some of the questions that are still unanswered.

RESECTABILITY

Surgery remains the only potential cure for patients with PC. Determining if the disease is resectable or not at the time of diagnosis is crucial, but often subjective to the interpretation of preoperative imaging tests. Resectability is usually determined using a combination of imaging tests and laparoscopic assessment of the peritoneal cavity to rule out small hepatic or peritoneal metastases that might be missed even with high-quality contrast enhanced computerized tomography (CT scans) or magnetic resonance imaging (MRI) studies[2,13]. There are several definitions of tumor resectability that are summarized in Table 1[13-16]. All criteria currently used to identify patients with resectable disease are based on the degree of contact between the tumor and blood vessels adjacent to the pancreas in the absence of distant disease.

Table 1 Operational definitions of resectability of pancreatic cancer.
Classification of resectability of pancreatic cancerDefinition by AHPBA/SSO/SSATDefinition by MD Anderson Cancer Centre
ResectableThe tumor does not abut or encase any of the following vascular structures: the superior mesenteric vein or portal vein, superior mesenteric artery or common hepatic artery or celiac trunkThe tumor abuts or encases the superior mesenteric vein or portal vein without occluding the lumen. Absence of abutment or encasement of the superior mesenteric artery, common hepatic artery or celiac trunk
Borderline resectableAbutment, encasement or occlusion of the superior mesenteric vein or portal vein. Abutment of the superior mesenteric artery. Abutment or short segment encasement of the common hepatic artery. Absence or abutment or encasement of the celiac trunkTumor causing a short-segment occlusion of the superior mesenteric vein or portal vein. Presence of abutment of the superior mesenteric artery, abutment or encasement of a short segment of the common hepatic artery, absence of abutment or encasement of the celiac trunk
Locally advancedTumor located in the proximity of the superior mesenteric vein or portal vein and the superior mesenteric vein or portal vein are unable to be resected and reconstructed. Tumor encasing the superior mesenteric artery, or long-segment encasement of the common hepatic artery, or abutment of the celiac trunkTumor located in the proximity of the superior mesenteric vein or portal vein that are not reconstructible. Presence of tumor encasement of the superior mesenteric artery, long-segment encasement of the common hepatic artery and encasement of the celiac trunk
TREATMENT STRATEGIES

Until recently, the most accepted treatment paradigm for resectable PC was surgery followed by postoperative systemic chemotherapy or chemoradiation. In recent years, the use of systemic pre-operative chemotherapy alone or in combination with radiation therapy has been offered to an increasing number of patients with the main intent of reducing the size of the tumor, increase the likelihood of negative resection margins, and test the effects of cytotoxic medications in vivo[9]. Most patients who are treated with neoadjuvant chemotherapy or chemoradiation receive oral or intravenous medications for the duration of three to six months before undergoing surgery[17].

ADVANTAGES AND DISADVANTAGES OF NEOADJUVANT THERAPY

Neoadjuvant therapy has several theoretical benefits but also drawbacks (Table 2). It is usually well tolerated, does not increase the perioperative morbidity, reduces the interval between diagnosis and the initiation of systemic treatment[17] and has the potential benefit of facilitating radical resections by lessening the size of the tumors before surgery. Despite these advantages, postponing surgery for neoadjuvant treatment might give enough time for the tumor to progress and become unresectable[17,18].

Table 2 Summary of the benefits and drawbacks of neo-adjuvant and adjuvant therapies for the treatment of patients with resectable pancreatic cancer.
Neo-adjuvant therapyAdjuvant therapy
AdvantagesDisadvantagesAdvantagesDisadvantages

In comparison to the strategy of adjuvant chemotherapy or chemoradiation therapy where up to 50% of patients who undergo surgery cannot complete their therapy due to complications or decline of their function, neoadjuvant strategy has been shown to be well tolerated by the majority of patients and therefore a greater proportion receive systemic therapyNeoadjuvant therapy requires the placement of biliary stents to decompress the biliary obstruction prior to surgery of patients with jaundice. The placement of biliary stents before surgery increases the risk of infections in the perioperative periodOne of the advantages of surgery first approach is that patients have a short period of time between when they are diagnosed and when they undergo resections of their tumor. This might have some benefits on patients’ and their families’ anxietyAbout 20%-50% of patients will not be able to complete their postoperative therapy due to surgical complications or overall decline of their performance status
The use of neo-adjuvant therapy might sterilize the presence of small metastatic disease and reduce the size of the primary tumor. Downsizing the primary tumor might increase the likelihood of negative resection marginsPre-operative therapy delays surgery and increases the risk of progression of the disease to the point of becoming unresectableSince patients undergo surgery as soon as possible after their diagnosis, their risk of tumor progression is smaller than patients who wait a longer time before being operated onOne of the risk of undergoing surgery first for pancreatic cancer is that, some patients will undergo a major operation without the benefit of being cured as they might already have micrometastases
Treating patients before surgery, gives physicians some time to identify the tumors with poor prognosis that do not respond to the therapy. The identification of those patients who are likely to experience early metastases is very important because prevents them to undergo unnecessary surgeryThe use of neoadjuvant therapies might increase the risk of perioperative morbidity and mortality due to the side effects of chemotherapy or chemoradiationPatients who undergo surgery first do not routinely need the placement of biliary stents to release their jaundice before undergoing resectionPatients who undergo surgery first have a higher risk of positive resection margins
One of the advantages of using chemotherapy or chemoradiation therapy before surgery is that the blood supply to the pancreatic tumor is not compromised by the ligation of vessels. Therefore, chemotherapy agents can be delivered to the pancreatic tumor in higher concentrations
RECENT STUDIES

Table 3 summarizes details of the latest phase I and II trials reporting the outcomes of patients treated with neoadjuvant chemotherapy or chemoradiation for radiologically resectable PC. In all these studies, tumor response was evaluated differently as some investigators reported radiographic or clinical response before surgical exploration and others the histopathological response observed in the surgical specimen.

Table 3 Phase I and Phase II studies assessing the outcomes of patients with resectable pancreatic cancer treated with neoadjuvant therapies.
Author (yr)/ journal/trial/institutionNo. of patientsClinical stage/ duration of neoadjuvant therapyStudy designChemotherapy/chemoradiationRadiological responseResection rate (%)Negative resection margins (%)Median overall survival (mo)
Hoffman (1998)/J Clin Oncol/ECOG53Resectable PC/2.8 moPhase II, prospective study, November 1991 to September 19935-FU (1000 mg/m2) per day + Mitomycin C (10 mg/m2) + RT (50 Gy)Partial response 8%; Stable disease 78%; Progression 16%456715 with surgery; without surgery 8; 10.9 for the entire cohort
PistersPister (2002)/J Clin Oncol/MD Anderson Cancer Centre35Resectable PC/1.8 moPhase II, prospective study, timeframe not specifiedPaclitaxel (60 mg/m2) weekly, RT (30 Gy)Partial response 4%; Stable disease 23%; Progression 20%576812 for the entire cohort; 19 with surgery; 10 without surgery
Joensuu (2004)/Int J Radiat Oncol Biol Phys/Helsinki University28Resectable PC/3.5 moPhase I-II prospective study, November 1999 to December 2001Gemcitabine (20 mg/m2vs 50 mg/m2vs 100 mg/m2) twice a week + RT (50 GY)NA71NA13.6 for the entire cohort
Talamonti (2006)/Ann Surg Oncol/Northwestern University20Resectable PC/3.8 moPhase II prospective, multi-institutional study, April 2002 to October 2003Gemcitabine (1000 mg/m2 weekly) + RT (36 Gy)Partial response 15%; Stable disease 80%; Progression 5%859426 mo with surgery
Palmer (2007)/Ann Surg Oncol/University of Birmingham24Resectable PC/4 moPhase II, prospective study, November 1999 to May 2003Gemcitabine (1000 mg/m2 weekly)Partial Response 0%; Stable Disease 29%; Progression 4%; Unable to measure 4%387528.4 with surgery; 9.9 for the entire cohort
Palmer (2007)/Ann Surg Oncol/University of Birmingham26Resectable PC/4 moPhase II, prospective study, November 1999 to May 2003Gemcitabine (1000 mg/m2 weekly) + Cisplatin (25 mg/m2)Partial Response 0%; Stable Disease 66%; Progression 21%; Unable to measure 4%707528.4 with surgery; 9.9 for the entire cohort
Le Scodan (2009)/Ann Oncol/SFRO-FFCD41Resectable PC/3 moPhase II, prospective study, January 1998 to March 2003RT (50 Gy) + 5-FU (300 mg/m2 daily) + Cisplatin (20 mg/m2)Partial response 10%; Stable Disease 65%; Progression 25%638111.7 with surgery; 9.4 for the entire cohort
Heinrich (2008)/Ann Surg/University Hospital of Zurich28Resectable PC/2 moPhase II, prospective study, August 2001 to April 2007Gemcitabine (1000 mg/m2 twice weekly) + Cisplatin (50 mg/m2)Partial response 4%; Stable Disease 61%; Progression 13%898019.1 mo with surgery
Evans (2008)/J Clin Oncol/MD Anderson Cancer Centre80Resectable PC/3 moPhase II, prospective study, July 1998 to October 2001Gemcitabine (400 mg/m2 weekly) + RT (30 Gy)NA858234 mo with surgery; 22.7 mo for the entire cohort; 7 mo without surgery
Varadhachari (2008)/J Clin Oncol/MD Anderson Cancer Centre90Resectable PC/4.3 moPhase II, prospective study, October 2002 to February 2006Gemcitabine (750 mg/m2 weekly) + Cisplatin (30 mg/m2) every 2 wk + RT (30 Gy)NA589631.0 mo with surgery; 17.4 mo for the entire cohort; 10.5 mo without surgery
Turrini (2009)/Oncology /University Mediterranean34Resectable PC/2.1 moPhase II, prospective study, May 2003 to July 2005Docetazel (30 mg/m2) weekly + RT (45 GY)Partial response 9%; Stable disease 59%; Progression 32%6810032 mo with surgery; 15.5 mo for entire cohort; 11 mo without surgery
Landry (2010)/J Surg Oncol/Emory University/Multicenter ECOG21Resectable PC/3 moPhase II, prospective two-arm study, October 2013 to June 2015Arm A: Gemcitabine (500 mg/m2) weekly + RT (50 Gy) Arm B: Gemcitabine (175 mg/m2) + Cisplatin (20 mg/m2) + 5-FU (600 mg/m2) + RT (50 Gy)Arm A: Partial response 10%, Arm B: Partial response 18.2%NANAArm A: Entire cohort 19.4 mo. Arm B: entire cohort 13.4 mo. 26.3 mo with surgery
Wo (2014)/Radiother Oncol/Multicentric10Resectable PCPhase I, prospective studyCapecitabine (1650 mg/m2) over 10 d + RT (30 Gy)NA80NANA
Shinoto (2013)/Cancer/Japan26Resetable PCPhase I, prospective study, April 2003 to December 2010RT (30 Gy)Partial response 3.8%; Stable disease 96.1%819018.6 mo for entire cohort; NA for patients who underwent surgery
O'Reilly (2014)/Ann Surg/Memorial Sloan Kettering Cancer Centre38Resectable PCPhase II, prospective study, July 2007 to December 2011Gemcitabine (1000 mg/m2) + Oxaliplatin (80 mg/m2) every 2 wkPartial response 10.5%; Stable disease 73.7%; Progression 7.9%; NA 7.9%777427.2 mo for the enire cohort; 22 mo progression free survival with surgery
Golcher (2015)/Strahlenther Onkol/Germany66 (33 patients allocated to surgery + 33 patients allocated to chemoradiation followed by surgery)Resectable PCPhase II, prospective randomized trial with two arms: Primary surgery vs preoperative chemoradiation followed by surgery. June 2003 to December 2009Gemcitabine (300 mg/m2) + Cisplatin (30 mg /m2) + RT (50.4 Gy) (Preoperative for patients enrolled in Arm A)NAPreoperative chemoradiation: 69% Surgery first: 57%Arm A (preoperative chemoradition): 48. Arm B (surgery first): 51Arm A (preoperative chemoradiation): 18.9 mo. Arm B (surgery first): 25.0 mo
Van Buren (2013)/Ann Surg Oncol/Multicenter/United States59Resectable PCPhase II, prospective study, February 2007 to February 2011Gemcitabine (1500 mg/m2) ever 2 wk + Bevacizumab (10 mg/kg) + RT (30 Gy)Partial response 8.4%; Stable disease 73.7%; Progression 7.9%748819.7 mo with surgery; 16.8 mo for the entire cohort

Gillen et al[17] published the first systematic meta-analysis on the effects of preoperative therapy in PC. The authors reviewed 515 studies, but only 111 trials were included with a total of 4394 patients. Among these studies, 15 were a phase I, 13 were a phase I/II, 28 were phase II, 14 were cohort studies, and 41 were case series. Most the studies were prospective (No. 78). Chemotherapy was used as neoadjuvant therapy in 107 (96%) and radiotherapy in 104 (94%) with doses ranging from 24 to 63 Gy. In 13 trials, patients received intraoperative radiation therapy with doses between 10 and 30 Gy.

Six studies stated that the RECIST criteria were used to assess the preoperative radiological response to neoadjuvant therapy. The criteria used to evaluate tumor response were clearly stated in 44 studies, while in 61 studies the criteria used were not adequately reported. Pooled results of patients with resectable cancers at the time of diagnosis showed a complete response in 3.6%, partial response in 30.6%, progression in 20.9% and stable disease in 42.1%. Resections were performed in 73.6% (95%CI: 65.9%-80.6%) of patients. Perioperative morbidity occurred in 26.7% (95%CI: 20.7%-33.3%) and mortality in 3.9% (95%CI: 2.2%-6.0%) which were comparable to the outcomes of patients undergoing surgery first. Negative resection margins (R0) were observed in 82.1% of patients (95%CI: 73.1%-89.6%) with a median survival of 23.3 mo (range 12-54). Analysis of trials with monotherapy vs poly-chemotherapy revealed higher rates of complete or partial response when multiple chemotherapy agents were used. Higher response rates, however, did not translate into higher resection rates.

One year later, Assifi et al[19], published a second systematic review and meta-analysis of only phase II neoadjuvant therapy trials. Out of 397 studies published from 1993 to 2010, 14 trials were included with a total of 536 patients. All studies were prospective, with 12 out of 14 (86%) being a single arm. Patients who had resectable tumors were 402 (75% of the sample). Gemcitabine was used in 8 trials, while the remaining 6 used 5-FU. Radiotherapy was given in 12 of 14 studies (85%) with doses ranging between 30 and 50.4 Gy. In patients with resectable disease at diagnosis, complete radiological response was observed in 0.8% (95%CI: 0.0%-2.6%), partial response in 9.5% (95%CI: 2.9%-19.4%), stable disease in 73.9% (95%CI: 63.2%-83.3%) and progression in 17.0% (95%CI: 11.9%-22.7%). After neoadjuvant therapy, the resection rate was 65.8% (95%CI: 55.4%-75.6%) and negative resection margins were observed in 85.1% (95%CI: 76.8%-91.9%). Median survival was 23.0 mo (range 11.7-34.0). The most significant finding of these two meta-analyses was that even if safe, neoadjuvant therapy did not seem to add any substantial survival advantage[18].

Due to the heterogeneity of these studies, no conclusion can be drawn regarding the overall impact on survival and what are the most effective chemotherapy agents or the best combination of chemotherapy agents for resectable PC.

More recently, D’Angelo et al[20] completed another systematic review of randomized controlled trials on adjuvant and neoadjuvant therapies for resectable PC. Fifteen studies were included covering a period of 30 years (1985 to 2015). Their analysis suggested that despite all the best efforts, the question whether neoadjuvant therapy provides a better overall survival than adjuvant therapy remains unanswered.

DECISION ANALYSES

VanHouten et al[21] used a decision analysis model to assess what is the best treatment strategy for resectable PC. A survival advantage of 7 mo was found in patients who underwent neoadjuvant therapy in comparison to surgery first (27.2 mo vs 19.9 mo).

Another Markov decision analysis by de Geus et al[22] supported the use of neoadjuvant chemotherapy that provided longer overall survival (32 mo vs 27 mo) and quality-adjusted life expectancy (25 mo vs 21 mo) in comparison to surgery followed by adjuvant chemotherapy. Sensitivity analysis of the model showed that if the probability of surgical resection after neoadjuvant therapy was lower than 57%, upfront surgery was the best treatment option.

Another group led by Sharma et al[23] compared the efficacy of neoadjuvant-based chemotherapy with adjuvant treatment with an intention-to-treat analysis using a two-arms Markov model. In the neoadjuvant group, patients were treated with an average of 3 mo of neoadjuvant therapy followed by surgery. After surgery, patients who received preoperative chemotherapy did not receive any adjuvant treatment. On the other hand, patients who underwent surgery first, underwent chemotherapy after they recovered from their operations. In this model, the median overall survival was longer for the neoadjuvant cohort (22 mo) in comparison to the adjuvant group (20 mo), and the cumulative quality-adjusted survival for patients who underwent the neoadjuvant strategy was 19.8 mo compared to 18.4 mo for patients who had adjuvant therapy. One-way sensitivity analysis showed that surgery first provided higher quality-adjusted survival rates if more than 44% of patients treated with neoadjuvant therapy experienced progression of their disease and failed to undergo surgical resection.

All these models provided evidence that neoadjuvant therapies have better overall survival and quality of life in comparison to surgery first, although the differences were clinically quite small.

PERSISTENT CONTROVERSY

For borderline or locally advanced PC, the use of neoadjuvant therapy makes sense, and it is desirable for both patients and physicians. For patients’ perspective, neoadjuvant treatments might decrease the tumor burden and give them the chance of becoming resectable. Similarly, for the surgeons’ perspective, any reduction of the tumor size is welcomed as it facilitates the technical aspect of the resection around critical vascular structures such as the superior mesenteric-portal vein junction or superior mesenteric artery.

However, this is not the case for resectable PC. Neoadjuvant therapy does not facilitate surgery, as the tumor is resectable at the time of diagnosis. Preoperative therapy might increase the rate of negative margins; however, this needs to be proven in randomized controlled trials, as the current evidence is not sufficient. Furthermore, for patients’ perspective, there is a considerable risk of missing out the only opportunity of being cured with surgery as the tumor might progress to become unresectable while neoadjuvant therapies are delivered.

Because the current evidence is inadequate, there are no unequivocal criteria able to assist health-care providers to select the strategy with the best long-term survival for resectable PC. Physicians are left to decide whether to use neoadjuvant therapy and whether to use of one or multiple pre-operative chemotherapeutic agents or chemoradiation is worth the risk of toxicities and the possibility of disease progression. In theory, neoadjuvant treatments would be unanimously recommended for patients at high risk of positive resection margins, as their surgery would not be curative. The selection of these patients is not easy. To overcome this concerns, Bao et al[24] developed a predictive module to maximize the probability of identifying patients with true resectable tumors by using commonly available preoperative imaging modalities. With this model, the authors could classify patients with low-risk and high-risk for noncurative resections and concluded that until better evidence is available, patients who are unlikely to have R0 margins should be treated with neoadjuvant therapy.

FUTURE DIRECTIONS

D’Angelo et al[20] pointed out that the current literature is biased because the likelihood that radiologically resectable PCs is indeed unresectable at the time of surgery is only about 40%[25]. Therefore, the only way to find out if there is any benefit from neoadjuvant therapy is to complete an intention to treat randomized controlled trial where one arm entails surgery followed by adjuvant therapy (current standard of care) and the second arm involves neoadjuvant therapy followed by surgery followed by adjuvant therapy (experimental group).

Recent chemotherapy regimens, such as FOLFI RINOX [folinic acid (leucovorin)/5-FU/Irinotecan/Oxaliplatin], have already demonstrated promising results in a small group of patients with borderline resectable tumors[26,27]. Given these findings, several ongoing prospective studies are examining the role of FOLFIRINOX in a neoadjuvant setting for resectable disease (Table 4). Other studies include NEOPAC, NEONAX, NCT01660711, and NCT02172976. NEOPAC (Adjuvant vs Neoadjuvant Plus Adjuvant Chemotherapy in Resectable Pancreatic Cancer) will compare neoadjuvant gemcitabine and oxaliplatin plus adjuvant gemcitabine vs adjuvant gemcitabine alone. NEONAX, (Neoadjuvant Plus Adjuvant or Only Adjuvant Nab-Paclitaxel Plus Gemcitabine for Resectable Pancreatic Cancer) will assess the effects of neoadjuvant plus adjuvant Nab-Paclitaxel plus gemcitabine vs adjuvant only Nab-Paclitaxel plus gemcitabine. Other ongoing trials are a single-arm nonrandomized trial evaluating preoperative and postoperative FOLFIRINOX in patients with resectable disease (NCT01660711) and the multicenter German randomized trial investigating adjuvant gemcitabine compared with neoadjuvant and adjuvant FOLFIRINOX (NCT02172976).

Table 4 List of ongoing phase II and phase III trials comparing neoadjuvant therapies vs adjuvant strategies for resectable pancreatic adenocarcinoma.
StudyDesignNo. of patients neededTherapyPrimary outcome
NEOPAC (NCT01314027)Phase III Enrollment 2009-2014350Neoadjuvant gemcitabineoxaliplatin + adjuvant gemcitabine vs Adjuvant gemcitabineProgression free survival
NEOPAC (NCT01521702)Phase III Initiated in 2011310Preoperative FOLFIRINOX, followed by adjuvant gemcitabine after surgery vs adjuvant gemcitabine after resectionFive-year progression free survival
NCT01900327Phase III410Neoadjuvant gemcitabine-based chemoradiation therapy followed by adjuvant gemcitabine vs adjuvant gemcitabineThree-year overall survival
NCT01771146Phase II100Neoadjuvant FOLFIRINOXProgression free survival
NEONAX (NCT02047513)Randomized phase II166Neoadjuvant gemcitabine + nab-paclitaxel followed by adjuvant gemcitabine + nab-paclitaxel vs adjuvant gemcitabine + nab-paclitaxelDisease-free survival at 18 mo
NCT01150630Randomized phase II/III370Adjuvant PEXG vs adjuvant gemcitabine vs neoadjuvant PEXG - followed by surgery and then adjuvant PEXGOne year event-free survival
ACOSOG-Z5041 (NCT00733746)Phase II123Neoadjuvant gemcitabine + erlotinib (completed; results pending)Two-year overall survival
NCT00727441Phase II87Neoadjuvant GVAX +/- IV or oral cyclophosphamide followed by adjuvant gemcitabine + CRTSafety, feasibility, and immune response
NCT02178709Phase II48Neoadjuvant FOLFIRINOXPathologic complete response
GEMCAD1003 (NCT01389440)Phase II24Neoadjuvant gemcitabine + erlotinibR0 resection rate
NCT02562716Phase II Enrollment 2015-2019112Neoadjuvant and adjuvant mFOLFIRINOX vs neoadjuvant and adjuvant Nab-paclitaxel and gemcitabineOverall survival
NCT02243007Randomized phase II112Neoadjuvant FOLFIRINOX vs gemcitabine + nab-paclitaxel18-mo overall survival
NCT02030860Pilot15Neoadjuvant gemcitabine + nab-paclitaxel ± paricalcitolNumber of adverse events
NCT02305186Randomized phase Ib/II56Neoadjuvant capecitabine-based CRT ± pembrolizumab (MK-3745)Safety and immune response
CONCLUSION

Based on the current literature, there is still insufficient evidence to fully support the use of neoadjuvant therapy for all patients with radiologically resectable PC. Randomized controlled trials are urgently needed to address many of the questions that are still unanswered. Until then, clinicians need to weigh the pros and cons of the two treatment strategies and guide their patients. Ideally, patients should be educated on the advantages, and detrimental effects associated with each of the two possible therapies and their preferences should be elicited. Since each patient is unique, proposing neoadjuvant therapy with one-size-fits-all approach should be discouraged, and patients should become active participants and share with their physicians the responsibility of selecting the treatment strategy that fits best with their goals and values.

ACKNOWLEDGMENTS

The authors would like to acknowledge: Stefanie Condon-Oldreive founder and director of Craig’s Cause Pancreatic Cancer Society (http://www.craigscause.ca) for the research scholarship that supported Dr. Sheikh Hasibur Raman while working on this project. The authors thank Melissa Connell for her administrative support and technical and language editing that helped improving the quality of the manuscript and the quality of the audio file.

Footnotes

Manuscript source: Unsolicited manuscript

Specialty type: Oncology

Country of origin: United States

Peer-review report classification

Grade A (Excellent): 0

Grade B (Very good): B, B

Grade C (Good): C

Grade D (Fair): D

Grade E (Poor): 0

P- Reviewer: Kim SM, Munoz M, Nakai Y, Sun XT S- Editor: Ji FF L- Editor: A E- Editor: Lu YJ

References
1.  Cameron JL, He J. Two thousand consecutive pancreaticoduodenectomies. J Am Coll Surg. 2015;220:530-536.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 336]  [Cited by in F6Publishing: 384]  [Article Influence: 42.7]  [Reference Citation Analysis (0)]
2.  Sharma C, Eltawil KM, Renfrew PD, Walsh MJ, Molinari M. Advances in diagnosis, treatment and palliation of pancreatic carcinoma: 1990-2010. World J Gastroenterol. 2011;17:867-897.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 146]  [Cited by in F6Publishing: 149]  [Article Influence: 11.5]  [Reference Citation Analysis (5)]
3.  Yeo TP. Demographics, epidemiology, and inheritance of pancreatic ductal adenocarcinoma. Semin Oncol. 2015;42:8-18.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 95]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
4.  Hurton S, MacDonald F, Porter G, Walsh M, Molinari M. The current state of pancreatic cancer in Canada: incidence, mortality, and surgical therapy. Pancreas. 2014;43:879-885.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 19]  [Article Influence: 1.9]  [Reference Citation Analysis (1)]
5.  Bilimoria KY, Bentrem DJ, Ko CY, Stewart AK, Winchester DP, Talamonti MS. National failure to operate on early stage pancreatic cancer. Ann Surg. 2007;246:173-180.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 428]  [Cited by in F6Publishing: 404]  [Article Influence: 23.8]  [Reference Citation Analysis (0)]
6.  Raval MV, Bilimoria KY, Talamonti MS. Quality improvement for pancreatic cancer care: is regionalization a feasible and effective mechanism? Surg Oncol Clin N Am. 2010;19:371-390.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 18]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
7.  Roland CL, Yang AD, Katz MH, Chatterjee D, Wang H, Lin H, Vauthey JN, Pisters PW, Varadhachary GR, Wolff RA. Neoadjuvant therapy is associated with a reduced lymph node ratio in patients with potentially resectable pancreatic cancer. Ann Surg Oncol. 2015;22:1168-1175.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 89]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
8.  Cloyd JM, Katz MH, Prakash L, Varadhachary GR, Wolff RA, Shroff RT, Javle M, Fogelman D, Overman M, Crane CH. Preoperative Therapy and Pancreatoduodenectomy for Pancreatic Ductal Adenocarcinoma: a 25-Year Single-Institution Experience. J Gastrointest Surg. 2017;21:164-174.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 100]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
9.  Sutton JM, Abbott DE. Neoadjuvant therapy for pancreas cancer: past lessons and future therapies. World J Gastroenterol. 2014;20:15564-15579.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 32]  [Cited by in F6Publishing: 33]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
10.  Wilkowski R, Wolf M, Heinemann V. Primary advanced unresectable pancreatic cancer. Recent Results Cancer Res. 2008;177:79-93.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 26]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
11.  Valeri S, Borzomati D, Nappo G, Perrone G, Santini D, Coppola R. Complete pathological response after FOLFIRINOX for locally advanced pancreatic cancer. The beginning of a new era? Case report and review of the literature. Pancreatology. 2014;14:425-430.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
12.  Springett GM, Hoffe SE. Borderline resectable pancreatic cancer: on the edge of survival. Cancer Control. 2008;15:295-307.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 36]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
13.  Murakami Y, Satoi S, Sho M, Motoi F, Matsumoto I, Kawai M, Honda G, Uemura K, Yanagimoto H, Shinzeki M. National Comprehensive Cancer Network Resectability Status for Pancreatic Carcinoma Predicts Overall Survival. World J Surg. 2015;39:2306-2314.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 40]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
14.  Katz MH, Landry J, Kindler HL. Current controversies in the stage-specific multidisciplinary management of pancreatic cancer. Am Soc Clin Oncol Educ Book. 2014;e157-e164.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 5]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
15.  Katz MH, Merchant NB, Brower S, Branda M, Posner MC, William Traverso L, Abrams RA, Picozzi VJ, Pisters PW; American College of Surgeons Oncology Group. Standardization of surgical and pathologic variables is needed in multicenter trials of adjuvant therapy for pancreatic cancer: results from the ACOSOG Z5031 trial. Ann Surg Oncol. 2011;18:337-344.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 62]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
16.  Katz MH, Varadhachary GR, Fleming JB, Wolff RA, Lee JE, Pisters PW, Vauthey JN, Abdalla EK, Sun CC, Wang H. Serum CA 19-9 as a marker of resectability and survival in patients with potentially resectable pancreatic cancer treated with neoadjuvant chemoradiation. Ann Surg Oncol. 2010;17:1794-1801.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 113]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
17.  Gillen S, Schuster T, Meyer Zum Büschenfelde C, Friess H, Kleeff J. Preoperative/neoadjuvant therapy in pancreatic cancer: a systematic review and meta-analysis of response and resection percentages. PLoS Med. 2010;7:e1000267.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1085]  [Cited by in F6Publishing: 1082]  [Article Influence: 77.3]  [Reference Citation Analysis (0)]
18.  Verma V, Li J, Lin C. Neoadjuvant Therapy for Pancreatic Cancer: Systematic Review of Postoperative Morbidity, Mortality, and Complications. Am J Clin Oncol. 2016;39:302-313.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 68]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
19.  Assifi MM, Lu X, Eibl G, Reber HA, Li G, Hines OJ. Neoadjuvant therapy in pancreatic adenocarcinoma: a meta-analysis of phase II trials. Surgery. 2011;150:466-473.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 150]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
20.  D’Angelo FA, Antolino L, La Rocca M, Petrucciani N, Magistri P, Aurello P, Ramacciato G. Adjuvant and neoadjuvant therapies in resectable pancreatic cancer: a systematic review of randomized controlled trials. Med Oncol. 2016;33:28.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 15]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
21.  VanHouten JP, White RR, Jackson GP. A decision model of therapy for potentially resectable pancreatic cancer. J Surg Res. 2012;174:222-230.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 12]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
22.  de Geus SW, Evans DB, Bliss LA, Eskander MF, Smith JK, Wolff RA, Miksad RA, Weinstein MC, Tseng JF. Neoadjuvant therapy versus upfront surgical strategies in resectable pancreatic cancer: A Markov decision analysis. Eur J Surg Oncol. 2016;42:1552-1560.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 41]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
23.  Sharma G, Whang EE, Ruan DT, Ito H. Efficacy of Neoadjuvant Versus Adjuvant Therapy for Resectable Pancreatic Adenocarcinoma: A Decision Analysis. Ann Surg Oncol. 2015;22 Suppl 3:S1229-S1237.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 24]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
24.  Bao P, Potter D, Eisenberg DP, Lenzner D, Zeh HJ, Lee Iii KK, Hughes SJ, Sanders MK, Young JL, Moser AJ. Validation of a prediction rule to maximize curative (R0) resection of early-stage pancreatic adenocarcinoma. HPB (Oxford). 2009;11:606-611.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 25]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
25.  Allen VB, Gurusamy KS, Takwoingi Y, Kalia A, Davidson BR. Diagnostic accuracy of laparoscopy following computed tomography (CT) scanning for assessing the resectability with curative intent in pancreatic and periampullary cancer. Cochrane Database Syst Rev. 2013;CD009323.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 33]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
26.  Denost Q, Laurent C, Adam JP, Capdepont M, Vendrely V, Collet D, Cunha AS. Pancreaticoduodenectomy following chemoradiotherapy for locally advanced adenocarcinoma of the pancreatic head. HPB (Oxford). 2013;15:716-723.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 21]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
27.  Ducreux M, Cuhna AS, Caramella C, Hollebecque A, Burtin P, Goéré D, Seufferlein T, Haustermans K, Van Laethem JL, Conroy T. Cancer of the pancreas: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2015;26 Suppl 5:v56-v68.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 905]  [Cited by in F6Publishing: 844]  [Article Influence: 93.8]  [Reference Citation Analysis (0)]